1
|
Singh R, Patel K, Xu H, Adeniyi A, Upshaw JN, Van Buren P, Kaufman PA, Dittus K, Landry KK. Cardio-Oncology and Breast Cancer Therapies. Curr Treat Options Oncol 2025:10.1007/s11864-025-01311-x. [PMID: 40257669 DOI: 10.1007/s11864-025-01311-x] [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: 03/17/2025] [Indexed: 04/22/2025]
Abstract
OPINION STATEMENT Assessing cardiac risk prior to initiating breast cancer treatment, monitoring cardiac function during treatment, and implementing appropriate follow-up strategies are essential components of managing cardiotoxicity in breast cancer patients. A comprehensive cardiovascular evaluation should be conducted before treatment, including a detailed medical history, physical examination, and baseline cardiac imaging. Risk stratification tools can aid in determining the individual patient's risk profile. Close monitoring of cardiac function, including regular assessment of left ventricular ejection fraction (LVEF) and monitoring for signs and symptoms of cardiac dysfunction, is crucial during treatment. Prompt action should be taken if an adverse cardiovascular event is detected, including considering discontinuing or modifying the treatment regimen. Appropriate follow-up care is essential to monitor for long-term cardiac effects and optimize cardiovascular health in breast cancer survivors. Regular cardiovascular assessments, lifestyle modifications, and collaboration between healthcare professionals are important in managing cardiotoxicity effectively.
Collapse
Affiliation(s)
- Rohit Singh
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Krina Patel
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Haze Xu
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Aderonke Adeniyi
- Division of Cardiology, Department of Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Jenica N Upshaw
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Peter Van Buren
- Division of Cardiology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Peter A Kaufman
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- University of Vermont Cancer Center, Burlington, VT, USA
| | - Kim Dittus
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Kara K Landry
- Division of Hematology/Oncology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- University of Vermont Cancer Center, Burlington, VT, USA.
| |
Collapse
|
2
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
3
|
Ryan TD, Bates JE, Kinahan KE, Leger KJ, Mulrooney DA, Narayan HK, Ness K, Okwuosa TM, Rainusso NC, Steinberger J, Armenian SH. Cardiovascular Toxicity in Patients Treated for Childhood Cancer: A Scientific Statement From the American Heart Association. Circulation 2025; 151:e926-e943. [PMID: 40104841 DOI: 10.1161/cir.0000000000001308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
The field of cardio-oncology has expanded over the past 2 decades to address the ever-increasing issues related to cardiovascular disease in patients with cancer and survivors. There is increasing recognition that nearly all cancer treatments pose some short- or long-term risk for development of cardiovascular disease and that pediatric patients with cancer may be especially vulnerable to cardiovascular disease because of young age at treatment and expected long life span afterward. Anthracycline chemotherapy and chest-directed radiotherapy are the most well-studied cardiotoxic therapies, and dose reduction, use of cardioprotection for anthracyclines, and modern radiotherapy approaches have contributed to improved cardiovascular outcomes for survivors. Newer treatments such as small-molecule inhibitors, antibody-based cytotoxic therapy, and immunotherapy have expanded options for previously difficult-to-treat cancers but have also revealed new cardiotoxic profiles. Application of effective surveillance strategies in patients with cancer and survivors has been a focus of practitioners and researchers, whereas the prevention and treatment of extant cardiovascular disease is still developing. Incorporation of new strategies in an equitable manner and appropriate transition from pediatric to adult care will greatly influence long-term health-related outcomes in the growing population of childhood cancer survivors at risk for cardiovascular disease.
Collapse
|
4
|
Anderson E, Choi Y, Buchsbaum RJ, Klein A, Ky B, Landsburg D, Durani U, Ruddy KJ, Yu AF, Leong D, Asnani A, Neilan TG, Ganatra S, Bloom M, Barac A, Yang EH, Deswal A, Cheng RK, Weiss M, Evens AM, Kahl B, Friedberg JW, Parsons SK, Upshaw JN. Hematology-oncology provider perspectives regarding lymphoma treatment and cardioprotective strategies in patients with lymphoma at high risk for heart failure. Leuk Lymphoma 2025:1-10. [PMID: 40195874 DOI: 10.1080/10428194.2025.2484367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
The optimal treatment of patients with diffuse large B-cell lymphoma (DLBCL) or Hodgkin lymphoma (HL) with preexisting cardiomyopathy is uncertain. An anonymous, electronic survey was distributed by e-mail to three US lymphoma cooperative groups, two community hospitals, and twelve academic medical systems, and distributed at one international lymphoma meeting. Fifty hematology-oncology providers caring for patients with lymphoma were included. In response to a vignette of a 67-yo with Stage III DLBCL with LVEF of 40-45%, 15 (30%) would use non-anthracycline regimens, 13 (26%) R-CHOP with liposomal doxorubicin instead of doxorubicin, 11 (22%) R-CHOP without modification and 6 (12%) R-CHOP with a continuous doxorubicin infusion. In a second vignette of a patient with HL in remission after frontline treatment with doxorubicin cumulative dose 300 mg/m2, 16 (32%) would order an echocardiogram after treatment. There was substantial variability in preferred treatment regimens with preexisting cardiomyopathy and in cardiac monitoring after anthracycline.
Collapse
Affiliation(s)
- Emily Anderson
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Yun Choi
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
| | | | - Andreas Klein
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Landsburg
- Division of Hematology-Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Kathryn J Ruddy
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Anthony F Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Darryl Leong
- Department of Medicine and the Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tomas G Neilan
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Sarju Ganatra
- Division of Cardiology, Lahey Clinic, Burlington, MA, USA
| | - Michelle Bloom
- Division of Cardiology, NYU Langone Health, New York, NY, USA
| | - Ana Barac
- Division of Cardiology, Inova Schar Heart and Vascular, Fairfax, VA, USA
| | - Eric H Yang
- Division of Cardiology, UCLA Cardio-Oncology Program, University of California, Los Angeles Medical Center, Los Angeles, CA, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard K Cheng
- Division of Cardiology, University of Washington Medical Center, Seattle, WA, USA
| | | | | | - Brad Kahl
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Susan K Parsons
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
- Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Boston, MA, USA
| | - Jenica N Upshaw
- Tufts Medical Center, Institute for Clinical Research and Health Policy Studies, Boston, MA, USA
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
5
|
Safiullina AA, Potievskaya VI, Vitsenya MV, Cheremisina IA. A Multicenter Prospective Observational Study to Examine the Experience of Using Phosphocreatine in Combination Therapy for Heart Failure Caused by Cancer Treatment. Rationale and Design of the Study. KARDIOLOGIIA 2025; 65:21-25. [PMID: 40195775 DOI: 10.18087/cardio.2025.3.n2870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/04/2025] [Indexed: 04/09/2025]
Abstract
Enhanced cancer treatment efficacy has resulted in a significant increase in the number of cancer survivors after the cure of malignant tumors. However, cardiovascular morbidity, including chronic heart failure, has become the leading cause of death and decreased life expectancy among cancer survivors. This is due, in particular, to the cardiotoxic effects of anticancer drugs and associated factors. Cardioprotective approaches aim to reduce the incidence and severity of cardiotoxicity through the use of cardioprotective agents (e.g., dexrazoxane), liposomal drug delivery systems (e.g., liposomal doxorubicin), and optimization of drug administration schedules. Reducing the cardiotoxicity of cancer treatments is a clinically important goal. Phosphocreatine-based therapy represents a potentially valuable new strategy in this area. In this regard, the study "Multicenter prospective observational study to investigate the experience of using phosphocreatine in combination therapy for heart failure caused by cancer treatment" was initiated. This publication presents the protocol of the observational non-interventional NEOCARD study.
Collapse
Affiliation(s)
- A A Safiullina
- Chazov National Medical Research Center of Cardiology, Moscow
| | | | - M V Vitsenya
- Chazov National Medical Research Center of Cardiology, Moscow
| | | |
Collapse
|
6
|
Im C, Hasan H, Stene E, Monick S, Rader RK, Sheade J, Wolfe H, Lu Z, Spector LG, McDonald AJ, Nolan V, Arnold MA, Conces MR, Moskowitz CS, Henderson TO, Robison LL, Armstrong GT, Yasui Y, Nanda R, Oeffinger KC, Neglia JP, Blaes A, Turcotte LM. Treatment, toxicity, and mortality after subsequent breast cancer in female survivors of childhood cancer. Nat Commun 2025; 16:3088. [PMID: 40164623 PMCID: PMC11958683 DOI: 10.1038/s41467-025-58434-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
Childhood cancer survivors, particularly those who received chest radiotherapy, are at high risk for developing subsequent breast cancer. Minimizing long-term toxicity risks associated with additional radiotherapy and chemotherapy is a priority, but therapeutic tradeoffs have not been comprehensively characterized and their impact on survival is unknown. In this study, 431 female childhood cancer survivors with subsequent breast cancer from a multicenter retrospective cohort study were evaluated. Compared with one-to-one matched females with first primary breast cancer, survivors are as likely to be prescribed guideline-concordant treatment (N = 344 pairs; survivors: 94%, controls: 93%), but more frequently undergo mastectomy (survivors: 81%, controls: 60%) and are less likely to be treated with anthracyclines (survivors: 47%, controls: 66%) or radiotherapy (survivors: 18%, controls: 61%). Despite this, survivors have nearly 3.5-fold (95% CI = 2.17-5.57) greater mortality risk. Here, we show survivors with subsequent breast cancer face excess mortality despite therapeutic tradeoffs and require specialized treatment guidelines.
Collapse
Affiliation(s)
- Cindy Im
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hasibul Hasan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Emily Stene
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sarah Monick
- Department of Hematology/Oncology, Mayo Clinic Arizona, Phoenix, AZ, 85054, USA
| | - Ryan K Rader
- Department of Medicine, University of Kansas, Westwood, KS, 66205, USA
| | - Jori Sheade
- Department of Hematology/Oncology, Northwestern Medicine Lake Forest Hospital, Lurie Cancer Center Affiliate Network, Lake Forest, IL, 60045, USA
| | - Heather Wolfe
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhanni Lu
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Logan G Spector
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Aaron J McDonald
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Vikki Nolan
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael A Arnold
- Department of Pathology and Laboratory Medicine, Children's Hospital Colorado, and Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Miriam R Conces
- Department of Pathology & Laboratory Medicine, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Chaya S Moskowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 10017, USA
| | - Tara O Henderson
- Department of Pediatrics, University of Chicago, Chicago, IL, 60637, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Rita Nanda
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | | | - Joseph P Neglia
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Anne Blaes
- Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Lucie M Turcotte
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
7
|
Zheng H, Zhan H. Dexrazoxane makes doxorubicin-induced heart failure a rare event in sarcoma patients receiving high cumulative doses. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:29. [PMID: 40108682 PMCID: PMC11921489 DOI: 10.1186/s40959-025-00323-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025]
Abstract
Doxorubicin remains a cornerstone in sarcoma treatment, but its dose-dependent cardiotoxicity limits its clinical use and therapeutic potential. Dexrazoxane, the only FDA-approved cardioprotective agent, has demonstrated substantial efficacy in preventing doxorubicin-induced cardiotoxicity. However, despite its proven benefits, dexrazoxane remains underutilized not only in clinical practice but also in contemporary trials. This review examines the role of dexrazoxane in recent oncology trials involving sarcoma patients treated with high cumulative doses of doxorubicin. The LMS 04 trial, a contemporary phase 3 sarcoma trial in which dexrazoxane use was prohibited, reported a 5.4% heart failure incidence at cumulative doxorubicin doses of 360-450 mg/m². In contrast, the trials, where dexrazoxane was used early or upfront, demonstrated rare heart failure incidences even at cumulative doses exceeding 600 mg/m², which is well beyond the conventional maximal limit. Additionally, dexrazoxane enables the safe administration of cumulative doxorubicin doses exceeding 1000 mg/m² without increasing cardiotoxicity. Concerns about secondary malignancies and reduced anti-tumor efficacy have not been supported by clinical trials and meta-analyses. The routine upfront use of dexrazoxane should be considered with doxorubicin treatment, especially in those requiring high cumulative doses or patients at high risk of cardiotoxicity, as each dose of doxorubicin incrementally contributes to the development of cardiotoxicity. Dexrazoxane not only mitigates cardiotoxicity but also allows for extended doxorubicin dosing, maximizing its therapeutic potential. Awareness and guideline updates are necessary to ensure its broader adoption in clinical practice.
Collapse
Affiliation(s)
- Haoyi Zheng
- The Heart Center, Saint Francis Hospital, 100 Port Washington Blvd, Roslyn, NY, 11576, USA.
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA
- Medical Service, Northport VA Medical Center, Northport, NY, USA
| |
Collapse
|
8
|
Shen C, Wang W, Wei D, Yang X, Jiang C, Sheng Y, Chen Y, Sun J, Li X, Li G, Ye S, Chen J. PCR array analysis reveals a novel expression profile of ferroptosis-related genes in idiopathic pulmonary fibrosis. BMC Pulm Med 2025; 25:98. [PMID: 40022042 PMCID: PMC11869717 DOI: 10.1186/s12890-025-03555-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic, irreversible, and fatal disease characterized by progressive interstitial lung fibrosis. Given its insidious onset and poor outcome, there is an urgent need to elucidate the molecular mechanisms underlying IPF and identify effective therapeutic targets and diagnosis and prognosis biomarkers. Ferroptosis is an iron-dependent form of programmed cell death that occurs as lipid peroxides accumulate. Growing evidence suggests that ferroptosis is important in IPF. METHODS Human ferroptosis PCR array was performed on IPF and control lung tissue. The differentially expressed ferroptosis-related genes (DE-FRGs) were identified, underwent functional enrichment analyses, protein-protein interaction network construction, and potential drug target prediction. The DE-FRGs were validated and their value as diagnostic and prognostic blood biomarkers were evaluated using the Gene Expression Omnibus dataset GSE28042. RESULTS The array identified 13 DE-FRGs. Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that the DE-FRGs were mainly related to iron ion transport, blood microparticles, and oxidoreductase activity, and were involved in porphyrin metabolism, necroptosis, and the p53 signaling pathway in addition to ferroptosis. The 13 DE-FRGs were analyzed using the Drug-Gene Interaction Database to explore novel IPF therapeutic agents, yielding 42 potential drugs. Four DE-FRGs (BBC3, STEAP3, EPRS, SLC39A8) in the peripheral blood of IPF patients from the GSE28042 dataset demonstrated the same expression pattern as that observed in the lung tissue array. The receiver operating characteristic analysis demonstrated that the area under the curve of STEAP3 and EPRS were > 0.75. The survival analysis demonstrated that STEAP3 and EPRS were significantly different between the IPF and control groups. CONCLUSIONS The FRG expression profiles in IPF and control lung tissue were characterized. The findings provided valuable ideas to elucidate the role of ferroptosis in IPF and aided the identification of novel IPF therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Chenyou Shen
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Wei Wang
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Dong Wei
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Xusheng Yang
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Cheng Jiang
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Yating Sheng
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Yuan Chen
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Jie Sun
- Department of Scientific Research, The Affiliated Wuxi People's Hospital of Nanjing Medical, University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Xiaoshan Li
- Organ Donation and Transplant Management Office, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, WuxiJiangsu, 214023, China
| | - Guirong Li
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China.
| | - Shugao Ye
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China.
| | - Jingyu Chen
- Lung Transplant Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
9
|
Avagimyan A, Pogosova N, Rizzo M, Sarrafzadegan N. Doxorubicin-induced cardiometabolic disturbances: what can we do? FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2025; 6:1537699. [PMID: 40027901 PMCID: PMC11868090 DOI: 10.3389/fcdhc.2025.1537699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025]
Affiliation(s)
- Ashot Avagimyan
- Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
- Isfahan Cardiovascular Research Centre, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nana Pogosova
- Sciences and Preventive Cardiology, National Medical Research Centre of Cardiology after Acad. E. I. Chazov, Moscow, Russia
- Evidence-Based Medicine Department, Institute of Medicine, Patrice Lumumba Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Manfredi Rizzo
- Cardiometabolic Medicine Department, University of Palermo, Palermo, Italy
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Centre, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
Siaravas KC, Moula AI, Tzourtzos IS, Ballas CE, Katsouras CS. Acute and Chronic Cardiovascular Adverse Events in Patients with Acute Myeloid Leukemia: A Systematic Review. Cancers (Basel) 2025; 17:541. [PMID: 39941907 PMCID: PMC11817404 DOI: 10.3390/cancers17030541] [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: 12/25/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Patients with acute myeloid leukemia (AML) have a higher propensity for adverse cardiovascular outcomes, primarily due to the toxic effects of chemotherapeutic agents. The purpose of this systematic review is to explore the association of acute myeloid leukemia treatment with adverse cardiovascular events. Methods: We systematically screened the literature for studies providing comparative data on cardiovascular toxicities in patients treated for acute myeloid leukemia. After the initial search, 3649 papers were screened and a final total number of 46 were included for the review process. Results: Common chemotherapeutic agents used in AML may cause cardiovascular (CV) toxicities. A plethora of pathophysiological mechanisms are incriminated for these effects. Drug combinations may increase the risk in a synergistic way. In addition, common mutations of AML, personal history of previous cardiovascular disease and impaired heart function carry an increased complication risk. Biomarkers, as well as multimodality imaging, may be used for the early detection of cardiovascular toxicities. Conclusions: Increased risks of CV toxicity and comorbidities are observed among AML patients. With all the available diagnostic modalities, early detection and CV prevention strategies can improve the patient's prognosis and quality of life.
Collapse
Affiliation(s)
- Konstantinos C. Siaravas
- Department of Cardiology, University Hospital of Ioannina, 45500 Ioannina, Greece; (K.C.S.); (I.S.T.)
| | - Amalia I. Moula
- Achilopouleio General Hospital of Volos, 38222 Volos, Greece;
| | - Ioannis S. Tzourtzos
- Department of Cardiology, University Hospital of Ioannina, 45500 Ioannina, Greece; (K.C.S.); (I.S.T.)
| | - Christos E. Ballas
- Department of Thoracic Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Christos S. Katsouras
- Department of Cardiology, University Hospital of Ioannina, 45500 Ioannina, Greece; (K.C.S.); (I.S.T.)
| |
Collapse
|
11
|
Liu X, Li Z. The role and mechanism of epigenetics in anticancer drug-induced cardiotoxicity. Basic Res Cardiol 2025; 120:11-24. [PMID: 38724618 DOI: 10.1007/s00395-024-01054-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is the main factor contributing to the global burden of diseases, and the cardiotoxicity caused by anticancer drugs is an essential component that cannot be ignored. With the development of anticancer drugs, the survival period of cancer patients is prolonged; however, the cardiotoxicity caused by anticancer drugs is becoming increasingly prominent. Currently, cardiovascular disease has emerged as the second leading cause of mortality among long-term cancer survivors. Anticancer drug-induced cardiotoxicity has become a frontier and hot topic. The discovery of epigenetics has given the possibility of environmental changes in gene expression, protein synthesis, and traits. It has been found that epigenetics plays a pivotal role in promoting cardiovascular diseases, such as heart failure, coronary heart disease, and hypertension. In recent years, increasing studies have underscored the crucial roles played by epigenetics in anticancer drug-induced cardiotoxicity. Here, we provide a comprehensive overview of the role and mechanisms of epigenetics in anticancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xuening Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zijian Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
12
|
Moreno-Arciniegas A, Cádiz L, Galán-Arriola C, Clemente-Moragón A, Ibáñez B. Cardioprotection strategies for anthracycline cardiotoxicity. Basic Res Cardiol 2025; 120:71-90. [PMID: 39249555 PMCID: PMC11790697 DOI: 10.1007/s00395-024-01078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Thanks to the fantastic progress in cancer therapy options, there is a growing population of cancer survivors. This success has resulted in a need to focus much effort into improving the quality of life of this population. Cancer and cardiovascular disease share many common risk factors and have an interplay between them, with one condition mechanistically affecting the other and vice versa. Furthermore, widely prescribed cancer therapies have known toxic effects in the cardiovascular system. Anthracyclines are the paradigm of efficacious cancer therapy widely prescribed with a strong cardiotoxic potential. While some cancer therapies cardiovascular toxicities are transient, others are irreversible. There is a growing need to develop cardioprotective therapies that, when used in conjunction with cancer therapies, can prevent cardiovascular toxicity and thus improve long-term quality of life in survivors. The field has three main challenges: (i) identification of the ultimate mechanisms leading to cardiotoxicity to (ii) identify specific therapeutic targets, and (iii) more sensible diagnostic tools to early identify these conditions. In this review we will focus on the cardioprotective strategies tested and under investigation. We will focus this article into anthracycline cardiotoxicity since it is still the agent most widely prescribed, the one with higher toxic effects on the heart, and the most widely studied.
Collapse
Affiliation(s)
| | - Laura Cádiz
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Agustín Clemente-Moragón
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
13
|
Linschoten M, Geels J, van Werkhoven E, Visser-Wisselaar H, Chamuleau MED, Teske AJ, Robbers L, Oerlemans S, Crommelin H, Breems-de Ridder M, Schut A, Asselbergs FW, van Rhenen A. Rationale and design of the HOVON 170 DLBCL-ANTICIPATE trial: preventing anthracycline-induced cardiac dysfunction with dexrazoxane. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:8. [PMID: 39875951 PMCID: PMC11773844 DOI: 10.1186/s40959-025-00303-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Dexrazoxane has been studied for its ability to prevent anthracycline-induced cardiac dysfunction (AICD) in several trials but its use in clinical practice remains limited. This is related to the low to moderate quality of the generated evidence, safety concerns and restricted prescribing indications. Additional randomized trials are needed before this drug can be routinely integrated into cardio-oncology clinical practice. OBJECTIVES To describe the rationale and design of the HOVON 170 DLBCL - ANTICIPATE trial. This trial aims to establish the efficacy and safety of dexrazoxane for the primary prevention of AICD in patients diagnosed with Diffuse Large B-Cell Lymphoma (DLBCL) treated with six cycles R-CHOP21 chemo-immunotherapy. METHODS This is a multicenter, parallel-group, open-label, phase III trial, randomizing 324 patients between either no cardioprotective treatment or dexrazoxane from the first R-CHOP cycle. The primary and co-primary endpoints are the incidence of AICD within 12 months of registration and the percentage of patients with complete metabolic remission at the end-of-treatment PET-CT respectively. The trial is registered at the EU Clinical Trials Register (EU-CT number 2023-505377-32) and ClinicalTrials.gov (NCT06220032). RESULTS The medical research ethics committee approved the trial in May 2024. Recruitment has started in September 2024 and is expected to last for three years. CONCLUSIONS This trial is poised to contribute crucial evidence concerning the efficacy and safety on the use of dexrazoxane in the primary prevention of AICD. The trial is anticipated to address critical knowledge gaps and offer important insights into the value of dexrazoxane in cardio-oncology practice.
Collapse
Affiliation(s)
- Marijke Linschoten
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
- Netherlands Heart Institute, Utrecht, The Netherlands.
| | - Jesse Geels
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik van Werkhoven
- HOVON Foundation, Rotterdam, The Netherlands
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | | | - Martine E D Chamuleau
- Department of Hematology, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Arco J Teske
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lourens Robbers
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone Oerlemans
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Heleen Crommelin
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Astrid Schut
- The Dutch Network for Cardiovascular Research (WCN), Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Institute of Health Informatics, University College London, London, UK
- Biomedical Research Center, The National Institute for Health Research University College London Hospitals, University College London, London, UK
| | - Anna van Rhenen
- Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
14
|
Fabiani I, Chianca M, Cipolla CM, Cardinale DM. Anthracycline-induced cardiomyopathy: risk prediction, prevention and treatment. Nat Rev Cardiol 2025:10.1038/s41569-025-01126-1. [PMID: 39875555 DOI: 10.1038/s41569-025-01126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Anthracyclines are the cornerstone of treatment for many malignancies. However, anthracycline cardiotoxicity is a considerable concern given that it can compromise the clinical effectiveness of the treatment and patient survival despite early discontinuation of therapy or dose reduction. Patients with cancer receiving anthracycline treatment can have a reduction in their quality of life and likelihood of survival due to cardiotoxicity, irrespective of their oncological prognosis. Increasing knowledge about anthracycline cardiotoxicity has enabled the identification of patients who are candidates for anthracycline regimens and those who might develop anthracycline-induced cardiomyopathy. Anthracycline cardiotoxicity is a unique and evolving phenomenon that begins with myocardial cell damage, progresses to reduced left ventricular ejection fraction, and culminates in symptomatic heart failure if it is not promptly detected and treated. Early risk stratification can be guided by imaging or biomarkers. In this Review, we present a comprehensive and clinically useful approach to cardiomyopathy related to anthracycline therapy, encompassing its epidemiology, definition, mechanisms, novel classifications, risk factors and patient risk stratification, diagnostic approaches (including imaging and biomarkers), treatment guidelines algorithms, and the role of new cardioprotective drugs that are used for the treatment of heart failure.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Michela Chianca
- Division of Cardiology, Cardiothoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
15
|
Qaed E, Almoiliqy M, Liu W, Al-Mashriqi HS, Alyafeai E, Aldahmash W, Mahyoub MA, Tang Z. Protective effects of phosphocreatine against Doxorubicin-Induced cardiotoxicity through mitochondrial function enhancement and apoptosis suppression via AMPK/PGC-1α signaling pathway. Int Immunopharmacol 2025; 144:113677. [PMID: 39580863 DOI: 10.1016/j.intimp.2024.113677] [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: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Doxorubicin (DOX), a potent chemotherapy drug, is limited by its cardiotoxic effects, which can lead to heart damage. This study explores the cardioprotective potential of Phosphocreatine (PCr) in vitro and in vivo models, focusing on its impact on the AMPK and PGC-1α pathways, apoptosis reduction, and mitochondrial function preservation. Advanced methodologies, including high-resolution respirometry (HRR), were employed to assess mitochondrial bioenergetics, AMPK activity, and apoptotic rates in cardiomyocytes. Electrocardiography (ECG) and echocardiography (echo) were used to monitor cardiac function in vivo. Results showed that PCr significantly activated the AMPK and PGC-1α pathways, reduced apoptosis, and stabilized mitochondrial function in cardiomyocytes exposed to DOX. There was an upregulation of AMPK and PGC-1α target genes, stabilization of mitochondrial membranes, and improvements in cellular energy production and antioxidant defenses. PCr also markedly reduced apoptotic markers, enhancing cardiomyocyte viability. ECG and echocardiography revealed that PCr preserved cardiac function, indicated by improved heart rate variability, reduced QT interval prolongation, and enhanced ejection fraction. These findings highlight PCr's potential in mitigating DOX-induced cardiotoxicity by enhancing mitochondrial function and reducing apoptosis. The study underscores the promise of PCr as an agent to reduce chemotherapy-related cardiac injuries, paving the way for further research to improve patient outcomes in cancer treatment.
Collapse
Affiliation(s)
- Eskandar Qaed
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China; State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Marwan Almoiliqy
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | - Wu Liu
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | | | - Eman Alyafeai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Waleed Aldahmash
- Zoology Department, College of Science, King Saud University, P. O. Box 2455, 11451 Riyadh, Saudi Arabia
| | - Mueataz A Mahyoub
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zeyao Tang
- Collage of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China.
| |
Collapse
|
16
|
Liu D, Liu J, Xiao R, Deng A, Liu W. Safety Evaluation of the Combination with Dexrazoxane and Anthracyclines: A Disproportionality Analysis Based on the Food and Drug Administration Adverse Event Reporting System Database. Pharmaceuticals (Basel) 2024; 17:1739. [PMID: 39770581 PMCID: PMC11678267 DOI: 10.3390/ph17121739] [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: 11/26/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: As one of the important interventions to alleviate anthracycline-related cardiotoxicity (ARC), the safety assessment of dexrazoxane in clinical practice is particularly important. This study aims to evaluate the actual efficacy and potential adverse effects of dexrazoxane in clinical practice by analyzing the reports of adverse events (AEs) related to the combination with dexrazoxane and anthracyclines. Methods: We utilized four disproportionality analysis methods to analyze AE reports of the combination with dexrazoxane and anthracyclines in the Food and Drug Administration Adverse Event Reporting System (FAERS) database from the third quarter of 2014 to the first quarter of 2024. Results: Under the three backgrounds, a large number of preferred terms (PTs) such as cardiac failure disappeared in the combined group, and the PTs with significant signal values were mainly concentrated in infections and infestations. For patients under 18, some PTs associated with infections and infestations disappeared after the combination of the two drugs. Conclusions: Dexrazoxane can effectively alleviate ARC, but it may also increase the risk of infection. For infections and infestations, children under 18 years old are more likely to benefit from the combination therapy. More attention should be paid to infectious AEs in the clinical use of dexrazoxane, though disproportionality analysis is a hypothesis-generating approach.
Collapse
Affiliation(s)
| | | | | | | | - Wei Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; (D.L.); (J.L.); (R.X.); (A.D.)
| |
Collapse
|
17
|
Marwick TH, Dewar E, Nolan M, Shirazi M, Dias P, Wright L, Fitzgerald B, Kearney L, Srivastava P, Atherton J, Negishi K, Sverdlov AL, Wahi S, Otton J, Selvanayagam J, Thomas L, Thavendiranathan P. Strain surveillance during chemotherapy to improve cardiovascular outcomes: the SUCCOUR-MRI trial. Eur Heart J 2024; 45:4414-4424. [PMID: 39217601 PMCID: PMC11542702 DOI: 10.1093/eurheartj/ehae574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/17/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS The detection of cancer therapy-related cardiac dysfunction (CTRCD) by reduction of left ventricular ejection fraction (LVEF) during chemotherapy usually triggers the initiation of cardioprotective therapy. This study addressed whether the same approach should be applied to patients with worsening of global longitudinal strain (GLS) without attaining thresholds of LVEF. METHODS Strain surveillance during chemotherapy for improving cardiovascular outcomes (SUCCOUR-MRI) was a prospective multicentre randomized controlled trial involving 14 sites. Of 355 patients receiving anthracyclines with normal baseline LVEF, 333 patients (age 59 ± 13 years, 79% women) with at least one other CTRCD risk factor, able to undergo magnetic resonance imaging (MRI), GLS, and three-dimensional echocardiography were tracked over 12 months. A total of 105 patients (age 59 ± 13 years, 75% women, 69% breast cancer) developing GLS-CTRCD (>12% relative reduction of GLS without a change in LVEF) were randomized to cardioprotection with neurohormonal antagonists vs. usual care. The primary endpoint was 12-month change in MRI-LVEF; the secondary endpoint was MRI-LVEF-defined CTRCD. RESULTS During follow-up, two patients died, and two developed heart failure. Most patients were randomized at 3 months (62%). Median doses of angiotensin inhibition/blockade and beta-blockade were 75% and 50% of respective targets; 21 (43%) had side-effects attributed to cardioprotection. Due to a smaller LVEF change from baseline with cardioprotection than usual care (-2.5 ± 5.4% vs. -5.6 ± 5.9%, P = .009), follow-up LVEF was higher after cardioprotection (59 ± 5% vs. 55 ± 6%, P < .0001). After adjustment for baseline LVEF, the mean (95% confidence interval) difference in the change in LVEF between the two groups was -3.6% (-1.8% to -5.5%, P < .001). After cardioprotection, 1/49 patients developed 12-month LVEF-CTRCD, compared to 6/56 in usual care (P = .075). Global longitudinal strain improved at 3 months post-randomization in the cardioprotection group, with little change with usual care. CONCLUSIONS In patients with isolated GLS reduction after anthracyclines, cardioprotection is associated with better preservation of 12-month MRI-LVEF compared with usual care.
Collapse
Affiliation(s)
- Thomas H Marwick
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardiovascular Imaging, Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Elizabeth Dewar
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Nolan
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardio-Oncology Section, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mitra Shirazi
- Department of Cardiology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Peter Dias
- Advara Heart Care, Murdoch, WA, Australia
| | - Leah Wright
- Imaging Research, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | | | | | | | - John Atherton
- University of Queensland Faculty of Medicine, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
| | - Kazuaki Negishi
- Cardiology Department, Nepean Hospital, Kingswood, NSW, Australia
- Nepean Clinical School, University of Sydney, Kingswood, NSW, Australia
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, The University of Newcastle, Hunter Medical Research Institute, Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia
| | - Sudhir Wahi
- Princess Alexandra Hospital, Brisbane, Australia
| | - James Otton
- Liverpool Hospital, Liverpool, NSW, Australia
| | | | - Liza Thomas
- Westmead Clinical School, University of Sydney and University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
18
|
Raisi-Estabragh Z, Murphy AC, Ramalingam S, Scherrer-Crosbie M, Lopez-Fernandez T, Reynolds KL, Aznar M, Lin AE, Libby P, Cordoba R, Bredsen-Masley C, Wechalekar A, Apperley J, Cheng RK, Manisty CH. Cardiovascular Considerations Before Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2024; 6:631-654. [PMID: 39479317 PMCID: PMC11520216 DOI: 10.1016/j.jaccao.2024.07.017] [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: 09/11/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024] Open
Abstract
Baseline cardiovascular assessment before the initiation of potentially cardiotoxic cancer therapies is a key component of cardio-oncology, aiming to reduce cardiovascular complications and morbidity in patients and survivors. Recent clinical practice guidelines provide both general and cancer therapy-specific recommendations for baseline cardiovascular toxicity risk assessment and management, including the use of dedicated risk scores, cardiovascular imaging, and biomarker testing. However, the value of such interventions in altering disease trajectories has not been established, with many recommendations based on expert opinion or Level of Evidence: C, studies with a potential for high risk of bias. Advances in understanding underlying mechanisms of cardiotoxicity and the increased availability of genetic and immunologic profiling present new opportunities for personalized risk assessment. This paper evaluates the existing evidence on cardiovascular care of cancer patients before cardiotoxic cancer therapy and highlights gaps in evidence and priorities for future research.
Collapse
Affiliation(s)
- Zahra Raisi-Estabragh
- William Harvey Research Institute, National Institute for Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
| | | | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez-Fernandez
- Hospital Universitario La Paz, Instituto de Investigación La Paz-IdiPaz, Madrid, Spain
- Hospital Universitario Quiron Pozuelo, Madrid, Spain
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marianne Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Amy E. Lin
- Department of Medicine, Division of Cardiology, Section of Cardio-Oncology and Immunology, University of California-San Francisco, San Francisco, California, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Christine Bredsen-Masley
- Division of Hematology/Oncology, Department of Medicine, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Ashu Wechalekar
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Jane Apperley
- Department of Clinical Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Richard K. Cheng
- Division of Cardiology, University of Washington Medical Center, Seattle, Washington, USA
| | - Charlotte H. Manisty
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
19
|
Camilli M, Cipolla CM, Dent S, Minotti G, Cardinale DM. Anthracycline Cardiotoxicity in Adult Cancer Patients: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:655-677. [PMID: 39479333 PMCID: PMC11520218 DOI: 10.1016/j.jaccao.2024.07.016] [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: 03/26/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 11/02/2024] Open
Abstract
Since their introduction in the 1960s, anthracyclines have been a significant breakthrough in oncology, introducing dramatic changes in the treatment of solid and hematologic malignancies. Although new-generation targeted drugs and cellular therapies are revolutionizing contemporary oncology, anthracyclines remain the cornerstone of treatment for lymphomas, acute leukemias, and soft tissue sarcomas. However, their clinical application is limited by a dose-dependent cardiotoxicity that can reduce cardiac performance and eventually lead to overt heart failure. The field of cardio-oncology has emerged to safeguard the cardiovascular health of cancer patients receiving these therapies. It focuses on controlling risk factors, implementing preventive strategies, ensuring appropriate surveillance, and managing complications. This state-of-the-art review summarizes the current indications for anthracyclines in modern oncology, explores recent evidence on pathophysiology and epidemiology, and discusses advances in cardioprotection measures in the anthracycline-treated patient. Additionally, it highlights key clinical challenges and research gaps in this area.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology IRCCS, Milan, Italy
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Giorgio Minotti
- Università e Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
20
|
Tetterton-Kellner J, Jensen BC, Nguyen J. Navigating cancer therapy induced cardiotoxicity: From pathophysiology to treatment innovations. Adv Drug Deliv Rev 2024; 211:115361. [PMID: 38901637 PMCID: PMC11534294 DOI: 10.1016/j.addr.2024.115361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Every year, more than a million people in the United States undergo chemotherapy or radiation therapy for cancer, as estimated by the CDC. While chemotherapy has been an instrumental tool for treating cancer, it also causes severe adverse effects. The more commonly acknowledged adverse effects include hair loss, fatigue, and nausea, but a more severe and longer lasting side effect is cardiotoxicity. Cardiotoxicity, or heart damage, is a common complication of cancer treatments. It can range from mild to severe, and it can affect some patients temporarily or others permanently, even after they are cured of cancer. Dexrazoxane is the only FDA-approved drug for treating anthracycline induced cardiotoxicity, but it also has drawbacks and adverse effects. There is no other type of chemotherapy induced cardiotoxicity that has an approved treatment option. In this review, we discuss the pathophysiology of chemotherapeutic-induced cardiotoxicity, methods and guidelines of diagnosis, methods of treatment and mitigation, and current drug delivery approaches in therapeutic development.
Collapse
Affiliation(s)
- Jessica Tetterton-Kellner
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Cardiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
21
|
Thonusin C, Osataphan N, Leemasawat K, Nawara W, Sriwichaiin S, Supakham S, Gunaparn S, Apaijai N, Somwangprasert A, Phrommintikul A, Chattipakorn SC, Chattipakorn N. Changes in blood metabolomes as potential markers for severity and prognosis in doxorubicin-induced cardiotoxicity: a study in HER2-positive and HER2-negative breast cancer patients. J Transl Med 2024; 22:398. [PMID: 38685030 PMCID: PMC11059746 DOI: 10.1186/s12967-024-05088-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/12/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND We aimed to compare the changes in blood metabolomes and cardiac parameters following doxorubicin treatment in HER2-positive and HER2-negative breast cancer patients. Additionally, the potential roles of changes in blood metabolomes as severity and prognostic markers of doxorubicin-induced cardiotoxicity were determined. METHODS HER2-positive (n = 37) and HER2-negative (n = 37) breast cancer patients were enrolled. Cardiac function assessment and blood collection were performed at baseline and 2 weeks after completion of doxorubicin treatment in all patients, as well as at three months after completion of doxorubicin treatment in HER2-negative breast cancer patients. Blood obtained at all three-time points was processed for measuring cardiac injury biomarkers. Blood obtained at baseline and 2 weeks after completion of doxorubicin treatment were also processed for measuring systemic oxidative stress and 85 metabolome levels. RESULTS Cardiac injury and systolic dysfunction 2 weeks after completion of doxorubicin treatment were comparable between these two groups of patients. However, only HER2-negative breast cancer patients exhibited increased systemic oxidative stress and cardiac autonomic dysfunction at this time point. Moreover, 33 and 29 blood metabolomes were altered at 2 weeks after completion of doxorubicin treatment in HER2-positive and HER2-negative breast cancer patients, respectively. The changes in most of these metabolomes were correlated with the changes in cardiac parameters, both at 2 weeks and 3 months after completion of doxorubicin treatment. CONCLUSIONS The changes in blood metabolomes following doxorubicin treatment were dependent on HER2 status, and these changes might serve as severity and prognostic markers of doxorubicin-induced cardiotoxicity. TRIAL REGISTRATION The study was conducted under ethical approval from the Institutional Review Board of the Faculty of Medicine, Chiang Mai University (Registration number: MED-2563-07001; Date: April 28, 2020). The study also complied with the Declaration of Helsinki.
Collapse
Affiliation(s)
- Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nichanan Osataphan
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Krit Leemasawat
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wichwara Nawara
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn Supakham
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriluck Gunaparn
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | | | - Arintaya Phrommintikul
- Division of Cardiology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
22
|
Du R, Xiao N, Han L, Guo K, Li K, Chen Z, Zhang H, Zhou Z, Huang Y, Zhao X, Bian H. Dexrazoxane inhibits the growth of esophageal squamous cell carcinoma by attenuating SDCBP/MDA-9/syntenin-mediated EGFR-PI3K-Akt pathway activation. Sci Rep 2024; 14:9167. [PMID: 38649770 PMCID: PMC11035576 DOI: 10.1038/s41598-024-59665-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Syndecan-binding protein (SDCBP) was reported to stimulate the advancement of esophageal squamous cell carcinoma (ESCC) and could potentially be a target for ESCC treatment. There is a growing corpus of research on the anti-tumor effects of iron chelators; however, very few studies have addressed the involvement of dexrazoxane in cancer. In this study, structure-based virtual screening was employed to select drugs targeting SDCBP from the Food and Drug Administration (FDA)-approved drug databases. The sepharose 4B beads pull-down assay revealed that dexrazoxane targeted SDCBP by interacting with its PDZ1 domain. Additionally, dexrazoxane inhibited ESCC cell proliferation and anchorage-independent colony formation via SDCBP. ESCC cell apoptosis and G2 phase arrest were induced as measured by the flow cytometry assay. Subsequent research revealed that dexrazoxane attenuated the binding ability between SDCBP and EGFR in an immunoprecipitation assay. Furthermore, dexrazoxane impaired EGFR membrane localization and inactivated the EGFR/PI3K/Akt pathway. In vivo, xenograft mouse experiments indicated that dexrazoxane suppressed ESCC tumor growth. These data indicate that dexrazoxane might be established as a potential anti-cancer agent in ESCC by targeting SDCBP.
Collapse
Affiliation(s)
- Ruijuan Du
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China.
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China.
| | - Nan Xiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Li Han
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - KeLei Guo
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - Kai Li
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - Zhiguo Chen
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
| | - Hui Zhang
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China
| | - Zijun Zhou
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
| | - Yunlong Huang
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China
| | - Xulin Zhao
- Oncology Department, Nanyang First People's Hospital, Nan Yang, 473004, Henan, People's Republic of China
| | - Hua Bian
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, 473004, Henan, People's Republic of China.
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, No. 80, Changjiang Road, Nanyang, 473004, Henan, People's Republic of China.
| |
Collapse
|
23
|
Zheng H, Zhan H. Preventing Anthracycline-Associated Heart Failure: What Is the Role of Dexrazoxane?: JACC: CardioOncology Controversies in Cardio-Oncology. JACC CardioOncol 2024; 6:318-321. [PMID: 38774009 PMCID: PMC11103021 DOI: 10.1016/j.jaccao.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024] Open
Affiliation(s)
- Haoyi Zheng
- Saint Francis Hospital, The Heart Center, Roslyn, New York, USA
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, New York, USA
- Medical Service, Northport VA Medical Center, Northport, New York, USA
| |
Collapse
|
24
|
Upshaw JN, Parson SK, Buchsbaum RJ, Schlam I, Ruddy KJ, Durani U, Epperla N, Leong DP. Dexrazoxane to Prevent Cardiotoxicity in Adults Treated with Anthracyclines: JACC: CardioOncology Controversies in Cardio-Oncology. JACC CardioOncol 2024; 6:322-324. [PMID: 38773999 PMCID: PMC11103024 DOI: 10.1016/j.jaccao.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024] Open
Affiliation(s)
- Jenica N. Upshaw
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Susan K. Parson
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Rachel J. Buchsbaum
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Ilana Schlam
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Kathryn J. Ruddy
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Urshila Durani
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Darryl P. Leong
- Department of Medicine and the Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Upshaw JN, Nelson J, Sweigart B, Rodday AM, Kumar AJ, Konstam MA, Wong JB, Ky B, Karmiy S, Friedberg JW, Evens AM, Kent DM, Parsons SK. Impact of Preexisting Heart Failure on Treatment and Outcomes in Older Patients With Hodgkin Lymphoma. JACC CardioOncol 2024; 6:200-213. [PMID: 38774008 PMCID: PMC11103040 DOI: 10.1016/j.jaccao.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 05/24/2024] Open
Abstract
Background Older patients with Hodgkin lymphoma (HL) often have comorbid cardiovascular disease; however, the impact of pre-existing heart failure (HF) on the management and outcomes of HL is unknown. Objectives The aim of this study was to assess the prevalence of pre-existing HF in older patients with HL and its impact on treatment and outcomes. Methods Linked Surveillance, Epidemiology, and End Results (SEER) and Medicare data from 1999 to 2016 were used to identify patients 65 years and older with newly diagnosed HL. Pre-existing HF, comorbidities, and cancer treatment were ascertained from billing codes and cause-specific mortality from SEER. The associations between pre-existing HF and cancer treatment were estimated using multivariable logistic regression. Cause-specific Cox proportional hazards models adjusted for comorbidities and cancer treatment were used to estimate the association between pre-existing HF and cause-specific mortality. Results Among 3,348 patients (mean age 76 ± 7 years, 48.6% women) with newly diagnosed HL, pre-existing HF was present in 437 (13.1%). Pre-existing HF was associated with a lower likelihood of using anthracycline-based chemotherapy regimens (OR: 0.42; 95% CI: 0.29-0.60) and a higher likelihood of lymphoma mortality (HR: 1.25; 95% CI: 1.06-1.46) and cardiovascular mortality (HR: 2.57; 95% CI: 1.96-3.36) in models adjusted for comorbidities. One-year lymphoma mortality cumulative incidence was 37.4% (95% CI: 35.5%-39.5%) with pre-existing HF and 26.3% (95% CI: 25.0%-27.6%) without pre-existing HF. The cardioprotective medications dexrazoxane and liposomal doxorubicin were used in only 4.2% of patients. Conclusions Pre-existing HF in older patients with newly diagnosed HL is common and associated with higher 1-year mortality. Strategies are needed to improve lymphoma and cardiovascular outcomes in this high-risk population.
Collapse
Affiliation(s)
- Jenica N. Upshaw
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jason Nelson
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Benjamin Sweigart
- Clinical and Translational Science Institute, Tufts University, Boston, Massachusetts, USA
| | - Angie Mae Rodday
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Anita J. Kumar
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Marvin A. Konstam
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
| | - John B. Wong
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Clinical Decision Making, Tufts Medical Center, Boston, Massachusetts, USA
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Samuel Karmiy
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jonathan W. Friedberg
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | | | - David M. Kent
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Susan K. Parsons
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Zhao P, Li Y, Xu X, Yang H, Li X, Fu S, Guo Z, Zhang J, Li H, Tian J. Neutrophil extracellular traps mediate cardiomyocyte ferroptosis via the Hippo-Yap pathway to exacerbate doxorubicin-induced cardiotoxicity. Cell Mol Life Sci 2024; 81:122. [PMID: 38456997 PMCID: PMC10923748 DOI: 10.1007/s00018-024-05169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Doxorubicin-induced cardiotoxicity (DIC), which is a cardiovascular complication, has become the foremost determinant of decreased quality of life and mortality among survivors of malignant tumors, in addition to recurrence and metastasis. The limited ability to accurately predict the occurrence and severity of doxorubicin-induced injury has greatly hindered the prevention of DIC, but reducing the dose to mitigate side effects may compromise the effective treatment of primary malignancies. This has posed a longstanding clinical challenge for oncologists and cardiologists. Ferroptosis in cardiomyocytes has been shown to be a pivotal mechanism underlying cardiac dysfunction in DIC. Ferroptosis is influenced by multiple factors. The innate immune response, as exemplified by neutrophil extracellular traps (NETs), may play a significant role in the regulation of ferroptosis. Therefore, the objective of this study was to investigate the involvement of NETs in doxorubicin-induced cardiomyocyte ferroptosis and elucidate their regulatory role. This study confirmed the presence of NETs in DIC in vivo. Furthermore, we demonstrated that depleting neutrophils effectively reduced the occurrence of doxorubicin-induced ferroptosis and myocardial injury in DIC. Additionally, our findings showed the pivotal role of high mobility group box 1 (HMGB1) as a critical molecule implicated in DIC and emphasized its involvement in the modulation of ferroptosis subsequent to NETs inhibition. Mechanistically, we obtained preliminary evidence suggesting that doxorubicin-induced NETs could modulate yes-associated protein (YAP) activity by releasing HMGB1, which subsequently bound to toll like receptor 4 (TLR4) on the cardiomyocyte membrane, thereby influencing cardiomyocyte ferroptosis in vitro. Our findings suggest that doxorubicin-induced NETs modulate cardiomyocyte ferroptosis via the HMGB1/TLR4/YAP axis, thereby contributing to myocardial injury. This study offers a novel approach for preventing and alleviating DIC by targeting alterations in the immune microenvironment.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, 150001, China
| | - You Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xiangli Xu
- Department of Ultrasound, The Second Hospital of Harbin City, Harbin, 150001, China
| | - Haobo Yang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, 150001, China
| | - Xintong Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, 150001, China
| | - Shuai Fu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Zihong Guo
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jianing Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hairu Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
27
|
Shi H, Duan L, Tong L, Pu P, Wei L, Wang L, Hu D, Tang H. Research Progress on Flavonoids in Traditional Chinese Medicine to Counteract Cardiotoxicity Associated with Anti-Tumor Drugs. Rev Cardiovasc Med 2024; 25:74. [PMID: 39076949 PMCID: PMC11263839 DOI: 10.31083/j.rcm2503074] [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: 08/13/2023] [Revised: 11/12/2023] [Accepted: 11/21/2023] [Indexed: 07/31/2024] Open
Abstract
The development of anti-tumor drugs has notably enhanced the survival rates and quality of life for patients with malignant tumors. However, the side effects of these drugs, especially cardiotoxicity, significantly limit their clinical application. The cardiotoxicity associated with anti-tumor drugs has been a subject of extensive attention and research. Traditional to mitigate these side effects have included reducing drug dosages, shortening treatment duration, modifying administration methods, and opting for drugs with lower toxicity. However, either approach may potentially compromise the anti-tumor efficacy of the medications. Therefore, exploring other effective methods for anti-cardiotoxicity will be the focus of future research. The potential of traditional Chinese medicine (TCM) in managing cardiovascular diseases and cancer treatment has gained widespread recognition. TCM is valued for its minimal side effects, affordability, and accessibility, offering promising avenues in the prevention and treatment of cardiotoxicity caused by anti-tumor drugs. Among its constituents, flavonoids, which are present in many TCMs, are particularly notable. These monomeric compounds with distinct structural components have been shown to possess both cardiovascular protective properties and anti-tumor capabilities. In this discussion, we will delve into the classification of anti-tumor drugs and explore the underlying mechanisms of their associated cardiotoxicity. Additionally, we will examine flavonoids found in TCM and investigate their mechanisms of cardiovascular protection. This will include an analysis of how these natural compounds can mitigate the cardiac side effects of anti-tumor therapies while potentially enhancing overall patient health and treatment outcomes.
Collapse
Affiliation(s)
- Hongwei Shi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
- Department of Oncology, Renmin Hospital of Wuhan University, 430064 Wuhan, Hubei, China
| | - Lian Duan
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Li Tong
- Department of Pharmacy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Peng Pu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Lai Wei
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Desheng Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Heng Tang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| |
Collapse
|
28
|
Bertorello N, Luksch R, Bisogno G, Haupt R, Spallarossa P, Cenna R, Fagioli F. Reply to Comment on: Cardiotoxicity in children with cancer treated with anthracyclines: A position statement on dexrazoxane. Pediatr Blood Cancer 2023; 70:e30690. [PMID: 37737681 DOI: 10.1002/pbc.30690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/23/2023]
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
|
29
|
Kontoghiorghes GJ. Drug Selection and Posology, Optimal Therapies and Risk/Benefit Assessment in Medicine: The Paradigm of Iron-Chelating Drugs. Int J Mol Sci 2023; 24:16749. [PMID: 38069073 PMCID: PMC10706143 DOI: 10.3390/ijms242316749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The design of clinical protocols and the selection of drugs with appropriate posology are critical parameters for therapeutic outcomes. Optimal therapeutic protocols could ideally be designed in all diseases including for millions of patients affected by excess iron deposition (EID) toxicity based on personalised medicine parameters, as well as many variations and limitations. EID is an adverse prognostic factor for all diseases and especially for millions of chronically red-blood-cell-transfused patients. Differences in iron chelation therapy posology cause disappointing results in neurodegenerative diseases at low doses, but lifesaving outcomes in thalassemia major (TM) when using higher doses. In particular, the transformation of TM from a fatal to a chronic disease has been achieved using effective doses of oral deferiprone (L1), which improved compliance and cleared excess toxic iron from the heart associated with increased mortality in TM. Furthermore, effective L1 and L1/deferoxamine combination posology resulted in the complete elimination of EID and the maintenance of normal iron store levels in TM. The selection of effective chelation protocols has been monitored by MRI T2* diagnosis for EID levels in different organs. Millions of other iron-loaded patients with sickle cell anemia, myelodysplasia and haemopoietic stem cell transplantation, or non-iron-loaded categories with EID in different organs could also benefit from such chelation therapy advances. Drawbacks of chelation therapy include drug toxicity in some patients and also the wide use of suboptimal chelation protocols, resulting in ineffective therapies. Drug metabolic effects, and interactions with other metals, drugs and dietary molecules also affected iron chelation therapy. Drug selection and the identification of effective or optimal dose protocols are essential for positive therapeutic outcomes in the use of chelating drugs in TM and other iron-loaded and non-iron-loaded conditions, as well as general iron toxicity.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
30
|
Faggiano A, Gherbesi E, Avagimyan A, Ruscica M, Donisi L, Fedele MA, Cipolla CM, Vicenzi M, Carugo S, Cardinale D. Melatonin mitigates oxidative damage induced by anthracycline: a systematic-review and meta-analysis of murine models. Front Cardiovasc Med 2023; 10:1289384. [PMID: 38075951 PMCID: PMC10701532 DOI: 10.3389/fcvm.2023.1289384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Oxidative stress induced by the excessive production of reactive oxygen species is one of the primary mechanisms implicated in anthracycline (ANT)-induced cardiotoxicity. There is a strong clinical need for a molecule capable of effectively preventing and reducing the oxidative damage caused by ANT. In vitro and in vivo studies conducted in mice have shown that melatonin stimulates the expression of antioxidative agents and reduces lipid peroxidation induced by ANT. METHODS We investigated this issue through a meta-analysis of murine model studies. The outcome of the meta-analysis was to compare oxidative damage, estimated by products of lipid peroxidation (MDA = Malondialdehyde) and markers of oxidative stress (SOD = Superoxide Dismutase, GSH = Glutathione), along with a marker of cardiac damage (CK-MB = creatine kinase-myocardial band), assessed by measurements in heart and/or blood samples in mice undergoing ANT chemotherapy and assuming melatonin vs. controls. The PubMed, OVID-MEDLINE and Cochrane library databases were analysed to search English-language review papers published from the inception up to August 1st, 2023. Studies were identified by using Me-SH terms and crossing the following terms: "melatonin", "oxidative stress", "lipid peroxidation", "anthracycline", "cardiotoxicity". RESULTS The metanalysis included 153 mice administered melatonin before, during or immediately after ANT and 153 controls from 13 studies. Compared with controls, the levels of all oxidative stress markers were significantly better in the pooled melatonin group, with standardized mean differences (SMD) for MDA, GSH and SOD being -8.03 ± 1.2 (CI: -10.43/-5.64, p < 0.001), 7.95 ± 1.8 (CI: 4.41/11.5, p < 0.001) and 3.94 ± 1.6 (CI: 0.77/7.12, p = 0.015) respectively. Similarly, compared with controls, CK-MB levels reflecting myocardial damage were significantly lower in the pooled melatonin group, with an SMD of -4.90 ± 0.5 (CI: -5.82/-3.98, p < 0.001). CONCLUSION Melatonin mitigates the oxidative damage induced by ANT in mouse model. High-quality human clinical studies are needed to further evaluate the use of melatonin as a preventative/treatment strategy for ANT-induced cardiotoxicity.
Collapse
Affiliation(s)
- Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Elisa Gherbesi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ashot Avagimyan
- Department of Anatomical Pathology and Clinical Morphology, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Massimiliano Ruscica
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, Milan, Italy
| | - Luca Donisi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Maria Antonia Fedele
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Marco Vicenzi
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Daniela Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| |
Collapse
|
31
|
Tai P, Chen X, Jia G, Chen G, Gong L, Cheng Y, Li Z, Wang H, Chen A, Zhang G, Zhu Y, Xiao M, Wang Z, Liu Y, Shan D, He D, Li M, Zhan T, Khan A, Li X, Zeng X, Li C, Ouyang D, Ai K, Chen X, Liu D, Liu Z, Wei D, Cao K. WGX50 mitigates doxorubicin-induced cardiotoxicity through inhibition of mitochondrial ROS and ferroptosis. J Transl Med 2023; 21:823. [PMID: 37978379 PMCID: PMC10655295 DOI: 10.1186/s12967-023-04715-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Doxorubicin (DOX)-induced cardiotoxicity (DIC) is a major impediment to its clinical application. It is indispensable to explore alternative treatment molecules or drugs for mitigating DIC. WGX50, an organic extract derived from Zanthoxylum bungeanum Maxim, has anti-inflammatory and antioxidant biological activity, however, its function and mechanism in DIC remain unclear. METHODS We established DOX-induced cardiotoxicity models both in vitro and in vivo. Echocardiography and histological analyses were used to determine the severity of cardiac injury in mice. The myocardial damage markers cTnT, CK-MB, ANP, BNP, and ferroptosis associated indicators Fe2+, MDA, and GPX4 were measured using ELISA, RT-qPCR, and western blot assays. The morphology of mitochondria was investigated with a transmission electron microscope. The levels of mitochondrial membrane potential, mitochondrial ROS, and lipid ROS were detected using JC-1, MitoSOX™, and C11-BODIPY 581/591 probes. RESULTS Our findings demonstrate that WGX50 protects DOX-induced cardiotoxicity via restraining mitochondrial ROS and ferroptosis. In vivo, WGX50 effectively relieves doxorubicin-induced cardiac dysfunction, cardiac injury, fibrosis, mitochondrial damage, and redox imbalance. In vitro, WGX50 preserves mitochondrial function by reducing the level of mitochondrial membrane potential and increasing mitochondrial ATP production. Furthermore, WGX50 reduces iron accumulation and mitochondrial ROS, increases GPX4 expression, and regulates lipid metabolism to inhibit DOX-induced ferroptosis. CONCLUSION Taken together, WGX50 protects DOX-induced cardiotoxicity via mitochondrial ROS and the ferroptosis pathway, which provides novel insights for WGX50 as a promising drug candidate for cardioprotection.
Collapse
Affiliation(s)
- Panpan Tai
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xinyu Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guihua Jia
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Guanjun Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lian Gong
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yaxin Cheng
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhuan Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, China
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, China
- Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, 410013, China
| | - Heng Wang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Aiyan Chen
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ganghua Zhang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuxing Zhu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Mengqing Xiao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yunqing Liu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongyong Shan
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Dong He
- Staff Hospital of Central South University, Central South University, Changsha, China
| | - Moying Li
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Abbas Khan
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China
| | - Chaopeng Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, China
| | - Dongsheng Ouyang
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd, Changsha, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xuan Chen
- College of Horticulture, Hunan Agricultural University, Changsha, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha, China
- National Research Center of Engineering Technology for Utilization Ingredients From Botanicals, Changsha, China
| | - Dongbo Liu
- College of Horticulture, Hunan Agricultural University, Changsha, China
- State Key Laboratory of Subhealth Intervention Technology, Changsha, China
- National Research Center of Engineering Technology for Utilization Ingredients From Botanicals, Changsha, China
| | - Zhonghua Liu
- National Research Center of Engineering Technology for Utilization Ingredients From Botanicals, Changsha, China
| | - Dongqing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
32
|
Wang L, Liu Y, Li S, Zha Z, Chen Y, Wang Q, Zhou S, Huang X, Xu M. Capsaicin alleviates doxorubicin-induced acute myocardial injury by regulating iron homeostasis and PI3K-Akt signaling pathway. Aging (Albany NY) 2023; 15:11845-11859. [PMID: 37916995 PMCID: PMC10683596 DOI: 10.18632/aging.205138] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Capsaicin (CAP), a frequently occurring alkaloid component found in spicy peppers, has demonstrated therapeutic potential against tumors, metabolic disease, and cardiovascular disorders. Doxorubicin (DOX), a widely used anthracycline drug in chemotherapy, is notorious for its cardiotoxicity. This study aimed to investigate the potential of CAP in mitigating DOX toxicity in mouse hearts and H9C2 cells, as well as to explore the underlying mechanisms. METHODS In our study, we conducted experiments on both mice and H9C2 cells. The mice were divided into four groups and treated with different substances: normal saline, CAP, DOX and CAP+DOX. We evaluated the induction of ferroptosis by DOX and the remission of ferroptosis by CAP using various methods, including echocardiography, Hematoxylin and Eosin (H&E) staining, Masson's trichrome staining, and determination of ferroptosis metabolites, genes and proteins. Additionally, we employed RNA-seq to identify the inhibitory effect of CAP on DOX-induced myocardial apoptosis, which was further confirmed through western blotting. Similar approaches were applied to H9C2 cells, yielding reliable results. RESULTS Our study demonstrated that treatment with CAP improved the survival rate of DOX-treated mice and reduced myocardial injury. Mechanistically, CAP downregulated transferrin (Trf) and upregulated solute carrier family 40 member 1 (SLC40A1), which helped maintain iron levels in the cells and prevent ferroptosis. Furthermore, CAP inhibited DOX-induced apoptosis by modulating the phosphoinositide 3-kinase (PI3K)- protein kinase B (Akt) signaling pathway. Specifically, CAP activated the PI3K-Akt pathway and regulated downstream BCL2 and BAX to mitigate DOX-induced apoptosis. Therefore, our results suggest that CAP effectively alleviates acute myocardial injury induced by DOX. CONCLUSION Our findings demonstrate that CAP has the potential to alleviate DOX-induced ferroptosis by regulating iron homeostasis. Additionally, it can inhibit DOX-induced apoptosis by activating PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Longbin Wang
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ying Liu
- Department of Cardiology, Sixth Medical Center, PLA General Hospital, Beijing, China
| | - Si Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhijian Zha
- Chinese Internal Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yu Chen
- School of Tropical Agriculture and Forestry, Hainan University, Haikou, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary, China
| | - Xufeng Huang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary, China
| | - Ming Xu
- College of Clinical Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
33
|
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: 1.0] [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
|
34
|
Zhao J, Zhang N, Ma X, Li M, Feng H. The dual role of ferroptosis in anthracycline-based chemotherapy includes reducing resistance and increasing toxicity. Cell Death Discov 2023; 9:184. [PMID: 37344500 DOI: 10.1038/s41420-023-01483-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023] Open
Abstract
In conjunction with previous studies, we have noted that ferroptosis, as an emerging mode of regulated cell death (RCD), is intimately related to anthracycline pharmacotherapy. Not only does ferroptosis significantly modulate tumour resistance and drug toxicity, which are core links of the relevant chemotherapeutic process, but it also appears to play a conflicting role that has yet to be appreciated. By targeting the dual role of ferroptosis in anthracycline-based chemotherapy, this review aims to focus on the latest findings at this stage, identify the potential associations and provide novel perspectives for subsequent research directions and therapeutic strategies.
Collapse
Affiliation(s)
- Jiazheng Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei, 050011, China
| | - Ning Zhang
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei, 050011, China
| | - Xiaowei Ma
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China
| | - Ming Li
- Department of Orthopedics, The Second Hospital of Hebei Medical University, 215 Heping Road, Shijia-zhuang, Hebei, China
| | - Helin Feng
- Departments of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
35
|
Wong-Siegel JR, Kim Y, Stitziel NO, Javaheri A. Genetic Testing in Evaluating Risk of Anthracycline Cardiomyopathy: Are We There Yet? JACC CardioOncol 2023; 5:406-408. [PMID: 37397085 PMCID: PMC10308055 DOI: 10.1016/j.jaccao.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Affiliation(s)
| | - Yuri Kim
- Brigham and Women’s Hospital, Cardiovascular Division, Boston, Massachusetts, USA
| | - Nathan O. Stitziel
- Washington University School of Medicine, Department of Medicine, St. Louis, Missouri, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
- Washington University School of Medicine, Department of Genetics, St. Louis, Missouri, USA
| | - Ali Javaheri
- Brigham and Women’s Hospital, Cardiovascular Division, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Henriksen PA, Rankin S, Lang NN. Cardioprotection in Patients at High Risk of Anthracycline-Induced Cardiotoxicity: JACC: CardioOncology Primer. JACC CardioOncol 2023; 5:292-297. [PMID: 37397086 PMCID: PMC10308056 DOI: 10.1016/j.jaccao.2023.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Affiliation(s)
- Peter A. Henriksen
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen Rankin
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Ninian N. Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
37
|
Ferrera A, Fiorentini V, Reale S, Solfanelli G, Tini G, Barbato E, Volpe M, Battistoni A. Anthracyclines-Induced Cardiac Dysfunction: What Every Clinician Should Know. Rev Cardiovasc Med 2023; 24:148. [PMID: 39076747 PMCID: PMC11273047 DOI: 10.31083/j.rcm2405148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/12/2023] [Accepted: 01/29/2023] [Indexed: 07/31/2024] Open
Abstract
Chemotherapies have changed the prognosis of patients affected by cancer over the last 20 years, with a significant increase in survival rates. However, they can cause serious adverse effects that may limit their use. In particular, anthracyclines, widely used to treat both hematologic cancers and solid cancers, may cause cardiac toxicity, leading to the development of heart failure in some cases. This review aims to explore current evidence with regards to anthracyclines' cardiotoxicity, with particular focus on the classifications and underlying molecular mechanisms, in order to provide an overview on the current methods of its diagnosis, treatment, and prevention. An attentive approach and a prompt management of patients undergoing treatment with anthracyclines is imperative to avoid preventable antineoplastic drug discontinuation and is conducive to improving both short-term and long-term cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Armando Ferrera
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| | - Vincenzo Fiorentini
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| | - Simone Reale
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| | - Giorgio Solfanelli
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| | - Giacomo Tini
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| | - Emanuele Barbato
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| | - Massimo Volpe
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
- IRCCS San Raffaele, 00163 Rome, Italy
| | - Allegra Battistoni
- Clinical and Molecular Medicine Department, Sapienza University of Rome,
00198 Rome, Italy
| |
Collapse
|
38
|
Al-hussaniy HA, Alburghaif AH, alkhafaje Z, AL-Zobaidy MAHJ, Alkuraishy HM, Mostafa-Hedeab G, Azam F, Al-Samydai AM, Al-tameemi ZS, Naji MA. Chemotherapy-induced cardiotoxicity: a new perspective on the role of Digoxin, ATG7 activators, Resveratrol, and herbal drugs. J Med Life 2023; 16:491-500. [PMID: 37305823 PMCID: PMC10251384 DOI: 10.25122/jml-2022-0322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/22/2022] [Indexed: 06/13/2023] Open
Abstract
Cancer is a major public health problem, and chemotherapy plays a significant role in the management of neoplastic diseases. However, chemotherapy-induced cardiotoxicity is a serious side effect secondary to cardiac damage caused by antineoplastic's direct and indirect toxicity. Currently, there are no reliable and approved methods for preventing or treating chemotherapy-induced cardiotoxicity. Understanding the mechanisms of chemotherapy-induced cardiotoxicity may be vital to improving survival. The independent risk factors for developing cardiotoxicity must be considered to prevent myocardial damage without decreasing the therapeutic efficacy of cancer treatment. This systematic review aimed to identify and analyze the evidence on chemotherapy-induced cardiotoxicity, associated risk factors, and methods to decrease or prevent it. We conducted a comprehensive search on PubMed, Google Scholar, and Directory of Open Access Journals (DOAJ) using the following keywords: "doxorubicin cardiotoxicity", "anthracycline cardiotoxicity", "chemotherapy", "digoxin decrease cardiotoxicity", "ATG7 activators", retrieving 59 articles fulfilling the inclusion criteria. Therapeutic schemes can be changed by choosing prolonged infusion application over boluses. In addition, some agents like Dexrazoxane can reduce chemotherapy-induced cardiotoxicity in high-risk groups. Recent research found that Digoxin, ATG7 activators, Resveratrol, and other medical substances or herbal compounds have a comparable effect on Dexrazoxane in anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
- Hany Akeel Al-hussaniy
- Department of Pharmacy, Bilad Alrafidain University College, Diyala, Iraq
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| | | | - Zahraa alkhafaje
- Department of Pharmacy, Alfarahidi University College, Baghdad, Iraq
| | | | - Hayder Mutair Alkuraishy
- Department of Clinical Pharmacology, College of Medicine, Almustansria University, Baghdad, Iraq
| | - Gomaa Mostafa-Hedeab
- Pharmacology Department & Health Research Unit, Medical College, Jouf University, Jouf, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Uniazah, Saudi Arabia
| | - Ali Mahmoud Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zahraa Salam Al-tameemi
- Department of Pharmacy, Bilad Alrafidain University College, Diyala, Iraq
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| | - Meena Akeel Naji
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| |
Collapse
|
39
|
Lisi DM. Comment on: AGS And NIA bench-to bedside conference summary: Cancer and cardiovascular disease. J Am Geriatr Soc 2023; 71:1670-1675. [PMID: 36929848 DOI: 10.1111/jgs.18332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Affiliation(s)
- Donna M Lisi
- Independent Consultant, Somerset, New Jersey, USA
| |
Collapse
|
40
|
Grosu L, Grosu AI, Crisan D, Zlibut A, Perju-Dumbrava L. Parkinson's disease and cardiovascular involvement: Edifying insights (Review). Biomed Rep 2023; 18:25. [PMID: 36846617 PMCID: PMC9944619 DOI: 10.3892/br.2023.1607] [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: 12/11/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative illnesses, and is a major healthcare burden with prodigious consequences on life-quality, morbidity, and survival. Cardiovascular diseases are the leading cause of mortality worldwide and growing evidence frequently reports their co-existence with PD. Cardiac dysautonomia due to autonomic nervous system malfunction is the most prevalent type of cardiovascular manifestation in these patients, comprising orthostatic and postprandial hypotension, along with supine and postural hypertension. Moreover, many studies have endorsed the risk of patients with PD to develop ischemic heart disease, heart failure and even arrhythmias, but the underlying mechanisms are not entirely clear. As importantly, the medication used in treating PD, such as levodopa, dopamine agonists or anticholinergic agents, is also responsible for cardiovascular adverse reactions, but further studies are required to elucidate the underlying mechanisms. The purpose of this review was to provide a comprehensive overview of current available data regarding the overlapping cardiovascular disease in patients with PD.
Collapse
Affiliation(s)
- Laura Grosu
- Department of Neurology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Neurology, Municipal Clinical Hospital, 400139 Cluj-Napoca, Romania
| | - Alin Ionut Grosu
- Department of Internal Medicine, 5th Medical Clinic, Iuliu Hatieganu University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania
- Department of Cardiology, Municipal Clinical Hospital, 400139 Cluj-Napoca, Romania
| | - Dana Crisan
- Department of Internal Medicine, 5th Medical Clinic, Iuliu Hatieganu University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania
- Department of Internal Medicine, Municipal Clinical Hospital, 400139 Cluj-Napoca, Romania
| | - Alexandru Zlibut
- Department of Internal Medicine, 5th Medical Clinic, Iuliu Hatieganu University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania
- Department of Cardiology, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania
| | - Lacramioara Perju-Dumbrava
- Department of Neurology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
41
|
Hamblin MR. Photobiomodulation Therapy for Treatment of Extravasation Injuries in Cancer Chemotherapy. Photobiomodul Photomed Laser Surg 2023; 41:1-2. [PMID: 36629901 DOI: 10.1089/photob.2022.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg-Doornfontein Campus, Doornfontein, South Africa
| |
Collapse
|
42
|
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
|