1
|
Kim B, Choi JS, Zhu Y, Kim J, Kim YS, Parra A, Locke PA, Kim JH, Herron T, Kim DH. Effect of electrochemical topology on detection sensitivity in MEA assay for drug-induced cardiotoxicity screening. Biosens Bioelectron 2025; 272:117082. [PMID: 39778241 PMCID: PMC11773427 DOI: 10.1016/j.bios.2024.117082] [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: 08/15/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Cardiotoxicity remains a major challenge in drug development, accounting for 45% of medication withdrawals due to cardiac ischemia and arrhythmogenicity. To overcome the limitations of traditional multielectrode array (MEA)-based cardiotoxicity assays, we developed a Nafion-coated NanoMEA platform with decoupled reference electrodes, offering enhanced sensitivity for electrophysiological measurements. The 'Decoupled' configuration significantly reduced polarization resistance (Rp) from 12.77 MΩ to 3.41 MΩ, improving charge transfer efficiency as demonstrated by electrochemical impedance spectroscopy and cyclic voltammetry. Additionally, the limit of detection significantly decreased from 0.175 MΩ (Coupled) to 0.040 MΩ (Decoupled), underscoring the system's enhanced sensitivity. Using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we evaluated the effects of three proarrhythmic drugs: Ranolazine, Domperidone, and Sotalol. Under the decoupled condition, the platform exhibited reductions in IC50 values for Domperidone (0.71 μM-0.29 μM), Sotalol (7.61 μM-0.27 μM), and Ranolazine (53.08 μM-5.89 μM), demonstrating significantly improved drug detection sensitivity. Longitudinal analysis revealed significant alterations in key electrophysiological parameters, including beating period (BP), field potential duration (FPD), spike slope, and amplitude, which were consistent with the known pharmacological actions of these drugs. Further validation through action potential (AP) waveform analysis showed enhanced repolarization dynamics, confirming the platform's predictive capabilities. Our findings highlight the critical role of electrochemical topology in optimizing MEA performance. The NanoMEA system, featuring decoupled Nafion-coated electrodes, represents a robust and sensitive platform for cardiotoxicity screening, setting a new standard for preclinical drug safety assessment and advancing bioelectronic device design for cardiac research.
Collapse
Affiliation(s)
- Byunggik Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, United States
| | - Jong Seob Choi
- Division of Advanced Materials Engineering, Kongju National University, Budaedong 275, Seobuk-gu, Cheonan-si, Chungnam, 31080, Republic of Korea; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Yiguang Zhu
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Juhyun Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Ye Seul Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Andres Parra
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Paul A Locke
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Todd Herron
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, United States; Department of Medicine, Johns Hopkins University, Baltimore, MD, 21205, United States; Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, 21218, United States; Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, United States.
| |
Collapse
|
2
|
Fujii K, Fujiwara-Tani R, Nukaga S, Ohmori H, Luo Y, Nishida R, Sasaki T, Miyagawa Y, Nakashima C, Kawahara I, Ogata R, Ikemoto A, Sasaki R, Kuniyasu H. Involvement of Ferroptosis Induction and Oxidative Phosphorylation Inhibition in the Anticancer-Drug-Induced Myocardial Injury: Ameliorative Role of Pterostilbene. Int J Mol Sci 2024; 25:3015. [PMID: 38474261 DOI: 10.3390/ijms25053015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 02/25/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
Patients with cancer die from cardiac dysfunction second only to the disease itself. Cardiotoxicity caused by anticancer drugs has been emphasized as a possible cause; however, the details remain unclear. To investigate this mechanism, we treated rat cardiomyoblast H9c2 cells with sunitinib, lapatinib, 5-fluorouracil, and cisplatin to examine their effects. All anticancer drugs increased ROS, lipid peroxide, and iron (II) levels in the mitochondria and decreased glutathione peroxidase-4 levels and the GSH/GSSG ratio. Against this background, mitochondrial iron (II) accumulates through the unregulated expression of haem oxygenase-1 and ferrochelatase. Anticancer-drug-induced cell death was suppressed by N-acetylcysteine, deferoxamine, and ferrostatin, indicating ferroptosis. Anticancer drug treatment impairs mitochondrial DNA and inhibits oxidative phosphorylation in H9c2 cells. Similar results were observed in the hearts of cancer-free rats treated with anticancer drugs in vitro. In contrast, treatment with pterostilbene inhibited the induction of ferroptosis and rescued the energy restriction induced by anticancer drugs both in vitro and in vivo. These findings suggest that induction of ferroptosis and inhibition of oxidative phosphorylation are mechanisms by which anticancer drugs cause myocardial damage. As pterostilbene ameliorates these mechanisms, it is expected to have significant clinical applications.
Collapse
Grants
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 21K06926 Ministry of Education, Culture, Sports, Science and Technology
- 21K11223 Ministry of Education, Culture, Sports, Science and Technology
- 22K11423 Ministry of Education, Culture, Sports, Science and Technology
- 23K16547 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Shota Nukaga
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Yi Luo
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ryoichi Nishida
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Yoshihiro Miyagawa
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Chie Nakashima
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Ayaka Ikemoto
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Rika Sasaki
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Nara, Japan
| |
Collapse
|
3
|
McGlynn MC, Brady K, Healey JM, Dharnidharka VR, Ybarra AM, Stoll J, Sweet S, Hayashi RJ. Late effects in survivors of post-transplant lymphoproliferative disease. Pediatr Blood Cancer 2024; 71:e30777. [PMID: 37988230 DOI: 10.1002/pbc.30777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/12/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Treatment of post-transplant lymphoproliferative disease (PTLD) varies, with only some patients receiving chemotherapy. Concern for chemotherapy toxicities may influence treatment decisions as little is known regarding the late effects (LE) in PTLD survivors. This report characterizes LE in PTLD survivors at our institution. PROCEDURE Pediatric patients (0-18 years old) diagnosed with PTLD from 1990 to 2020 were examined. All patients included survived 6 months after completing chemotherapy or were 6 months from diagnosis if received no chemotherapy. Treatment with anti-CD20 antibody (rituximab) alone was not considered chemotherapy. Toxicities were classified per Common Terminology Criteria for Adverse Events Version 5.0. Chi-square tests assessed differences between categorical groups, or Fischer's exact test or the Fischer-Freeman-Halton exact test for limited sample sizes. RESULTS Of the 44 patients included, 24 (55%) were treated with chemotherapy. Twenty-four (55%) were alive at last follow-up. Chemotherapy was not associated with differences in survival (odds ratio [OR] 1.40, confidence interval [CI]: 0.42-4.63; p = .31). All patients experienced LE. Grade 3 toxicity or higher was experienced by 82% of patients with no difference in incidence (OR 1.20, CI: 0.27-5.80; p > .99) or median toxicity grade (3.00 vs. 4.00, p = .21) between treatment groups. Patients who received chemotherapy were more likely to experience blood and lymphatic toxicity (58% vs. 25%, p = .03) and cardiac toxicity (46% vs. 15%, p = .03), but less likely to have infections (54% vs. 85%, p = .03). CONCLUSIONS Survivors of PTLD experience LE including late mortality regardless of chemotherapy exposure. Further investigation to better understand LE could optimize upfront therapy for children with PTLD and improve outcomes.
Collapse
Affiliation(s)
- Mary Claire McGlynn
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Kassidy Brady
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Jessica M Healey
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Vikas R Dharnidharka
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - A Marion Ybarra
- Division of Pediatric Cardiology, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Janis Stoll
- Division of Pediatric Gastroenterology, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Stuart Sweet
- Division of Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Robert J Hayashi
- Division of Pediatric Hematology Oncology, Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Battisha A, Sawalha K, Obeidat Y, Patel B. Role of Cardiac Biomarkers in Monitoring Cardiotoxicity in Chemotherapy Patients. Crit Pathw Cardiol 2023; 22:83-87. [PMID: 37607037 DOI: 10.1097/hpc.0000000000000314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
PURPOSE This review aims to highlight the different types of chemotherapy-induced cardiotoxicity and will discuss the evidence base behind the use of different cardiac biomarkers to predict cardiovascular complications. Additionally, we will review the use of cardiac biomarkers to monitor cardiac outcomes and the role of cardioprotective medications in reducing cardiovascular side effects. RECENT FINDINGS Chemotherapy has been linked to an increased risk of cardiotoxicity and heart failure. Currently, patients receiving chemotherapy undergo echocardiogram before starting chemotherapy and every 6 months to monitor for any decline in cardiac function. We reviewed the current evidence and practice guidelines of monitoring chemotherapy cardiotoxicity. SUMMARY Cardio-oncology is a rapidly evolving subspecialty in cardiology, especially with the advent of new chemotherapeutic agents, which have cardiovascular side effects. Early detection of these effects is crucial to prevent life-threatening and irreversible cardiovascular outcomes. Monitoring troponin, pro-brain natriuretic peptide, and other cardiac biomarkers during chemotherapy will help to early detect cardiotoxicity.
Collapse
Affiliation(s)
- Ayman Battisha
- From the Department of Medicine, University of Massachusetts Chan Medical School-Baystate, Springfield, MA
| | - Khalid Sawalha
- From the Department of Medicine, University of Massachusetts Chan Medical School-Baystate, Springfield, MA
| | - Yasin Obeidat
- From the Department of Medicine, University of Massachusetts Chan Medical School-Baystate, Springfield, MA
| | - Brijesh Patel
- Department of Cardiology, Heart and Vascular Institute, West Virginia University, Morgantown, WV
| |
Collapse
|
5
|
Ho CC, Wen PC, Yu WC, Hu YW, Yang CC. Pre-existing chronic kidney disease and hypertension increased the risk of cardiotoxicity among colorectal cancer patients treated with anticancer drugs. J Chin Med Assoc 2021; 84:877-884. [PMID: 34320515 DOI: 10.1097/jcma.0000000000000590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND This population-based study was to investigate the potential risk factors of cardiotoxicity among colorectal cancer (CRC) patients treated with anticancer drugs. METHODS This was a retrospective cohort study using the National Health Insurance Research Database to identify the CRC patients receiving chemotherapy (CT) alone or CT combined with targeted therapies between 2000 and 2013. The patients enrolled were those who had the first diagnosis of CRC established ≥20 years and had no cancer history three years before the incident diagnosis of CRC. The outcomes of cardiotoxicity were defined by the diagnosis of acute myocarditis, cardiomyopathy, heart failure, hypertensive heart disease, and so on. RESULTS A total of 11 819 CRC patients were identified and 3781 were eligible; 556 (14.7%) patients developed cardiotoxicity after receiving anticancer treatment. Patients showed a similar risk of having primary outcome (hazard ratio [HR], 0.7; p = 0.3662) between CT and CT combined with targeted therapy groups, whereas the risk of developing secondary outcome was significantly different between the two groups (HR, 0.7; p = 0.0339). The hazard was found to be increased with age (60-69, HR 2.1, p = 0.0236; 70-79, HR 3.3, p = 0.0003; and ≥80, HR 3.7, p < 0.0001). CRC patients who had a prior history of hypertension exhibited a higher risk than those without hypertension (HR 1.6, p < 0.0001). The hazard of having cardiotoxicity among patients with a prior history of severe chronic kidney disease was 2.4 times than that in those without renal dysfunction, regardless of the stage of cancer (HR 2.4, p < 0.0001). CONCLUSION CRC patients over 60 years of age run a higher risk of developing cardiotoxicity when treated with anticancer drugs. For CRC patients who have a previous history of hypertension or chronic kidney disease, physicians must be careful in evaluating the risk of anticancer drugs-related cardiotoxicity. Prescribe drugs may prevent cardiotoxicity if necessary.
Collapse
Affiliation(s)
- Chin-Chin Ho
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan, ROC
- Institute of Environmental and Occupational Health Sciences, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Pin-Chun Wen
- Institute of Environmental and Occupational Health Sciences, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Wen-Chung Yu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Wen Hu
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chen-Chang Yang
- Institute of Environmental and Occupational Health Sciences, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Division of Clinical Toxicology and Occupational Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
6
|
Rotz SJ, Ryan TD, Hayek SS. Cardiovascular disease and its management in children and adults undergoing hematopoietic stem cell transplantation. J Thromb Thrombolysis 2021; 51:854-869. [PMID: 33230704 PMCID: PMC8085022 DOI: 10.1007/s11239-020-02344-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cell transplantation (HSCT) is a potentially curative treatment for many malignancies, hemoglobinopathies, metabolic diseases, bone marrow failure syndromes, and primary immune deficiencies. Despite the significant improvement in survival afforded by HSCT, the therapy is associated with major short and long-term morbidity and mortality. Cardiovascular complications such as cardiomyopathy, arrhythmias, pulmonary hypertension, and pericardial effusions are increasingly recognized as potential outcomes following HSCT. The incidence of cardiac complications is related to various factors such as age, co-morbid medical conditions, whether patients received cardiotoxic chemotherapy prior to HSCT, the type of HSCT (autologous versus allogeneic), and the specific conditioning regimen. Thus, the cardiovascular evaluation has become a core component of the pre-transplant assessment, however, the practice differs from center to center as national guidelines and contemporary high-quality studies are lacking. We review the incidence of cardiotoxicity in pediatric and adult HSCT, potential mechanisms of injury, and effects on long-term outcomes. We also discuss the possible therapeutic approaches when disease arises, as well as the indications and need for surveillance before, during, and after transplantation.
Collapse
Affiliation(s)
- Seth J Rotz
- Department of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Pediatric Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| | - Thomas D Ryan
- Department of Pediatrics, University of Cincinnati College of Medicine, and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
7
|
Chung R, Tyebally S, Chen D, Kapil V, Walker JM, Addison D, Ismail-Khan R, Guha A, Ghosh AK. Hypertensive Cardiotoxicity in Cancer Treatment-Systematic Analysis of Adjunct, Conventional Chemotherapy, and Novel Therapies-Epidemiology, Incidence, and Pathophysiology. J Clin Med 2020; 9:jcm9103346. [PMID: 33081013 PMCID: PMC7603211 DOI: 10.3390/jcm9103346] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiotoxicity is the umbrella term for cardiovascular side effects of cancer therapies. The most widely recognized phenotype is left ventricular dysfunction, but cardiotoxicity can manifest as arrhythmogenic, vascular, myocarditic and hypertensive toxicities. Hypertension has long been regarded as one of the most prevalent and modifiable cardiovascular risk factors in the general population, but its relevance during the cancer treatment journey may be underestimated. Hypertensive cardiotoxicity occurs de novo in a substantial proportion of treated cancer patients. The pathology is incompletely characterized—natriuresis and renin angiotensin system interactions play a role particularly in conventional treatments, but in novel therapies endothelial dysfunction and the interaction between the cancer and cardiac kinome are implicated. There exists a treatment paradox in that a significant hypertensive response not only mandates anti-hypertensive treatment, but in fact, in certain cancer treatment scenarios, hypertension is a predictor of cancer treatment efficacy and response. In this comprehensive review of over 80,000 patients, we explored the epidemiology, incidence, and mechanistic pathophysiology of hypertensive cardiotoxicity in adjunct, conventional chemotherapy, and novel cancer treatments. Conventional chemotherapy, adjunct treatments, and novel targeted therapies collectively caused new onset hypertension in 33–68% of treated patients. The incidence of hypertensive cardiotoxicity across twenty common novel therapies for any grade hypertension ranged from 4% (imatinib) to 68% (lenvatinib), and high grade 3 or 4 hypertension in <1% (imatinib) to 42% (lenvatinib). The weighted average effect was all-grade hypertension in 24% and grade 3 or 4 hypertension in 8%.
Collapse
Affiliation(s)
- Robin Chung
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London EC1A 7BE, UK; (R.C.); (S.T.); (D.C.)
- Cardio-Oncology Service, University College London Hospital, London WC1E 6HX, UK;
| | - Sara Tyebally
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London EC1A 7BE, UK; (R.C.); (S.T.); (D.C.)
| | - Daniel Chen
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London EC1A 7BE, UK; (R.C.); (S.T.); (D.C.)
- Cardio-Oncology Service, University College London Hospital, London WC1E 6HX, UK;
- Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK
| | - Vikas Kapil
- Barts Blood Pressure Centre of Excellence, Barts Heart Centre, St Bartholomew’s Hospital, London EC1A 7BE, UK;
- Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - J. Malcolm Walker
- Cardio-Oncology Service, University College London Hospital, London WC1E 6HX, UK;
- Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK
| | - Daniel Addison
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH 43210, USA; (D.A.); (A.G.)
| | - Roohi Ismail-Khan
- Cardio-oncology Program, H. Lee Moffitt Cancer Center, Tampa, FL 33559, USA;
| | - Avirup Guha
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH 43210, USA; (D.A.); (A.G.)
- Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Arjun K Ghosh
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew’s Hospital, London EC1A 7BE, UK; (R.C.); (S.T.); (D.C.)
- Cardio-Oncology Service, University College London Hospital, London WC1E 6HX, UK;
- Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK
- Correspondence: ; Tel.: +44-20-7377-7000
| |
Collapse
|
8
|
Balakrishnan KR, Rao KGS, Subramaniam G, Sharma D. Transplantation for chemotherapy-induced cardiomyopathy-case series and review of current practice. Indian J Thorac Cardiovasc Surg 2020; 36:287-293. [PMID: 33061213 DOI: 10.1007/s12055-020-01018-8] [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: 04/24/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 10/23/2022] Open
Abstract
Multimodality therapies have improved the survival after tumors like Ewing's sarcoma and breast cancer. However, cardiotoxicity following chemotherapy remains an important concern. We report a case series of four patients who presented to our heart team with severe dilated cardiomyopathy along with biventricular involvement. Two of the patients were females and had breast cancer for which they were treated with trastuzumab and had developed chemotherapy induced cardiomyopathy (CCMP). The other two patients were males who had Ewing's sarcoma who developed CCMP following treatment with doxorubicin.
Collapse
Affiliation(s)
| | | | | | - Dhruva Sharma
- Department of Cardiothoracic and Vascular Surgery, SMS Medical College & attached hospitals, Jaipur, Rajasthan India
| |
Collapse
|
9
|
Yu CX, Zhang YY, Wu XY, Tang HX, Liang XQ, Xue ZM, Xue YD, Li J, Zhu H, Huo R, Ban T. Transient receptor potential melastatin 4 contributes to early-stage endothelial injury induced by arsenic trioxide. Toxicol Lett 2019; 312:98-108. [DOI: 10.1016/j.toxlet.2019.04.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/03/2019] [Accepted: 04/30/2019] [Indexed: 01/09/2023]
|
10
|
Gill JH, Rockley KL, De Santis C, Mohamed AK. Vascular Disrupting Agents in cancer treatment: Cardiovascular toxicity and implications for co-administration with other cancer chemotherapeutics. Pharmacol Ther 2019; 202:18-31. [PMID: 31173840 DOI: 10.1016/j.pharmthera.2019.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/30/2019] [Indexed: 02/08/2023]
Abstract
Destruction of the established tumour vasculature by a class of compound termed Vascular Disrupting Agents (VDAs) is showing considerable promise as a viable approach for the management of solid tumours. VDAs induce a rapid shutdown and collapse of tumour blood vessels, leading to ischaemia and consequent necrosis of the tumour mass. Their efficacy is hindered by the persistence of a viable rim of tumour cells, supported by the peripheral normal vasculature, necessitating their co-administration with additional chemotherapeutics for maximal therapeutic benefit. However, a major limitation for the use of many cancer therapeutics is the development of life-threatening cardiovascular toxicities, with significant consequences for treatment response and the patient's quality of life. The aim of this review is to outline VDAs as a cancer therapeutic approach and define the mechanistic basis of cardiovascular toxicities of current chemotherapeutics, with the overall objective of discussing whether VDA combinations with specific chemotherapeutic classes would be good or bad in terms of cardiovascular toxicity.
Collapse
Affiliation(s)
- Jason H Gill
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK; School of Pharmacy, Faculty of Medical Sciences, Newcastle University, UK.
| | - Kimberly L Rockley
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| | - Carol De Santis
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| | - Asma K Mohamed
- Northern Institute for Cancer Research (NICR), Faculty of Medical Sciences, Newcastle University, UK
| |
Collapse
|
11
|
Tsai E, Mouhayar E, Lenihan D, Song J, Durand JB, Fadol A, Massey M, Harrison C, Basen-Engquist K. Feasibility and Outcomes of an Exercise Intervention for Chemotherapy-Induced Heart Failure. J Cardiopulm Rehabil Prev 2019; 39:199-203. [PMID: 31022003 PMCID: PMC6492623 DOI: 10.1097/hcr.0000000000000388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE Cancer treatment-related heart failure (HF) is an emerging health concern, as the number of survivors is increasing rapidly, and cardiac health issues are a leading cause of mortality in this population. While there is general evidence for the efficacy of exercise rehabilitation interventions, more research is needed on exercise rehabilitation interventions for patients specifically with treatment-induced HF and whether such interventions are safe and well-accepted. This study provides feasibility and health outcomes of a pilot exercise intervention for cancer survivors with chemotherapy-induced HF. METHODS Twenty-five participants were randomized to a clinic-based exercise intervention or a wait-list control group or, alternatively, allowed to enroll in a home-based exercise intervention if they declined the randomized study. For purposes of analysis, both types of exercise programs were combined into a single intervention group. Repeated-measures analysis of variance was conducted to assess for significant time and treatment group main effects separately and time × treatment group interaction effects. RESULTS Significant improvements in maximum oxygen uptake ((Equation is included in full-text article.)O2max) were observed in the intervention group. Intervention satisfaction and adherence were high for both clinic- and home-based interventions, with no reported serious adverse events. Enrollment was initially low for the clinic-based intervention, necessitating the addition of the home-based program as an intervention alternative. CONCLUSIONS Results suggest that exercise rehabilitation interventions are feasible in terms of safety, retention, and satisfaction and have the potential to improve (Equation is included in full-text article.)O2max. To maximize adherence and benefits while minimizing participant burden, an ideal intervention may incorporate elements of both clinic-based supervised exercise sessions and a home-based program.
Collapse
Affiliation(s)
- Edward Tsai
- Department of Health Promotion & Behavioral Science, The University of Texas School of Public Health, Houston, TX, USA
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elie Mouhayar
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Lenihan
- Cardiovascular Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jaejoon Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Bernard Durand
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anecita Fadol
- Department of Nursing, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mona Massey
- Clinical Research Support Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carol Harrison
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karen Basen-Engquist
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Chistyakova MV, Goncharova EV. Early diagnosis of cardiotoxic complications of chemotherapy: the possibility of radiation research methods. ACTA ACUST UNITED AC 2018; 58:11-17. [PMID: 30625104 DOI: 10.18087/cardio.2512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 12/26/2018] [Indexed: 11/18/2022]
Abstract
Oncological diseases are the main causes of death in the world. Modern treatment of cancer patients contributes to an increase in survival rate due to strong chemotherapeutic drugs, the use of which is accompanied by toxic effects on cardiomyocytes. The main manifestations of cardiotoxicity are left ventricular dysfunction, myocardial ischemia, thromboembolic complications, chronic heart failure. As a result, the risk of cardiovascular mortality may be higher than the risk of death from the tumor process. An important task of oncologists and cardiologists is the early diagnosis of cardiotoxic complications in order to start treatment in time and reduce mortality from cardiovascular pathology in cancer patients.
Collapse
|
13
|
Abstract
Although many cancer survivors diagnosed with early-stage disease will outlive their cancer, they may continue to experience long-term and/or latent side effects due to cancer treatment. Many of these side effects are common and contribute to worse quality of life, morbidity, and mortality for cancer survivors. This article summarizes the treatment side effects for several of the most prevalent cancers in the United States.
Collapse
Affiliation(s)
- Nana Gegechkori
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1087, New York, NY 10029, USA
| | - Lindsay Haines
- Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1087, New York, NY 10029, USA
| | - Jenny J Lin
- Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1087, New York, NY 10029, USA.
| |
Collapse
|
14
|
Biomy R, Abdelshafy M, Abdelmonem A, Abu-Elenin H, Ghaly G. Effect of Chronic Hepatitis C Virus Treatment by Combination Therapy on Cardiovascular System. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2017; 11:1179546817713204. [PMID: 28804248 PMCID: PMC5484549 DOI: 10.1177/1179546817713204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/16/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The prevalence of hepatitis C virus (HCV) in Egypt is quite high, and the combined oral direct-acting antiviral agents (DAAs) may have impressive results. OBJECTIVE To assess the cardiovascular effects of DAAs in patients with HCV. METHODS A total of 170 patients with HCV were divided into 2 groups: first group (100 patients) received triple combination therapy (pegylated interferon alfa, sofosbuvir, and ribavirin, whereas the second group (70 patients) received dual combination therapy (sofosbuvir and simeprevir). Group 1 patients were followed up for 1 year more than 3 visits, whereas group 2 patients were followed up for 6 months more than 2 visits; and the end point of the study was the development of a major cardiovascular event (eg, congestive heart failure, echocardiographic evidence of left ventricular dysfunction, occurrence of significant arrhythmias, or acute coronary syndrome). The following parameters were accomplished: medical history and clinical examination, electrocardiogram, echo-Doppler study, and laboratory investigations. RESULTS No significant differences were found between the 2 study groups regarding demographic criteria. None of the both group patients had developed any major cardiac event. No significant changes were observed regarding ST-T wave abnormalities, arrhythmias, or QT interval. None of the both group patients developed echocardiographic regional wall motion abnormalities at baseline or at study end. Systolic function parameters showed minute nonsignificant changes over study visits. Diastolic function parameters showed nonsignificant changes between baseline and 6-month and 12-month visits. CONCLUSIONS The DAAs used in combination regimen with interferon or used orally in combination do not significantly affect the cardio-vascular system.
Collapse
Affiliation(s)
- Reda Biomy
- Faculty of Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | | | | | | | - George Ghaly
- Al-Sahel Teaching Hospital, Cardiology Department Cairo, Egypt
| |
Collapse
|
15
|
Madeddu C, Deidda M, Piras A, Cadeddu C, Demurtas L, Puzzoni M, Piscopo G, Scartozzi M, Mercuro G. Pathophysiology of cardiotoxicity induced by nonanthracycline chemotherapy. J Cardiovasc Med (Hagerstown) 2016; 17 Suppl 1:e12-e18. [PMID: 27183520 DOI: 10.2459/jcm.0000000000000376] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The risk and mechanism of chemotherapy-induced cardiotoxicity (CTX) vary depending on the type and intensity of the anticancer regimen. Myriad chemotherapeutic drugs produce adverse cardiovascular effects such as arterial hypertension, heart failure, and thromboembolic events. Among the numerous classes of these drugs, anthracyclines have been studied most extensively because of their overt cardiovascular effects and the high associated incidence of heart failure. However, CTX might also be caused by other types of chemotherapeutic agents, including alkylating agents (cyclophosphamide, ifosfamide), platinum agents, antimetabolites (5-fluorouracil, capecitabine), antibiotics (mitoxantrone, mitomycin, bleomycin), and antimicrotubule agents (taxanes). Here, we review the incidence, clinical impact, and potential mechanisms of CTX associated with nonanthracycline chemotherapy used for cancer patients. The published data support a marked increase in CTX risk, particularly with certain drugs such as 5-fluorouracil and cisplatin. Each anticancer regimen is associated with distinct modes of heart damage, both symptomatic and asymptomatic. However, the underlying mechanisms of CTX have been established only in a few cases, and only few nonanthracycline chemotherapeutics (mitoxantrone, mitomycin, ifosfamide) act through a recognizable mechanism and show a predictable dose dependence. Lastly, nonanthracycline chemotherapy can induce both chronic lesions, such as systolic dysfunction, and acute lesions, such as the ischemia that occurs within hours or days after treatment. An increased understanding of the incidence, mechanisms, and potential therapeutic targets of CTX induced by various nonanthracycline chemotherapeutic agents is clearly required.
Collapse
Affiliation(s)
- Clelia Madeddu
- aDepartment of Medical Sciences Mario Aresu, Unit of Medical Oncology bDepartment of Medical Sciences Mario Aresu, Unit of Cardiology and Angiology, University Hospital Cagliari, University of Cagliari, Cagliari cDivision of Cardiology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale' - IRCCS, Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Gavila J, Seguí MÁ, Calvo L, López T, Alonso JJ, Farto M, Sánchez-de la Rosa R. Evaluation and management of chemotherapy-induced cardiotoxicity in breast cancer: a Delphi study. Clin Transl Oncol 2016; 19:91-104. [PMID: 27101413 PMCID: PMC5215075 DOI: 10.1007/s12094-016-1508-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/25/2016] [Indexed: 12/03/2022]
Abstract
Purpose While much progress has been made in the treatment of breast cancer, cardiac complications resulting from therapy remain a significant concern. Both anthracyclines and novel targeted agents can inflict cardiac damage. The present study aimed to evaluate the difference between what it is currently done and what standards of care should be used to minimizing and managing cardiac toxicity in breast cancer survivors. Methods A two-round multicenter Delphi study was carried out. The panel consisted of 100 oncologists who were asked to define the elected therapies for breast cancer patients, the clinical definition and patterns of cancer drug-derived cardiac toxicity, and those protocols focused on early detection and monitoring of cardiovascular outcomes. Results Experts agreed a more recent definition of cardiotoxicity. Around 38 % of patients with early-stage disease, and 51.3 % cases with advanced metastatic breast cancer had preexisting risk factors for cardiotoxicity. Among risk factors, cumulative dose of anthracycline ≥450 mg/m2 and its combination with other anticancer drugs, and a preexisting cardiovascular disease were considered the best predictors of cardiotoxicity. Echocardiography and radionuclide ventriculography have been the proposed methods for monitoring changes in cardiac structure and function. Breast cancer is generally treated with anthracyclines (80 %), so that the panel strongly stated about the need to plan a strategy to managing cardiotoxicity. A decline of left ventricular ejection fraction (LVEF) >10 %, to an LVEF value <53 % was suggested as a criterion for changing the dose schedule of anthracyclines, or suspending the treatment of chemotherapy plus trastuzumab until the normalization of the left ventricular function. The use of liposomal anthracyclines was strongly suggested as a treatment option for breast cancer patients. Conclusions The present report is the first to produce a set of statements on the prevention, evaluation and monitoring of chemotherapy-induced cardiac toxicity in breast cancer patients.
Collapse
Affiliation(s)
- J Gavila
- Servicio de Oncología Médica, Fundación Instituto Valenciano de Oncología, Calle del Profesor Beltrán Bàguena, 8, 46009, Valencia, Spain.
| | - M Á Seguí
- Servicio de Oncología Médica, Corporació Sanitaria ParcTaulí, Barcelona, Spain
| | - L Calvo
- Servicio de Oncología Médica, Complejo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - T López
- Servicio de Cardiología, Hospital Universitario La Paz, Madrid, Spain
| | - J J Alonso
- Servicio de Cardiología, Hospital Universitario de Getafe, Madrid, Spain
| | - M Farto
- Medical Department, TEVA Pharma, Madrid, Spain
| | | |
Collapse
|
17
|
Dorokhina EI, Magomedova AU, Shevelev AA, Kulikov SM, Gitis MK, Vedernikov AV, Vorobyev AI, Kravchenko SK. [Late cardiotoxicity of high-dose chemotherapy according to the modified NHL-BFM-90 program in adult patients with diffuse large B-cell lymphoma]. TERAPEVT ARKH 2015; 87:51-57. [PMID: 26390725 DOI: 10.17116/terarkh201587751-57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM To evaluate the late cardiotoxicity (CT) of high-dose chemotherapy (CT) according to the modified NHL-BFM-90 (mNHL-BFM-90) protocol in adult patients with diffuse large B-cell lymphoma (DLBCL). SUBJECTS AND METHODS The results of electrocardiography (ECG) and echocardiography (echoCG) were analyzed in 40 DLBCL patients treated according to the mNHL-BFM-90 program in the Hematology Research Center (HRC), Russian Academy of Medical Sciences (RAMS), in 2002 to 2009. A study group consisted of 20 men and 20 women whose age was 31 to 76 years; median age was 56.5 years at the time of their examination and the median follow-up time after therapy was 6 years. The individual cumulative dose of doxorubicin was 150-300 mg/M2. A comparison group included 19 patients receiving CHOP/R-CHOP CT in HRC, RAMS, in 2002 to 2009. Out of them, there were 8 men and 11 women whose age was 39 to 78 years median age was 70 years at the time of their examination. The individual cumulative dose of doxorubicin was 200-400 mg/M2. ECG and echoCG were carried out before and 5 years or more after CT. RESULTS Out of the 40 patients with DLBCL, the signs of subclinical cardiomyopathy (CMP) were detected in 24 (60%) patients; no clinical manifestations of congestive heart failure (CHF) were found in any patient. In the comparison group of 19 patients receiving CHOP/R-CHOP CT, 14 (74%) patients were found to have signs of subclinical CMP and no clinical signs of CHF. The summary toxicity index significantly depended on age (p=0.03) and a history of heart disease (p=0.3); it was significantly higher after CHOP/R-CHOP CT (p=0.05). There was a statistically significant relationship of the risk of subclinical CMP to the history of heart diseases (p=0.05). CONCLUSION Late cardiotoxicity of the mNHL-BFM-90 program does not exceed the toxicity of standard CHOP/R-CHOP therapy. Post-CT Echo-CG and ECG findings showed that the patients with the most marked subclinical signs of CMP in both groups had cardiotoxicity risk factors, such as coronary heart disease, hypertensive disease, or diabetes in their history. No clinically significant CHF was identified in any patient.
Collapse
Affiliation(s)
- E I Dorokhina
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A U Magomedova
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A A Shevelev
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - S M Kulikov
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - M K Gitis
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A V Vedernikov
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A I Vorobyev
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - S K Kravchenko
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
18
|
Piper SE, McDonagh TA. Chemotherapy-related Cardiomyopathy. Eur Cardiol 2015; 10:19-24. [PMID: 30310418 PMCID: PMC6159418 DOI: 10.15420/ecr.2015.10.01.19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/23/2015] [Indexed: 11/04/2022] Open
Abstract
Advances in chemotherapeutic agents have resulted in significantly improved cancer survival rates. Cardiac toxicity, however, has emerged as a leading cause of morbidity, both during and years after treatment. One of the most common manifestations of cardiotoxicity is that of heart failure and left ventricular systolic dysfunction. In this review, current opinions and guidelines in this field are discussed, with particular focus on the most common culprits, the anthracyclines, and the monoclonal antibody, trastuzumab.
Collapse
Affiliation(s)
- Susan E Piper
- King's College London, The James Black Centre, London, UK; Kings College Hospital NHS Foundation Trust, London, UK
| | - Theresa A McDonagh
- King's College London, The James Black Centre, London, UK; Kings College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
19
|
Abstract
Advances in chemotherapeutic agents over the past two decades have resulted in significantly improved cancer survival rates. Cardiac toxicity, however, has emerged as a leading cause of morbidity, both during and years after treatment. One of the most common manifestations of cardiotoxicity is that of heart failure and left ventricular systolic dysfunction. Consequently, the field of cardio-oncology is a rapidly emerging field of sub-specialty, with growing research interests in all aspects of management. In this review, current opinions and guidelines in this field are discussed, with particular focus on the most common culprits, the anthracyclines and the monoclonal antibody, trastuzumab.
Collapse
Affiliation(s)
- Susan Piper
- Department of Cardiovascular Research, King's College London, The James Black Center, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Theresa McDonagh
- Department of Cardiovascular Research, King's College London, The James Black Center, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
20
|
Blaes A, Duprez D, Defor T, Shanley R, Beckwith H, Haddad T, Potter D, Yee D, Sanghavi K, Jacobson P. Angiotensin Converting Enzyme Inhibitors (ACEI) and doxorubicin pharmacokinetics in women receiving adjuvant breast cancer treatment. SPRINGERPLUS 2015; 4:32. [PMID: 25646154 PMCID: PMC4309801 DOI: 10.1186/s40064-015-0802-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 01/03/2023]
Abstract
Purpose Doxorubicin (DOX) chemotherapy can cause cardiac complications. Angiotensin converting enzyme inhibitors (ACEI) may protect against these complications. We performed a pharmacokinetics (PK) study to determine whether DOX levels are altered in the presence of ACEI. Methods In this randomized, cross-over, single-blinded drug-drug interaction study, 19 women with breast cancer prescribed DOX and cyclophosphamide every 14 days received one cycle of DOX chemotherapy with ACEI enalapril 10 mg daily and another cycle of DOX with placebo. Blood samples for DOX and doxorubicinol were drawn at baseline, 0.5, 1.0, 2.0, 4.0, 24.0 and 48.0 hours after infusion with and without ACEI enalapril. Correlative laboratories were also obtained. PK data was analyzed using non compartmental methods and DOX and doxorubicinol area under the curve (AUC) 0 to infinity, Cmax and half-life were estimated. Paired t-tests were used to determine whether DOX and its metabolite were altered with the use of enalapril (P < 0.05). Results 17 women (median age 45 years) received 60 mg/m2 DOX every two weeks for four cycles. Mean (SD) AUC0- ∞ for DOX and doxorubicinol with enalapril exposure was 1185.56 (44.64) hr*ng/ml and 1040 (80.6) hr*ng/ml, respectively. AUC0- ∞ for DOX and doxobubicinol without enalapril was 1167.73 (45.26) hr*ng/ml and 1056.32 (92.03) hr*ng/ml, respectively. There is no interaction between DOX and enalapril. Enalapril was tolerated (33% grade 1 dizziness). Conclusion ACEI, enalapril, does not appear to alter the PK of DOX. Ongoing efforts to determine the effectiveness of ACEI as a cardioprotective agent in women receiving DOX chemotherapy should be continued.
Collapse
Affiliation(s)
- Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, 420 Delaware Street, S.E., MMC 480, Minneapolis, MN 55455 USA
| | - Daniel Duprez
- University of Minnesota, Division of Cardiology, Minneapolis, MN USA
| | - Todd Defor
- Biostatistics Core, University of Minnesota, Minneapolis, MN USA
| | - Ryan Shanley
- Biostatistics Core, University of Minnesota, Minneapolis, MN USA
| | - Heather Beckwith
- Division of Hematology/Oncology/Transplantation, University of Minnesota, 420 Delaware Street, S.E., MMC 480, Minneapolis, MN 55455 USA
| | | | - David Potter
- Division of Hematology/Oncology/Transplantation, University of Minnesota, 420 Delaware Street, S.E., MMC 480, Minneapolis, MN 55455 USA
| | - Douglas Yee
- Division of Hematology/Oncology/Transplantation, University of Minnesota, 420 Delaware Street, S.E., MMC 480, Minneapolis, MN 55455 USA
| | - Kinjal Sanghavi
- College of Pharmacy, University of Minnesota, Minneapolis, MN USA
| | - Pamala Jacobson
- College of Pharmacy, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
21
|
Christenson ES, James T, Agrawal V, Park BH. Use of biomarkers for the assessment of chemotherapy-induced cardiac toxicity. Clin Biochem 2014; 48:223-35. [PMID: 25445234 DOI: 10.1016/j.clinbiochem.2014.10.013] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 10/25/2014] [Accepted: 10/28/2014] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To review the evidence for the use of various biomarkers in the detection of chemotherapy associated cardiac damage. DESIGN AND METHODS Pubmed.gov was queried using the search words chemotherapy and cardiac biomarkers with the filters of past 10years, humans, and English language. An emphasis was placed on obtaining primary research articles looking at the utility of biomarkers for the detection of chemotherapy-mediated cardiac injury. RESULTS Biomarkers may help identify patients undergoing treatment who are at high risk for cardiotoxicity and may assist in identification of a low risk cohort that does not necessitate continued intensive screening. cTn assays are the best studied biomarkers in this context and may represent a promising and potentially valuable modality for detecting cardiac toxicity in patients undergoing chemotherapy. Monitoring cTnI levels may provide information regarding the development of cardiac toxicity before left ventricular dysfunction becomes apparent on echocardiography or via clinical symptoms. A host of other biomarkers have been evaluated for their utility in the field of chemotherapy related cardiac toxicity with intermittent success; further trials are necessary to determine what role they may end up playing for prediction and prognostication in this setting. CONCLUSIONS Biomarkers represent an exciting potential complement or replacement for echocardiographic monitoring of chemotherapy related cardiac toxicity which may allow for earlier realization of the degree of cardiac damage occurring during treatment, creating the opportunity for more timely modulation of therapy.
Collapse
Affiliation(s)
- Eric S Christenson
- Johns Hopkins University, Department of Medicine, Baltimore, MD 21287, USA.
| | - Theodore James
- Johns Hopkins University, Department of Medicine, Baltimore, MD 21287, USA
| | - Vineet Agrawal
- Johns Hopkins University, Department of Medicine, Baltimore, MD 21287, USA
| | - Ben H Park
- Johns Hopkins University, Department of Oncology, Baltimore, MD 21287, USA
| |
Collapse
|
22
|
Accordino MK, Neugut AI, Hershman DL. Cardiac effects of anticancer therapy in the elderly. J Clin Oncol 2014; 32:2654-61. [PMID: 25071122 DOI: 10.1200/jco.2013.55.0459] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cancer incidence increases with age, and as life expectancy increases, the number of elderly patients with cancer is increasing. Cancer treatments, including chemotherapy and radiotherapy, have significant short- and long-term effects on cardiovascular function. These cardiotoxic effects can be acute, such as changes in electrocardiogram (ECG), arrhythmias, ischemia, and pericarditis and/or myocarditis-like syndromes, or they can be chronic, such as ventricular dysfunction. Anticancer therapies can also have indirect effects, such as alterations in blood pressure, or can cause metabolic abnormalities that subsequently increase risk for cardiac events. In this review, we explore both observational and clinical trial evidence of cardiac risk in the elderly. In both observational and clinical trial data, risk of cardiotoxicity with anthracycline-based chemotherapy increases with age. However, it is less clear whether the association between age and cardiotoxicity exists for newer treatments. The association may not be well demonstrated as a result of under-representation of elderly patients in clinical trials and avoidance of these therapies in this population. In addition, we discuss strategies for surveillance and prevention of cardiotoxicity in the elderly. In the elderly, it is important to be aware of the potential for cardiotoxicity during long-term follow-up and to consider both prevention and surveillance of these late effects.
Collapse
Affiliation(s)
| | - Alfred I Neugut
- All authors: Columbia University Medical Center, New York, NY
| | - Dawn L Hershman
- All authors: Columbia University Medical Center, New York, NY.
| |
Collapse
|
23
|
Locatelli M, Criscitiello C, Esposito A, Minchella I, Goldhirsch A, Cipolla C, Curigliano G. QTc prolongation induced by targeted biotherapies used in clinical practice and under investigation: a comprehensive review. Target Oncol 2014; 10:27-43. [DOI: 10.1007/s11523-014-0325-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 06/12/2014] [Indexed: 12/14/2022]
|
24
|
Lipshultz SE, Adams MJ, Colan SD, Constine LS, Herman EH, Hsu DT, Hudson MM, Kremer LC, Landy DC, Miller TL, Oeffinger KC, Rosenthal DN, Sable CA, Sallan SE, Singh GK, Steinberger J, Cochran TR, Wilkinson JD. Long-term cardiovascular toxicity in children, adolescents, and young adults who receive cancer therapy: pathophysiology, course, monitoring, management, prevention, and research directions: a scientific statement from the American Heart Association. Circulation 2013; 128:1927-95. [PMID: 24081971 DOI: 10.1161/cir.0b013e3182a88099] [Citation(s) in RCA: 382] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
25
|
Abstract
Modern cancer therapies are highly effective in the treatment of various malignancies, but their use is limited by the potential for cardiotoxicity. The most frequent and typical clinical manifestation of cardiotoxicity is left ventricular dysfunction, induced not only by cytotoxic conventional cancer therapy like anthracyclines, but also by new antitumor targeted therapy such as trastuzumab. The current standard for monitoring cardiac function, based on periodic assessment of left ventricular ejection fraction detects cardiotoxicity only when a functional impairment has already occurred, precluding any chance of preventing its development. A novel approach, based on the use of cardiac biomarkers has emerged in the last decade, resulting in a cost-effective diagnostic tool for early, real-time identification, assessment and monitoring of cardiotoxicity. In particular, prophylactic treatment with enalapril in patients with an early increase in troponin after chemotherapy has been shown to be very effective in preventing left ventricular dysfunction and associated cardiac events. In patients developing cancer treatment induced-cardiomyopathy, complete left ventricular ejection fraction recovery and a reduction of cardiac events may be achieved only when left ventricular dysfunction is detected early after the end of cancer treatment and treatment with angiotensin-converting enzyme inhibitors, possibly in combination with beta-blockers, is promptly initiated.
Collapse
|
26
|
Abstract
OPINION STATEMENT The increase in survivorship of cancer patients makes the understanding of the available options for prevention and treatment of cardiotoxicity induced by antineoplastic agents a crucial topic both for cardiologists and oncologists. The most frequent and typical clinical manifestation of cardiotoxicity is asymptomatic or symptomatic left ventricular dysfunction, which may progress to overt heart failure. It may be induced not only by conventional cancer therapy, like anthracyclines, but also by new antitumoral targeted therapy such as trastuzumab. The current standard for monitoring cardiac damage during antineoplastic treatment, mainly based on the quantification of left ventricular ejection fraction, detects cardiac toxicity only when a functional impairment has already occurred. Evaluation of cardiac biomarkers such as troponin, however, has shown excellent sensitivity in the early detection of cardiotoxicity by the identification of patients with subclinical cardiac injury that precedes the development of cardiac dysfunction. The use of angiotensin-converting enzyme inhibitors in patients with troponin elevation during chemotherapy may be an effective tool to prevent left ventricular ejection fraction reduction and late cardiac events. There are no well established recommendations for treatment of cancer patients who develop cardiac dysfunction. Angiotensin-converting enzyme inhibitors and beta-blockers have proven to be effective in this setting. However, there are concerns in using these medications in cancer patients, and therefore the tendency is to treat patients only if symptomatic. However, the clinical benefit of these medications may be more evident in asymptomatic patients, and the recovery of cardiac function strongly depends on the amount of time elapsed from the end of chemotherapy to the start of heart failure therapy. This observation suggests that the early detection of cardiac damage is crucial and early use of angiotensin-converting enzyme inhibitors and beta-blockers should be considered in patients with left ventricular dysfunction induced by antineoplastic agents.
Collapse
|
27
|
Colombo A, Cardinale D. Using cardiac biomarkers and treating cardiotoxicity in cancer. Future Cardiol 2013; 9:105-18. [PMID: 23259478 DOI: 10.2217/fca.12.73] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cardiotoxicity is a frequent and serious adverse effect of both conventional and novel anticancer treatments, affecting patient survival and quality of life. The current standard for cardiac monitoring during cancer therapy, mainly based on left ventricular ejection fraction assessment, detects myocardial damage only when a functional impairment has already occurred, not allowing for early preventive strategies. Measurement of cardiospecific biomarkers has proven to have higher prognostic value than imaging modalities. In particular, cardiac troponin elevation during chemotherapy allows the identification of patients who are more prone to develop myocardial dysfunction and cardiac events during follow-up. In these patients, the use of an angiotensin-converting enzyme inhibitor such as enalapril has shown to be effective in improving clinical outcome, giving the chance for a cardioprotective strategy in a selected population. Once left ventricular dysfunction occurs, heart failure therapies currently used for other forms of left ventricular dysfunction, particularly angiotensin-converting enzyme inhibitors and β-blockers, seem to be effective. However, their use in cancer patients is still undervalued.
Collapse
Affiliation(s)
- Alessandro Colombo
- Cardiology Division, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| | | |
Collapse
|
28
|
Bonsignore A, Warburton D. The mechanisms responsible for exercise intolerance in early-stage breast cancer: What role does chemotherapy play? Hong Kong Physiother J 2013. [DOI: 10.1016/j.hkpj.2013.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
29
|
Almawardy R, Elhammady W, Mousa N, Abotaleb S. Is combination therapy for chronic hepatitis C toxic for cardiac function? HEPATITIS MONTHLY 2012; 12:e6254. [PMID: 23105944 PMCID: PMC3477652 DOI: 10.5812/hepatmon.6254] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/14/2012] [Accepted: 07/27/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Many types of cardiovascular complications such as; cardiac arrhythmias, impaired cardiac function, myocardial ischemia and decreased left ventricular function, have been attributed to interferon therapy. OBJECTIVES The aim of this study was to evaluate the effects of combination therapy pegylated interferon and ribavirin on left ventricular systolic and diastolic functions in patients with a chronic hepatitis C infection. PATIENTS AND METHODS A total of 120 patients, eligible for hepatitis C virus (HCV) treatment with pegylated interferon and ribavirin, were included in this study. All patients underwent a full cardiovascular baseline examination including; detailed medical history, thorough clinical examination, 12 lead electrocardiogram (ECG), and echocardiography. A cardiac evaluation was performed at the beginning and six months after starting combination therapy. RESULTS No significant changes regarding cardiac symptoms including; shortness of breath, cough, palpitations, chest pain and hypertension, were found during or six months after starting the combined therapy. ECG findings showed statistically non-significant decreases in the QT interval, while corrected QT showed statistically non-significant increases six months after beginning combined therapy, when compared to their values before treatment. Also with regard to the echocardiography findings, there was no statistically significant difference found between any of the echocardiography parameters six months after starting combined therapy compared to their values before treatment. CONCLUSIONS The results of our study suggest that, combination therapy does not cause a significant deterioration in cardiac function in patients with a chronic hepatitis C infection, and it may be used safely in patients without cardiac disease.
Collapse
Affiliation(s)
| | | | - Nasser Mousa
- Tropical Department, Mansoura University, Mansoura, Egypt
- Corresponding author: Nasser Mousa, Tropical Medicine Department, Mansoura University, Mansoura, Egypt.Tel.: +20-573606440, Fax: E-mail:
| | | |
Collapse
|
30
|
Hedhli N, Russell KS. Cardiotoxicity of molecularly targeted agents. Curr Cardiol Rev 2011; 7:221-33. [PMID: 22758623 PMCID: PMC3322440 DOI: 10.2174/157340311799960636] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Revised: 06/08/2011] [Accepted: 12/07/2011] [Indexed: 02/06/2023] Open
Abstract
Cardiac toxicity of molecularly targeted cancer agents is increasingly recognized as a significant side effect of chemotherapy. These new potent therapies may not only affect the survival of cancer cells, but have the potential to adversely impact normal cardiac and vascular function. Unraveling the mechanisms by which these therapies affect the heart and vasculature is crucial for improving drug design and finding alternative therapies to protect patients predisposed to cardiovascular disease. In this review, we summarize the classification and side effects of currently approved molecularly targeted chemotherapeutics.
Collapse
Affiliation(s)
- Nadia Hedhli
- Department of Internal Medicine-Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kerry S Russell
- Department of Internal Medicine-Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
31
|
Abstract
Cancer treatment has improved extraordinarily in recent years. The development of targeted therapies has widened the cardiotoxic spectrum of antineoplastic drugs. Optimum management of cardiovascular disease before and during antineoplastic treatment is essential to reduce morbidity and mortality in cancer patients. This article reviews the incidence and characteristics of cardiotoxic effects of antineoplastic drugs with special focus on the pathophysiological mechanisms. It also emphasizes the importance of early detection and correction of cardiovascular risk factors and the relevance of close cardiac monitoring during antineoplastic treatment in order to reduce cardiotoxicity.
Collapse
Affiliation(s)
- I Brana
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | | |
Collapse
|
32
|
Cardinale D. Treatment and prevention of cardiotoxicity due to anticancer therapy. J Cardiovasc Echogr 2011. [DOI: 10.1016/j.jcecho.2011.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
33
|
Abstract
Cardiac complications resulting from chemotherapy and radiation pose a significant risk for morbidity and mortality to the cancer survivor. Cardiac side effects may progress over time and are a concern for patients treated during childhood. Long-term pulmonary complications are relatively infrequent, and acute respiratory effects of drugs (mostly bleomycin) or radiation normally resolve early after therapy. Although most cardiovascular risk statistics and clinical experience are derived from patients treated before 1985, the modern radiation approach that limits the exposure of the heart and reduces the total dose seems to attenuate the previously observed cardiovascular risk. Potential preventive measures for high-risk patients are of increasing interest but remain experimental.
Collapse
Affiliation(s)
- Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA.
| | | |
Collapse
|
34
|
Jarkowski A, Glode AE, Spangenthal EJ, Wong MKK. Heart Failure Caused by Molecularly Targeted Therapies for Cancer. Pharmacotherapy 2011; 31:62-75. [DOI: 10.1592/phco.31.1.62] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
35
|
Tolentino GP, Battaglini CL, Araújo SS, Otano AS, Conde DM, Evans ES, de Oliveira RJ. Cardiorespiratory fitness and quality-of-life analysis posttreatment in breast cancer survivors. J Psychosoc Oncol 2010; 28:381-98. [PMID: 20623414 DOI: 10.1080/07347332.2010.484831] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This study compared VO(2peak), treatment frequency and dosage, and QOL between 22 post-treated breast cancer survivors (CATotal) and 22 apparently healthy, age-matched women (CO). The CATotal group included 11 with no history of any other co-morbidities (CA) and 11 with controlled hypertension (CA + H). VO(2peak) was measured using the Bruce Protocol. QOL was measured using the SF-36 survey. Significant differences were observed in VO(2peak) between CATotal and CO (p = 0.014), CA and CA + H (p = 0.001), and CA + H and CO (p = 0.001). Physical, emotional, and mental health domains of the SF-36 were significantly different between CATotal and CO (p = 0.006, 0.001, and 0.05 respectively). These results suggest that breast cancer survivors with controlled hypertension can experience a significant reduction in VO(2peak) when compared to apparently healthy, age-matched controls. Also, treatment frequency and dosage affected QOL in this group of survivors.
Collapse
|
36
|
|
37
|
Cardinale D, Colombo A, Torrisi R, Sandri MT, Civelli M, Salvatici M, Lamantia G, Colombo N, Cortinovis S, Dessanai MA, Nolè F, Veglia F, Cipolla CM. Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol 2010; 28:3910-6. [PMID: 20679614 DOI: 10.1200/jco.2009.27.3615] [Citation(s) in RCA: 443] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Treatment of breast cancer with trastuzumab is complicated by cardiotoxicity in up to 34% of the patients. In most patients, trastuzumab-induced cardiotoxicity (TIC) is reversible: left ventricular ejection fraction (LVEF) improves after trastuzumab withdrawal and with, or sometimes without, initiation of heart failure (HF) therapy. The reversibility of TIC, however, is not foreseeable, and identification of patients at risk and of those who will not recover from cardiac dysfunction is crucial. The usefulness of troponin I (TNI) in the identification of patients at risk for TIC and in the prediction of LVEF recovery has never been investigated. PATIENTS AND METHODS In total, 251 women were enrolled. TNI was measured before and after each trastuzumab cycle. LVEF was evaluated at baseline, every 3 months during trastuzumab therapy, and every 6 months afterward. In case of TIC, trastuzumab was discontinued, and HF treatment with enalapril and carvedilol was initiated. TIC was defined as LVEF decrease of > 10 units and below 50%. Recovery from TIC was defined as LVEF increase above 50%. RESULTS TIC occurred in 42 patients (17%) and was more frequent in patients with TNI elevation (TNI+; 62% v 5%; P < .001). Twenty-five patients (60%) recovered from TIC. LVEF recovery occurred less frequently in TNI+ patients (35% v 100%; P < .001). At multivariate analysis, TNI+ was the only independent predictor of TIC (hazard ratio [HR], 22.9; 95% CI, 11.6 to 45.5; P < .001) and of lack of LVEF recovery (HR, 2.88; 95% CI,1.78 to 4.65; P < .001). CONCLUSION TNI+ identifies trastuzumab-treated patients who are at risk for cardiotoxicity and are unlikely to recover from cardiac dysfunction despite HF therapy.
Collapse
|
38
|
Cardinale D, Colombo A, Cipolla CM. Prevention and treatment of cardiomyopathy and heart failure in patients receiving cancer chemotherapy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2010; 10:486-95. [PMID: 19026179 DOI: 10.1007/s11936-008-0041-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chemotherapy (CT)-induced cardiotoxicity remains an unresolved problem that strongly affects the quality of life and overall survival of cancer patients. The most typical form of cardiotoxicity, a dilated cardiomyopathy (CMP), usually becomes manifest late in the course of the disease and is classically considered to be refractory to therapy. Preventing cardiotoxicity remains the most important strategy, and several measures have been proposed, including cardiac function monitoring, limitation of CT dose, use of anthracycline analogues and cardioprotectants, and early detection of cardiotoxicity by biomarkers. The response to modern heart failure therapy of CT-induced CMP has never been evaluated in clinical trials, and no definite guidelines have been adopted. Although it is likely that medications used for other forms of CMP, particularly angiotensin-converting enzyme inhibitors and beta-blockers, may be highly effective, there is still some unjustified concern regarding their use in cancer patients.
Collapse
Affiliation(s)
- Daniela Cardinale
- Cardiology Unit, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| | | | | |
Collapse
|
39
|
Raschi E, Vasina V, Ursino MG, Boriani G, Martoni A, De Ponti F. Anticancer drugs and cardiotoxicity: Insights and perspectives in the era of targeted therapy. Pharmacol Ther 2010; 125:196-218. [DOI: 10.1016/j.pharmthera.2009.10.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 10/05/2009] [Indexed: 01/01/2023]
|
40
|
Kaufman KR, Chhabra S, Levitt M, Sood R. Arsenic trioxide and olanzapine co-administration: case analysis. J Oncol Pharm Pract 2009; 17:260-4. [DOI: 10.1177/1078155209352912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
QTc prolongation is associated with arsenic trioxide (ATO) treatment of acute promyelocytic leukemia (APL). Olanzapine was safely co-administered with ATO to treat co-morbid psychiatric diagnoses. It is important to closely monitor for drug—drug interactions and cumulative drug adverse effects in patients receiving oncology agents and psychotropics. Further research is indicated to determine risk/benefit profiles of psychotropics co-administered with ATO. In light of current limited data, co-administration of psychotropics with ATO should be reported presenting both instances wherein no interactions are noted and those with adverse effects.
Collapse
Affiliation(s)
- Kenneth R Kaufman
- Department of Psychiatry, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA; Department of Neurology, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA; Department of Anesthesiology, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA, /
| | - Saurabh Chhabra
- Department of Medicine, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Michael Levitt
- Department of Medicine, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Rahul Sood
- Department of Anesthesiology, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
41
|
Aksnes LH, Bauer HCF, Dahl AA, Fosså SD, Hjorth L, Jebsen N, Lernedal H, Hall KS. Health status at long-term follow-up in patients treated for extremity localized Ewing Sarcoma or osteosarcoma: a Scandinavian sarcoma group study. Pediatr Blood Cancer 2009; 53:84-9. [PMID: 19343771 DOI: 10.1002/pbc.22027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND The purpose of this study was to evaluate late effects and symptom complaints in long-term survivors (>5 years) of Extremity Bone Sarcoma (EBS survivors). The results were compared with findings in age- and gender-matched individuals from the general population (NORMs). PATIENTS AND METHODS Among 155 EBS survivors approached, 133 (86%) were included, and 110 of them (83%) attended an outpatient examination. Health status was evaluated by a mailed questionnaire concerning demographic and current health issues, and physical examinations at the outpatient clinic. Age- and gender-adjusted normative controls were drawn from participants of the Health Study of Nord-Trøndelag County (HUNT 2). RESULTS Median age at follow-up was 29 (15-57) years. Median follow-up was 12 (6-22) years. Of EBS survivors 42% had > or =1 somatic disease, 33% had ototoxicity and 13% had reduced renal function. EBS survivors were more likely to have heart disease (odds ratio [OR], 7.9; 95% confidence interval [95% CI], 2.5-25.3; P = 0.001), hypertension (OR, 3.4; 95% CI, 1.1-10.1; P = 0.03) and thyroid disease (OR, 3.0; 95% CI, 1.1-8.3; P = 0.04) compared to NORMs. EBS survivors reported more diarrhoea (29% vs. 19%, P = 0.02), palpitations (23% vs. 13%, P = 0.01) and shortness of breath (11% vs. 5%, P = 0.01) than NORMs. CONCLUSIONS EBS survivors have poorer health status compared to age- and gender-matched controls. Long-term follow-up of these patients is therefore mandatory.
Collapse
Affiliation(s)
- L H Aksnes
- Department of Oncology, The Norwegian Radium Hospital, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wong MKK, Jarkowski A. Response to sorafenib after sunitinib-induced acute heart failure in a patient with metastatic renal cell carcinoma: case report and review of the literature. Pharmacotherapy 2009; 29:473-8. [PMID: 19323623 DOI: 10.1592/phco.29.4.473] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cardiotoxicity is an emerging concern with a new class of drugs known as targeted agents, which include trastuzumab and sunitinib. Sunitinib is a small molecule that inhibits multiple tyrosine kinase receptors. This drug was approved by the United States Food and Drug Administration in 2006 for the treatment of clear cell metastatic renal cell carcinoma and advanced gastrointestinal stromal tumors. We describe a 65-year-old woman who was treated with sunitinib for metastatic clear cell renal cell carcinoma. After 5 months of therapy, she developed acute heart failure requiring hospitalization; sunitinib was immediately discontinued. The patient had classic symptoms of heart failure, including pleural effusion. An echocardiogram revealed a left ventricular ejection fraction of 30%. She received standard treatment for heart failure, including a beta-blocker, an angiotensin-converting enzyme inhibitor, and diuretics. Within 1 month, the patient's symptoms resolved, and subsequent cardiac evaluation showed that her left ventricular ejection fraction returned to normal. According to the Common Terminology Criteria for Adverse Events developed by the National Cancer Institute, her cardiac event associated with sunitinib was defined as grade III toxicity. One month later, sorafenib, another tyrosine kinase inhibitor, was started with the aim of continuing her previous response to sunitinib. After 7 months of sorafenib therapy, the patient had no evidence of heart failure, and her condition was responding to treatment. Clinicians should be aware that sunitinib-induced heart failure occurs occultly and that many--but not all--cases resolve with discontinuation of the drug. Use of sorafenib after sunitinib-induced heart failure appears to be safe and effective, which suggests that cardiotoxicity is not a general class effect of the tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Michael K K Wong
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | |
Collapse
|
43
|
Siemann DW, Chaplin DJ, Walicke PA. A review and update of the current status of the vasculature-disabling agent combretastatin-A4 phosphate (CA4P). Expert Opin Investig Drugs 2009; 18:189-97. [PMID: 19236265 DOI: 10.1517/13543780802691068] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular-disrupting strategies impair a tumor's blood vessel network, which is essential for tumor progression and metastasis. Vascular-disrupting agents (VDAs) cause a rapid and selective vascular shutdown in tumors to produce extensive secondary neoplastic cell death due to ischemia. A lead agent in this therapeutic strategy is the tubulin depolymerizing agent combretastatin-A4 phosphate (CA4P). Used alone, CA4P induces extensive necrosis in a wide variety of preclinical cancer models and significant blood flow reductions in the patient tumors. Preclinical and clinical data further indicate that CA4P can effectively be combined with chemotherapy or radiotherapy. Finally, the potential of combining VDAs with antiangiogenic therapies has shown considerable promise in preclinical models and such combinations are now beginning to be evaluated in patients.
Collapse
Affiliation(s)
- Dietmar W Siemann
- Department of Radiation Oncology, University of Florida, 2000 SW Archer Road, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
44
|
Barrett-Lee PJ, Dixon JM, Farrell C, Jones A, Leonard R, Murray N, Palmieri C, Plummer CJ, Stanley A, Verrill MW. Expert opinion on the use of anthracyclines in patients with advanced breast cancer at cardiac risk. Ann Oncol 2009; 20:816-27. [PMID: 19153118 DOI: 10.1093/annonc/mdn728] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Anthracyclines are considered to be among the most active agents for the treatment of breast cancer. However, their use is limited by cumulative, dose-related cardiotoxicity. Such cardiotoxicity results in a permanent loss of cardiac myocytes and a progressive reduction in cardiac function following each subsequent dose of anthracycline. Initially, damage to the heart is subclinical; however, increasingly impaired cardiac function can result in cardiovascular symptoms, with serious cardiac injury resulting in chronic heart failure. Since the early detection and treatment of cardiotoxicity can reduce its clinical effects, it is important that oncologists are aware of these adverse effects and manage them appropriately. This review examines the risk factors for anthracycline-associated cardiotoxicity and offers recommendations on strategies to reduce the cardiotoxicity of anthracyclines in the management of patients with advanced breast cancer.
Collapse
|
45
|
Cardinale D, Colombo A, Lamantia G, Colombo N, Civelli M, De Giacomi G, Pandini C, Sandri MT, Cipolla CM. Cardio-oncology: a new medical issue. Ecancermedicalscience 2008; 2:126. [PMID: 22275992 PMCID: PMC3234071 DOI: 10.3332/ecancer.2008.126] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Indexed: 11/18/2022] Open
Abstract
Due to the increasing number of long-term cancer survivors, the ageing of the population, as well as the increased incidence and prevalence of oncologic and cardiovascular diseases, the number of patients presenting oncologic and cardiologic co-morbidities are increasing. Accordingly, there is a rapidly growing need for a comprehensive and proficient management of patients in whom the two co-morbidities exist, and for cancer patients whose clinical history and oncologic treatment put them at higher risk for developing cardiovascular problems, in order to provide the optimal treatment in every situation, and to avoid the possibility that the development of the second disease does not lead to a reduction of therapeutic opportunities for the patient. A new discipline, cardio-oncology, has been created to deal with this need. Its aim is to investigate new strategies, collect new evidence-based indications and develop interdisciplinary expertise in order to manage this growing category of patients. Cardio-oncology deals with the following main clinical and research areas: early diagnosis of cardiotoxicity, risk stratification and preventions, treatment and monitoring of cardiotoxicity.
Collapse
Affiliation(s)
- D Cardinale
- Cardiology Unit and Laboratory Medicine Unit, European Institute of Oncology, IRCCS, 20141 Milan, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cruz-Monteagudo M, Munteanu CR, Borges F, Cordeiro MND, Uriarte E, Chou KC, González-Díaz H. Stochastic molecular descriptors for polymers. 4. Study of complex mixtures with topological indices of mass spectra spiral and star networks: The blood proteome case. POLYMER 2008. [DOI: 10.1016/j.polymer.2008.09.070] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Abstract
BACKGROUND The ancient drug, arsenic, has remarkable efficacy in the treatment of relapsed acute promyelocytic leukemia (APL) and this success has led to exploration of its use in other malignancies. OBJECTIVE To provide an overview of the mechanism of action of arsenic and summarize its development in the treatment of APL and other malignant disorders. METHODS A 20-year search of MEDLINE, EMBASE and Web of Science was conducted. RESULTS/CONCLUSIONS A series of clinical trials with arsenic trioxide has confirmed its benefit in the therapy of APL. Its role in the treatment of other malignancies remains to be determined. Careful attention to the clinical management of patients on arsenic trioxide therapy can significantly lessen the risk of major side effects.
Collapse
Affiliation(s)
- Mark R Litzow
- Mayo Clinic, Division of Hematology, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
48
|
Goldsmith YB, Roistacher N, Baum MS. Capecitabine-Induced Coronary Vasospasm. J Clin Oncol 2008; 26:3802-4. [DOI: 10.1200/jco.2008.17.1637] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yuliya B. Goldsmith
- Department of Cardiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Nancy Roistacher
- Department of Cardiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Michael S. Baum
- Department of Cardiology, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
49
|
Sarapa N, Britto MR. Challenges of characterizing proarrhythmic risk due to QTc prolongation induced by nonadjuvant anticancer agents. Expert Opin Drug Saf 2008; 7:305-18. [DOI: 10.1517/14740338.7.3.305] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Yahalom J, Portlock CS. Long-Term Cardiac and Pulmonary Complications of Cancer Therapy. Hematol Oncol Clin North Am 2008; 22:305-18, vii. [DOI: 10.1016/j.hoc.2008.01.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|