51
|
Barbosa RR, Bourguignon TB, Torres LD, Arruda LS, Jacques TDM, Serpa RG, Calil ODA, Barbosa LFM. Anthracycline-associated cardiotoxicity in adults: systematic review on the cardioprotective role of beta-blockers. ACTA ACUST UNITED AC 2019; 64:745-754. [PMID: 30673046 DOI: 10.1590/1806-9282.64.08.745] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 01/07/2018] [Indexed: 01/05/2023]
Abstract
OBJECTIVES This study aimed at assessing the role of beta-blockers on preventing anthracycline-induced cardiotoxicity in adults. METHODS A systematic review was performed on electronic databases, including relevant studies that analysed beta-blockers as cardioprotective agents before the use of anthracyclines by adult oncologic patients. RESULTS After application of eligibility and selection criteria, eight articles were considered as high quality, complying with the proposed theme; all eight clinical trials, four of them placebo-controlled, with a total number of 655 patients included. From this sample, 281 (42.9%) used beta-blocker as intervention, and carvedilol was the most frequent (167 patients - 25.5%). Six studies were considered positive regarding the cardioprotection role played by beta-blockers, although only four demonstrated significant difference on left ventricle ejection fraction after chemotherapy on groups that used beta-blockers compared to control groups. Carvedilol and nebivolol, but not metoprolol, had positive results regarding cardioprotection. Other beta-blockers were not analysed in the selected studies. CONCLUSIONS Despite the potential cardioprotective effect of beta-blockers, as demonstrated in small and unicentric clinical trials, its routine use on prevention of anthracycline-associated cardiotoxicity demands greater scientific evidence.
Collapse
Affiliation(s)
| | | | - Luíza Dias Torres
- . School of Sciences of Santa Casa de Misericórdia de Vitoria, Vitória, ES, Brasil
| | | | | | - Renato Giestas Serpa
- . School of Sciences of Santa Casa de Misericórdia de Vitoria, Vitória, ES, Brasil
| | | | | |
Collapse
|
52
|
Zhan T, Daniyal M, Li J, Mao Y. Preventive use of carvedilol for anthracycline-induced cardiotoxicity: a systematic review and meta-analysis of randomized controlled trials. Herz 2019; 45:1-14. [PMID: 30656389 DOI: 10.1007/s00059-018-4779-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 11/13/2018] [Accepted: 12/09/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Clinical or subclinical cardiotoxicity is a concern for cancer patients receiving anthracycline-based chemotherapy. Carvedilol is promising for preventing anthracycline-induced cardiotoxicity (AIC). This review appraised the preventive effects of carvedilol against AIC based on randomized controlled trials (RCTs). METHODS The Cochrane Collaboration Central Register of Controlled Trials, PubMed, and Embase databases were searched from inception to March 27, 2018. RCTs using carvedilol for the prevention of AIC were selected. Risk of bias and methodological quality were assessed. Meta-analysis was conducted, when applicable, for the trial endpoints; otherwise the data were analyzed descriptively. RESULTS Nine RCTs comprising 717 patients were selected. The risk of bias was unclear and the methodological quality differed substantially. Data pooling of five eligible studies indicated no decreased mortality in patients receiving carvedilol (risk difference = -0.02, 95% CI: -0.07-0.04, p = 0.57, I2 = 44%). The impact on the incidence of left ventricular systolic dysfunction (LVSD) was inconsistently reported but meta-analysis was not applicable due to discordant LVSD definitions. Data pooling of eight studies and a subgroup analysis indicated a higher left ventricular ejection fraction (LVEF) with substantial heterogeneity in the carvedilol group (mean difference [MD] = 5.23, 95% CI: 2.20-8.27, p = 0.0007, I2 = 95%, and MD = 4.65, 95% CI: 0.67-8.64, p = 0.02, I2 = 90%, respectively). Further analysis of echocardiographic parameters and biomarkers showed weak evidence of improvement in diastolic function and troponin I level by carvedilol administration. CONCLUSION Preventive use of carvedilol in patients undergoing anthracycline-based chemotherapy may be associated with a reduced incidence of LVSD, higher LVEF value, better diastolic function, and lower troponin I level. RCTs with larger sample size and longer follow-up are needed to verify these findings.
Collapse
Affiliation(s)
- T Zhan
- Hunan University of Chinese Medicine, 300 Xueshi Rd., Yuelu District, 410208, Changsha, Hunan, China
- Department of Integrated TCM and Western Medicine, The First Hospital of Changsha, 410005, Changsha, Hunan, China
| | - M Daniyal
- TCM and Ethnomedicine Innovation & Development Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, 410208, Changsha, Hunan, China
| | - J Li
- Hunan University of Chinese Medicine, 300 Xueshi Rd., Yuelu District, 410208, Changsha, Hunan, China.
| | - Y Mao
- Department of Geriatric Medicine, The Second Affiliated Hospital, Hunan University of Chinese Medicine, 233 North Cai'e Rd., Kaifu District, 410005, Changsha, Hunan, China.
| |
Collapse
|
53
|
Kheiri B, Abdalla A, Osman M, Haykal T, Chahine A, Ahmed S, Osman K, Hassan M, Bachuwa G, Bhatt DL. Meta-Analysis of Carvedilol for the Prevention of Anthracycline-Induced Cardiotoxicity. Am J Cardiol 2018; 122:1959-1964. [PMID: 30292333 DOI: 10.1016/j.amjcard.2018.08.039] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/07/2018] [Accepted: 08/13/2018] [Indexed: 01/04/2023]
Abstract
Anthracycline is a commonly prescribed antineoplastic agent. As a consequence of the growing number of cancer survivors, the incidence of anthracycline-induced cardiotoxicity is increasing. However, the optimal primary preventive strategy is lacking. Therefore, we conducted a meta-analysis of all randomized controlled trials to evaluate the efficacy of carvedilol for the primary prevention of anthracycline-induced cardiotoxicity. A comprehensive search of electronic databases was conducted. The primary and secondary outcomes were the occurrence of low left ventricular ejection fraction, and the absolute change in left ventricular ejection fraction (LVEF), respectively. We calculated the odds ratios for the primary outcome and the weighted mean differences for the secondary outcomes using a random-effects model. We included 8 randomized controlled trials (633 total patients). Our results showed significantly reduced rates of low LVEF favoring the carvedilol group (3.2% vs 5.8%; odds ratios: 0.42; 95% confidence interval: 0.18 to 0.99; p = 0.05). Furthermore, there were significantly smaller reductions in LVEF in carvedilol-treated patients than in placebo-treated patients (mean differences: 2.41%; 95% confidence interval: 0.01 to 4.81; p = 0.05). In conclusion, prophylactic administration of carvedilol in anthracycline-treated cancer patients may reduce the early onset of left ventricular dysfunction compared with placebo.
Collapse
Affiliation(s)
- Babikir Kheiri
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Ahmed Abdalla
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Mohammed Osman
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Tarek Haykal
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Adam Chahine
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Sahar Ahmed
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Khansa Osman
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Mustafa Hassan
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Ghassan Bachuwa
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart & Vascular Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
54
|
National Heart Foundation of Australia and Cardiac Society of Australia and New Zealand: Guidelines for the Prevention, Detection, and Management of Heart Failure in Australia 2018. Heart Lung Circ 2018; 27:1123-1208. [DOI: 10.1016/j.hlc.2018.06.1042] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
55
|
Abedinzadeh M, Neamatzadeh H, Jafari M, Forat-Yazdi M, Nasiri R, Farahnak S, Foroughi E, Zare-Shehneh M. Association of Interleukin-10 -1082A>G (rs1800896) Polymorphism with Predisposition to Breast Cancer: a Meta-Analysis based on 17 Case-Control Studies. Rev Assoc Med Bras (1992) 2018; 64:756-764. [DOI: 10.1590/1806-9282.64.08.756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/07/2018] [Indexed: 11/22/2022] Open
Abstract
SUMMARY INTRODUCTION The association between the between IL-10 -1082A>G (rs1800896) polymorphism and breast cancer has been evaluated by several number case-control studies. However, these studies might be underpowered to reveal the true association. OBJECTIVE We have performed a comprehensive meta-analysis to investigate the association IL-10 -1082A>G polymorphism and breast cancer. MATERIALS AND METHODS A systematic literature search was conducted using PubMed, Google Scholar, and Web of Science up to September 20, 2017. Data was analysed with CMA software to identify the strength of the association by pooled odds ratios (ORs) with corresponding 95% confidence intervals (CIs). RESULTS A total of 17 case-control studies involving 3275 cases and 3416 controls obtained from database searches were examined. Overall, there was no significant association between IL-10 -1082A>G polymorphism and breast cancer risk under all genetic models. No significant publication bias was found for the five genetic models (G vs. A OR = 1.184, 95% CI = 0.895-1.180, p= 0.230; GG vs. AA: OR = 1.430, 95% CI = 0.927-2.204, p= 0.106; GA vs. AA: OR = 0.966, 95% CI = 0.765-1.221, p= 0.774; GG+GA vs. AA: OR = 0.957, 95% CI = 0.697-1.314, p= 0.786; and GG vs. GA+AA: OR = 1.221, 95% CI = 0.981-1.518, p= 0.073). Moreover, there was no significant association between the IL-10 -1082A>G polymorphism and breast cancer risk by ethnicity. CONCLUSION Our findings indicated that IL-10 -1082A>G (rs1800896) polymorphism might not be a risk factor for the development of breast cancer.
Collapse
|
56
|
Avila MS, Ayub-Ferreira SM, de Barros Wanderley MR, das Dores Cruz F, Gonçalves Brandão SM, Rigaud VOC, Higuchi-Dos-Santos MH, Hajjar LA, Kalil Filho R, Hoff PM, Sahade M, Ferrari MSM, de Paula Costa RL, Mano MS, Bittencourt Viana Cruz CB, Abduch MC, Lofrano Alves MS, Guimaraes GV, Issa VS, Bittencourt MS, Bocchi EA. Carvedilol for Prevention of Chemotherapy-Related Cardiotoxicity: The CECCY Trial. J Am Coll Cardiol 2018. [PMID: 29540327 DOI: 10.1016/j.jacc.2018.02.049] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Anthracycline (ANT) chemotherapy is associated with cardiotoxicity. Prevention with β-blockers remains controversial. OBJECTIVES This prospective, randomized, double-blind, placebo-controlled study sought to evaluate the role of carvedilol in preventing ANT cardiotoxicity. METHODS The authors randomized 200 patients with HER2-negative breast cancer tumor status and normal left ventricular ejection fraction (LVEF) referred for ANT (240 mg/m2) to receive carvedilol or placebo until chemotherapy completion. The primary endpoint was prevention of a ≥10% reduction in LVEF at 6 months. Secondary outcomes were effects of carvedilol on troponin I, B-type natriuretic peptide, and diastolic dysfunction. RESULTS Primary endpoint occurred in 14 patients (14.5%) in the carvedilol group and 13 patients (13.5%) in the placebo group (p = 1.0). No differences in changes of LVEF or B-type natriuretic peptide were noted between groups. A significant difference existed between groups in troponin I levels over time, with lower levels in the carvedilol group (p = 0.003). Additionally, a lower incidence of diastolic dysfunction was noted in the carvedilol group (p = 0.039). A nonsignificant trend toward a less-pronounced increase in LV end-diastolic diameter during the follow-up was noted in the carvedilol group (44.1 ± 3.64 mm to 45.2 ± 3.2 mm vs. 44.9 ± 3.6 mm to 46.4 ± 4.0 mm; p = 0.057). CONCLUSIONS In this largest clinical trial of β-blockers for prevention of cardiotoxicity under contemporary ANT dosage, the authors noted a 13.5% to 14.5% incidence of cardiotoxicity. In this scenario, carvedilol had no impact on the incidence of early onset of LVEF reduction. However, the use of carvedilol resulted in a significant reduction in troponin levels and diastolic dysfunction. (Carvedilol Effect in Preventing Chemotherapy-Induced Cardiotoxicity [CECCY]; NCT01724450).
Collapse
Affiliation(s)
- Mônica Samuel Avila
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Silvia Moreira Ayub-Ferreira
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mauro Rogerio de Barros Wanderley
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Fatima das Dores Cruz
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sara Michelly Gonçalves Brandão
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vagner Oliveira Carvalho Rigaud
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Ludhmila Abrahão Hajjar
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | - Roberto Kalil Filho
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | - Paulo Marcelo Hoff
- Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | - Marina Sahade
- Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | - Marcela S M Ferrari
- Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | | | - Max Senna Mano
- Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | - Cecilia Beatriz Bittencourt Viana Cruz
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil
| | - Maria Cristina Abduch
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marco Stephan Lofrano Alves
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Guilherme Veiga Guimaraes
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Victor Sarli Issa
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marcio Sommer Bittencourt
- Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo-Universidade de São Paulo, São Paulo, Brazil; Center for Clinical and Epidemiological Research, University Hospital, University of São Paulo, São Paulo, Brazil
| | - Edimar Alcides Bocchi
- Heart Failure Department, Heart Institute (InCor) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
57
|
Gast KC, Viscuse PV, Nowsheen S, Haddad TC, Mutter RW, Wahner Hendrickson AE, Couch FJ, Ruddy KJ. Cardiovascular Concerns in BRCA1 and BRCA2 Mutation Carriers. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018; 20:18. [PMID: 29497862 DOI: 10.1007/s11936-018-0609-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW BRCA1 and BRCA2 mutation carriers can be at increased cardiovascular risk. The goal of this review is to provide information about factors associated with increased cardiovascular risk, methods to prevent cardiovascular toxicities, and recommended screening guidelines. RECENT FINDINGS BRCA1/2 mutation carriers who are diagnosed with cancer are often exposed to chemotherapy, chest radiotherapy, and/or HER2 directed therapies, all of which can be cardiotoxic. In addition, BRCA1/2 carriers often undergo prophylactic salpingoopherectomies, which may also increase cardiovascular risks. Understanding the potential for increased cardiovascular risk in individuals with a BRCA1 or BRCA2 mutation, as well as gold standard practices for prevention, detection, and treatment of cardiac concerns in this population, is important.
Collapse
Affiliation(s)
- Kelly C Gast
- Department of Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN, USA
| | - Paul V Viscuse
- Department of Internal Medicine, Mayo Clinic School of Graduate Medical Education, Rochester, MN, USA
| | - Somaira Nowsheen
- Mayo Clinic Graduate School of Biomedical Sciences, Medical Scientist Training Program, Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Tufia C Haddad
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55095, USA
| | - Robert W Mutter
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Andrea E Wahner Hendrickson
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55095, USA
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kathryn J Ruddy
- Department of Oncology, Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55095, USA.
| |
Collapse
|
58
|
Lee A, Djamgoz MBA. Triple negative breast cancer: Emerging therapeutic modalities and novel combination therapies. Cancer Treat Rev 2017; 62:110-122. [PMID: 29202431 DOI: 10.1016/j.ctrv.2017.11.003] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/11/2022]
Abstract
Triple negative breast cancer (TNBC) is a complex and aggressive subtype of breast cancer which lacks oestrogen receptors, progesterone receptors and HER2 amplification, thereby making it difficult to target therapeutically. In addition, TNBC has the highest rates of metastatic disease and the poorest overall survival of all breast cancer subtypes. Resultantly, development of targeted therapies for TNBC is urgently needed. Recent efforts aimed at molecular characterisation of TNBCs have revealed various emerging therapeutic targets including PARP1, receptor and non-receptor tyrosine kinases, immune-checkpoints, androgen receptor and epigenetic proteins. Key successes include that of the PARP inhibitor, olaparib, which prolonged progression-free survival in a trial of BRCA-mutated breast cancer and for which clinical approval (in this setting) appears imminent. Nevertheless, the heterogeneity of TNBC has limited the clinical benefits of many trialled therapies in 'unselected' patients. Further, drug resistance develops following use of many targeted monotherapies due to upregulation of compensatory signalling pathways. In this review, we evaluate the current status of investigational targeted treatments and present evidence for the role of novel biomarkers and combination therapies in increasing response rates and circumventing drug-induced resistance. Additionally, we discuss promising novel targets in metastatic TNBC identified through preclinical and/or epidemiological studies.
Collapse
Affiliation(s)
- Alice Lee
- Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Mustafa B A Djamgoz
- Neuroscience Solution to Cancer Research Group, Department of Life Sciences, Faculty of Natural Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| |
Collapse
|
59
|
Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, Barker AD, Bastani R, Bhatia S, Bluestone JA, Brawley O, Butte AJ, Coit DG, Davidson NE, Davis M, DePinho RA, Diasio RB, Draetta G, Frazier AL, Futreal A, Gambhir SS, Ganz PA, Garraway L, Gerson S, Gupta S, Heath J, Hoffman RI, Hudis C, Hughes-Halbert C, Ibrahim R, Jadvar H, Kavanagh B, Kittles R, Le QT, Lippman SM, Mankoff D, Mardis ER, Mayer DK, McMasters K, Meropol NJ, Mitchell B, Naredi P, Ornish D, Pawlik TM, Peppercorn J, Pomper MG, Raghavan D, Ritchie C, Schwarz SW, Sullivan R, Wahl R, Wolchok JD, Wong SL, Yung A. Future cancer research priorities in the USA: a Lancet Oncology Commission. Lancet Oncol 2017; 18:e653-e706. [PMID: 29208398 PMCID: PMC6178838 DOI: 10.1016/s1470-2045(17)30698-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022]
Abstract
We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US$2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.
Collapse
Affiliation(s)
| | - Chi Van Dang
- Ludwig Institute for Cancer Research New York, NY; Wistar Institute, Philadelphia, PA, USA.
| | - David B Agus
- University of Southern California, Beverly Hills, CA, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Alan Ashworth
- University of California San Francisco, San Francisco, CA, USA
| | | | - Roshan Bastani
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Sangeeta Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey A Bluestone
- University of California San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Atul J Butte
- University of California San Francisco, San Francisco, CA, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Mark Davis
- California Institute for Technology, Pasadena, CA, USA
| | | | | | - Giulio Draetta
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Lindsay Frazier
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Patricia A Ganz
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Levi Garraway
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA; Eli Lilly and Company, Boston, MA, USA
| | | | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Faculty of Medicine and IHPME, University of Toronto, Toronto, Canada
| | - James Heath
- California Institute for Technology, Pasadena, CA, USA
| | - Ruth I Hoffman
- American Childhood Cancer Organization, Beltsville, MD, USA
| | - Cliff Hudis
- Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Chanita Hughes-Halbert
- Medical University of South Carolina and the Hollings Cancer Center, Charleston, SC, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Hossein Jadvar
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Kavanagh
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| | - Rick Kittles
- College of Medicine, University of Arizona, Tucson, AZ, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | - Scott M Lippman
- University of California San Diego Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - David Mankoff
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Mardis
- The Institute for Genomic Medicine at Nationwide Children's Hospital Columbus, OH, USA; College of Medicine, Ohio State University, Columbus, OH, USA
| | - Deborah K Mayer
- University of North Carolina Lineberger Cancer Center, Chapel Hill, NC, USA
| | - Kelly McMasters
- The Hiram C Polk Jr MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dean Ornish
- University of California San Francisco, San Francisco, CA, USA
| | - Timothy M Pawlik
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | | | - Martin G Pomper
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Raghavan
- Levine Cancer Institute, Carolinas HealthCare, Charlotte, NC, USA
| | | | - Sally W Schwarz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Richard Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Sandra L Wong
- Department of Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alfred Yung
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
60
|
Effect of prophylactic betablocker or ACE inhibitor on cardiac dysfunction & heart failure during anthracycline chemotherapy ± trastuzumab. Breast 2017; 37:64-71. [PMID: 29101824 DOI: 10.1016/j.breast.2017.10.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/24/2017] [Accepted: 10/23/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Administration of anthracycline chemotherapy ± trastuzumab is associated with cardiac dysfunction. We sought to identify whether prophylactic administration of angiotensin converting enzyme (ACE) inhibitor or beta-blocker attenuates subsequent left ventricular dysfunction and heart failure. METHODS Electronic databases were searched for studies examining the effect of beta-blockers or ACE inhibitors on left ventricular ejection fraction (LVEF) and heart failure in patients receiving anthracycline chemotherapy ± trastuzumab. The end-point was change in LVEF or development of heart failure. RESULTS Eight studies (1048 patients) were identified. There was no difference in the change in LVEF (weighted mean difference (MWD) - 4.74 (95% confidence interval (CI): -12.6 to 3.1), p = 0.24) or risk of new heart failure diagnosis (Odds ratio (OR) 0.24 (95% CI: 0.03-1.73), p = 0.16) between patients receiving ACE inhibitor or control. Betablocker use was associated with a significant smaller drop in LVEF compared to control (MWD -3.28 (95% CI: -6.1 to -0.51), p = 0.02) but not in patients who received anthracycline chemotherapy alone (MWD - 3.05 (95% CI -7.22 to 1.12), p = 0.15). There was a significant reduction in new heart failure diagnosis in those receiving beta-blocker compared to those not (OR 0.33 (95% CI: 0.14-0.80), p = 0.01). CONCLUSIONS Prophylactic ACE inhibitor does not attenuate development of LV dysfunction or heart failure in patients receiving anthracycline chemotherapy ± trastuzumab. Beta-blocker attenuated reduction in LVEF and new heart failure diagnosis. However, the effect in patients only receiving anthracycline is less certain. Studies examining the effect on mortality are required.
Collapse
|
61
|
Shahul S, Ramadan H, Mueller A, Nizamuddin J, Nasim R, Lopes Perdigao J, Chinthala S, Tung A, Rana S. Abnormal mid-trimester cardiac strain in women with chronic hypertension predates superimposed preeclampsia. Pregnancy Hypertens 2017; 10:251-255. [PMID: 29111424 DOI: 10.1016/j.preghy.2017.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/23/2017] [Accepted: 10/21/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND Chronic hypertension (cHTN) affects 7% of all pregnancies. We hypothesized that cHTN during pregnancy would be associated with abnormal myocardial strain patterns and adverse perinatal outcomes. METHODS This was a retrospective cohort study of patients seen in a high-risk obstetrics clinic with cHTN. Parturients with a singleton pregnancy who had undergone an echocardiogram as part of routine clinical care were eligible. Clinical and demographic information was collected from medical records. Global peak longitudinal strain (GLS) was measured using automated software from stored echocardiographic images. RESULTS 60 patients were included in this analysis, of which 48 (80.0%) were African American. The median BMI was 40.6, age was 34 years, and the gestational age was 20.4 weeks at the time of the echo and 37.9 weeks at delivery. Thirty-four patients (56.7%) demonstrated abnormal strain, defined as a GLS <= -19%. Patients with abnormal strain were similar in age and BMI to patients with normal cardiac function. When compared to women with normal strain, those with abnormal strain had lower stroke volume (69.0 ml vs 81.5 ml; p = .001) and ejection fraction (49.6% vs 57.5%; p < .0001). Rates of superimposed preeclampsia were higher (38.2% vs 11.5%, p-value = .02) and a higher proportion of patients in the abnormal strain group delivered before 37 weeks (44.1% vs 19.2%; p = .04). CONCLUSION In a population of parturients with cHTN, we found that more than one-half demonstrated subclinical abnormal cardiac function. The presence of abnormal cardiac strain predates superimposed preeclampsia and preterm delivery. Further studies are needed to validate these findings.
Collapse
Affiliation(s)
- Sajid Shahul
- Department of Anesthesia and Critical Care, University of Chicago, IL, United States
| | - Hadi Ramadan
- Department of Obstetrics and Gynecology/Division of Maternal Fetal Medicine, University of Chicago, IL, United States
| | - Ariel Mueller
- Department of Obstetrics and Gynecology/Division of Maternal Fetal Medicine, University of Chicago, IL, United States; Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Junaid Nizamuddin
- Department of Anesthesia and Critical Care, University of Chicago, IL, United States
| | - Rabab Nasim
- Department of Obstetrics and Gynecology/Division of Maternal Fetal Medicine, University of Chicago, IL, United States
| | - Joana Lopes Perdigao
- Department of Obstetrics and Gynecology/Division of Maternal Fetal Medicine, University of Chicago, IL, United States
| | - Sireesha Chinthala
- Department of Obstetrics and Gynecology/Division of Maternal Fetal Medicine, University of Chicago, IL, United States
| | - Avery Tung
- Department of Anesthesia and Critical Care, University of Chicago, IL, United States
| | - Sarosh Rana
- Department of Obstetrics and Gynecology/Division of Maternal Fetal Medicine, University of Chicago, IL, United States.
| |
Collapse
|
62
|
Zhu C, Wang Y, Liu H, Mu H, Lu Y, Zhang J, Huang J. Oral administration of Ginsenoside Rg1 prevents cardiac toxicity induced by doxorubicin in mice through anti-apoptosis. Oncotarget 2017; 8:83792-83801. [PMID: 29137383 PMCID: PMC5663555 DOI: 10.18632/oncotarget.19698] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/28/2017] [Indexed: 11/25/2022] Open
Abstract
Although Ginsenoside Rg1 has been reported to have protective cardiac effects, its effects on cardiac toxicity induced by doxorubicin needs to be studied. The present study investigated the effects of oral administration of Rg1 on the heart in mice treated with doxorubicin and found improved fractional shortening and ejection fraction of the heart and decreased cardiac apoptosis in mice treated with doxorubicin. The underlying mechanisms include increased phosphorylation of Akt and Erk by Rg1, increased ratio of Bcl-2 and Bax, and decreased release of cytochrome c from mitochondria, thereby protecting the heart from doxorubicin-induced apoptosis. This phenotype suggested that the oral administration of Rg1 may be a potential method preventing the cardiac toxicity caused by doxorubicin in clinical practice.
Collapse
Affiliation(s)
- Chen Zhu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.,Graduated School of Jinzhou Medical University, Jinzhou, China
| | - Yi Wang
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hua Liu
- Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Haiman Mu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yue Lu
- Graduated School of Jinzhou Medical University, Jinzhou, China
| | - Jiayi Zhang
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jianhua Huang
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
63
|
Reddy P, Shenoy C, Blaes AH. Cardio-oncology in the older adult. J Geriatr Oncol 2017; 8:308-314. [PMID: 28499724 PMCID: PMC5776715 DOI: 10.1016/j.jgo.2017.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/21/2017] [Accepted: 04/20/2017] [Indexed: 12/28/2022]
Abstract
Heart disease and cancer are the leading causes of death in older adults. Many first-line cancer treatments have the potential for cardiotoxicity. Age-related risk factors, pre-existing cardiac disease, and a high prevalence of comorbidities are reasons for increased cardiotoxicity in older adults. Concerns regarding cardiotoxicity may lead to frailty bias and undertreatment, resulting in suboptimal outcomes. There is an urgent need for geriatric-specific evidence and guidelines to help tailor care for this vulnerable group. A multi-disciplinary approach based on close collaboration between oncologists, cardiologists, and geriatricians, among other specialist clinicians is essential.
Collapse
Affiliation(s)
- Prajwal Reddy
- Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN, USA.
| | - Chetan Shenoy
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Anne H Blaes
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN, USA
| |
Collapse
|
64
|
Abstract
Anthracycline chemotherapy maintains a prominent role in treating many forms of cancer. Cardiotoxic side effects limit their dosing and improved cancer outcomes expose the cancer survivor to increased cardiovascular morbidity and mortality. The basic mechanisms of cardiotoxicity may involve direct pathways for reactive oxygen species generation and topoisomerase 2 as well as other indirect pathways. Cardioprotective treatments are few and those that have been examined include renin angiotensin system blockade, beta blockers, or the iron chelator dexrazoxane. New treatments exploiting the ErbB or other novel pro-survival pathways, such as conditioning, are on the cardioprotection horizon. Even in the forthcoming era of targeted cancer therapies, the substantial proportion of today's anthracycline-treated cancer patients may become tomorrow's cardiac patient.
Collapse
Affiliation(s)
- John V McGowan
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Robin Chung
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Angshuman Maulik
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Izabela Piotrowska
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - J Malcolm Walker
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, WC1E 6HX, UK.
| |
Collapse
|
65
|
Hang P, Zhao J, Sun L, Li M, Han Y, Du Z, Li Y. Brain-derived neurotrophic factor attenuates doxorubicin-induced cardiac dysfunction through activating Akt signalling in rats. J Cell Mol Med 2016; 21:685-696. [PMID: 28098423 PMCID: PMC5345637 DOI: 10.1111/jcmm.13012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/18/2016] [Indexed: 12/21/2022] Open
Abstract
The clinical application of doxorubicin (Dox) is limited by its adverse effect of cardiotoxicity. Previous studies have suggested the cardioprotective effect of brain‐derived neurotrophic factor (BDNF). We hypothesize that BDNF could protect against Dox‐induced cardiotoxicity. Sprague Dawley rats were injected with Dox (2.5 mg/kg, 3 times/week, i.p.), in the presence or absence of recombinant BDNF (0.4 μg/kg, i.v.) for 2 weeks. H9c2 cells were treated with Dox (1 μM) and/or BDNF (400 ng/ml) for 24 hrs. Functional roles of BDNF against Dox‐induced cardiac injury were examined both in vivo and in vitro. Protein level of BDNF was reduced in Dox‐treated rat ventricles, whereas BDNF and its receptor tropomyosin‐related kinase B (TrkB) were markedly up‐regulated after BDNF administration. Brain‐derived neurotrophic factor significantly inhibited Dox‐induced cardiomyocyte apoptosis, oxidative stress and cardiac dysfunction in rats. Meanwhile, BDNF increased cell viability, inhibited apoptosis and DNA damage of Dox‐treated H9c2 cells. Investigations of the underlying mechanisms revealed that BDNF activated Akt and preserved phosphorylation of mammalian target of rapamycin and Bad without affecting p38 mitogen‐activated protein kinase and extracellular regulated protein kinase pathways. Furthermore, the beneficial effect of BDNF was abolished by BDNF scavenger TrkB‐Fc or Akt inhibitor. In conclusion, our findings reveal a potent protective role of BDNF against Dox‐induced cardiotoxicity by activating Akt signalling, which may facilitate the safe use of Dox in cancer treatment.
Collapse
Affiliation(s)
- Pengzhou Hang
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin, China.,Heilongjiang Academy of Medical Science, Harbin, China
| | - Jing Zhao
- Department of Cardiology, The First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin, China
| | - Li Sun
- Department of Cardiology, The First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin, China
| | - Minghui Li
- Department of Cardiology, The First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin, China
| | - Yu Han
- Department of Cardiology, The First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin, China
| | - Zhimin Du
- Institute of Clinical Pharmacology, The Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin, China
| | - Yue Li
- Heilongjiang Academy of Medical Science, Harbin, China.,Department of Cardiology, The First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin, China
| |
Collapse
|
66
|
Saddic LA, Muehlschlegel JD. Sarco"MiR" friend or foe: a perspective on the mechanisms of doxorubicin-induced cardiomyopathy. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:203. [PMID: 27294099 DOI: 10.21037/atm.2016.05.30] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Anthracyclines are a class of chemotherapeutics used to treat a variety of human cancers including both solid tumors such as breast, ovarian, and lung, as well as malignancies of the blood including leukemia and lymphoma. Despite being extremely effective anti-cancer agents, the application of these drugs is offset by side effects, most notably cardiotoxicity. Many patients treated with doxorubicin (DOX), one of the most common anthracyclines used in oncology, will develop radiographic signs and/or symptoms of cardiomyopathy. Since more and more patients treated with these drugs are surviving their malignancies and manifesting with heart disease, there is particular interest in understanding the mechanisms of anthracycline-induced injury and developing ways to prevent and treat its most feared complication, heart failure. MicroRNAs (miRNAs) are small noncoding RNAs that regulate the expression of mRNAs. Since miRNAs can regulate many mRNAs in a single network they tend to play a crucial role in the pathogenesis of several diseases, including heart failure. Here we present a perspective on a recent work by Roca-Alonso and colleagues who demonstrate a cardioprotective function of the miR-30 family members following DOX-induced cardiac injury. They provide evidence for direct targeting of these miRNAs on key elements of the β-adrenergic pathway and further show that this interaction regulates cardiac function and apoptosis. These experiments deliver fresh insights into the biology of toxin-induced cardiomyopathy and suggest the potential for novel therapeutic targets.
Collapse
Affiliation(s)
- Louis A Saddic
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jochen D Muehlschlegel
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
67
|
Cardioprotective Potentials of Plant-Derived Small Molecules against Doxorubicin Associated Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5724973. [PMID: 27313831 PMCID: PMC4893565 DOI: 10.1155/2016/5724973] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/02/2016] [Accepted: 04/20/2016] [Indexed: 12/17/2022]
Abstract
Doxorubicin (DOX) is a potent and widely used anthracycline antibiotic for the treatment of several malignancies. Unfortunately, the clinical utility of DOX is often restricted due to the elicitation of organ toxicity. Particularly, the increased risk for the development of dilated cardiomyopathy by DOX among the cancer survivors warrants major attention from the physicians as well as researchers to develop adjuvant agents to neutralize the noxious effects of DOX on the healthy myocardium. Despite these pitfalls, the use of traditional cytotoxic drugs continues to be the mainstay treatment for several types of cancer. Recently, phytochemicals have gained attention for their anticancer, chemopreventive, and cardioprotective activities. The ideal cardioprotective agents should not compromise the clinical efficacy of DOX and should be devoid of cumulative or irreversible toxicity on the naïve tissues. Furthermore, adjuvants possessing synergistic anticancer activity and quelling of chemoresistance would significantly enhance the clinical utility in combating DOX-induced cardiotoxicity. The present review renders an overview of cardioprotective effects of plant-derived small molecules and their purported mechanisms against DOX-induced cardiotoxicity. Phytochemicals serve as the reservoirs of pharmacophore which can be utilized as templates for developing safe and potential novel cardioprotective agents in combating DOX-induced cardiotoxicity.
Collapse
|