1
|
Chang HM, Hsu JY, Ahn C, Yeh ETH. Prevention of Heart Failure Induced by Doxorubicin with Early Administration of Dexrazoxane (PHOENIX Study): dose response and time course of dexrazoxane-induced degradation of topoisomerase 2b. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:42. [PMID: 40317097 DOI: 10.1186/s40959-025-00339-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Dexrazoxane, a putative iron chelator, is effective in preventing doxorubicin-induced cardiotoxicity. However, dexrazoxane is also a catalytic inhibitor of topoisomerase 2b (Top2b), a key mediator of doxorubicin toxicity. Preclinical studies have shown that dexrazoxane induces Top2b degradation, and early administration (8 h before doxorubicin) can prevent doxorubicin-induced cardiotoxicity. In this study, we investigated the dose-response relationship and time course of dexrazoxane-induced Top2b degradation in human volunteers. METHODS Twenty-five healthy female volunteers received an intravenous infusion of dexrazoxane at doses ranging from 100 mg/m2 to 500 mg/m2. Blood samples were collected hourly from time zero to 12 h, as well as at 24- and 48-h post-infusion. Peripheral blood mononuclear cells (PBMCs) were isolated, nuclear fractions were extracted, and Top2b expression was analyzed by western blot using Lamin B1 as a control. A linear mixed-effects model was used to assess differences among the five dose groups. RESULTS Dexrazoxane infusion led to a rapid and sustained reduction of Top2b in PBMCs, lasting up to 12 h. Statistical analysis revealed a significant difference in Top2b levels among the five dose groups (p = 0.0002). Subgroup analysis identified a significant difference between the 100 mg/m2 and 500 mg/m2 groups (p = 0.005). However, topoisomerase 2a (Top2a), the molecular target of doxorubicin's tumor-killing effect, remained unchanged following dexrazoxane infusion. CONCLUSIONS Findings from this dose-response and time-course study can inform the design of future clinical trials investigating the efficacy of early dexrazoxane administration in preventing doxorubicin-induced cardiotoxicity while minimizing the risk of tumor protection. TRIAL REGISTRATION (Funded by the National Institute of Health, RO1HL151993; PHOENIX trials, ClinicalTrials.gov number, NCT03930680.).
Collapse
Affiliation(s)
- Hui-Ming Chang
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
- W. P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| | - Jinn-Yuan Hsu
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Chul Ahn
- School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward T H Yeh
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
- W. P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| |
Collapse
|
2
|
Reyna-Luna J, Soriano-Agueda L, Vera CJ, Franco-Pérez M. Insights into the coordination chemistry of antineoplastic doxorubicin with 3d-transition metal ions Zn 2+, Cu 2+, and VO 2+: a study using well-calibrated thermodynamic cycles and chemical interaction quantum chemistry models. J Comput Aided Mol Des 2023; 37:279-299. [PMID: 37245168 DOI: 10.1007/s10822-023-00506-4] [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: 04/20/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
We present a computational strategy based on thermodynamic cycles to predict and describe the chemical equilibrium between the 3d-transition metal ions Zn2+, Cu2+, and VO2+ and the widely used antineoplastic drug doxorubicin. Our method involves benchmarking a theoretical protocol to compute gas-phase quantities using DLPNO Coupled-Cluster calculations as reference, followed by estimating solvation contributions to the reaction Gibbs free energies using both explicit partial (micro)solvation steps for charged solutes and neutral coordination complexes, as well as a continuum solvation procedure for all solutes involved in the complexation process. We rationalized the stability of these doxorubicin-metal complexes by inspecting quantities obtained from the topology of their electron densities, particularly the bond critical points and non-covalent interaction index. Our approach allowed us to identify representative species in solution phase, infer the most likely complexation process for each case, and identify key intramolecular interactions involved in the stability of these compounds. To the best of our knowledge, this is the first study reporting thermodynamic constants for the complexation of doxorubicin with transition metal ions. Unlike other methods, our procedure is computationally affordable for medium-sized systems and provides valuable insights even with limited experimental data. Furthermore, it can be extended to describe the complexation process between 3d-transition metal ions and other bioactive ligands.
Collapse
Affiliation(s)
- Julieta Reyna-Luna
- Departamento de Física y Química Teórica, Facultad de Química, Universidad Nacional Autónoma de México, Cd. Universitaria, 04510, Ciudad de Mexico, México
| | - Luis Soriano-Agueda
- Donostia International Physics Center (DIPC), 20018, Donostia, Euskadi, Spain
| | - Christiaan Jardinez Vera
- Laboratorio de Modelado y Simulación Computacional en Nanomedicina, Escuela Superior de Apan, Universidad Autónoma del Estado de Hidalgo, Carretera Apan-Calpulalpan S/N, Colonia, 43920, Chimalpa Tlalayote, Hgo, México
| | - Marco Franco-Pérez
- Departamento de Física y Química Teórica, Facultad de Química, Universidad Nacional Autónoma de México, Cd. Universitaria, 04510, Ciudad de Mexico, México.
| |
Collapse
|
3
|
Kassaian SE, Gandhi B, Barac A. Cardio-oncology: Implications for Clinical Practice for Women. Curr Cardiol Rep 2022; 24:1685-1698. [PMID: 36112292 DOI: 10.1007/s11886-022-01779-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/21/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Clinical cardio-oncology considerations specific to women span across many areas and are particularly relevant for management of patients with sex-specific cancers, such as breast cancer. RECENT FINDINGS Major improvement in breast cancer survivorship over the last decade and the recognition of CV disease as the second leading cause of death among survivors point to the relevance of long-term cardiovascular (CV) safety. This review summarizes the CV effects associated with multimodality breast cancer treatments and contemporary approach to CV risk stratification, prevention, early detection, monitoring, and management at the time of cancer diagnosis, during and after completion of treatment. We highlight the growing role of a multidisciplinary, team-based approach for comprehensive CV and oncology care through the entire cancer treatment continuum, from diagnosis through survivorship.
Collapse
Affiliation(s)
- Seyed Ebrahim Kassaian
- J.D. Murphy Jr. Cardio-Oncology Fellowship Program, MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Georgetown University, 110 Irving Street, NW, Suite 1A130, Washington, DC, 20010, USA
| | - Bhumika Gandhi
- Cancer Survivorship Program, MedStar Georgetown University Hospital, 3800 Reservoir Road, Washington, DC, 20007, USA
| | - Ana Barac
- MedStar Heart and Vascular Institute, MedStar Washington Hospital Center, Georgetown University, 110 Irving Street, NW, Suite 1A130, Washington, DC, 20010, USA.
| |
Collapse
|
4
|
Sayed A, Abdelfattah OM, Munir M, Shazly O, Awad AK, Ghaith HS, Moustafa K, Gerew M, Guha A, Barac A, Fradley MG, Abela GS, Addison D. Long-term effectiveness of empiric cardio-protection in patients receiving cardiotoxic chemotherapies: A systematic review & bayesian network meta-analysis. Eur J Cancer 2022; 169:82-92. [PMID: 35524992 DOI: 10.1016/j.ejca.2022.03.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cardioprotective therapies represent an important avenue to reduce treatment-limiting cardiotoxicities in patients receiving chemotherapy. However, the optimal duration, strategy and long-term efficacy of empiric cardio-protection remains unknown. METHODS Leveraging the MEDLINE/Pubmed, CENTRAL and clinicaltrials.gov databases, we identified all randomised controlled trials investigating cardioprotective therapies from inception to November 2021 (PROSPERO-ID:CRD42021265006). Cardioprotective classes included ACEIs, ARBs, Beta-blockers, dexrazoxane (DEX), statins and mineralocorticoid receptor antagonists. The primary end-point was new-onset heart failure (HF). Secondary outcomes were the mean difference in left ventricular ejection fraction (LVEF) change, hypotension and all-cause mortality. Network meta-analyses were used to assess the cardioprotective effects of each therapy to deduce the most effective therapies. Both analyses were performed using a Bayesian random effects model to estimate risk ratios (RR) and 95% credible intervals (95% CrI). RESULTS Overall, from 726 articles, 39 trials evaluating 5931 participants (38.0 ± 19.1 years, 72.0% females) were identified. The use of any cardioprotective strategy associated with reduction in new-onset HF (RR:0.32; 95% CrI:0.19-0.55), improved LVEF (mean difference: 3.92%; 95% CrI:2.81-5.07), increased hypotension (RR:3.27; 95% CrI:1.38-9.87) and no difference in mortality. Based on control arms, the number-needed-to-treat for 'any' cardioprotective therapy to prevent one incident HF event was 45, including a number-needed-to-treat of 21 with ≥1 year of therapy. Dexrazoxane was most effective at HF prevention (Surface Under the Cumulative Ranking curve: 81.47%), and mineralocorticoid receptor antagonists were most effective at preserving LVEF (Surface Under the Cumulative Ranking curve: 99.22%). CONCLUSION Cardiotoxicity remains a challenge for patients requiring anticancer therapies. The initiation of extended duration cardioprotection reduces incident HF. Additional head-to-head trials are needed.
Collapse
Affiliation(s)
- Ahmed Sayed
- Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Omar M Abdelfattah
- Department of Medicine, Morristown Medical Center, Atlantic Health System, Morristown, NJ, USA.
| | - Malak Munir
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Omar Shazly
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed K Awad
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Khaled Moustafa
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Maria Gerew
- Department of Medicine, Morristown Medical Center, Atlantic Health System, Morristown, NJ, USA
| | - Avirup Guha
- Cardio-Oncology Program, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Cardio-Oncology Program, Division of Cardiology, Ohio State University, Columbus, OH, USA
| | - Ana Barac
- Cardio-Oncology Program, Medstar Heart and Vascular Institute, Georgetown University, Washington, DC, USA
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - George S Abela
- Department of Cardiovascular Medicine, Michigan State University, East Lansing, MI, USA
| | - Daniel Addison
- Cardio-Oncology Program, Division of Cardiology, Ohio State University, Columbus, OH, USA; Division of Cancer Control and Prevention, James Cancer Hospital and Solove Research Institute at the Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Saleh Y, Abdelkarim O, Herzallah K, Abela GS. Anthracycline-induced cardiotoxicity: mechanisms of action, incidence, risk factors, prevention, and treatment. Heart Fail Rev 2021; 26:1159-1173. [PMID: 32410142 DOI: 10.1007/s10741-020-09968-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Anthracycline is a mainstay in treatment of many cancers including lymphoma and breast cancer among many others. However, anthracycline treatment can be cardiotoxic. Although anthracycline-induced cardiotoxicity is dose dependent, it can also occur early at the onset of treatment and even up to several years following completion of treatment. This review article focuses on the understanding of mechanisms of anthracycline-induced cardiotoxicity, the treatments, and recommended follow-up and preventive approaches.
Collapse
Affiliation(s)
- Yehia Saleh
- Department of Internal Medicine, Michigan State University, East Lansing, MI, USA
| | - Ola Abdelkarim
- Department of Internal Medicine, Cardiology, Michigan State University, 788 service road, Room B-208, Clinical Center, East Lansing, MI, USA
| | - Khader Herzallah
- Department of Internal Medicine, Michigan State University, East Lansing, MI, USA
| | - George S Abela
- Department of Internal Medicine, Cardiology, Michigan State University, 788 service road, Room B-208, Clinical Center, East Lansing, MI, USA.
| |
Collapse
|
6
|
Benjamin RS, Minotti G. Doxorubicin-Dexrazoxane from Day 1 for Soft-tissue Sarcomas: The Road to Cardioprotection. Clin Cancer Res 2021; 27:3809-3811. [PMID: 33990361 DOI: 10.1158/1078-0432.ccr-21-1376] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
Doxorubicin cardiac toxicity is widely misunderstood, largely preventable, and starts with the first dose. This article reviews the history of doxorubicin cardiac toxicity and strategies for minimizing it. Dexrazoxane cardioprotection can safely be initiated on day 1 without compromising antitumor activity, allowing doxorubicin administration beyond the reported maximum lifetime dose.See related articles by Van Tine et al., p. 3854 and Jones et al., p. 3861.
Collapse
Affiliation(s)
- Robert S Benjamin
- Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Giorgio Minotti
- Department of Medicine, Center for Integrated Research and Unit of Drug Sciences, University Campus Bio-Medico of Rome, Rome, Italy.
| |
Collapse
|
7
|
Anthracycline-Related Heart Failure: Certain Knowledge and Open Questions : Where Do we Stand with Chemotherapyinduced Cardiotoxicity? Curr Heart Fail Rep 2020; 17:357-364. [PMID: 32964378 PMCID: PMC7683464 DOI: 10.1007/s11897-020-00489-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/24/2022]
Abstract
In the last decade, cardio-oncology has become a discipline on its own, with tremendous research going on to unravel the mechanisms underpinning different manifestations of cardiotoxicity caused by anticancer drugs. Although this domain is much broader than the effect of chemotherapy alone, a lot of questions about anthracycline-induced cardiotoxicity remain unknown. In this invited review, we provide insights in molecular mechanisms behind anthracycline-induced cardiotoxicity and put it in a clinical framework emphasizing the need for patients to understand, detect, and treat this detrimental condition.
Collapse
|
8
|
Abstract
Anthracycline-associated cardiomyopathy and peripartum cardiomyopathy are nonischemic cardiomyopathies that often afflict previously healthy young patients; both diseases have been well described since at least the 1970s and both occur in the settings of predictable stressors (ie, cancer treatment and pregnancy). Despite this, the precise mechanisms and the ability to reliably predict who exactly will go on to develop cardiomyopathy and heart failure in the face of anthracycline exposure or childbirth have proven elusive. For both cardiomyopathies, recent advances in basic and molecular sciences have illuminated the complex balance between cardiomyocyte and endothelial homeostasis via 3 broad pathways: reactive oxidative stress, interference in apoptosis/growth/metabolism, and angiogenic imbalance. These advances have already shown potential for specific, disease-altering therapies, and as our mechanistic knowledge continues to evolve, further clinical successes are expected to follow.
Collapse
Affiliation(s)
- Joshua A Cowgill
- From the Department of Cardiovascular Medicine, Maine Medical Center, Portland
| | - Sanjeev A Francis
- From the Department of Cardiovascular Medicine, Maine Medical Center, Portland
| | - Douglas B Sawyer
- From the Department of Cardiovascular Medicine, Maine Medical Center, Portland
| |
Collapse
|
9
|
Abstract
Toxic injury is one of the many ways by which the functional integrity of the heart may become compromised. Any of the subcellular elements may be the target of toxic injury, including all of the various membranes and organelles. Understanding the mechanisms underlying cardiotoxicity may lead to treatment of the toxicity or to its prevention. Doxorubicin and its analogs are very important cancer chemotherapeutic agents that can cause cardiotoxicity. Other agents which are cardiotoxic and which have profound public health implications include the alkaloid emetine in ipecac syrup, cocaine, and ethyl alcohol. The most important cardiotoxic mechanisms proposed for doxorubicin include oxidative stress with its resultant damage to myocardial elements, changes in calcium homeostasis, decreased ability to produce ATP, and systemic release of cardiotoxic humoral mediators from tissue mast cells. Each of the first 3 mechanisms can lead to each of the other 2, and the causal relationships between all of these mechanisms are not clear. New evidence suggests that doxorubicinol, one of the metabolites of doxorubicin may be the moiety responsible for cardiotoxicity. Several other potential mechanisms also have been proposed for doxorubicin. Emetine in ipecac syrup is the first aid treatment of choice for many acute toxic oral ingestions and the alkaloid, itself, is used to treat amebiasis. Cardiotoxicity occurs following chronic exposure, such as occurs therapeutically in amebiasis and with ipecac abuse by bulemics. A number of mechanisms are proposed for emetine cardiotoxicity, but the current mechanistic literature is quite scarce. Cocaine abuse recently has caught the public interest, in particular because of the drug-related sudden deaths of certain athletes. Cocaine can cause hypertension, arrhythmias, and reduced coronary blood flow, each of which can contribute to its lethality. However, it may be possible that cocaine sudden death episodes are more related to hyperthermia and convulsive seizures, rather than to cardiovascular toxicity. Chronic alcohol use leads to dilated cardiomyopathy and failure as part of the general physical degeneration that occurs with alcoholism. Several mechanisms are proposed for the cardiomyopathy, but only 2 things seem clear. The cardiotoxicity is due to an intrinsic effect of alcohol, rather than to malnutrition or co-toxicity, and abstinence is the only effective treatment for the cardiomyopathy. Recent articles indicate that very moderate use of alcohol may be beneficial and protect against cardiovascular-related morbidity. One explanation for these findings seems to be that the non-drinking groups, against whom the moderate drinking comparisons were made, were enriched in former drinkers with significant alcohol-related cardiovascular pathology.
Collapse
|
10
|
Macedo AVS, Hajjar LA, Lyon AR, Nascimento BR, Putzu A, Rossi L, Costa RB, Landoni G, Nogueira-Rodrigues A, Ribeiro ALP. Efficacy of Dexrazoxane in Preventing Anthracycline Cardiotoxicity in Breast Cancer. JACC: CARDIOONCOLOGY 2019; 1:68-79. [PMID: 34396164 PMCID: PMC8352186 DOI: 10.1016/j.jaccao.2019.08.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 01/23/2023]
Abstract
Objectives The authors performed a systematic review and meta-analysis of randomized and nonrandomized trials on the efficacy of dexrazoxane in patients with breast cancer who were treated with anthracyclines with or without trastuzumab. Background Breast cancer treatment with anthracyclines and trastuzumab is associated with an increased risk of cardiotoxicity. Among the various strategies to reduce the risk of cardiotoxicity, dexrazoxane is an option for primary prevention, but it is seldom used in clinical practice. Methods Online databases were searched from January 1990 up to March 1, 2019, for clinical trials on the use of dexrazoxane for the prevention of cardiotoxicity in patients with breast cancer receiving anthracyclines with or without trastuzumab. Risk ratios (RRs) with 95% confidence intervals (CIs) were calculated using a random-effects model meta-analysis. Results Seven randomized trials and 2 retrospective trials with a total of 2,177 patients were included. Dexrazoxane reduced the risk of clinical heart failure (RR: 0.19; 95% CI: 0.09 to 0.40; p < 0.001) and cardiac events (RR: 0.36; 95% CI: 0.27 to 0.49; p < 0.001) irrespective of previous exposure to anthracyclines. The rate of a partial or complete oncological response, overall survival, and progression-free survival were not affected by dexrazoxane. Conclusions Dexrazoxane reduced the risk of clinical heart failure and cardiac events in patients with breast cancer undergoing anthracycline chemotherapy with or without trastuzumab and did not significantly impact cancer outcomes. However, the quality of available evidence is low, and further randomized trials are warranted before the systematic implementation of this therapy in clinical practice.
Collapse
Affiliation(s)
- Ariane V S Macedo
- Department of Cardiology of Hospital das Clínicas, Federal University of Minas Gerais and Department of Internal Medicine, School of Medicine of Federal University of Minas Gerais, Belo Horizonte, Brazil.,Oncoclínicas Group, Belo Horizonte, Brazil
| | - Ludhmila A Hajjar
- Department of Cardiopneumology of InCor and Division of Cardio-Oncology, Cancer Institute of Sao Paulo, School of Medicine of São Paulo University, São Paulo, Brazil
| | - Alexander R Lyon
- Cardio-oncology Service, Royal Brompton Hospital and Imperial College London, United Kingdom
| | - Bruno R Nascimento
- Department of Cardiology of Hospital das Clínicas, Federal University of Minas Gerais and Department of Internal Medicine, School of Medicine of Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alessandro Putzu
- Division of Anesthesiology, Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Lorenzo Rossi
- Institute of Oncology of Southern Switzerland, Bellinzona, Switzerland
| | | | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Angélica Nogueira-Rodrigues
- Division of Oncology of Hospital das Clínicas, Federal University of Minas Gerais and Department of Internal Medicine, School of Medicine of Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Antonio L P Ribeiro
- Department of Cardiology of Hospital das Clínicas, Federal University of Minas Gerais and Department of Internal Medicine, School of Medicine of Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Filomena D, Versacci P, Cimino S, Mattiucci C, Maestrini V, Cantisani D, Petronilli V, Agati L, Schiavetti A. Echocardiographic long-term follow-up of adult survivors of pediatric cancer treated with Dexrazoxane-Anthracyclines association. Int J Cardiol 2019; 299:271-275. [PMID: 31422879 DOI: 10.1016/j.ijcard.2019.07.096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/28/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022]
Abstract
AIMS Cardiovascular disease is a well-recognized cause of increased late morbidity and mortality among survivors of childhood cancer treated with anthracyclines. Co-administration of Dexrazoxane has been shown to significantly reduce short-term and mid-term cardiotoxicity. Aim of this study was to assess cardiac function in long-term (>10 years) survivors of childhood tumors treated with dexrazoxane/anthracycline association. METHODS AND RESULTS Twenty cancer survivors previously treated with co-administration of anthracyclines-dexrazoxane for childhood renal tumors or sarcoma and a control group of 20 healthy subjects were enrolled in the study. Echocardiographic measurements included 3D left ventricular (LV) ejection fraction (LVEF) and LV and right ventricular (RV) global longitudinal strain (GLS). Among cancer survivors group the median age at diagnosis was 5 years (1-17) and they were evaluated at median follow-up time of 21.5 years (10-26). No evidence of cardiac toxicity, as defined by current guidelines, was reported in all survivors. No significant differences in standard and deformation imaging parameters were observed between survivors and controls (3D LVEF 58 ± 3% vs 60 ± 5% p = NS; LV GLS -21 ± 1% vs -21 ± 2% p = NS; RV GLS -23 ± 2% vs -23 ± 5% p = NS). No second tumor was registered in dexrazoxane-treated survivors. CONCLUSIONS Our findings may support the role of dexrazoxane as a useful strategy for cardio-protection in children undergoing anthracycline based treatment. However, large randomized trials are needed to confirm the cardio-protective role of dexrazoxane in pediatric setting at long-term follow-up.
Collapse
Affiliation(s)
- D Filomena
- Department of Cardiovascular, Respiratory, Nephrological, Aenesthesiological and Geriatric Sciences, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy.
| | - P Versacci
- Department of Pediatrics, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - S Cimino
- Department of Cardiovascular, Respiratory, Nephrological, Aenesthesiological and Geriatric Sciences, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - C Mattiucci
- Department of Pediatrics, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - V Maestrini
- Department of Cardiovascular, Respiratory, Nephrological, Aenesthesiological and Geriatric Sciences, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - D Cantisani
- Department of Cardiovascular, Respiratory, Nephrological, Aenesthesiological and Geriatric Sciences, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - V Petronilli
- Department of Cardiovascular, Respiratory, Nephrological, Aenesthesiological and Geriatric Sciences, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - L Agati
- Department of Cardiovascular, Respiratory, Nephrological, Aenesthesiological and Geriatric Sciences, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| | - A Schiavetti
- Department of Pediatrics, "Sapienza" University of Rome, Policlinico Umberto I, Rome, Italy
| |
Collapse
|
12
|
Abstract
Doxorubicin-induced cardiotoxicity in childhood cancer survivors is a growing problem. The population of patients at risk for cardiovascular disease is steadily increasing, as five-year survival rates for all types of childhood cancers continue to improve. Doxorubicin affects the developing heart differently from the adult heart and in a subset of exposed patients, childhood exposure leads to late, irreversible cardiomyopathy. Notably, the prevalence of late-onset toxicity is increasing in parallel with improved survival. By the year 2020, it is estimated that there will be 500,000 childhood cancer survivors and over 50,000 of them will suffer from doxorubicin-induced cardiotoxicity. The majority of the research to-date, concentrated on childhood cancer survivors, has focused mostly on clinical outcomes through well-designed epidemiological and retrospective cohort studies. Preclinical studies have elucidated many of the cellular mechanisms that elicit acute toxicity in cardiomyocytes. However, more research is needed in the areas of early- and late-onset cardiotoxicity and more importantly improving the scientific understanding of how other cells present in the cardiac milieu are impacted by doxorubicin exposure. The overall goal of this review is to succinctly summarize the major clinical and preclinical studies focused on doxorubicin-induced cardiotoxicity. As the prevalence of patients affected by doxorubicin exposure continues to increase, it is imperative that the major gaps in existing research are identified and subsequently utilized to develop appropriate research priorities for the coming years. Well-designed preclinical research models will enhance our understanding of the pathophysiology of doxorubicin-induced cardiotoxicity and directly lead to better diagnosis, treatment, and prevention. © 2019 American Physiological Society. Compr Physiol 9:905-931, 2019.
Collapse
Affiliation(s)
- Trevi R. Mancilla
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Brian Iskra
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Gregory J. Aune
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| |
Collapse
|
13
|
Fimasartan for Remodeling after Myocardial Infarction. J Clin Med 2019; 8:jcm8030366. [PMID: 30875971 PMCID: PMC6463200 DOI: 10.3390/jcm8030366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/02/2019] [Accepted: 03/07/2019] [Indexed: 11/17/2022] Open
Abstract
An angiotensin receptor blocker (ARB) mitigates cardiac remodeling after myocardial infarction (MI). Here, we investigated the effect of fimasartan, a new ARB, on cardiac remodeling after MI. Sprague–Dawley rats were assigned into 3 groups: surgery only (sham group, n = 7), MI without (MI-only group, n = 13), and MI with fimasartan treatment (MI + Fima group, n = 16). MI was induced by the permanent ligation of the left anterior descending artery. Treatment with fimasartan (10 mg/kg) was initiated 24 h after MI and continued for 7 weeks. Rats in the MI + Fima group had a higher mean ejection fraction (66.3 ± 12.5% vs. 51.3 ± 14.8%, P = 0.002) and lower left ventricular end-diastolic diameter (9.14 ± 1.11 mm vs. 9.91 ± 1.43 mm, P = 0.045) than those in the MI-only group at 7 weeks after MI. The infarct size was lower in the MI + Fima than in the MI group (P < 0.05). A microarray analysis revealed that the expression of genes related to the lipid metabolism and mitochondrial membrane ion transporters were upregulated, and those involved in fibrosis and inflammation were downregulated by fimasartan. Fimasartan attenuates cardiac remodeling and dysfunction in rats after MI and may prevent the progression to heart failure after MI.
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW We review the cardiotoxic chemotherapeutic agents, the clinical and subclinical presentations and progression of their cardiotoxicity, and the management of the subsequent cardiovascular disease in survivors of childhood cancer. We discuss various preventive measures, especially the cardioprotectant, dexrazoxane, whose use with anthracycline chemotherapy, including doxorubicin, is based on strong evidence. Most treatment recommendations for this unique population are based on expert opinion, not on empirical evidence. RECENT FINDINGS As patients with childhood cancers live longer, morbidity from the cardiac side effects of chemotherapy is increasing. Treatment-related cardiac damage is irreversible and often progressive. It is imperative that such damage be prevented with strategies such as limiting the cumulative anthracycline dose, the use of anthracycline structural analogues and the use of cardioprotective agents. SUMMARY A deeper understanding of the mechanisms of their cardiotoxicity reveals that there is no 'safe' dose of anthracyclines. However, certain risk factors, such as higher lifetime anthracycline cumulative doses, higher anthracycline dose rates, female sex, longer follow-up, younger age at anthracycline treatment and cardiac irradiation, are associated with more severe cardiotoxicity. We advocate the use of dexrazoxane to limit the cardiotoxic effects of anthracycline chemotherapy.
Collapse
|
15
|
Wahab MH, Akoul ES, Abdel-Aziz AA. Modulatory Effects of Melatonin and Vitamin E on Doxorubicin-Induced Cardiotoxicity in Ehrlich Ascites Carcinoma-Bearing Mice. TUMORI JOURNAL 2018; 86:157-62. [PMID: 10855855 DOI: 10.1177/030089160008600210] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Doxorubicin (Dox), an anthracycline antibiotic, has a wide spectrum of antitumor activity with dose-limiting cardiotoxicity. The drug's toxicity is known to be closely related to the generation of active oxygen free radicals. In our study the normal cardiac tissue contents of total protein, glutathione (GSH) and malondialdehyde (MDA) were significantly decreased, by 25%, 33% and 92%, respectively, in the group of mice bearing Ehrlich ascites carcinoma (EAC) and treated with Dox (4 mg/kg/week x 2, ip). Administration of melatonin (5 mg/kg/day x 15, po) starting 24 hours prior to Dox treatment significantly increased the cardiac contents of total protein and GSH as well as the superoxide dismutase (SOD) activity, by 31%, 36% and 39%, respectively, compared to treatment with Dox only, while the content of MDA was decreased by 26%. Similarly, administration of vitamin E (250 mg/kg/day x 15, po) starting 24 hours prior to Dox treatment significantly increased the cardiac contents of total protein, GSH and SOD, by 23%, 26% and 42%, respectively, while the cardiac content of MDA was decreased by 35% compared with the Dox-only-treated group. As to the oncolytic activity of Dox, pretreatment of EAC-bearing mice with melatonin (5 mg/kg/day x 30, po) or vitamin E (250 mg/kg/day x 30, po) 24 hours prior to Dox administration (4 mg/kg/week x 4, ip) improved the antitumor activity of Dox as indicated by the increase in the average life span of the animals and the number of long-term survivors as well as the decrease in body weight loss induced by Dox treatment. It is clear from these results that administration of melatonin not only protects against the cardiotoxicity induced by Dox treatment but also enhances its antitumor activity to a more significant extent than does vitamin E.
Collapse
Affiliation(s)
- M H Wahab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | | | | |
Collapse
|
16
|
Nogueira SS, Sousa MG, Gava FN, Rosa FA, Melo GD, Dittrich G, Machado GF, Camacho AA. Matrix metalloproteinases 2 and 9 in rabbits with doxorubicin-induced cardiomyopathy. PESQUISA VETERINARIA BRASILEIRA 2018. [DOI: 10.1590/1678-5150-pvb-4990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
ABSTRACT: Some studies have shown the role played by matrix metalloproteinases and their inhibitors in doxorubicin cardiotoxicity. In this study, we sought to investigate how plasma and myocardial MMP 2 and 9 perform in rabbits with doxorubicin-induced cardiomyopathy, searching for a correlation between the activity of these collagenases and cardiac remodeling. Cardiomyopathy was induced by doxorubicin given intravenously twice a week for six consecutive weeks. Plasma MMP activity and the echocardiogram were assessed at baseline, and at 15 and 45 days after first injection of doxorubicin. The myocardial activity of these enzymes was solely evaluated in nine rabbits at 45 days, and results were compared with nine healthy controls. We only identified the full-length forms of both MMP 2 and 9 throughout the study. The plasma pro-MMP 2 reduced along the deterioration of cardiac function, while the pro-MMP 9 increased significantly at T45 as compared to baseline and T15. A negative significant correlation was found to exist between the plasma activity of pro-MMP 2 and mitral E-to-mitral septal annular early diastolic velocity ratio, which is an estimate of mean left atrial pressure and congestion. Only pro-MMP 2 was found in myocardial samples, and mean activity of such enzyme was statistically lower than that recorded for healthy controls. Although no active form was documented for either collagenase, the duration of the treatment with doxorubicin played a role in the alteration of plasma pro-forms activity. However, these changes could not be associated with most echocardiographic parameters that are supportive of cardiac remodeling.
Collapse
|
17
|
Lee JH, Wendorff TJ, Berger JM. Resveratrol: A novel type of topoisomerase II inhibitor. J Biol Chem 2017; 292:21011-21022. [PMID: 29074616 DOI: 10.1074/jbc.m117.810580] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/18/2017] [Indexed: 02/04/2023] Open
Abstract
Resveratrol, a polyphenol found in various plant sources, has gained attention as a possible agent responsible for the purported health benefits of certain foods, such as red wine. Despite annual multi-million dollar market sales as a nutriceutical, there is little consensus about the physiological roles of resveratrol. One suggested molecular target of resveratrol is eukaryotic topoisomerase II (topo II), an enzyme essential for chromosome segregation and DNA supercoiling homeostasis. Interestingly, resveratrol is chemically similar to ICRF-187, a clinically approved chemotherapeutic that stabilizes an ATP-dependent dimerization interface in topo II to block enzyme activity. Based on this similarity, we hypothesized that resveratrol may antagonize topo II by a similar mechanism. Using a variety of biochemical assays, we find that resveratrol indeed acts through the ICRF-187 binding locus, but that it inhibits topo II by preventing ATPase domain dimerization rather than stabilizing it. This work presents the first comprehensive analysis of the biochemical effects of both ICRF-187 and resveratrol on the human isoforms of topo II, and reveals a new mode for the allosteric regulation of topo II through modulation of ATPase status. Natural polyphenols related to resveratrol that have been shown to impact topo II function may operate in a similar manner.
Collapse
Affiliation(s)
- Joyce H Lee
- From the Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 and
| | - Timothy J Wendorff
- the Biophysics Graduate Program, University of California, Berkeley, California 94720
| | - James M Berger
- From the Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 and
| |
Collapse
|
18
|
Bansal N, Amdani S, Lipshultz ER, Lipshultz SE. Chemotherapy-induced cardiotoxicity in children. Expert Opin Drug Metab Toxicol 2017; 13:817-832. [DOI: 10.1080/17425255.2017.1351547] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Neha Bansal
- Department of Pediatrics, Wayne State University School of Medicine and Children’s Hospital of Michigan, Detroit, MI, USA
| | - Shahnawaz Amdani
- Department of Pediatrics, Wayne State University School of Medicine and Children’s Hospital of Michigan, Detroit, MI, USA
| | - Emma R. Lipshultz
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven E. Lipshultz
- Department of Pediatrics, Wayne State University School of Medicine and Children’s Hospital of Michigan, Detroit, MI, USA
- Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
19
|
Hutchins KK, Siddeek H, Franco VI, Lipshultz SE. Prevention of cardiotoxicity among survivors of childhood cancer. Br J Clin Pharmacol 2017; 83:455-465. [PMID: 27591829 PMCID: PMC6396850 DOI: 10.1111/bcp.13120] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/22/2022] Open
Abstract
LINKED ARTICLES This article is part of a joint Themed section with the British Journal of Pharmacology on Cardiotoxicity. The rest of the Themed section will appear in a future issue of BJP and will be available at http://onlinelibrary.wiley.com/journal/10.1111/(ISSN)1476-5381 The number of survivors of childhood cancers has increased exponentially over the past few decades. However, these survivors are also at substantially increased long-term risk of morbidity and mortality, especially from treatment-related cardiotoxicity. Preventing these risks is now a priority when treating children and adolescents with cancer. Dexrazoxane reduces the risk of anthracycline-induced cardiotoxicity among adults and children with cancer without reducing its antineoplastic effects or event-free survival. Thus, it should be strongly considered as a part of therapy for children and adolescents treated with anthracyclines.
Collapse
Affiliation(s)
- Kelley K. Hutchins
- Department of Pediatric Hematology/OncologyChildren's Hospital of Michigan3901 Beaubien BoulevardDetroitMichigan48201USA
| | - Hani Siddeek
- Department of PediatricsChildren's Hospital of Michigan3901 Beaubien BoulevardDetroitMichigan48201USA
| | - Vivian I. Franco
- Department of PediatricsWayne State University School of Medicine3901 Beaubien Boulevard, Suite 1K40DetroitMichigan48201USA
| | - Steven E. Lipshultz
- Department of PediatricsWayne State University School of Medicine3901 Beaubien Boulevard, Suite 1K40DetroitMichigan48201USA
- Karmanos Cancer InstituteChildren's Hospital of Michigan
| |
Collapse
|
20
|
Atteya R, Ashour ME, Ibrahim EE, Farag MA, El-Khamisy SF. Chemical screening identifies the β-Carboline alkaloid harmine to be synergistically lethal with doxorubicin. Mech Ageing Dev 2017; 161:141-148. [PMID: 27282658 DOI: 10.1016/j.mad.2016.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/16/2016] [Accepted: 04/30/2016] [Indexed: 12/18/2022]
Abstract
Despite being an invaluable chemotherapeutic agent for several types of cancer, the clinical utility of doxorubicin is hampered by its age-related and dose-dependent cardiotoxicity. Co-administration of dexrazoxane as a cardioprotective agent has been proposed, however recent studies suggest that it attenuates doxorubicin-induced antitumor activity. Since compounds of natural origin present a rich territory for drug discovery, we set out to identify putative natural compounds with the view to mitigate or minimize doxorubicin cardiotoxicity. We identify the DYRK1A kinase inhibitor harmine, which phosphorylates Tau that is deregulated in Alzheimer's disease, as a potentiator of cell death induced by non-toxic doses of doxorubicin. These observations suggest that harmine or other compounds that target the DYRK1A kinase my offer a new therapeutic opportunity to suppress doxorubicin age-related and dose-dependent cardiotoxicity.
Collapse
Affiliation(s)
- Reham Atteya
- Center of Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed E Ashour
- Center of Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Elsayed E Ibrahim
- Center of Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed A Farag
- Deparrtment of Pharamcognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Sherif F El-Khamisy
- Center of Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt; Krebs Institute, University of Sheffield, Sheffield, S10 2TN, UK.
| |
Collapse
|
21
|
Schuler MK, Gerdes S, West A, Richter S, Busemann C, Hentschel L, Lenz F, Kopp HG, Ehninger G, Reichardt P, Pink D. Efficacy and safety of Dexrazoxane (DRZ) in sarcoma patients receiving high cumulative doses of anthracycline therapy - a retrospective study including 32 patients. BMC Cancer 2016; 16:619. [PMID: 27507014 PMCID: PMC4977890 DOI: 10.1186/s12885-016-2654-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/29/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Anthracyclines, as the most effective therapy, are the cornerstone of advanced stage sarcoma treatment. However, anthracyclines can also contribute to myocardial dysfunction and congestive heart failure, ultimately limiting the therapeutic potential of the drug. Coadministration of Dexrazoxane has been shown to effectively reduce cardiotoxicity, however primarily in patients suffering in diseases other than sarcoma. METHODS The aim of this retrospective analysis was to evaluate safety and efficacy of chemotherapy with high cumulative doses of anthracyclines in combination with Dexrazoxane. The medical charts of 32 patients treated in four institutions were analyzed. Reasons for coadministration were rechallenge, reaching the cumulative anthracycline dose and preexisting heart failure. RESULTS The median age was 54 years [18-68 years]. The median cumulative anthracycline dose before adding DRZ was 450 mg/m(2) and after administration of last anthracycline containing therapy 750 mg/m(2). Either during treatment or follow up, 2/27 patients (7 %) without preexisting major cardiac findings developed anthracycline-induced cardiotoxicity. The median overall survival (OS) from start of the first anthracycline containing chemotherapy was 46 months and 17 months from the initial coadministration of DRZ. At rechallenge, the median progression free survival (PFS) with DRZ was 7 months. In continuous therapy, the median PFS was 13 months from beginning of chemotherapy and 9 months from the addition of DRZ. CONCLUSION Chemotherapy with high cumulative doses of anthracyclines in addition with DRZ demonstrated a remarkable OS in these advanced disease patients. Cardiac side-effects due to high cumulative doses of anthracyclines requiring discontinuation of anthracycline treatment were rare. A PFS of 9 months from the beginning of the coadministration of DRZ indicates that continuing anthracycline therapy beyond established cumulative doses is a promising therapeutic option.
Collapse
Affiliation(s)
- Markus K Schuler
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany. .,Department of Internal Medicine II, HELIOS Clinic Emil von Behring, Walterhöferstr. 11, 14165, Berlin, Germany.
| | - Sebastian Gerdes
- Institute of Medical Informatics and Biometry, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Antje West
- Department of Hematology and Oncology, HELIOS Clinic Bad Saarow, Pieskower Straße 33, 15526, Bad Saarow, Germany
| | - Stephan Richter
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Christoph Busemann
- Department of Internal Medicine C, University Hospital Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| | - Leopold Hentschel
- University Cancer Center, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Felicitas Lenz
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Hans-Georg Kopp
- Department of Internal Medicine II, University Hospital Tübingen, Eberhard Karls University Tübingen, Geissweg 3, 72076, Tübingen, Germany
| | - Gerhard Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany.,University Cancer Center, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Peter Reichardt
- Department of Interdisciplinary Oncology, HELIOS Clinic Berlin-Buch, Berlin, Germany
| | - Daniel Pink
- Department of Hematology and Oncology, HELIOS Clinic Bad Saarow, Pieskower Straße 33, 15526, Bad Saarow, Germany.,Department of Internal Medicine C, University Hospital Greifswald, Sauerbruchstraße, 17475, Greifswald, Germany
| |
Collapse
|
22
|
Zhang HY, Lu X. Coenzyme complex decreased cardiotoxicity when combined with chemotherapy in treating elderly patients with gastrointestinal cancer. Asian Pac J Cancer Prev 2016; 16:4045-9. [PMID: 25987084 DOI: 10.7314/apjcp.2015.16.9.4045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To investigate the effect of coenzyme complex on decreasing cardiotoxicity in elderly patients with gastrointestinal cancer who were treated by chemotherapy. METHODS From September 2011 to February 2015, we recruited 54 elderly (with more than 70 years of age) patients with gastrointestinal cancer, with advanced disease. Then treated with chemotherapy combined with or without coenzyme complex. After two cycles of treatment, the effect of coenzyme complex on decreasing cardiotoxicity were evaluated. RESULTS Chemotherapy was combined with coenzyme complex in 32 patients (22man, 10 woman; median age: 74 years, range: 70-87 years) without coenzyme complex in 22 patients (15man, 7 woman; median age: 73 years, range: 70-80 years) with gastrointestinal cancer. Cardiac event was significantly lower in patients treated with chemotherapy combined with coenzyme complex (p<0.01). CONCLUSIONS Coenzyme Complex decreased cardiotoxicity when combined with chemotherapy in treating elderly patients with gastrointestinal cancer.
Collapse
Affiliation(s)
- Hai-Yan Zhang
- Department of Cardiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China E-mail :
| | | |
Collapse
|
23
|
Asselin BL, Devidas M, Chen L, Franco VI, Pullen J, Borowitz MJ, Hutchison RE, Ravindranath Y, Armenian SH, Camitta BM, Lipshultz SE. Cardioprotection and Safety of Dexrazoxane in Patients Treated for Newly Diagnosed T-Cell Acute Lymphoblastic Leukemia or Advanced-Stage Lymphoblastic Non-Hodgkin Lymphoma: A Report of the Children's Oncology Group Randomized Trial Pediatric Oncology Group 9404. J Clin Oncol 2015; 34:854-62. [PMID: 26700126 DOI: 10.1200/jco.2015.60.8851] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
PURPOSE To determine the oncologic efficacy, cardioprotective effectiveness, and safety of dexrazoxane added to chemotherapy that included a cumulative doxorubicin dose of 360 mg/m(2) to treat children and adolescents with newly diagnosed T-cell acute lymphoblastic leukemia (T-ALL) or lymphoblastic non-Hodgkin lymphoma (L-NHL). PATIENTS AND METHODS Patients were treated on Pediatric Oncology Group Protocol POG 9404, which included random assignment to treatment with or without dexrazoxane given as a bolus infusion immediately before every dose of doxorubicin. Cardiac effects were assessed by echocardiographic measurements of left ventricular function and structure. RESULTS Of 573 enrolled patients, 537 were eligible, evaluable, and randomly assigned to an arm with or without dexrazoxane. The 5-year event-free survival (with standard error) did not differ between groups: 77.2% (2.7%) for the dexrazoxane group versus 76.0% (2.7%) for the doxorubicin-only group (P = .9). The frequencies of severe grade 3 or 4 hematologic toxicity, infection, CNS events, and toxic deaths were similar in both groups (P ranged from .26 to .64). Of 11 second malignancies, eight occurred in patients who received dexrazoxane (P = .17). The mean left ventricular fractional shortening, wall thickness, and thickness-to-dimension ratio z scores measured 3 years after diagnosis were worse in the doxorubicin-alone group (n = 55 per group; P ≤ .01 for all comparisons). Mean fractional shortening z scores measured 3.5 to 6.4 years after diagnosis remained diminished and were lower in the 21 patients who received doxorubicin alone than in the 31 patients who received dexrazoxane (-2.03 v -0.24; P ≤ .001). CONCLUSION Dexrazoxane was cardioprotective and did not compromise antitumor efficacy, did not increase the frequencies of toxicities, and was not associated with a significant increase in second malignancies with this doxorubicin-containing chemotherapy regimen. We recommend dexrazoxane as a cardioprotectant for children and adolescents who have malignancies treated with anthracyclines.
Collapse
Affiliation(s)
- Barbara L Asselin
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI.
| | - Meenakshi Devidas
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Lu Chen
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Vivian I Franco
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Jeanette Pullen
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Michael J Borowitz
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Robert E Hutchison
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Yaddanapudi Ravindranath
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Saro H Armenian
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Bruce M Camitta
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| | - Steven E Lipshultz
- Barbara L. Asselin, University of Rochester School of Medicine and Wilmot Cancer Institute, Rochester; Robert E. Hutchison, State University of New York Upstate Medical Center, Syracuse, NY; Meenakshi Devidas, Children's Oncology Group and University of Florida, Gainesville, FL; Lu Chen, Children's Oncology Group, Monrovia; Saro H. Armenian, City of Hope National Medical Center, Duarte, CA; Vivian I. Franco, Yaddanapudi Ravindranath, and Steven E. Lipshultz, Wayne State University School of Medicine and Children's Hospital of Michigan, Detroit, MI; Jeanette Pullen, University of Mississippi Medical Center and Children's Hospital, Jackson, MS; Michael J. Borowitz, Johns Hopkins University School of Medicine and Johns Hopkins Hospital, Baltimore, MD; and Bruce M. Camitta, Medical College of Wisconsin, Midwest Center for Cancer and Blood Disorders, Milwaukee, WI
| |
Collapse
|
24
|
Petek BJ, Greenman C, Herrmann J, Ewer MS, Jones RL. Cardio-oncology: an ongoing evolution. Future Oncol 2015. [DOI: 10.2217/fon.15.89] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer survivorship has been greatly impacted with the development of modern cancer treatments. While significant strides have been made in managing many types of cancer, now physicians face new challenges. Over the past decades, cardiovascular events in cancer survivors have increased in prevalence, driving the development of multidisciplinary cardio-oncology programs. Additionally, as cancer patients live longer, their risk of developing secondary cardiovascular events increases. The rapid development of novel cancer therapies will continue to generate questions of cardiac risk and cardiac protection in cancer patients over time. We wish to outline the development of cardio-oncology in its present state, and provide future perspectives for the discipline.
Collapse
Affiliation(s)
- Bradley J Petek
- School of Medicine, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Chris Greenman
- Division of Cardiology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Joerg Herrmann
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Michael S Ewer
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Robin L Jones
- Sarcoma Unit, The Institute of Cancer Research & The Royal Marsden Hospital, Fulham Road, SW3 6JJ, London, UK
| |
Collapse
|
25
|
Chang SA, Lim BK, Lee YJ, Hong MK, Choi JO, Jeon ES. A Novel Angiotensin Type I Receptor Antagonist, Fimasartan, Prevents Doxorubicin-induced Cardiotoxicity in Rats. J Korean Med Sci 2015; 30:559-68. [PMID: 25931786 PMCID: PMC4414639 DOI: 10.3346/jkms.2015.30.5.559] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 02/13/2015] [Indexed: 11/20/2022] Open
Abstract
Angiotensin receptor blockers (ARBs) have organ-protective effects in heart failure and may be also effective in doxorubicin-induced cardiomyopathy (DOX-CMP); however, the efficacy of ARBs on the prevention of DOX-CMP have not been investigated. We performed a preclinical experiment to evaluate the preventive effect of a novel ARB, fimasartan, in DOX-CMP. All animals underwent echocardiography and were randomly assigned into three groups: treated daily with vehicle (DOX-only group, n=22), 5 mg/kg of fimasartan (Low-fima group, n=22), and 10 mg/kg of fimasartan (High-fima group, n=19). DOX was injected once a week for six weeks. Echocardiography and hemodynamic assessment was performed at the 8th week using a miniaturized conductance catheter. Survival rate of the High-fima group was greater (100%) than that of the Low-fima (75%) and DOX-only groups (50%). Echocardiography showed preserved left ventricular (LV) ejection fraction in the High-fima group, but not in the DOX-only group (P=0.002). LV dimensions increased in the DOX-only group; however, remodeling was attenuated in the Low-fima and High-fima groups. Hemodynamic assessment showed higher dP/dt in the High-fima group compared with the DOX-only group. A novel ARB, fimasartan, may prevent DOX-CMP and improve survival rate in a dose-dependent manner in a rat model of DOX-CMP and could be a treatment option for the prevention of DOX-CMP.
Collapse
Affiliation(s)
- Sung-A Chang
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cardiovascular Imaging Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung-Kwan Lim
- Department of Biomedical Science, Jungwon University, Goesan-gun, Korea
| | - You Jung Lee
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mi-Kyung Hong
- Cardiovascular Imaging Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin-Oh Choi
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cardiovascular Imaging Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun-Seok Jeon
- Division of Cardiology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cardiovascular Imaging Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
26
|
Seif AE, Walker DM, Li Y, Huang YSV, Kavcic M, Torp K, Bagatell R, Fisher BT, Aplenc R. Dexrazoxane exposure and risk of secondary acute myeloid leukemia in pediatric oncology patients. Pediatr Blood Cancer 2015; 62:704-9. [PMID: 24668949 PMCID: PMC4177031 DOI: 10.1002/pbc.25043] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/28/2014] [Indexed: 11/06/2022]
Abstract
BACKGROUND Dexrazoxane may reduce anthracycline-associated cardiotoxicity in pediatric cancer patients. However, concerns of secondary acute myeloid leukemia (AML) have led to restrictions on pediatric dexrazoxane use in Europe. Published data about dexrazoxane-associated secondary AML are limited and conflicting. We sought to estimate the secondary AML risk in children receiving dexrazoxane after anthracycline exposure. PROCEDURE A retrospective cohort of children with newly identified malignancies (excluding AML) receiving anthracyclines between January 1, 1999 and March 31, 2011 was established using the Pediatric Health Information System (PHIS). Patients were followed for all subsequent admissions to identify dexrazoxane exposures and secondary AML, defined by AML ICD-9 codes and AML induction chemotherapy. Logistic regression was used to model the association of dexrazoxane and secondary AML risk. A propensity score was used to adjust for measurable confounding. RESULTS Of 15,532 patients in the cohort exposed to anthracyclines, 1,406 received dexrazoxane. The secondary AML rate was 0.21% (3 of 1,046) in dexrazoxane-exposed and 0.55% (77 of 14,126) in unexposed patients. In a propensity score-adjusted multivariate analysis, dexrazoxane exposure was not associated with an increased risk of secondary AML, OR = 0.38, 95% CI 0.11-1.26. CONCLUSIONS Dexrazoxane was not associated with an increased risk of secondary AML in a large cohort of pediatric cancer patients receiving anthracyclines in US hospitals. While these data support dexrazoxane's safety in the general pediatric oncology population, additional studies are needed to confirm these findings and to quantify dexrazoxane's long-term cardioprotective effects.
Collapse
Affiliation(s)
- Alix E. Seif
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence to: Alix Seif, Division of Oncology, The Children’s Hospital of Philadelphia, 3501 Civic Center Boulevard, CTRB 3004 Philadelphia, PA 19104.
| | | | - Yimei Li
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yuan-Shung V. Huang
- The Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Kari Torp
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rochelle Bagatell
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian T. Fisher
- Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Center for Clinical Epidemiology and Biostatistics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,Division of Infectious Diseases, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Richard Aplenc
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Department of Pediatrics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania,The Center for Pediatric Clinical Effectiveness, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Center for Clinical Epidemiology and Biostatistics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Dong D, Gao W, Liu Y, Qi XR. Therapeutic potential of targeted multifunctional nanocomplex co-delivery of siRNA and low-dose doxorubicin in breast cancer. Cancer Lett 2015; 359:178-86. [DOI: 10.1016/j.canlet.2015.01.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/31/2014] [Accepted: 01/08/2015] [Indexed: 11/24/2022]
|
28
|
Abstract
For more than half a century, the different properties of dexrazoxane have captured the attention of scientists and clinicians. Presently, dexrazoxane is licensed in many parts of the world for two different indications: prevention of cardiotoxicity from anthracycline-based chemotherapy, and prevention of tissue injuries after extravasation of anthracyclines. This article reviews the historical, preclinical, and clinical background for the use of dexrazoxane for these indications.
Collapse
Affiliation(s)
- Seppo W Langer
- Thoracic and Neuroendocrine Section, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
29
|
Lipshultz SE, Franco VI, Sallan SE, Adamson PC, K. Steiner R, Swain SM, Gligorov J, Minotti G. Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence. PROGRESS IN PEDIATRIC CARDIOLOGY 2014. [DOI: 10.1016/j.ppedcard.2014.09.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Herman EH, Hasinoff BB, Steiner R, Lipshultz SE. A review of the preclinical development of dexrazoxane. PROGRESS IN PEDIATRIC CARDIOLOGY 2014. [DOI: 10.1016/j.ppedcard.2014.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
Vejpongsa P, Yeh ETH. Topoisomerase 2β: a promising molecular target for primary prevention of anthracycline-induced cardiotoxicity. Clin Pharmacol Ther 2013; 95:45-52. [PMID: 24091715 DOI: 10.1038/clpt.2013.201] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 09/20/2013] [Indexed: 02/08/2023]
Abstract
Anthracyclines are powerful chemotherapy agents that are still widely used today. However, their clinical use is limited by the development of dose-dependent cardiotoxicity. Recently, we showed that topoisomerase 2β (Top2β) is required for anthracycline to induce DNA double-strand breaks and changes in the transcriptome, leading to mitochondrial dysfunction and generation of reactive oxygen species. Furthermore, deleting Top2β from cardiomyocytes prevented the development of anthracycline-induced cardiotoxicity in mice. On the basis of this molecular insight, new strategies should be developed to prevent anthracycline-induced cardiotoxicity. First, Top2α-specific anthracyclines should be tested to determine whether they will spare the heart. Second, Top2β should be studied as a potential biomarker to predict risk of developing cardiotoxicity before anthracycline treatment. Third, inhibiting and deleting Top2β in the heart should also be tested as primary prevention strategies. We propose that Top2β is a promising molecular target that can be used to design interventions to prevent anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
- P Vejpongsa
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - E T H Yeh
- 1] Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA [2] Texas Heart Institute/St. Luke's Episcopal Hospital, Houston, Texas, USA
| |
Collapse
|
32
|
Jensen BC, McLeod HL. Pharmacogenomics as a risk mitigation strategy for chemotherapeutic cardiotoxicity. Pharmacogenomics 2013; 14:205-13. [PMID: 23327580 DOI: 10.2217/pgs.12.205] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Damage to the heart can result from both traditional chemotherapeutic agents, such as doxorubicin, and newer 'targeted' therapies, such as trastuzumab. This chemotherapeutic cardiotoxicity is potentially life-threatening and necessitates limiting or discontinuing an otherwise-effective cancer treatment. Clinical strategies focus on surveillance rather than prevention, although there are no specific therapies for this highly morbid adverse effect. Current models for prospectively predicting risk of chemotherapeutic cardiotoxicity are limited. Cardiotoxicity can occur idiosyncratically in patients without obvious demographic risk factors, suggesting a genetically determined susceptibility, and candidate-gene studies have identified a limited number of variants that increase risk. In this commentary we indicate a need for more powerful means to identify risk prospectively, and suggest that broad pharmacogenomic approaches may be fruitful.
Collapse
Affiliation(s)
- Brian C Jensen
- UNC School of Medicine, Division of Cardiology, 160 Dental Circle, CB 7075, Chapel Hill, NC 27599-7075, USA.
| | | |
Collapse
|
33
|
Sparano J. Cytotoxic Therapy and Other Nonhormonal Approaches for the Treatment of Metastatic Breast Cancer. Breast Cancer 2013. [DOI: 10.1201/b14039-16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
34
|
Carvalho FS, Burgeiro A, Garcia R, Moreno AJ, Carvalho RA, Oliveira PJ. Doxorubicin-Induced Cardiotoxicity: From Bioenergetic Failure and Cell Death to Cardiomyopathy. Med Res Rev 2013; 34:106-35. [DOI: 10.1002/med.21280] [Citation(s) in RCA: 349] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Filipa S. Carvalho
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
- Department of Life Sciences; University of Coimbra; 3004-517 Coimbra Portugal
| | - Ana Burgeiro
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
- IMAR-Institute of Marine Research; University of Coimbra; Portugal
| | - Rita Garcia
- IMAR-Institute of Marine Research; University of Coimbra; Portugal
| | - António J. Moreno
- Department of Life Sciences; University of Coimbra; 3004-517 Coimbra Portugal
- IMAR-Institute of Marine Research; University of Coimbra; Portugal
| | - Rui A. Carvalho
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
- Department of Life Sciences; University of Coimbra; 3004-517 Coimbra Portugal
| | - Paulo J. Oliveira
- CNC-Center for Neuroscience and Cell Biology; University of Coimbra; 3004-517 Coimbra Portugal
| |
Collapse
|
35
|
Doroshow JH. Dexrazoxane for the prevention of cardiac toxicity and treatment of extravasation injury from the anthracycline antibiotics. Curr Pharm Biotechnol 2013; 13:1949-56. [PMID: 22352729 DOI: 10.2174/138920112802273245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 02/05/2011] [Accepted: 05/04/2011] [Indexed: 12/11/2022]
Abstract
The cumulative cardiac toxicity of the anthracycline antibiotics and their propensity to produce severe tissue injury following extravasation from a peripheral vein during intravenous administration remain significant problems in clinical oncologic practice. Understanding of the free radical metabolism of these drugs and their interactions with iron proteins led to the development of dexrazoxane, an analogue of EDTA with intrinsic antineoplastic activity as well as strong iron binding properties, as both a prospective cardioprotective therapy for patients receiving anthracyclines and as an effective treatment for anthracycline extravasations. In this review, the molecular mechanisms by which the anthracyclines generate reactive oxygen species and interact with intracellular iron are examined to understand the cardioprotective mechanism of action of dexrazoxane and its ability to protect the subcutaneous tissues from anthracycline-induced tissue necrosis.
Collapse
Affiliation(s)
- James H Doroshow
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
36
|
Khouri MG, Douglas PS, Mackey JR, Martin M, Scott JM, Scherrer-Crosbie M, Jones LW. Cancer therapy-induced cardiac toxicity in early breast cancer: addressing the unresolved issues. Circulation 2012; 126:2749-63. [PMID: 23212997 PMCID: PMC3667651 DOI: 10.1161/circulationaha.112.100560] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
| | | | | | - Miguel Martin
- Hospital General Universitario Gregorio Maranon, Universidad Complutense, Madrid, Spain
| | | | | | | |
Collapse
|
37
|
Gaurav K, Goel RK, Shukla M, Pandey M. Glutamine: A novel approach to chemotherapy-induced toxicity. Indian J Med Paediatr Oncol 2012; 33:13-20. [PMID: 22754203 PMCID: PMC3385273 DOI: 10.4103/0971-5851.96962] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Treatment of cancer is associated with short- and long-term side-effects. Cancer produces a state of glutamine deficiency, which is further aggravated by toxic effects of chemotherapeutic agents leading to increased tolerance of tumor to chemotherapy as well as reduced tolerance of normal tissues to the side-effects of chemotherapy. This article reviews the possible role of glutamine supplementation in reducing the serious adverse events in patients treated with anticancer drugs. The literature related to the possible role of glutamine in humans with cancer and the supportive evidence from animal studies was reviewed. Searches were made and the literature was retrieved using PUBMED, MEDLINE, COCHRANE LIBRARY, CENAHL and EMBASE, with a greater emphasis on the recent advances and clinical trials. Glutamine supplementation was found to protect against radiation-induced mucositis, anthracycline-induced cardiotoxicity and paclitaxel-related myalgias/arthralgias. Glutamine may prevent neurotoxicity of paclitaxel, cisplatin, oxaplatin bortezomib and lenolidamide, and is beneficial in the reduction of the dose-limiting gastrointestinal toxic effects of irinotecan and 5-FU-induced mucositis and stomatitis. Dietary glutamine reduces the severity of the immunosuppressive effect induced by methotrexate and improves the immune status of rats recovering from chemotherapy. In patients with acute myeloid leukemia requiring parenteral nutrition, glycyl-glutamine supplementation could hasten neutrophil recovery after intensive myelosuppressive chemotherapy. Current data supports the usefulness of glutamine supplementation in reducing complications of chemotherapy; however, paucity of clinical trials weakens the clear interpretation of these findings.
Collapse
Affiliation(s)
- Kumar Gaurav
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | | | | | | |
Collapse
|
38
|
Effects of MnDPDP and ICRF-187 on Doxorubicin-Induced Cardiotoxicity and Anticancer Activity. Transl Oncol 2012; 5:252-9. [PMID: 22937177 DOI: 10.1593/tlo.11304] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 04/11/2012] [Accepted: 04/12/2012] [Indexed: 11/18/2022] Open
Abstract
Oxidative stress participates in doxorubicin (Dx)-induced cardiotoxicity. The metal complex MnDPDP and its metabolite MnPLED possess SOD-mimetic activity, DPDP and PLED have, in addition, high affinity for iron. Mice were injected intravenously with MnDPDP, DPDP, or dexrazoxane (ICRF-187). Thirty minutes later, mice were killed, the left atria were hung in organ baths and electrically stimulated, saline or Dx was added, and the contractility was measured for 60 minutes. In parallel experiments, 10 µM MnDPDP or MnPLED was added directly into the organ bath. The effect of MnDPDP on antitumor activity of Dx against two human tumor xenografts (MX-1 and A2780) was investigated. The in vitro cytotoxic activity was studied by co-incubating A2780 cells with MnDPDP, DPDP, and/or Dx. Dx caused a marked reduction in contractile force. In vivo treatment with MnDPDP and ICRF-187 attenuated the negative effect of Dx. When added directly into the bath, MnDPDP did not protect, whereas MnPLED attenuated the Dx effect by approximately 50%. MnDPDP or ICRF-187 did not interfere negatively with the anti-tumor activity of Dx, either in vivo or in vitro. Micromolar concentrations of DPDP but not MnDPDP displayed an in vitro cytotoxic activity against A2780 cells. The present results show that MnDPDP, after being metabolized to MnPLED, protects against acute Dx cardiotoxicity. Both in vivo and in vitro experiments show that cardioprotection takes place without interfering negatively with the anticancer activity of Dx. Furthermore, the results suggest that the previously described cytotoxic in vivo activity of MnDPDP is an inherent property of DPDP.
Collapse
|
39
|
Castel M, Despas F, Modesto A, Gales C, Honton B, Galinier M, Senard JM, Pathak A. [Cardiotoxicity of chemotherapies]. Presse Med 2012; 42:26-39. [PMID: 22727981 DOI: 10.1016/j.lpm.2012.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 04/11/2012] [Accepted: 04/12/2012] [Indexed: 12/18/2022] Open
Abstract
The spectrum of chemotherapy's cardiac side effect of chemotherapy has expanded with the new combinations of cytotoxic and targeted therapies over the past 10 years. Moreover, cancer therapy administrated to "new" populations, especially elderly patients or patients with cardiovascular disease and/or coronary artery disease history, has increased considerably. According to the American College of Cardiology and American Heart Association (ACC/AHA), patients receiving chemotherapy can be considered in the A group of heart failure. Many cardiovascular adverse effects appear with cancer therapy and suspend treatment purchase, or leading to an alteration of quality of life, and increasing mortality risks. The most clinically evident cardiotoxicity and best known is the anthracyclines adverse effect. Other cytotoxic are associated with a significant risk of cardiovascular complications include alkylating agents such as 5-fluorouracil and paclitaxel. Cardiovascular adverse effects are associated with the use of targeted therapies such as tyrosine kinase inhibitors: trastuzumab, bevacizumab. At the same time, drugs used to hematological malignancies, as acid all-trans-retinoic acid and arsenic trioxide are cardiotoxics. The most serious cardiac complications of cancer therapies is heart congestive failure, mainly due to the use of anthracyclines, cyclophosphamide and trastuzumab, usually at high doses. Myocardial ischemia is mainly caused by interferon and antimetabolites. Other side effects may occur such as hypotension, hypertension, arrhythmias and conduction disturbances, pericarditis, and thromboembolic complications.
Collapse
Affiliation(s)
- Marion Castel
- Institut des maladies métaboliques et cardiovasculaires de Rangueil, IFR31, UPS, université de Toulouse, Inserm, U1048, 31432 Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Oh E, Chim H, Soltanian HT. The effects of neoadjuvant and adjuvant chemotherapy on the surgical outcomes of breast reconstruction. J Plast Reconstr Aesthet Surg 2012; 65:e267-80. [PMID: 22633392 DOI: 10.1016/j.bjps.2012.04.053] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 03/24/2012] [Accepted: 04/30/2012] [Indexed: 12/26/2022]
Abstract
Breast reconstruction following mastectomy has become, in many centers the standard of care. An increasingly encountered trend is the use of neoadjuvant chemotherapy to downstage high stage tumors and to decrease tumor burden prior to definitive oncologic surgery. These agents clearly provide a survival benefit, but also have the potential to adversely affect the surgical course of immediate and delayed breast reconstruction. The use of new biologic and hormonal agents may also have effects on surgery and reconstruction. Furthermore, chemotherapeutic agents as a whole may impair cellular functions necessary for normal recovery from surgery. In this paper we present a concise review for the reconstructive surgeon on adverse effects of chemotherapeutic, hormonal and biologic agents used for treatment of breast cancer, important perioperative issues, and also discuss their potential effect on breast reconstruction.
Collapse
Affiliation(s)
- Eugene Oh
- Department of Plastic Surgery, Case School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
41
|
Roti Roti EC, Salih SM. Dexrazoxane ameliorates doxorubicin-induced injury in mouse ovarian cells. Biol Reprod 2012; 86:96. [PMID: 22190700 DOI: 10.1095/biolreprod.111.097030] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Doxorubicin (DXR) is a frontline chemotherapy agent implicated in unintended ovarian failure in female cancer survivors. The fertility preservation techniques currently available for cancer patients are often time and cost prohibitive and do not necessarily preserve endocrine function. There are no drug-based ovary protection therapies clinically available. This study provides the first investigation using dexrazoxane (Dexra) to limit DXR insult in ovarian tissue. In KK-15 granulosa cells, a 3-h DXR treatment increased double-strand (ds) DNA breaks 40%-50%, as quantified by the neutral comet assay, and dose-dependent cytotoxicity. Dexra exhibited low toxicity in KK-15 cells, inducing no DNA damage and less than 20% cell loss. Cotreating KK-15 cells with Dexra prevented acute DXR-induced dsDNA damage. Similarly, Dexra attenuated the DXR-induced 40%-65% increase in dsDNA breaks in primary murine granulosa cells and cells from in vitro cultured murine ovaries. DXR can cause DNA damage either through a topoisomerase II-mediated pathway, based on DXR intercalation into DNA, or through oxidative stress. Cotreating KK-15 cells with 2 μM Dexra was sufficient to prevent DXR-induced, but not H(2)O(2)-induced, DNA damage. These data indicated the protective effects are likely due to Dexra's inhibition of topoisomerase II catalytic activity. This putative protective agent attenuated downstream cellular responses to DXR, preventing H2AFX activation in KK-15 cells and increasing viability as demonstrated by increasing the DXR lethal dose in KK-15 cells 5- to 8-fold (LD(20)) and primary murine granulosa cells 1.5- to 2-fold (LD(50)). These data demonstrate Dexra protects ovarian cells from DXR insult and suggest that it is a promising tool to limit DXR ovarian toxicity in vivo.
Collapse
Affiliation(s)
- Elon C Roti Roti
- REI Division, Department of Obstetrics & Gynecology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | | |
Collapse
|
42
|
van Dalen EC, Caron HN, Dickinson HO, Kremer LCM. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev 2011; 2011:CD003917. [PMID: 21678342 PMCID: PMC6457676 DOI: 10.1002/14651858.cd003917.pub4] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Anthracyclines are among the most effective chemotherapeutic agents in the treatment of numerous malignancies. Unfortunately, their use is limited by a dose-dependent cardiotoxicity. In an effort to prevent this cardiotoxicity, different cardioprotective agents have been studied. OBJECTIVES The objective of this review was to assess the efficacy of different cardioprotective agents in preventing heart damage in cancer patients treated with anthracyclines. SEARCH STRATEGY We searched the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2010, Issue 10), MEDLINE (1966 to November 2010) and EMBASE (1980 to November 2010) databases. In addition, we handsearched reference lists, conference proceedings of the International Society of Paediatric Oncology (SIOP) and American Society of Clinical Oncology (ASCO) meetings (1998 to 2010) and ongoing trials registers. SELECTION CRITERIA Randomised controlled trials (RCTs) in which any cardioprotective agent was compared to no additional therapy or placebo in cancer patients (children and adults) receiving anthracyclines. DATA COLLECTION AND ANALYSIS Two review authors independently performed the study selection, risk of bias assessment and data extraction including adverse effects. MAIN RESULTS We identified RCTs for the eight cardioprotective agents N-acetylcysteine, phenethylamines, coenzyme Q10, a combination of vitamins E and C and N-acetylcysteine, L-carnitine, carvedilol, amifostine and dexrazoxane (mostly for adults with advanced breast cancer). All studies had methodological limitations and for the first seven agents there were too few studies to allow pooling of results. None of the individual studies showed a cardioprotective effect. The 10 included studies on dexrazoxane enrolled 1619 patients. The meta-analysis for dexrazoxane showed a statistically significant benefit in favour of dexrazoxane for the occurrence of heart failure (risk ratio (RR) 0.29, 95% CI 0.20 to 0.41). No evidence was found for a difference in response rate or survival between the dexrazoxane and control groups. The results for adverse effects were ambiguous. No significant difference in the occurrence of secondary malignancies was identified. AUTHORS' CONCLUSIONS No definitive conclusions can be made about the efficacy of cardioprotective agents for which pooling of results was impossible. Dexrazoxane prevents heart damage and no evidence for a difference in response rate or survival between the dexrazoxane and control groups was identified. The evidence available did not allow us to reach any definite conclusions about adverse effects. We conclude that if the risk of cardiac damage is expected to be high, it might be justified to use dexrazoxane in patients with cancer treated with anthracyclines. However, clinicians should weigh the cardioprotective effect of dexrazoxane against the possible risk of adverse effects for each individual patient.
Collapse
Affiliation(s)
- Elvira C van Dalen
- Emma Children's Hospital/Academic Medical CenterDepartment of Paediatric OncologyPO Box 22660 (room H4‐139)AmsterdamNetherlands1100 DD
| | - Huib N Caron
- Emma Children's Hospital/Academic Medical CenterDepartment of Paediatric OncologyPO Box 22660 (room H4‐139)AmsterdamNetherlands1100 DD
| | - Heather O Dickinson
- Newcastle UniversityInstitute of Health & Society21 Claremont PlaceNewcastle upon TyneUKNE2 4AA
| | - Leontien CM Kremer
- Emma Children's Hospital/Academic Medical CenterDepartment of Paediatric OncologyPO Box 22660 (room H4‐139)AmsterdamNetherlands1100 DD
| | | |
Collapse
|
43
|
Abstract
Cardiotoxicity of anticancer treatments has become an increasingly important clinical problem faced by cardiologists. Left ventricular systolic dysfunction and heart failure generate the most concern, but clinical features and prognosis vary considerably depending on the causative agent. Anthracycline-related cardiomyopathy differs fundamentally from effects associated with newer targeted agents, such as trastuzumab. Other forms of cardiovascular disease that occur as a result of cancer treatment include hypertension, thromboembolic disease, pericardial disease, arrhythmia, and myocardial ischemia. The approach to cardiovascular disease in patients with cancer is often different from that in the general population, not only because of distinct underlying mechanisms and clinical features of their heart disease, but also because of the potential ongoing need for additional cancer treatment as well as the altered duration of anticipated survival. In an effort to maximize both quality of life and survival, cardiologists and oncologists should collaborate with the aim of balancing the risks of cardiotoxicity with the benefits of oncologic therapy.
Collapse
|
44
|
L'Ecuyer TJ, Aggarwal S, Zhang JP, Van der Heide RS. Effect of hypothermia on doxorubicin-induced cardiac myoblast signaling and cell death. Cardiovasc Pathol 2011; 21:96-104. [PMID: 21489822 DOI: 10.1016/j.carpath.2011.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 01/10/2011] [Accepted: 02/04/2011] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Anthracyclines (AC) are useful chemotherapeutic agents whose principal limitation is cardiac toxicity, which may progress to heart failure, transplantation or even death. We have shown that this toxicity involves oxidative stress-induced activation of the DNA damage pathway. Hypothermia has been shown to be protective against other diseases involving oxidative stress but has not been studied in models of AC toxicity. METHODS In the current experiments, H9C2 cardiac myoblasts were treated with varying concentrations of the AC doxorubicin (DOX) during normothermia (37°C) or mild hypothermia (35°C). Total cell death was assayed using trypan blue exclusion and apoptosis by terminal deoxynucleotidyl transferase-mediated deoxyuridine-biotin nick end labeling (TUNEL) staining. Oxidative stress was assayed using the fluorescent indicator 2'7'-dichlorofluorescein diacetate. DNA damage pathway activation was assayed by immunostaining for H2AX and p53. Mitochondrial membrane potential was assayed by JC-1 staining. RESULTS At all concentrations of DOX examined (1, 2.5 and 5 μM), hypothermia reduced oxidative stress, activation of H2AX and p53, loss of mitochondrial membrane potential and total and apoptotic cell death (P=.001-.03 for each observation). CONCLUSIONS The reduction of oxidative stress-induced activation of the DNA damage pathway and consequent cell death by mild hypothermia supports a possible protective role to reduce the clinical impact of DOX-induced cardiac toxicity. Such an approach may allow expanded use of these effective chemotherapeutic agents to increase cancer cure rates.
Collapse
Affiliation(s)
- Thomas J L'Ecuyer
- Cardiology Division, Children's Hospital of Michigan, 3901 Beaubien Boulevard, Detroit, MI 48201, USA
| | | | | | | |
Collapse
|
45
|
Zhang L, Yang Y, Yu L, Wang Y, Liu L, Fan X. Cardioprotective effects of Glycyrrhiza uralensis extract against doxorubicin-induced toxicity. Int J Toxicol 2011; 30:181-9. [PMID: 21378374 DOI: 10.1177/1091581810393033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The purpose of the present study was to investigate the cardioprotective effects of Glycyrrhiza uralensis extract (GUE) against doxorubicin (DOX)-induced cardiotoxicity. Imprinting control region (ICR) mice were treated with saline, DOX (20 mg/kg intraperitoneal [ip] for once), GUE (100 mg/kg intragastric [ig] for 8 days), co-treatments with DOX and GUE (100 mg/kg ig for 8 days), and amifostine (100 mg/kg intravenous [iv] for once), respectively. Serum levels of lactate dehydrogenase (LDH) and creatine kinase isoenzyme (CK-MB), glutathione peroxidase (GSH-P(X)) activity, and glutathione (GSH) level in heart tissue were measured. Histopathologic analysis of heart tissue was also performed. Treatment with GUE significantly protected the mice from DOX-induced cardiotoxicity, indicated by decreased levels of serum LDH and CK-MB, improved heart morphology and increased GSH-P(X) activity and GSH level. Additionally, GUE did not compromise the tumor-inhibitory effect of DOX. In conclusion, our studies imply the potentially clinical application of GUE to overcome the cardiotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Ling Zhang
- Pharmaceutical Informatics Institute, Zhejiang University, Hangzhou, P R China
| | | | | | | | | | | |
Collapse
|
46
|
Sepe DM, Ginsberg JP, Balis FM. Dexrazoxane as a cardioprotectant in children receiving anthracyclines. Oncologist 2010; 15:1220-6. [PMID: 21051660 DOI: 10.1634/theoncologist.2010-0162] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Anthracyclines play a critical role in the treatment of a variety of childhood cancers. However, the cumulative cardiotoxic effects of anthracyclines limit the use of these agents in many treatment regimens. Dexrazoxane is a cardioprotectant that significantly reduces the incidence of adverse cardiac events in women with advanced breast cancer treated with doxorubicin-containing regimens. Clinical evidence for the efficacy of dexrazoxane as a cardioprotectant in children, especially from randomized clinical trials, is limited, but the available data support a short-term cardioprotective effect. Long-term follow-up in children treated with dexrazoxane has not been reported. Dexrazoxane's impact on the antitumor effect and toxicity profile of the anthracyclines and the role of dexrazoxane in the development of secondary malignant neoplasms in patients who received dexrazoxane are reviewed. Based on the available data, dexrazoxane appears to be a safe and effective cardioprotectant in children, and it does not appear to alter overall survival times in children with cancer. Continued follow-up from previous trials is needed to determine the long-term effect of dexrazoxane on cardiac outcomes and quality of life.
Collapse
Affiliation(s)
- Dana M Sepe
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
47
|
Janeway KA, Grier HE. Sequelae of osteosarcoma medical therapy: a review of rare acute toxicities and late effects. Lancet Oncol 2010; 11:670-8. [DOI: 10.1016/s1470-2045(10)70062-0] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
48
|
[High risk of cardiac dysfunction after treatment of secondary acute myeloid leukemia following chemotherapy and radiotherapy for breast cancer]. Bull Cancer 2010; 97:245-54. [PMID: 19819776 DOI: 10.1684/bdc.2009.0958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Secondary acute myeloid leukaemia (AML) occurring after breast cancer is a rare long-term complication of the chemo- and/or radiation therapy required to treat breast cancer. The usually recognized curative option of these secondary AML includes courses of anthracycline-based chemotherapy followed by haematopoietic stem cell transplantation (HSCT). Cardiac dysfunction during AML treatment of these patients previously treated with anthracyclines for breast cancer has not been reported to date. We evaluated the evolution of cardiac function in seven patients treated with anthracyclines and/or autologous or allogeneic bone marrow transplantation for secondary AML occurring after breast cancer. All of the patients who received a cumulative anthracycline dose above the cardiac toxicity threshold developed cardiac symptoms during AML chemotherapy courses. Moreover, four of the five transplanted patients developed severe heart failure among which two were fatal. Thus, the risk of severe cardiac dysfunction after treatment of secondary AML following breast cancer must be taken in account as part of the therapeutic strategy of those patients. As discussed here, an accurate evaluation of risk factors, the use of sensitive detection tests and of cardioprotective drugs as well as that of non-cardiotoxic chemotherapy might decrease the occurrence and severity of this life-threatening complication.
Collapse
|
49
|
Abstracts. Cancer Invest 2009. [DOI: 10.3109/07357909209053257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Gligorov J, Lotz JP. Optimal treatment strategies in postmenopausal women with hormone-receptor-positive and HER2-negative metastatic breast cancer. Breast Cancer Res Treat 2008; 112 Suppl 1:53-66. [PMID: 19101794 DOI: 10.1007/s10549-008-0232-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 10/20/2008] [Indexed: 01/09/2023]
Abstract
Metastatic breast cancer (MBC) is unfortunately still considered incurable; treatment aims to prolong progression-free and overall survival, relieve disease symptoms, and maintain quality of life. Treatment can include endocrine therapy, radiotherapy, chemotherapy, bisphosphonates, and/or targeted therapy; which is used depends on the characteristics of the disease [e.g., hormone receptor status, disease site(s), and response to previous treatment] and the patient (age, comorbidity, and personal preferences). For most patients with hormone-receptor-positive tumors, the first choice of treatment is further endocrine therapy, but endocrine resistance is a common problem in advanced disease. Several novel anticancer agents have been developed with the aim of overcoming endocrine resistance, many of which target intracellular signaling pathways implicated in disease progression or resistance. Among these, inhibitors of growth factor receptor tyrosine kinases and of mammalian target of rapamycin have shown the most promise in clinical trials. Chemotherapy is the cornerstone of MBC treatment in most women. Important considerations when choosing chemotherapy include the choice of agents, and whether to use single-agent or combination therapy. Anthracyclines are one of the most active cytotoxic agents currently used for the treatment of breast cancer, and for many women, further anthracycline therapy at progression or relapse would be the preferred option. However, lifetime exposure to anthracyclines is limited by cumulative cardiotoxicity, which often prevents rechallenge in later lines of therapy. Newer anthracycline formulations have been developed with lower cardiotoxicity than the conventional anthracycline doxorubicin, but these agents still impair cardiac function, and have maximum recommended lifetime doses. Recently, the concomitant use of cardioprotective agents, such as dexrazoxane, has emerged as an effective approach to reducing the cardiotoxic effects of anthracyclines, thus permitting retreatment. Bisphosphonates, which are not associated with the acute toxicities of cytotoxic chemotherapy drugs, are the established standard of care for patients with metastatic bone disease, and have greatly improved outcomes over the last decade. The search is ongoing for novel agents that will, hopefully, bring a cure closer to reality.
Collapse
Affiliation(s)
- Joseph Gligorov
- Department of Medical Oncology, CancerEst, APHP Tenon, University of Paris VI, 4 rue de la Chine, 75970, Paris Cedex 20, France.
| | | |
Collapse
|