1
|
Simon LH, Garritson J, Pullen N, Hayward R. Exercise Preconditioning Preserves Cardiac Function and Enhances Cardiac Recovery Following Dobutamine Stimulation in Doxorubicin-Treated Rat Hearts. J Cardiovasc Pharmacol 2024; 84:188-198. [PMID: 38814887 DOI: 10.1097/fjc.0000000000001583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 04/08/2024] [Indexed: 06/01/2024]
Abstract
ABSTRACT Exercise preconditioning has been shown to protect against doxorubicin (DOX)-induced cardiac dysfunction when hearts are maintained under resting conditions. However, it is unclear whether this exercise-induced protective effect is maintained when the heart is challenged with the β 1 -adrenergic receptor agonist dobutamine (DOB), which mimics acute exercise stress. Fischer 344 rats were randomly assigned to sedentary (SED) or voluntary wheel running (WR) groups for 10 weeks. At week 11, rats were treated with either 15 mg/kg DOX or saline. Five days later, ex vivo cardiac function was assessed using an isolated working heart model at baseline, during the infusion of 7.5 μg·kg -1 ·min -1 DOB, and during recovery. DOB infusion significantly increased left ventricular developed pressure (LVDP), maximal (dP/dt max ) and minimal (dP/dt min ) rate of left ventricular pressure development, and heart rate in all groups ( P < 0.05). SED + DOX also showed a lower baseline and recovery LVDP than WR + DOX (83 ± 12 vs. 109 ± 6 mm Hg baseline, 76 ± 11 vs. 100 ± 10 mm Hg recovery, P < 0.05). WR + DOX showed higher dP/dt max and lower dP/dt min when compared with SED + DOX during DOB infusion (7311 ± 1481 vs. 5167 ± 1436 mm Hg/s and -4059 ± 1114 vs.-3158 ± 1176 mm Hg/s, respectively). SED + DOX dP/dt max was significantly lower during baseline and during recovery when compared with all other groups ( P < 0.05). These data suggest that exercise preconditioning preserved cardiac function after DOX exposure even when the heart is challenged with DOB, and it appeared to preserve the heart's ability to recover from this functional challenge.
Collapse
Affiliation(s)
- Lea Haverbeck Simon
- Department of Kinesiology, Nutrition, and Dietetics, and the University of Northern Colorado Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO; and
| | - Jacob Garritson
- Department of Kinesiology, Nutrition, and Dietetics, and the University of Northern Colorado Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO; and
| | - Nicholas Pullen
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO
| | - Reid Hayward
- Department of Kinesiology, Nutrition, and Dietetics, and the University of Northern Colorado Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO; and
| |
Collapse
|
2
|
Belger C, Abrahams C, Imamdin A, Lecour S. Doxorubicin-induced cardiotoxicity and risk factors. IJC HEART & VASCULATURE 2024; 50:101332. [PMID: 38222069 PMCID: PMC10784684 DOI: 10.1016/j.ijcha.2023.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used as a chemotherapeutic agent to treat solid tumours and hematologic malignancies. Although useful in the treatment of cancers, the benefit of DOX is limited due to its cardiotoxic effect that is observed in a large number of patients. In the literature, there is evidence that the presence of various factors may increase the risk of developing DOX-induced cardiotoxicity. A better understanding of the role of these different factors in DOX-induced cardiotoxicity may facilitate the choice of the therapeutic approach in cancer patients suffering from various cardiovascular risk factors. In this review, we therefore discuss the latest findings in both preclinical and clinical research suggesting a link between DOX-induced cardiotoxicity and various risk factors including sex, age, ethnicity, diabetes, dyslipidaemia, obesity, hypertension, cardiovascular disease and co-medications.
Collapse
Affiliation(s)
| | | | - Aqeela Imamdin
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
3
|
Qiu Y, Jiang P, Huang Y. Anthracycline-induced cardiotoxicity: mechanisms, monitoring, and prevention. Front Cardiovasc Med 2023; 10:1242596. [PMID: 38173817 PMCID: PMC10762801 DOI: 10.3389/fcvm.2023.1242596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Anthracyclines are the most fundamental and important treatment of several cancers especially for lymphoma and breast cancer. However, their use is limited by a dose-dependent cardiotoxicity which may emerge early at the initiation of anthracycline administration or several years after termination of the therapy. A full comprehending of the mechanisms of anthracycline-induced cardiotoxicity, which has not been achieved and is currently under the efforts, is critical to the advance of developing effective methods to protect against the cardiotoxicity, as well as to early detect and treat it. Therefore, we review the recent progress of the mechanism underlying anthracycline-induced cardiotoxicity, as well as approaches to monitor and prevent this issue.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Piao Jiang
- Department of Oncology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Yingmei Huang
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
4
|
Das B. Pharmacotherapy for Cancer Treatment-Related Cardiac Dysfunction and Heart Failure in Childhood Cancer Survivors. Paediatr Drugs 2023; 25:695-707. [PMID: 37639193 DOI: 10.1007/s40272-023-00585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 08/29/2023]
Abstract
The number of childhood cancer survivors is increasing rapidly. According to American Association for Cancer Research, there are more than 750,000 childhood cancer survivors in the United States and Europe. As the number of childhood cancer survivors increases, so does cancer treatment-related cardiac dysfunction (CTRCD), leading to heart failure (HF). It has been reported that childhood cancer survivors who received anthracyclines are 15 times more likely to have late cancer treatment-related HF and have a 5-fold higher risk of death from cardiovascular (CV) disease than the general population. CV disease is the leading cause of death in childhood cancer survivors. The increasing need to manage cancer survivor patients has led to the rapid creation and adaptation of cardio-oncology. Cardio-oncology is a multidisciplinary science that monitors, treats, and prevents CTRCD. Many guidelines and position statements have been published to help diagnose and manage CTRCD, including those from the American Society of Clinical Oncology, the European Society of Cardiology, the Canadian Cardiovascular Society, the European Society of Medical Oncology, the International Late Effects of Childhood Cancer Guideline Harmonization Group, and many others. However, there remains a gap in identifying high-risk patients likely to develop cardiomyopathy and HF in later life, thus reducing primary and secondary measures being instituted, and when to start treatment when there is echocardiographic evidence of left ventricular (LV) dysfunctions without symptoms of HF. There are no randomized controlled clinical trials for treatment for CTRCD leading to HF in childhood cancer survivors. The treatment of HF due to cancer treatment is similar to the guidelines for general HF. This review describes the latest pharmacologic therapy for preventing and treating LV dysfunction and HF in childhood cancer survivors based on expert consensus guidelines and extrapolating data from adult HF trials.
Collapse
Affiliation(s)
- Bibhuti Das
- Division of Pediatric Cardiology, Department of Pediatrics, Baylor Scott and White McLane Children's Medical Center, Temple, TX, 76502, USA.
| |
Collapse
|
5
|
Kouwenberg TW, van Dalen EC, Feijen EAM, Netea SA, Bolier M, Slieker MG, Hoesein FAAM, Kremer LCM, Grotenhuis HB, Mavinkurve-Groothuis AMC. Acute and early-onset cardiotoxicity in children and adolescents with cancer: a systematic review. BMC Cancer 2023; 23:866. [PMID: 37710224 PMCID: PMC10500898 DOI: 10.1186/s12885-023-11353-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Cardiotoxicity is among the most important adverse effects of childhood cancer treatment. Anthracyclines, mitoxantrone and radiotherapy involving the heart are its main causes. Subclinical cardiac dysfunction may over time progress to clinical heart failure. The majority of previous studies have focused on late-onset cardiotoxicity. In this systematic review, we discuss the prevalence and risk factors for acute and early-onset cardiotoxicity in children and adolescents with cancer treated with anthracyclines, mitoxantrone or radiotherapy involving the heart. METHODS A literature search was performed within PubMed and reference lists of relevant studies. Studies were eligible if they reported on cardiotoxicity measured by clinical, echocardiographic and biochemical parameters routinely used in clinical practice during or within one year after the start of cancer treatment in ≥ 25 children and adolescents with cancer. Information about study population, treatment, outcomes of diagnostic tests used for cardiotoxicity assessment and risk factors was extracted and risk of bias was assessed. RESULTS Our PubMed search yielded 3649 unique publications, 44 of which fulfilled the inclusion criteria. One additional study was identified by scanning the reference lists of relevant studies. In these 45 studies, acute and early-onset cardiotoxicity was studied in 7797 children and adolescents. Definitions of acute and early-onset cardiotoxicity prove to be highly heterogeneous. Prevalence rates varied for different cardiotoxicity definitions: systolic dysfunction (0.0-56.4%), diastolic dysfunction (30.0-100%), combinations of echocardiography and/or clinical parameters (0.0-38.1%), clinical symptoms (0.0-25.5%) and biomarker levels (0.0-37.5%). Shortening fraction and ejection fraction significantly decreased during treatment. Cumulative anthracycline dose proves to be an important risk factor. CONCLUSIONS Various definitions have been used to describe acute and early-onset cardiotoxicity due to childhood cancer treatment, complicating the establishment of its exact prevalence. Our findings underscore the importance of uniform international guidelines for the monitoring of cardiac function during and shortly after childhood cancer treatment.
Collapse
Affiliation(s)
- Theodorus W Kouwenberg
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| | - Elvira C van Dalen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Elizabeth A M Feijen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Stejara A Netea
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Melissa Bolier
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Martijn G Slieker
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Cardiology, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | | - Leontien C M Kremer
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Heynric B Grotenhuis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Cardiology, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| | | |
Collapse
|
6
|
Bertorello N, Luksch R, Bisogno G, Haupt R, Spallarossa P, Cenna R, Fagioli F. Cardiotoxicity in children with cancer treated with anthracyclines: A position statement on dexrazoxane. Pediatr Blood Cancer 2023; 70:e30515. [PMID: 37355856 DOI: 10.1002/pbc.30515] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/26/2023]
Abstract
Cardiovascular disease is the leading cause of non-malignant morbidity and mortality in childhood cancer survivors (CCSs). Anthracyclines are included in many treatment regimens for paediatric cancer, but unfortunately, these compounds are cardiotoxic. One in 10 CCSs who has received an anthracycline will develop a symptomatic cardiac event over time. Given the crucial need to mitigate anthracycline-related cardiotoxicity (ARC), the authors critically examined published data to identify effective cardioprotective strategies. Based on their expert analysis of contemporary literature data, it was concluded that consideration should be given for routine use of dexrazoxane in children with cancer who are at risk of ARC.
Collapse
Affiliation(s)
- Nicoletta Bertorello
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
| | - Roberto Luksch
- Paediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gianni Bisogno
- Hematology and Oncology Division, Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Riccardo Haupt
- Epidemiology and Biostatistics Unit and DOPO clinic, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Rosita Cenna
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
| | - Franca Fagioli
- Paediatric Oncology Division, Regina Margherita Children's Hospital, AOU Città della Salute e della Scienza, Turin, Italy
- University of Turin, Turin, Italy
| |
Collapse
|
7
|
Boluda B, Solana-Altabella A, Cano I, Martínez-Cuadrón D, Acuña-Cruz E, Torres-Miñana L, Rodríguez-Veiga R, Navarro-Vicente I, Martínez-Campuzano D, García-Ruiz R, Lloret P, Asensi P, Osa-Sáez A, Aguero J, Rodríguez-Serrano M, Buendía-Fuentes F, Megías-Vericat JE, Martín-Herreros B, Barragán E, Sargas C, Salas M, Wooddell M, Dharmani C, Sanz MA, De la Rubia J, Montesinos P. Incidence and Risk Factors for Development of Cardiac Toxicity in Adult Patients with Newly Diagnosed Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:2267. [PMID: 37190195 PMCID: PMC10136564 DOI: 10.3390/cancers15082267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The incidence of cardiac morbimortality in acute myeloid leukemia (AML) is not well known. We aim to estimate the cumulative incidence (CI) of cardiac events in AML patients and to identify risk factors for their occurrence. Among 571 newly diagnosed AML patients, 26 (4.6%) developed fatal cardiac events, and among 525 treated patients, 19 (3.6%) experienced fatal cardiac events (CI: 2% at 6 months; 6.7% at 9 years). Prior heart disease was associated with the development of fatal cardiac events (hazard ratio (HR) = 6.9). The CI of non-fatal cardiac events was 43.7% at 6 months and 56.9% at 9 years. Age ≥ 65 (HR = 2.2), relevant cardiac antecedents (HR = 1.4), and non-intensive chemotherapy (HR = 1.8) were associated with non-fatal cardiac events. The 9-year CI of grade 1-2 QTcF prolongation was 11.2%, grade 3 was 2.7%, and no patient had grade 4-5 events. The 9-year CI of grade 1-2 cardiac failure was 1.3%, grade 3-4 was 15%, and grade 5 was 2.1%; of grade 1-2, arrhythmia was 1.9%, grade 3-4 was 9.1%, and grade 5 was 1%. Among 285 intensive therapy patients, median overall survival decreased in those experiencing grade 3-4 cardiac events (p < 0.001). We observed a high incidence of cardiac toxicity associated with significant mortality in AML.
Collapse
Affiliation(s)
- Blanca Boluda
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Antonio Solana-Altabella
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Pharmacy Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Isabel Cano
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - David Martínez-Cuadrón
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Evelyn Acuña-Cruz
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Laura Torres-Miñana
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Rebeca Rodríguez-Veiga
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Irene Navarro-Vicente
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - David Martínez-Campuzano
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Raquel García-Ruiz
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Pilar Lloret
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Pedro Asensi
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
| | - Ana Osa-Sáez
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Jaume Aguero
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | | | | | | | - Beatriz Martín-Herreros
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Eva Barragán
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Claudia Sargas
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Maribel Salas
- Daiichi Sankyo, Inc., Basking Ridge, NJ 07920, USA
- Center for Real-World Effectiveness and Safety of Therapeutics (CREST), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | - Miguel A. Sanz
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier De la Rubia
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Internal Medicine, School of Medicine and Dentistry, Catholic University of Valencia, 46001 Valencia, Spain
| | - Pau Montesinos
- Hematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain (I.N.-V.)
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Mir A, Badi Y, Bugazia S, Nourelden AZ, Fathallah AH, Ragab KM, Alsillak M, Elsayed SM, Hagrass AI, Bawek S, Kalot M, Brumberger ZL. Efficacy and safety of cardioprotective drugs in chemotherapy-induced cardiotoxicity: an updated systematic review & network meta-analysis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:10. [PMID: 36804940 PMCID: PMC9938608 DOI: 10.1186/s40959-023-00159-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Cancer patients receiving chemotherapy have an increased risk of cardiovascular complications. This limits the widespread use of lifesaving therapies, often necessitating alternate lower efficacy regimens, or precluding chemotherapy entirely. Prior studies have suggested that using common cardioprotective agents may attenuate chemotherapy-induced cardiotoxicity. However, small sample sizes and conflicting outcomes have limited the clinical significance of these results. HYPOTHESIS A comprehensive network meta-analysis using updated and high-quality data can provide more conclusive information to assess which drug or drug class has the most significant effect in the management of chemotherapy-induced cardiotoxicity. METHODS We performed a literature search for randomized controlled trials (RCTs) investigating the effects of cardioprotective agents in patients with chemotherapy-induced cardiotoxicity. We used established analytical tools (netmeta package in RStudio) and data extraction formats to analyze the outcome data. To obviate systematic bias in the selection and interpretation of RCTs, we employed the validated Cochrane risk-of-bias tools. Agents included were statins, aldosterone receptor antagonists (MRAs), ACEIs, ARBs, and beta-blockers. Outcomes examined were improvement in clinical and laboratory parameters of cardiac function including a decreased reduction in left ventricular ejection fraction (LVEF), clinical HF, troponin-I, and B-natriuretic peptide levels. RESULTS Our study included 33 RCTs including a total of 3,285 patients. Compared to control groups, spironolactone therapy was associated with the greatest LVEF improvement (Mean difference (MD) = 12.80, [7.90; 17.70]), followed by enalapril (MD = 7.62, [5.31; 9.94]), nebivolol (MD = 7.30, [2.39; 12.21]), and statins (MD = 6.72, [3.58; 9.85]). Spironolactone was also associated with a significant reduction in troponin elevation (MD = - 0.01, [- 0.02; - 0.01]). Enalapril demonstrated the greatest BNP reduction (MD = - 49.00, [- 68.89; - 29.11]), which was followed by spironolactone (MD = - 16.00, [- 23.9; - 8.10]). Additionally, patients on enalapril had the lowest risk of developing clinical HF compared to the control population (RR = 0.05, [0.00; 0.75]). CONCLUSION Our analysis reaffirmed that statins, MRAs, ACEIs, and beta-blockers can significantly attenuate chemotherapy-induced cardiotoxicity, while ARBs showed no significant effects. Spironolactone showed the most robust improvement of LVEF, which best supports its use among this population. Our analysis warrants future clinical studies examining the cardioprotective effects of cardiac remodeling therapy in cancer patients treated with chemotherapeutic agents.
Collapse
Affiliation(s)
- Ali Mir
- grid.273335.30000 0004 1936 9887Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY USA
| | - Yasra Badi
- grid.517786.aAll Saints University School of Medicine, Roseau, Dominica
| | - Seif Bugazia
- grid.490189.d0000 0004 0433 2862Henry Ford Macomb Hospital, Clinton Township, Macomb County, MI USA
| | | | | | - Khaled Mohamed Ragab
- grid.411806.a0000 0000 8999 4945Faculty of Medicine, Minia University, Minia, Egypt
| | - Mohammed Alsillak
- grid.417218.90000 0004 0451 9790Woodhull Medical and Mental Health Center Program, Brooklyn, NY USA
| | - Sarah Makram Elsayed
- grid.412319.c0000 0004 1765 2101Faculty of Medicine, October 6 University, Giza, Egypt
| | | | - Sawyer Bawek
- grid.273335.30000 0004 1936 9887Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY USA
| | - Mohamad Kalot
- grid.273335.30000 0004 1936 9887Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY USA
| | - Zachary L. Brumberger
- grid.273335.30000 0004 1936 9887Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY USA
| |
Collapse
|
9
|
Salau VF, Erukainure OL, Olofinsan KA, Msomi NZ, Ijomone OK, Islam MS. Ferulic acid mitigates diabetic cardiomyopathy via modulation of metabolic abnormalities in cardiac tissues of diabetic rats. Fundam Clin Pharmacol 2023; 37:44-59. [PMID: 35841183 PMCID: PMC10086938 DOI: 10.1111/fcp.12819] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 01/25/2023]
Abstract
Cardiovascular abnormalities have been reported as a major contributor of diabetic mortality. The protective effect of ferulic acid on diabetic cardiomyopathy in fructose-streptozotocin induced type 2 diabetes (T2D) rat model was elucidated in this study. Type 2 diabetic rats were treated by oral administration of low (150 mg/kg b.w) and high (300 mg/kg b.w) doses of ferulic acid. Metformin was used as the antidiabetic drug. Rats were humanely euthanized after 5 weeks of treatment, and their blood and hearts were collected. Induction of T2D depleted the levels of reduced glutathione, glycogen, and HDL-cholesterol and the activities of superoxide dismutase, catalase, ENTPDase, and 5'nucleotidase. It simultaneously triggered increase in the levels of malondialdehyde, total cholesterol, triglyceride, LDL-cholesterol, creatinine kinase-MB as well as activities of acetylcholinesterase, angiotensin converting enzyme (ACE), ATPase, glucose-6-phopsphatase, fructose-1,6-bisphophatase, glycogen phosphorylase, and lipase. T2D induction further revealed an obvious degeneration of cardiac muscle morphology. However, treatment with ferulic acid markedly reversed the levels and activities of these biomarkers with concomitant improvement in myocardium structural morphology, which had favorable comparison with the standard drug, metformin. Additionally, T2D induction led to the depletion of 40%, 75%, and 33% of fatty acids, fatty esters, and steroids, respectively, with concomitant generation of eicosenoic acid, gamolenic acid, and vitamin E. Ferulic acid treatment restored eicosanoic acid, 2-hydroxyethyl ester, with concomitant generation of 6-octadecenoic acid, (Z)-, cis-11-eicosenoic acid, tridecanedioic acid, octadecanoic acid, 2-hydroxyethyl ester, ethyl 3-hydroxytridecanoate, dipalmitin, cholesterol isocaproate, cholest-5-ene, 3-(1-oxobuthoxy)-, cholesta-3,5-diene. These results suggest the cardioprotective potential of ferulic acid against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Veronica F Salau
- Department of Biochemistry, University of KwaZulu-Natal, Durban, South Africa.,Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | | | - Nontokozo Z Msomi
- Department of Biochemistry, University of KwaZulu-Natal, Durban, South Africa
| | - Olayemi K Ijomone
- Department of Anatomy, University of Medical Sciences, Ondo City, Nigeria
| | - Md Shahidul Islam
- Department of Biochemistry, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
10
|
Hegazy M, Ghaleb S, Das BB. Diagnosis and Management of Cancer Treatment-Related Cardiac Dysfunction and Heart Failure in Children. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10010149. [PMID: 36670699 PMCID: PMC9856743 DOI: 10.3390/children10010149] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
It is disheartening for parents to discover that their children have long-term cardiac dysfunction after being cured of life-threatening childhood cancers. As the number of childhood cancer survivors increases, early and late oncology-therapy-related cardiovascular complications continues to rise. It is essential to understand that cardiotoxicity in childhood cancer survivors is persistent and progressive. A child's cancer experience extends throughout his lifetime, and ongoing care for long-term survivors is recognized as an essential part of the cancer care continuum. Initially, there was a lack of recognition of late cardiotoxicities related to cancer therapy. About 38 years ago, in 1984, pioneers like Dr. Lipshultz and others published anecdotal case reports of late cardiotoxicities in children and adolescents exposed to chemotherapy, including some who ended up with heart transplantation. At that time, cardiac tests for cancer survivors were denied by insurance companies because they did not meet appropriate use criteria. Since then, cardio-oncology has been an emerging field of cardiology that focuses on the early detection of cancer therapy-related cardiac dysfunction occurring during and after oncological treatment. The passionate pursuit of many healthcare professionals to make life better for childhood cancer survivors led to more than 10,000 peer-reviewed publications in the last 40 years. We synthesized the existing evidence-based practice and described our experiences in this review to share our current method of surveillance and management of cardiac dysfunction related to cancer therapy. This review aims to discuss the pathological basis of cancer therapy-related cardiac dysfunction and heart failure, how to stratify patients prone to cardiotoxicity by identifying modifiable risk factors, early detection of cardiac dysfunction, and prevention and management of heart failure during and after cancer therapy in children. We emphasize serial longitudinal follow-ups of childhood cancer survivors and targeted intervention for high-risk patients. We describe our experience with the new paradigm of cardio-oncology care, and collaboration between cardiologist and oncologist is needed to maximize cancer survival while minimizing late cardiotoxicity.
Collapse
Affiliation(s)
- Mohamed Hegazy
- University of Mississippi Medical Center Program, Jackson, MS 39216, USA
| | - Stephanie Ghaleb
- Division of Pediatric Cardiology, Department of Pediatrics, Children’s of Mississippi Heart Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bibhuti B Das
- Division of Pediatric Cardiology, Department of Pediatrics, McLane Children’s Baylor Scott and White Medical Center, Baylor College of Medicine-Temple, Temple, TX 76502, USA
- Correspondence: ; Tel.: +1-254-935-4980
| |
Collapse
|
11
|
Bennati E, Girolami F, Spaziani G, Calabri GB, Favre C, Parrini I, Lucà F, Tamburini A, Favilli S. Cardio-Oncology in Childhood: State of the Art. Curr Oncol Rep 2022; 24:1765-1777. [PMID: 36181610 DOI: 10.1007/s11912-022-01329-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Cardio-oncology is an increasingly important field of cardiology that focuses on the detection, monitoring, and treatment of cardiovascular disease (CVD) occurring during and after oncological treatments. The survival rate for childhood cancer patients has dramatically increased thanks to new treatment protocols and cardiovascular (CV) sequelae represent the third most frequent cause of mortality in surviving patients. This study aims to provide a complete and updated review of all the main aspects of cardio-oncology in childhood and to highlight the critical issues. RECENT FINDINGS The problem of CV complications in childhood cancer survivors raises the need to make an early diagnosis of cardiotoxicity by the new imaging and laboratory techniques in order to intervene promptly and to implement pharmacological strategies and lifestyle changes to reduce or even to prevent cardiac injury. Furthermore, a stratification of CV risk, also including new predisposing factors such as the presence of some genetic mutations, is of paramount importance before undertaking oncological treatments. Besides, a systematic and personalized planning of long-term follow-up is fundamental to ensure a transition from pediatric to adult hospital and to avoid missed or late diagnosis of cardiomyopathy. We reviewed the main risk factors for cardiotoxicity in children, both traditional and emerging ones: the mechanisms of toxicity of both old and new antineoplastic therapies, the techniques for detecting cardiac damage, and the current evidence regarding pharmacological cardioprotection. At the end, we focused our attention on the existing guidelines and strategies about the long-term follow-up of childhood cancer survivors.
Collapse
Affiliation(s)
- Elena Bennati
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy.
| | - Francesca Girolami
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Gaia Spaziani
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | | | - Claudio Favre
- Department of Pediatric Hematology-Oncology, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Iris Parrini
- Cardiology Unit, Mauriziano Umberto I Hospital, Corso Turati 62, Turin, Italy
| | - Fabiana Lucà
- Department of Cardiology, Grande Ospedale Metropolitano, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Angela Tamburini
- Department of Pediatric Hematology-Oncology, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Silvia Favilli
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| |
Collapse
|
12
|
Cardiovascular Complications in Hematopoietic Stem Cell Transplanted Patients. J Pers Med 2022; 12:jpm12111797. [PMID: 36579521 PMCID: PMC9692512 DOI: 10.3390/jpm12111797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only curative treatment for many patients suffering from hematologic malignancies, solid tumors, inborn errors of metabolism or genetic disorders. Despite decades of successful HSCT, clinical outcomes are still far from satisfactory due to treatment-related complications, including graft-versus-host disease (GvHD) and cardiovascular complications (CVC). CVC may affect patients in the acute period post-HSCT; however, the occurrence is far higher among long-term survivors. Induction treatment using cardiotoxic treatments, e.g., anthracyclines and radiotherapy, conditioning regimens containing cyclophosphamide, and post-HSCT comorbidities, including GvHD, are factors contributing to CVC. Cardiac function evaluation prior to and post-transplantation is an important strategy for choosing the proper conditioning regimen, HSCT protocol and post-HSCT supportive care. Cardiac systolic function evaluation by echocardiography, in addition to serum cardiac biomarkers, such as troponins and brain natriuretic peptides, is recommended as a routine follow-up for HSCT patients. Angiotensin-converting enzyme inhibitors, angiotensin-II-receptor blockers, and beta-blockers, which are mostly used for the treatment of chemotherapy-induced cardiotoxicity, might be used as treatments for HSCT-related CVC. In summary, the present review reveals the urgent need for further investigations concerning HSCT-related CVC both at the preclinical and clinical levels due to the lack of knowledge about CVC and its underlying mechanisms.
Collapse
|
13
|
Hao W, Shi YY, Qin YN, Sun CP, Chen LY, Wu CY, Bao YJ, Liu S. Cardioprotective effect of Chinese herbal medicine for anthracycline-induced cardiotoxicity in cancer patients: A meta-analysis of prospective studies. Medicine (Baltimore) 2022; 101:e29691. [PMID: 35905252 PMCID: PMC9333524 DOI: 10.1097/md.0000000000029691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND To assess the benefits and harmful effects of Chinese herbal medicine (CHM) formulations in preventing anthracyclines (ANT)-induced cardiotoxicity. METHOD The Cochrane Library, Pubmed and EMBASE databases were electronically searched for relevant randomized controlled trials (RCTs) published till December 2021 in English or Chinese-language, in addition to manual searches through the reference lists of the selected papers, and the Chinese Conference Papers Database. Data was extracted by 2 investigators independently. RESULT Seventeen RCTs reporting 11 different CHMs were included in this meta-analysis. The use of CHM reduced the occurrence of clinical heart failure (RR 0.48, 95% CI 0.39 to 0.60, P < .01) compared to the control group. Data on subclinical heart failure in terms of LVEF values showed that CHM reduced the occurrence of subclinical heart failure (RR 0.47, 95% CI 0.35 to 0.62, P < .01) as well. CONCLUSION CHM is an effective and safe cardioprotective intervention that can potentially prevent ANT-induced cardiotoxicity. However, due to the insufficient quality of the included trials, our results should be interpreted with cautious.
Collapse
Affiliation(s)
- Wei Hao
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - You-Yang Shi
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue-Nong Qin
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen-Ping Sun
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Ying Chen
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chun-Yu Wu
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Jia Bao
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Long Hua Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Sheng Liu, South Wanping Road, No. 725 (e-mail: )
| |
Collapse
|
14
|
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: 2.5] [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
|
15
|
Chen DS, Yan J, Yang PZ. Cardiomyocyte Atrophy, an Underestimated Contributor in Doxorubicin-Induced Cardiotoxicity. Front Cardiovasc Med 2022; 9:812578. [PMID: 35282350 PMCID: PMC8913904 DOI: 10.3389/fcvm.2022.812578] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022] Open
Abstract
Left ventricular (LV) mass loss is prevalent in doxorubicin (DOX)-induced cardiotoxicity and is responsible for the progressive decline of cardiac function. Comparing with the well-studied role of cell death, the part of cardiomyocyte atrophy (CMA) playing in the LV mass loss is underestimated and the knowledge of the underlying mechanism is still limited. In this review, we summarized the recent advances in the DOX-induced CMA. We found that the CMA caused by DOX is associated with the upregulation of FOXOs and “atrogenes,” the activation of transient receptor potential canonical 3-NADPH oxidase 2 (TRPC3-Nox2) axis, and the suppression of IGF-1-PI3K signaling pathway. The imbalance of anabolic and catabolic process may be the common final pathway of these mechanisms. At last, we provided some strategies that have been demonstrated to alleviate the DOX-induced CMA in animal models.
Collapse
Affiliation(s)
- De-Shu Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
| | - Jing Yan
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
- Jing Yan
| | - Ping-Zhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, China
- Heart Center of Zhujiang Hospital, Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, China
- *Correspondence: Ping-Zhen Yang
| |
Collapse
|
16
|
Jefferies JL, Mazur WM, Howell CR, Plana JC, Ness KK, Li Z, Joshi VM, Green DM, Mulrooney DA, Towbin JA, Martinez HR, Goldberg JF, Howell RM, Srivastava DK, Robison LL, Hudson MM, Armstrong GT. Cardiac remodeling after anthracycline and radiotherapy exposure in adult survivors of childhood cancer: A report from the St Jude Lifetime Cohort Study. Cancer 2021; 127:4646-4655. [PMID: 34411296 DOI: 10.1002/cncr.33860] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/03/2021] [Accepted: 06/25/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Limited data exist regarding left ventricular remodeling patterns observed in adult survivors of childhood cancer after therapy. METHODS Among 1190 adult survivors diagnosed with childhood cancer (median age at diagnosis, 9 years [interquartile range (IQR), 3.8-14.4 years]; age at evaluation, 35.6 years [IQR, 29.5-42.8 years]), treatment exposures included anthracyclines (n = 346), chest radiotherapy (n = 174), both (n = 245), or neither (n = 425). Prospective echocardiographic assessment compared survivors with 449 noncancer controls classified according to left ventricle geometric patterns. Associations between left ventricle geometric patterns and decreased exercise tolerance were assessed. RESULTS Overall, 28.2% of survivors (95% confidence interval [CI], 25.6%-30.8%) exhibited concentric remodeling, 2.4% (95% CI, 1.6%-3.5%) exhibited eccentric hypertrophy, and 1.1% (95% CI, 0.6%-1.9%) exhibited concentric hypertrophy. A greater proportion of survivors who received only chest radiotherapy (41%) had concentric remodeling compared with those who received only anthracyclines (24%), both (27%), or neither (27%; all P < .001), and all were greater than the proportions in noncancer controls (18%; all P < .05). Concentric remodeling was associated with radiation exposure, but not with anthracycline exposure, in multivariable models. Survivors who had concentric remodeling were more likely to have a maximal oxygen uptake peak <85% compared with those who had normal geometry (81.0% vs 66.3%; odds ratio, 1.75; 95% CI, 1.15-2.68). CONCLUSIONS Chest radiation therapy, but not anthracycline therapy, increased the risk for concentric remodeling in survivors of childhood cancer. The presence of concentric remodeling was associated with increased exercise intolerance.
Collapse
Affiliation(s)
- John L Jefferies
- The Cardiovascular Institute, University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Carrie R Howell
- Department of Medicine, Division of Preventive Medicine, University of Alabama Birmingham, Birmingham, Alabama
| | - Juan C Plana
- Division of Cardiology, Baylor College of Medicine, Houston, Texas
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Zhenghong Li
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Vijaya M Joshi
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Daniel M Green
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Daniel A Mulrooney
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Jeffrey A Towbin
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hugo R Martinez
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jason F Goldberg
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Rebecca M Howell
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deo Kumar Srivastava
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Melissa M Hudson
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee.,Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
17
|
Cardiac complications associated with hematopoietic stem-cell transplantation. Bone Marrow Transplant 2021; 56:2637-2643. [PMID: 34381168 DOI: 10.1038/s41409-021-01427-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022]
Abstract
Advances in chemotherapy and supportive therapy have resulted in improved clinical outcomes in patients with hematological malignancies undergoing hematopoietic stem-cell transplantation (HSCT). However, the association between HSCT and early- and late-onset cardiotoxicity remains controversial as these cardiac complications, including acute heart failure and arrhythmia, such as atrial fibrillation, can occasionally be lethal. Although the overall pathophysiology has not been elucidated, initial/salvage chemotherapy before HSCT, such as anthracycline-combined regimens, conditioning regimens, thoracic radiotherapy, and pre-existing personal risk factors, could be associated with an increased risk of cardiac events. Routine monitoring of cardiac function using global longitudinal strain or left ventricular ejection fraction in echocardiogram and serum biomarkers could be an option to detect early changes in cardiac status before irreversible cardiac complications develop. While beta-blockers and angiotensin-converting enzyme inhibitors are commonly used for cardioprotection, their clinical benefit has not been fully established in HSCT-associated cardiotoxicity. In the future, genetic analysis to reveal individual vulnerability to cardiotoxicity and prospective trials assessing the clinical benefit of early interventions, including novel agents such as angiotensin receptor-neprilysin inhibitor, are warranted. Collaboration between oncologists and cardiologists is crucial to establishing a strategy to prevent cardiac complications.
Collapse
|
18
|
Lee M, Chung WB, Lee JE, Park CS, Park WC, Song BJ, Youn HJ. Candesartan and carvedilol for primary prevention of subclinical cardiotoxicity in breast cancer patients without a cardiovascular risk treated with doxorubicin. Cancer Med 2021; 10:3964-3973. [PMID: 33998163 PMCID: PMC8209607 DOI: 10.1002/cam4.3956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND There is no proven primary preventive strategy for doxorubicin-induced subclinical cardiotoxicity (DISC), especially among patients without a cardiovascular (CV) risk. We investigated the primary preventive effect on DISC of the concomitant use of angiotensin receptor blockers (ARBs) or beta-blockers (BBs), especially among breast cancer patients without a CV risk. METHODS A total of 385 patients who were scheduled for doxorubicin chemotherapy were screened. Among them, 195 patients of the study populations were included and were randomly divided into two groups [candesartan 4 mg q.d. vs. carvedilol 3.125 mg q.d.] and patients who were unwilling to take one of the medications were evaluated as controls. The primary outcomes were the incidence of early DISC (DISC developing within 6 months after chemotherapy), and late DISC (DISC developing only at least 12 months after chemotherapy). RESULT Compared with the control group (8 out of 43 patients (18.6%)), only the candesartan group (4 out of 82 patients (4.9%)) showed a significantly lower incidence of early DISC (p = 0.022). Compared with the control group, the candesartan group demonstrated a significantly reduced decrease in left ventricular ejection fraction (LVEF) throughout the study period [-1.0% vs. -3.00 (p < 0.001) at the first follow-up, -1.10% vs. -3.40(p = 0.009) at the second follow-up]. CONCLUSIONS Among breast cancer patients without a CV risk treated with doxorubicin-containing chemotherapy, subclinical cardiotoxicity is prevalent and concomitant administration of low-dose candesartan might be effective to prevent an early decrease in LVEF. Further large-scale, randomized controlled trials will be needed to confirm our findings.
Collapse
Affiliation(s)
- Myunhee Lee
- Division of Cardiology, Department of Internal Medicine, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Baek Chung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji-Eun Lee
- Division of Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan-Seok Park
- Division of Cardiology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Chan Park
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung-Joo Song
- Division of Breast-Thyroid surgery, Department of Surgery, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho-Joong Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
19
|
Temtanakitpaisan Y, Saengnipanthkul S. Monitoring of Metabolic Syndrome and Cardiovascular Disease in Childhood Cancer Survivors. J Adolesc Young Adult Oncol 2021; 11:17-26. [PMID: 33989069 DOI: 10.1089/jayao.2021.0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Advances in cancer treatment have significantly improved childhood cancer survival, although metabolic syndrome and cardiovascular disease are common long-term complications that may occur years after treatment. Childhood cancer survivors may not receive appropriate follow-up due to lack of communication between oncologists and primary care physicians, or, from lack of awareness of possible long-term metabolic and cardiovascular complications after cancer treatment. We, therefore, reviewed current evidence on long-term effects of cancer therapy, and appropriate monitoring for long-term treatment effects in childhood cancer survivors that could lead to early detection and prompt treatment to prevent future cardiovascular events.
Collapse
Affiliation(s)
- Yutthapong Temtanakitpaisan
- Division of Cardiology, Bangkok Hospital Khon Kaen, Khon Kaen, Thailand.,Faculty of Medicine, Mahasarakham University, Mahasarakham, Thailand
| | - Suchaorn Saengnipanthkul
- Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
20
|
Gonciar D, Mocan L, Zlibut A, Mocan T, Agoston-Coldea L. Cardiotoxicity in HER2-positive breast cancer patients. Heart Fail Rev 2021; 26:919-935. [PMID: 33405000 DOI: 10.1007/s10741-020-10072-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 01/22/2023]
Abstract
Due to the recent advances in diagnosis and management of patients with HER2-positive breast cancer, especially through novel HER2-targeted agents, cardiotoxicity becomes an emerging problem. Although chemotherapy significantly increases survival, the risk of cardiovascular disease development is high and still underestimated and could imply treatment discontinuation. Frequently, due to lack of rigorous diagnosis strategies, cardiotoxicity assessment is delayed, and, moreover, the efficacy of current therapy options in restoring heart function is questionable. For a comprehensive risk assessment, it is vital to characterize the clinical spectrum of HER2-targeted agents and anthracyclines, as well as their pathogenic pathways involved in cardiotoxicity. Advanced cardiovascular multimodal imaging and circulating biomarkers plays primary roles in early assessing cardiotoxicity and also in guiding specific preventive measures. Even though the knowledge in this field is rapidly expanding, there are still questions that arise regarding the optimal approach in terms of timing and methods. The aim of the current review aims to providean overview of currently available data.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lucian Mocan
- 3rd Surgery Department, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Alexandru Zlibut
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Teodora Mocan
- Physiology Department, Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca, Romania
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| |
Collapse
|
21
|
Hoeger CW, Turissini C, Asnani A. Doxorubicin Cardiotoxicity: Pathophysiology Updates. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020. [DOI: 10.1007/s11936-020-00842-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
22
|
Ayuna A, Abidin N. The role of neurohormonal blockers in the primary prevention of acute-, early-, and late-onset anthracycline-induced cardiotoxicity. Egypt Heart J 2020; 72:59. [PMID: 32915331 PMCID: PMC7486348 DOI: 10.1186/s43044-020-00090-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/27/2020] [Indexed: 11/28/2022] Open
Abstract
Background Anthracycline-induced cardiotoxicity has been classified based on its onset into acute, early, and late. It may have a significant burden on the quality and quantity of life of those exposed to this class of medication. Currently, there are several ongoing debates on the role of different measures in the primary prevention of cardiotoxicity in cancer survivors. Our article aims to focus on the role of neurohormonal blockers in the primary prevention of anthracycline-induced cardiotoxicity, whether it is acute, early, or late onset. Main body of the abstract PubMed and Google Scholar database were searched for the relevant articles; we reviewed and appraised 15 RCTs, and we found that angiotensin-converting enzyme inhibitors (ACEI) and B-blockers were the most commonly used agents. Angiotensin II receptor blockers (ARBs) and mineralocorticoid receptor antagonists (MRAs) were used in a few other trials. The follow-up period was on the range of 1–156 weeks (mode 26 weeks). Left ventricular ejection fraction (LVEF), left ventricular diameters, and diastolic function were assessed by either echocardiogram or occasionally by cardiac magnetic resonance imaging (MRI). The occurrence of myocardial injury was assessed by troponin I. It was obvious that neurohormonal blockers reduced the occurrence of LVEF and myocardial injury in 14/15 RCTs. Short conclusion Beta-blockers, especially carvedilol and ACEI, especially enalapril, should be considered for the primary prevention of acute- and early-onset cardiotoxicity. ARB and MRA are suitable alternatives when patients are intolerant to ACE-I and B-blockers. We recommend further studies to explore and establish the role of neurohormonal blockers in the primary prevention of the acute-, early-, and late-onset cardiotoxicity.
Collapse
Affiliation(s)
- Ahmed Ayuna
- Salford Royal NHS Foundation Trust, Manchester, UK.
| | - Nik Abidin
- Salford Royal NHS Foundation Trust, Manchester, UK
| |
Collapse
|
23
|
Zhang Y, Liu J, Li Y, Tan N, Du K, Zhao H, Wang J, Zhang J, Wang W, Wang Y. Protective Role of Enalapril in Anthracycline-Induced Cardiotoxicity: A Systematic Review. Front Pharmacol 2020; 11:788. [PMID: 32536868 PMCID: PMC7266978 DOI: 10.3389/fphar.2020.00788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Background Evidence of the preventive and therapeutic effects of enalapril on cardiotoxicity caused by chemotherapy needs to be further confirmed and updated. Methods We performed a systematic review of studies from electronic databases that were searched from inception to January 29, 2019, and included relevant studies analyzing enalapril as a cardioprotective agent before or during the use of anthracyclines by oncology patients. Homogeneous results from different studies were pooled using RevMan 5.3 software. The Cochrane risk-of-bias tool was used to determine the quality of the studies. Results We examined and screened 626 studies according to specific criteria and ultimately included seven studies that were relevant to the indicated topic. Among them, three studies reported the incidence of death during 6- and 12-month follow-up periods. Six of the seven included studies showed possible positive results, suggesting that enalapril plays a cardioprotective role, while five of these studies showed that there was a significant difference in the left ventricular ejection fraction (LVEF) between an enalapril group and a control group (weighted mean difference (WMD) = 7.18, 95% CI: 2.49–11.87, I2 = 96%, P < .001). Moreover, enalapril was beneficial in reducing troponin I (TnI), creatine kinase myocardial band (CK-MB) and N-terminal pro-b-type natriuretic peptide (NT-proBNP) levels in cancer patients treated with anthracycline. Conclusions Although a protective effect of enalapril on myocardial toxicity was observed in terms of the LVEF values and TnI, CK-MB and NT-proBNP levels, its use in the prevention and treatment of cardiotoxicity caused by anthracycline needs to be investigated by more scientific research.
Collapse
Affiliation(s)
- Yili Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junjie Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kangjia Du
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huihui Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Ministry of Education Key Laboratory of TCM Syndrome and Formula & Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
| | - Juan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Ministry of Education Key Laboratory of TCM Syndrome and Formula & Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
| | - Jian Zhang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Ministry of Education Key Laboratory of TCM Syndrome and Formula & Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
24
|
Sobczuk P, Czerwińska M, Kleibert M, Cudnoch-Jędrzejewska A. Anthracycline-induced cardiotoxicity and renin-angiotensin-aldosterone system-from molecular mechanisms to therapeutic applications. Heart Fail Rev 2020; 27:295-319. [PMID: 32472524 PMCID: PMC8739307 DOI: 10.1007/s10741-020-09977-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Few millions of new cancer cases are diagnosed worldwide every year. Due to significant progress in understanding cancer biology and developing new therapies, the mortality rates are decreasing with many of patients that can be completely cured. However, vast majority of them require chemotherapy which comes with high medical costs in terms of adverse events, of which cardiotoxicity is one of the most serious and challenging. Anthracyclines (doxorubicin, epirubicin) are a class of cytotoxic agents used in treatment of breast cancer, sarcomas, or hematological malignancies that are associated with high risk of cardiotoxicity that is observed in even up to 30% of patients and can be diagnosed years after the therapy. The mechanism, in which anthracyclines cause cardiotoxicity are not well known, but it is proposed that dysregulation of renin-angiotensin-aldosterone system (RAAS), one of main humoral regulators of cardiovascular system, may play a significant role. There is increasing evidence that drugs targeting this system can be effective in the prevention and treatment of anthracycline-induced cardiotoxicity what has recently found reflection in the recommendation of some scientific societies. In this review, we comprehensively describe possible mechanisms how anthracyclines affect RAAS and lead to cardiotoxicity. Moreover, we critically review available preclinical and clinical data on use of RAAS inhibitors in the primary and secondary prevention and treatment of cardiac adverse events associated with anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.,Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Magdalena Czerwińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Kleibert
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
25
|
Schwach V, Slaats RH, Passier R. Human Pluripotent Stem Cell-Derived Cardiomyocytes for Assessment of Anticancer Drug-Induced Cardiotoxicity. Front Cardiovasc Med 2020; 7:50. [PMID: 32322588 PMCID: PMC7156610 DOI: 10.3389/fcvm.2020.00050] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/16/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiotoxicity is a major cause of high attrition rates among newly developed drugs. Moreover, anti-cancer treatment-induced cardiotoxicity is one of the leading reasons of mortality in cancer survivors. Cardiotoxicity screening in vitro may improve predictivity of cardiotoxicity by novel drugs, using human pluripotent stem cell (hPSC)-derived-cardiomyocytes. Anthracyclines, including Doxorubicin, are widely used and highly effective chemotherapeutic agents for the treatment of different forms of malignancies. Unfortunately, anthracyclines cause many cardiac complications early or late after therapy. Anthracyclines exhibit their potent anti-cancer effect primarily via induction of DNA damage during the DNA replication phase in proliferative cells. In contrast, studies in animals and hPSC-cardiomyocytes have revealed that cardiotoxic effects particularly arise from (1) the generation of oxidative stress inducing mitochondrial dysfunction, (2) disruption of calcium homeostasis, and (3) changes in transcriptome and proteome, triggering apoptotic cell death. To increase the therapeutic index of chemotherapeutic Doxorubicin therapy several protective strategies have been developed or are under development, such as (1) reducing toxicity through modification of Doxorubicin (analogs), (2) targeted delivery of anthracyclines specifically to the tumor tissue or (3) cardioprotective agents that can be used in combination with Doxorubicin. Despite continuous progress in the field of cardio-oncology, cardiotoxicity is still one of the major complications of anti-cancer therapy. In this review, we focus on current hPSC-cardiomyocyte models for assessing anthracycline-induced cardiotoxicity and strategies for cardioprotection. In addition, we discuss latest developments toward personalized advanced pre-clinical models that are more closely recapitulating the human heart, which are necessary to support in vitro screening platforms with higher predictivity. These advanced models have the potential to reduce the time from bench-to-bedside of novel antineoplastic drugs with reduced cardiotoxicity.
Collapse
Affiliation(s)
- Verena Schwach
- Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, Netherlands
| | - Rolf H Slaats
- Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, Netherlands
| | - Robert Passier
- Applied Stem Cell Technologies, TechMed Centre, University of Twente, Enschede, Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
26
|
Ketterl TG, Latham GJ. Perioperative Cardiothoracic and Vascular Risk in Childhood Cancer and its Survivors. J Cardiothorac Vasc Anesth 2020; 35:162-175. [PMID: 32360009 DOI: 10.1053/j.jvca.2020.02.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/11/2020] [Accepted: 02/28/2020] [Indexed: 11/11/2022]
Abstract
CHILDREN with cancer and survivors of childhood cancer have an increased risk of cardiovascular disease, and this risk in the perioperative period must be understood. During diagnosis and treatment of pediatric cancer, multiple acute cardiovascular morbidities are possible, including anterior mediastinal mass, tamponade, hypertension, cardiomyopathy,and heart failure. Childhood cancer survivors reaching late childhood and adulthood experience substantially increased rates of cardiomyopathy, heart failure, valvular disease, pericardiac disease, ischemia, and arrhythmias. Despite considerable advances in the understanding and therapeutic options of pediatric malignancies, cardiac disease remains the most common treatment-related, noncancer cause of death in childhood cancer survivors. Increasingly, molecularly targeted agents, including small molecule inhibitors, are being incorporated into pediatric oncology. The acute and chronic risks associated with these newer therapeutic options in children are not yet well-described, which poses challenges for clinicians caring for these patients. In the present review, the unique risks factors, prevention strategies, and treatment of cardiovascular toxicities of the child with cancer and the childhood cancer survivor are examined, with an emphasis on the perioperative period.
Collapse
Affiliation(s)
- Tyler G Ketterl
- Department of Pediatric Hematology and Oncology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA
| | - Gregory J Latham
- Department of Anesthesiology and Pain Medicine, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
27
|
Michel L, Mincu RI, Mrotzek SM, Korste S, Neudorf U, Rassaf T, Totzeck M. Cardiac biomarkers for the detection of cardiotoxicity in childhood cancer-a meta-analysis. ESC Heart Fail 2020; 7:423-433. [PMID: 32069386 PMCID: PMC7160486 DOI: 10.1002/ehf2.12589] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/12/2019] [Accepted: 11/25/2019] [Indexed: 01/04/2023] Open
Abstract
AIMS Childhood cancer therapy is associated with a significant risk of therapy-related cardiotoxicity. This meta-analysis aims to evaluate cardiac biomarkers for the detection of cancer therapy-related left ventricular (LV) dysfunction in childhood cancer patients. METHODS AND RESULTS PubMed, Cochrane Library, Wiley Library, and Web of Science were screened for studies investigating brain natriuretic peptide (BNP)/N-terminal proBNP (NT-proBNP) or cardiac troponin in childhood cancer patients. The odds ratios (OR) for elevation of cardiac biomarkers and association with LV dysfunction were calculated using a random-effects model. Data from 27 studies with 1651 subjects were included. BNP/NT-proBNP levels were higher post-treatment compared with controls or pre-treatment values [standardized mean difference = 1.0; 95% confidence interval (CI) = 0.6-1.4; n = 320; P < 0.001]. LV dysfunction was present in 11.76% of included patients, and risk for LV dysfunction was increased in patients with elevated BNP/NT-proBNP (OR = 7.1; 95% CI = 2.0-25.5; n = 350; P = 0.003). The sensitivity of BNP/NT-proBNP for the detection of LV dysfunction was 33.3%, and the specificity was 91.5%. Sensitivity increased when selecting for studies that assessed patients < 5 years after anthracycline exposure and for studies including high cumulative anthracycline doses. Anthracycline chemotherapy was associated with an increased frequency of elevated troponin (OR = 3.7; 95% CI = 2.1-6.5; n = 348; P < 0.001). The available evidence on the association between elevated troponin and LV dysfunction was insufficient for an adequate analysis. In five included studies, the frequency of LV dysfunction was not increased in patients with elevated troponin (OR = 2.5; 95% CI = 0.5-13.2; n = 179; P = 0.53). CONCLUSIONS BNP/NT-proBNP is associated with cardiotoxicity in paediatric cancer patients receiving anthracycline therapy, but owing to low sensitivity, BNP/NT-proBNP has to be evaluated in the context of further parameters including clinical assessment and echocardiography. Future studies are needed to determine whether troponin serves as a marker for cardiotoxicity in children. Standardized recommendations for the application of cardiac biomarkers in children undergoing cardiotoxic cancer therapy may benefit management and clinical outcome.
Collapse
Affiliation(s)
- Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Raluca I Mincu
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Simone M Mrotzek
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Sebastian Korste
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Ulrich Neudorf
- Department of Pediatrics, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| |
Collapse
|
28
|
Fang K, Zhang Y, Liu W, He C. Effects of angiotensin-converting enzyme inhibitor/angiotensin receptor blocker use on cancer therapy-related cardiac dysfunction: a meta-analysis of randomized controlled trials. Heart Fail Rev 2020; 26:101-109. [PMID: 31900787 DOI: 10.1007/s10741-019-09906-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Angiotensin-converting enzyme inhibitor (ACEI) and angiotensin receptor blocker (ARB) may attenuate cancer therapy-related cardiac dysfunction (CTRCD). However, results of the previous studies were not consistent. We performed a meta-analysis to evaluate the influence of ACEI/ARB on CTRCD. Randomized controlled trials (RCTs) were obtained by searching of PubMed, Embase, and Cochrane's Library databases. A random-effect model was used to pool the results. Nine RCTs with 1095 cancer patients that underwent chemotherapy with anthracycline and/or trastuzumab were included. Using of ACEI/ARB significantly preserved left ventricular ejection fraction (LVEF, weighed mean difference = 4.24%, p = 0.002) compared with controls. Subgroup analyses showed that the benefits of ACEI/ARB on LVEF following chemotherapy were consistent and independent of study characteristics including study design, sample size, cancer type, chemotherapy protocols, preventative medications of ACEI or ARB, methods for LVEF measurement, and follow-up durations. The benefits on LVEF following chemotherapy were more remarkable in studies using ACEI and followed ≤ 12 months (p for subgroup difference = 0.04 and 0.02). Use of ACEI/ARB did not significantly reduce the risk of cardiotoxicity events (risk ratio [RR] = 0.63, p = 0.22) but increased the risk of hypotension in these patients (RR = 3.94, p = 0.008). These results indicated that using of ACEI/ARB may moderately attenuate CTRCD following chemotherapy with anthracycline and/or trastuzumab. Large-scale RCTs are needed to evaluate whether the benefits of ACEI/ARB on LVEF are clinically relevant.
Collapse
Affiliation(s)
- Kuaifa Fang
- Department of Cardiology, the Sixth People's Hospital of Huizhou City, Sothern Medical University, Huizhou, 516211, China.
| | - Yihui Zhang
- Department of Cardiology, the Sixth People's Hospital of Huizhou City, Sothern Medical University, Huizhou, 516211, China
| | - Wenbin Liu
- Department of Cardiology, the Sixth People's Hospital of Huizhou City, Sothern Medical University, Huizhou, 516211, China
| | - Cuifang He
- Department of Cardiology, the Sixth People's Hospital of Huizhou City, Sothern Medical University, Huizhou, 516211, China
| |
Collapse
|
29
|
Chen C, Jiang L, Zhang M, Pan X, Peng C, Huang W, Jiang Q. Isodunnianol alleviates doxorubicin-induced myocardial injury by activating protective autophagy. Food Funct 2020; 10:2651-2657. [PMID: 31025676 DOI: 10.1039/c9fo00063a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recurrent cardiotoxicity limits the clinical application of doxorubicin (DOX); however the detailed molecular mechanism of DOX cardiotoxicity remains unclear. In the current study, we found that a natural product extracted from Illicium verum, isodunnianol (IDN), mitigates DOX-induced cardiotoxicity by regulating autophagy and apoptosis both in vitro and in vivo. DOX suppressed protective autophagy and induced apoptosis in H9C2 cardiac myoblasts. Additionally, IDN demonstrated up-regulated autophagy and reduced apoptosis through the activation of the AMPK-ULK1 pathway. In addition, the beneficial effects of IDN on DOX which induced myocardial injury were dependent on AMPK and ULK1 phosphorylation. Similar results were also observed in a DOX-induced cardiotoxicity rat model. The combination of IDN and DOX resulted in decreased apoptosis and inflammatory myocardial fibrosis compared to the DOX mono-treatment group. In summary, our findings provide novel insights into the prevention of DOX-related toxicity by isodunnianol, a food source natural product, warranting further investigation.
Collapse
Affiliation(s)
- Can Chen
- The First Affiliated Hospital, Sichuan Province College Key Laboratory of Structure-Specific Small Molecule Drugs, School of Pharmacy, Chengdu Medical College, Chengdu 610500, China.
| | | | | | | | | | | | | |
Collapse
|
30
|
Bansal N, Adams MJ, Ganatra S, Colan SD, Aggarwal S, Steiner R, Amdani S, Lipshultz ER, Lipshultz SE. Strategies to prevent anthracycline-induced cardiotoxicity in cancer survivors. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2019; 5:18. [PMID: 32154024 PMCID: PMC7048046 DOI: 10.1186/s40959-019-0054-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022]
Abstract
Cancer diagnostics and therapies have improved steadily over the last few decades, markedly increasing life expectancy for patients at all ages. However, conventional and newer anti-neoplastic therapies can cause short- and long-term cardiotoxicity. The clinical implications of this cardiotoxicity become more important with the increasing use of cardiotoxic drugs. The implications are especially serious among patients predisposed to adverse cardiac effects, such as youth, the elderly, those with cardiovascular comorbidities, and those receiving additional chemotherapies or thoracic radiation. However, the optimal strategy for preventing and managing chemotherapy-induced cardiotoxicity remains unknown. The routine use of neurohormonal antagonists for cardioprotection is not currently justified, given the marginal benefits and associated adverse events, particularly with long-term use. The only United States Food and Drug Administration and European Medicines Agency approved treatment for preventing anthracycline-related cardiomyopathy is dexrazoxane. We advocate administering dexrazoxane during cancer treatment to limit the cardiotoxic effects of anthracycline chemotherapy.
Collapse
Affiliation(s)
- Neha Bansal
- Division of Pediatric Cardiology, Children’s Hospital at Montefiore, Bronx, NY USA
| | - M. Jacob Adams
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
- Cardio-Oncology Program, Dana-Farber Cancer Institute / Brigham and Women’s Hospital, Boston, MA USA
| | - Steven D. Colan
- Department of Pediatric Cardiology, Boston Children’s Hospital, Boston, MA USA
| | - Sanjeev Aggarwal
- Division of Pediatric Cardiology, Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI USA
| | | | - Shahnawaz Amdani
- Division of Pediatric Cardiology, Cleveland Clinic Children’s Hospital, Cleveland, OH USA
| | - Emma R. Lipshultz
- Dana-Farber Cancer Institute, Boston, MA USA
- University of Miami Miller School of Medicine, Miami, FL USA
| | - Steven E. Lipshultz
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Oishei Children’s Hospital, 1001 Main Street, Buffalo, NY 14203 USA
- Oishei Children’s Hospital, Buffalo, NY USA
- Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| |
Collapse
|
31
|
Li X, Li Y, Zhang T, Xiong X, Liu N, Pang B, Ruan Y, Gao Y, Shang H, Xing Y. Role of cardioprotective agents on chemotherapy-induced heart failure: A systematic review and network meta-analysis of randomized controlled trials. Pharmacol Res 2019; 151:104577. [PMID: 31790821 DOI: 10.1016/j.phrs.2019.104577] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/07/2019] [Accepted: 11/26/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although previous clinical randomized controlled trials (RCTs) have tested the effect of a variety of cardioprotective agents on cancer therapy-induced cardiotoxicity, the number of included patients was limited, and the results remained controversial. In this study, we aimed to evaluate the preventive or therapeutic effects of cardioprotective agents on heart failure (HF) caused by cardiotoxicity induced by cancer therapy. METHODS We included trials of the following cardioprotective drugs: Angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, beta-blockers, aldosterone antagonists and stains. We extracted the relevant information with predefined data extraction forms, and assessed the risk of bias in randomized controlled trials with the Cochrane risk of bias tool. The primary outcome was the left ventricular ejection fraction of patients after chemotherapy. We used the random-effects model to carry out pair-wise meta-analysis, and then carry out the random-effects network meta-analysis within the Bayesian framework. RESULTS Twenty-two relevant RCTs, including 1 916 patients (79.6 % women) with a mean age of 48.4 years, were included. Based on the evaluation of all drug species from 20 studies (26 comparisons), the analysis found that 4 therapies, aldosterone antagonists (MD, 12.78 [95 % CI, 2.87-22.69] and MD, 13.75 [95 % CI, 2.21-25.30]), ACEIs (MD, 6.79 [95 % CI, 2.11-11.48] and MD, 7.76 [95 % CI, 2.64-12.88]), statin (MD, 8.35 [95 % CI, 1.11-15.59]), and beta-blockers (MD, 4.00 [95 % CI, 0.87-7.14]), had a higher efficacy than placebo and/or control, suggesting an LVEF protective effect of cardioprotective therapy. In the analysis classified by single drug or drug combination, based on 22 studies (31 comparisons), spironolactone (MD, 12.77 [95 % CI, 1.76-23.79] and MD, 14.62 [95 % CI, 1.70-27.55]), a combination of candesartan and carvedilol (MD, 12.40 [95 % CI, 0.99-23.81]), enalapril (MD, 7.35 [95 % CI, 1.16-13.54] and MD, 9.20 [95 % CI, 2.61-15.79]), and statin (MD, 8.36 [95 % CI, 0.36-16.36]) showed significant benefits in protecting left ventricular (LV) systolic function compared with the placebo and/or control. CONCLUSION When classified according to drug type, aldosterone antagonists, ACEIs, statins, and beta-blockers could substantially improve the LV systolic function. In the analysis classified by single drug or drug combination, spironolactone, enalapril, and statin have a significant cardioprotective effect. However, ARBs have no cardioprotective effect and fail to improve the LVEF.
Collapse
Affiliation(s)
- Xinye Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Beijing University of Chinese Medicine, Beijing, China
| | - Yanda Li
- Institute of Basic Research In Clinical Medicine, China Academy Of Chinese Medical Sciences, Beijing, China
| | - Tiansong Zhang
- Jing'an District Center Hospital, Fudan Univetsity, Shanghai, China
| | - Xingjiang Xiong
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nian Liu
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bing Pang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Ruan
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China.
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
32
|
Ryan TD, Border WL, Baker-Smith C, Barac A, Bock MJ, Canobbio MM, Choueiter NF, Chowdhury D, Gambetta KE, Glickstein JS, Kondapalli L, Mital S, Peiris V, Schiff RJ, Spicer RL, Towbin JA, Chen MH. The landscape of cardiovascular care in pediatric cancer patients and survivors: a survey by the ACC Pediatric Cardio-Oncology Work Group. CARDIO-ONCOLOGY 2019; 5:16. [PMID: 32154022 PMCID: PMC7048086 DOI: 10.1186/s40959-019-0051-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
Abstract
Objective To enhance the understanding of cardiovascular care delivery in childhood cancer patients and survivors. Study design A 20-question survey was created by the Pediatric Cardio-oncology Work Group of the American College of Cardiology (ACC) Cardio-oncology Section to assess the care, management, and surveillance tools utilized to manage pediatric/young adult cardio-oncology patients. The survey distribution was a collaborative effort between Cardio-oncology Section and membership of the Adult Congenital and Pediatric Cardiology Section (ACPC) of the ACC. Results Sixty-five individuals, all self-identified as physicians, responded to the survey. Most respondents (n = 58,89%) indicated childhood cancer patients are regularly screened prior to and during cancer therapy at their centers, predominantly by electrocardiogram (75%), standard echocardiogram (58%) and advanced echocardiogram (50%) (i.e. strain, stress echo). Evaluation by a cardiologist prior to/during therapy was reported by only 8(12%) respondents, as compared to post-therapy which was reported by 28 (43%, p < 0.01). The most common indications for referral to cardiology at pediatric centers were abnormal test results (n = 31,48%) and history of chemotherapy exposure (n = 27,42%). Of note, during post-treatment counseling, common cardiovascular risk-factors like blood pressure (31,48%), lipid control (22,34%), obesity & smoking (30,46%) and diet/exercise/weight loss (30,46%) were addressed by fewer respondents than was LV function (72%). Conclusions The survey data demonstrates that pediatric cancer patients are being screened by EKG and/or imaging prior to/during therapy at most centers. Our data, however, highlight the potential for greater involvement of a cardiovascular specialist for pre-treatment evaluation process, and for more systematic cardiac risk factor counseling in posttreatment cancer survivors.
Collapse
Affiliation(s)
- Thomas D Ryan
- Heart Institute, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, 3333 Burnet Ave, MLC 2003, Cincinnati, OH 45229 USA
| | - William L Border
- 2Children's Sibley Heart Center, Children's Healthcare of Atlanta, Atlanta, GA USA
| | - Carissa Baker-Smith
- 3Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD USA
| | - Ana Barac
- 4MedStar Heart and Vascular Institute, Georgetown University, Washington, DC, USA
| | - Matthew J Bock
- 5Loma Linda University Children's Hospital, Loma Linda, CA USA
| | - Mary M Canobbio
- 6Ahmanson/UCLA ACHD Center, UCLA Health, Los Angeles, CA USA
| | - Nadine F Choueiter
- 7The Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY USA
| | | | | | - Julie S Glickstein
- 10Morgan Stanley Children's Hospital of New York, Columbia University Vagelos College of Physicians and Surgeons, New York, NY USA
| | - Lavanya Kondapalli
- 11UC Health Heart & Vascular Center - Anschutz, University of Colorado School of Medicine, Aurora, CO USA
| | - Seema Mital
- 12The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Vasum Peiris
- Department of Health and Human Services, Food and Drug Administration Center for Devices and Radiological Health, Children's National Health System and the George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Russell J Schiff
- Cohen Children's Medical Center of New York - Northwell Health, New Hyde Park and Huntington, NY USA
| | - Robert L Spicer
- 15Children's Hospital and Medical Center, University of Nebraska Medical Center, Omaha, NE USA
| | - Jeffrey A Towbin
- Le Bonheur Children's Hospital, The University of Tennessee Health Science Center, St. Jude Children's Research Hospital, Memphis, TN USA
| | - Ming Hui Chen
- Departments of Cardiology and Pediatrics, Boston Children's Hospital, Dana-Farber Cancer Institute/Harvard Cancer Center, and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115 USA
| |
Collapse
|
33
|
Rozga-Wijas K, Sierant M. Daunorubicin-silsesquioxane conjugates (POSS-DAU) for theranostic drug delivery system: Characterization, biocompatibility and drug release study. REACT FUNCT POLYM 2019. [DOI: 10.1016/j.reactfunctpolym.2019.104332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
34
|
Michel L, Rassaf T, Totzeck M. Biomarkers for the detection of apparent and subclinical cancer therapy-related cardiotoxicity. J Thorac Dis 2018; 10:S4282-S4295. [PMID: 30701097 DOI: 10.21037/jtd.2018.08.15] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Progress in cancer therapy over the past decades improved long-term survival but increased cancer therapy-related cardiotoxicity. Many novel treatment options have been implemented with yet incompletely characterized cardiovascular side effects including heart failure, coronary artery disease, arrhythmias, valvular disease, venous thromboembolism and myocarditis. Diagnosis of potential cardiotoxic side effects is essential for an optimal treatment but remains challenging. Cardiac biomarkers troponin and brain natriuretic peptide/N-terminal proBNP (BNP/NT-proBNP) have been extensively studied in heart failure and acute coronary syndromes. Emerging evidence implicates a significant role in the detection of cardiotoxicity and guidance of therapy in cancer patients. Elevated troponin or BNP/NT-proBNP levels were associated with increased all-cause mortality in cancer patients and have been shown to predict manifest heart failure. BNP/NT-proBNP may be useful for the prediction of cancer therapy-related heart failure and response to heart failure therapy in adult and pediatric cancer patients while troponin can indicate acute myocardial infarction in patients with cancer therapy-related risk for coronary artery disease. Furthermore, troponin may be used for the identification of immune checkpoint inhibitor-related myocarditis with very high sensitivity. Finally, even D-dimer levels have been shown to improve risk stratification and diagnosis in cancer-associated venous thromboembolism. This review aims to summarize the current knowledge about biomarkers in cancer therapy-related cardiotoxicity. We also outline possible clinical recommendations for the detection and treatment of subclinical and clinically apparent cardiotoxic effects using biomarkers.
Collapse
Affiliation(s)
- Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| |
Collapse
|