1
|
Moustafa HAM, El-Dakroury WA, Ashraf A, Abulsoud AI, Elshaer SS, Abdelmaksoud NM, Rizk NI, Mageed SSA, Zaki MB, Mansour RM, Mohammed OA, Abd-Elmawla MA, Abdel-Reheim MA, Doghish AS. SNP's use as a potential chemotoxicity stratification tool in breast cancer: from bench to clinic. Funct Integr Genomics 2025; 25:93. [PMID: 40261508 DOI: 10.1007/s10142-025-01602-4] [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: 01/15/2025] [Revised: 03/22/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Breast cancer (BC) remains one of the most prevalent malignancies affecting women worldwide, necessitating ongoing research to improve treatment outcomes and minimize adverse effects associated with chemotherapy. This article explores the role of genetic variations, particularly single nucleotide polymorphisms (SNPs), in influencing the efficacy and toxicity of chemotherapeutic agents used in BC treatment. It highlights the impact of polymorphisms in drug metabolism and transport genes, such as UDP-glucuronosyltransferase 1A1 (UGT1A1), carbonyl reductase 1 (CBR1), and ATP-binding cassette multidrug transporter (ABCB1) on the risk of adverse effects, including cardiotoxicity and hematological toxicities. By identifying specific SNPs associated with drug response and toxicity, this research underscores the potential for personalized medicine approaches to optimize treatment regimens, enhance therapeutic efficacy, and minimize side effects in BC patients. The findings advocate for the integration of genetic screening in clinical practice to improve patient outcomes and tailor chemotherapy based on individual genetic profiles.
Collapse
Affiliation(s)
- Hebatallah Ahmed Mohamed Moustafa
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Cairo, 11829, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Alaa Ashraf
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Cairo, 11829, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Shereen Saeid Elshaer
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, 11823, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat, Sadat, City, Menoufia, 32897, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Menoufia National University, km Cairo- Alexandria Agricultural Road, Menofia, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| |
Collapse
|
2
|
Fazzini L, Campana N, Cossu S, Deidda M, Madaudo C, Quagliariello V, Maurea N, Di Lisi D, Novo G, Zito C, Cadeddu Dessalvi C. Genetic Background in Patients with Cancer Therapy-Induced Cardiomyopathy. J Clin Med 2025; 14:1286. [PMID: 40004816 PMCID: PMC11856774 DOI: 10.3390/jcm14041286] [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: 01/12/2025] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Emerging evidence indicates that specific genetic variants are associated with an increased risk of toxicity from anticancer treatments and cancer-related cardiovascular complications. These genetic factors influence drug metabolism, efficacy, and susceptibility to adverse effects. For cancer patients, the genetic background can have two major cardiovascular implications, namely therapy-related cardiotoxicity and cancer-related cardiovascular complications. Baseline risk stratification is essential to identify higher-risk individuals and ensure they receive appropriate preventive and therapeutic interventions and more frequent follow-up. Current guidelines recommend stratification based on cardiovascular risk factors, but these factors alone cannot accurately define individual risk. Genetic background has been shown to enhance risk stratification. Beyond rare genetic variants, recent genome-wide association studies have identified single nucleotide polymorphisms implicated in cancer therapy toxicity. Despite their current limitations, polygenic risk scores are expected to play a significant role in risk stratification. This review aims to summarize the current evidence on the role of the genetic background of patients with cancer treated with potentially cardiotoxic drugs who develop cardiotoxicity, aiming to provide insights to refine risk stratification further and tailor the management of these patients.
Collapse
Affiliation(s)
- Luca Fazzini
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Nicola Campana
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Stefano Cossu
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| | - Cristina Madaudo
- Cardiology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital ‘Paolo Giaccone’, University of Palermo, 90133 Palermo, Italy (D.D.L.); (G.N.)
| | - Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (N.M.)
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (N.M.)
| | - Daniela Di Lisi
- Cardiology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital ‘Paolo Giaccone’, University of Palermo, 90133 Palermo, Italy (D.D.L.); (G.N.)
| | - Giuseppina Novo
- Cardiology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University Hospital ‘Paolo Giaccone’, University of Palermo, 90133 Palermo, Italy (D.D.L.); (G.N.)
| | - Concetta Zito
- Cardiology Unit, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, University of Messina, 98122 Messina, Italy;
| | - Christian Cadeddu Dessalvi
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (L.F.); (N.C.); (S.C.); (M.D.)
| |
Collapse
|
3
|
Wang B, Wang J, Liu C, Li C, Meng T, Chen J, Liu Q, He W, Liu Z, Zhou Y. Ferroptosis: Latest evidence and perspectives on plant-derived natural active compounds mitigating doxorubicin-induced cardiotoxicity. J Appl Toxicol 2025; 45:135-158. [PMID: 39030835 DOI: 10.1002/jat.4670] [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: 06/08/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy drug widely used in clinical settings, acting as a first-line treatment for various malignant tumors. However, its use is greatly limited by the cardiotoxicity it induces, including doxorubicin-induced cardiomyopathy (DIC). The mechanisms behind DIC are not fully understood, but its potential biological mechanisms are thought to include oxidative stress, inflammation, energy metabolism disorders, mitochondrial damage, autophagy, apoptosis, and ferroptosis. Recent studies have shown that cardiac injury induced by DOX is closely related to ferroptosis. Due to their high efficacy, availability, and low side effects, natural medicine treatments hold strong clinical potential. Currently, natural medicines have been shown to mitigate DOX-induced ferroptosis and ease DIC through various functions such as antioxidation, iron ion homeostasis correction, lipid metabolism regulation, and mitochondrial function improvement. Therefore, this review summarizes the mechanisms of ferroptosis in DIC and the regulation by natural plant products, with the expectation of providing a reference for future research and development of inhibitors targeting ferroptosis in DIC. This review explores the mechanisms of ferroptosis in doxorubicin-induced cardiomyopathy (DIC) and summarizes how natural plant products can alleviate DIC by inhibiting ferroptosis through reducing oxidative stress, correcting iron ion homeostasis, regulating lipid metabolism, and improving mitochondrial function.
Collapse
Affiliation(s)
- Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wang He
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Ye H, Wu L, Liu Y. Iron metabolism in doxorubicin-induced cardiotoxicity: From mechanisms to therapies. Int J Biochem Cell Biol 2024; 174:106632. [PMID: 39053765 DOI: 10.1016/j.biocel.2024.106632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Doxorubicin (DOX) is an anti-tumor agent for chemotherapy, but its use is often hindered by the severe and life-threatening side effect of cardiovascular toxicity. In recent years, studies have focused on dysregulated iron metabolism and ferroptosis, a unique type of cell death induced by iron overload, as key players driving the development of DOX-induced cardiotoxicity (DIC). Recent advances have demonstrated that DOX disturbs normal cellular iron metabolism, resulting in excessive iron accumulation and ferroptosis in cardiomyocytes. This review will explore how dysregulated iron homeostasis and ferroptosis drive the progression of DIC. We will also discuss the current approaches to target iron metabolism and ferroptosis to mitigate DIC. Besides, we will discuss the limitations and challenges for clinical translation for these therapeutic regimens.
Collapse
Affiliation(s)
- Hua Ye
- Department of Burns & Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China.
| | - Lin Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yanmei Liu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
5
|
Qi Y, Wei Y, Li L, Ge H, Wang Y, Zeng C, Ma F. Genetic factors in the pathogenesis of cardio-oncology. J Transl Med 2024; 22:739. [PMID: 39103883 DOI: 10.1186/s12967-024-05537-5] [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: 04/07/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024] Open
Abstract
In recent years, with advancements in medicine, the survival period of patients with tumours has significantly increased. The adverse effects of tumour treatment on patients, especially cardiac toxicity, have become increasingly prominent. In elderly patients with breast cancer, treatment-related cardiovascular toxicity has surpassed cancer itself as the leading cause of death. Moreover, in recent years, an increasing number of novel antitumour drugs, such as multitargeted agents, antibody‒drug conjugates (ADCs), and immunotherapies, have been applied in clinical practice. The cardiotoxicity induced by these drugs has become more pronounced, leading to a complex and diverse mechanism of cardiac damage. The risks of unintended cardiovascular toxicity are increased by high-dose anthracyclines, immunotherapies, and concurrent radiation, in addition to traditional cardiovascular risk factors such as smoking, hypertension, diabetes, hyperlipidaemia, and obesity. However, these factors do not fully explain why only a subset of individuals experience treatment-related cardiac toxicity, whereas others with similar clinical features do not. Recent studies indicate that genetics play a significant role in susceptibility to the development of cardiovascular toxicity from cancer therapies. These genes are involved in drug metabolism, oxidative damage, cardiac dysfunction, and other processes. Moreover, emerging evidence suggests that epigenetics also plays a role in drug-induced cardiovascular toxicity. We conducted a review focusing on breast cancer as an example to help oncologists and cardiologists better understand the mechanisms and effects of genetic factors on cardiac toxicity. In this review, we specifically address the relationship between genetic alterations and cardiac toxicity, including chemotherapy-related genetic changes, targeted therapy-related genetic changes, and immune therapy-related genetic changes. We also discuss the role of epigenetic factors in cardiac toxicity. We hope that this review will improve the risk stratification of patients and enable therapeutic interventions that mitigate these unintended adverse consequences of life-saving cancer treatments.
Collapse
Affiliation(s)
- Yalong Qi
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China
| | - Yuhan Wei
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China
| | - Lixi Li
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China
| | - Hewei Ge
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China
| | - Yuanyi Wang
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China
| | - Cheng Zeng
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China
| | - Fei Ma
- Department of Medical Oncology, Cancer Hospital, National Cancer Center, National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Pan jia yuan nan Road 17, Beijing, 100021, China.
| |
Collapse
|
6
|
Hwang HJ, Han SA, Sohn IS. Breast Cancer and Therapy-Related Cardiovascular Toxicity. J Breast Cancer 2024; 27:147-162. [PMID: 38769686 PMCID: PMC11221208 DOI: 10.4048/jbc.2024.0085] [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: 04/02/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
The global incidence of breast cancer is on the rise, a trend also observed in South Korea. However, thanks to the rapid advancements in anticancer therapies, survival rates are improving. Consequently, post-treatment health and quality of life for breast cancer survivors are emerging as significant concerns, particularly regarding treatment-related cardiotoxicity. In this review, we delve into the cardiovascular complications associated with breast cancer treatment, explore surveillance protocols for early detection and diagnosis of late complications, and discuss protective strategies against cardiotoxicity in breast cancer patients undergoing anticancer therapy, drawing from multiple guidelines.
Collapse
Affiliation(s)
- Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Sang-Ah Han
- Department of Surgery, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | - Il Suk Sohn
- Department of Cardiology, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Korea.
| |
Collapse
|
7
|
Wu L, Zhang Y, Wang G, Ren J. Molecular Mechanisms and Therapeutic Targeting of Ferroptosis in Doxorubicin-Induced Cardiotoxicity. JACC Basic Transl Sci 2024; 9:811-826. [PMID: 39070280 PMCID: PMC11282888 DOI: 10.1016/j.jacbts.2023.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 07/30/2024]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, has received increasing attention for its pathophysiologic contribution to the onset and development of doxorubicin-induced cardiotoxicity. Moreover, modulation of ferroptosis with specific inhibitors may provide new therapeutic opportunities for doxorubicin-induced cardiotoxicity. Here, we will review the molecular mechanisms and therapeutic promise of targeting ferroptosis in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Lin Wu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Guizhen Wang
- Department of Emergency, Shanghai Tenth People’s Hospital, School of Medicine Tongji University, Shanghai, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
8
|
Vaitiekiene A, Kulboke M, Bieseviciene M, Jankauskas A, Bartnykaite A, Rinkuniene D, Strazdiene I, Lidziute E, Jankauskaite D, Gaidamavicius I, Bucius P, Lapinskas T, Gerbutavicius R, Juozaityte E, Vaskelyte JJ, Vaitiekus D, Sakalyte G. T1 Mapping in Cardiovascular Magnetic Resonance-A Marker of Diffuse Myocardial Fibrosis in Patients Undergoing Hematopoietic Stem Cell Transplantation. J Pers Med 2024; 14:412. [PMID: 38673039 PMCID: PMC11051481 DOI: 10.3390/jpm14040412] [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: 03/11/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction: Hematopoietic stem cell transplantation (HSCT) recipients are at increased risk of cardiovascular diseases. In our study, we aimed to find subclinical changes in myocardial tissue after HSCT with the help of cardiovascular magnetic resonance (CMR) tissue imaging techniques. Methods: The data of 44 patients undergoing autologous and allogeneic HSCT in the Hospital of Lithuanian University of Health Sciences Kaunas Clinics from October 2021 to February 2023 were analyzed. Bioethics approval for the prospective study was obtained (No BE-2-96). CMR was performed two times: before enrolling for the HSCT procedure (before starting mobilization chemotherapy for autologous HSCT and before starting the conditioning regimen for allogeneic HSCT) and 12 ± 1 months after HSCT. LV end-diastolic volume, LV end-systolic volume, LV mass and values indexed to body surface area (BSA), and LV ejection fraction were calculated. T1 and T2 mapping values were measured. Results: There was a statistically significant change in T1 mapping values. Before HSCT, mean T1 mapping was 1226.13 ± 39.74 ms, and after HSCT, it was 1248.70 ± 41.07 ms (p = 0.01). The other parameters did not differ significantly. Conclusions: Increases in T1 mapping values following HSCT can show the progress of diffuse myocardial fibrosis and may reflect subclinical injury. T2 mapping values remain the same and do not show edema and active inflammation processes at 12 months after HSCT.
Collapse
Affiliation(s)
- Audrone Vaitiekiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Migle Kulboke
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Monika Bieseviciene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Antanas Jankauskas
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Agne Bartnykaite
- Oncology Research Laboratory, Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Diana Rinkuniene
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Igne Strazdiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Emilija Lidziute
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Darija Jankauskaite
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Ignas Gaidamavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Paulius Bucius
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Tomas Lapinskas
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Rolandas Gerbutavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Elona Juozaityte
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Jolanta Justina Vaskelyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Domas Vaitiekus
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Gintare Sakalyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| |
Collapse
|
9
|
Vaitiekiene A, Kulboke M, Bieseviciene M, Bartnykaite A, Kireilis B, Rinkuniene D, Jankauskas A, Zemaitis J, Gaidamavicius I, Gerbutavicius R, Vaitiekus D, Vaskelyte JJ, Sakalyte G. Early Impact of Mobilization Process on Cardiac Function and Size in Patients Undergoing Autologous Hematopoietic Stem Cell Transplantation. J Clin Med 2024; 13:773. [PMID: 38337467 PMCID: PMC10856069 DOI: 10.3390/jcm13030773] [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: 12/14/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Background: The hematopoietic stem cell transplantation (HSCT) process is known to cause cardiac toxicity of different grades. In this paper, we aimed to evaluate the impact of mobilization procedure of hematopoietic stem cells for autologous HSCT process for left and right ventricle sizes and functions. Material and Methods: The data of 47 patients undergoing autologous HSCT were analyzed. All patients underwent hematopoietic stem cell mobilization with chemotherapy and filgrastim at 10 µg/kg/d. Echocardiography was performed two times: before enrolling in the transplantation process and after mobilization before the conditioning regimen for transplantation. Changes in left and right ventricle (RV) diameter and systolic and diastolic function of the left ventricle and systolic function of the RV were measured. Results: A statistically significant difference was observed in the change of right ventricular function (S')-it slightly decreased. Mean S' before mobilization was 13.93 ± 2.85 cm/s, and after mobilization it was 12.19 ± 2.64 cm/s (p = 0.003). No statistically significant change in left ventricular diameter and systolic and diastolic function and RV diameter was observed. Conclusions: The mobilization procedure in patients undergoing autologous HSCT is associated with reduced RV systolic function. S' could be used as a reliable tool to evaluate early cardiotoxicity in HSCT patients and guide further follow-up.
Collapse
Affiliation(s)
- Audrone Vaitiekiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
| | - Migle Kulboke
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Monika Bieseviciene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
| | - Agne Bartnykaite
- Oncology Research Laboratory, Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Benas Kireilis
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Diana Rinkuniene
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Antanas Jankauskas
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| | - Justinas Zemaitis
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
| | - Ignas Gaidamavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Rolandas Gerbutavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Domas Vaitiekus
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Jolanta Justina Vaskelyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| | - Gintare Sakalyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| |
Collapse
|
10
|
Balaji S, Antony AK, Tonchev H, Scichilone G, Morsy M, Deen H, Mirza I, Ali MM, Mahmoud AM. Racial Disparity in Anthracycline-induced Cardiotoxicity in Breast Cancer Patients. Biomedicines 2023; 11:2286. [PMID: 37626782 PMCID: PMC10452913 DOI: 10.3390/biomedicines11082286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer has become the most common cancer in the US and worldwide. While advances in early detection and treatment have resulted in a 40% reduction in breast cancer mortality, this reduction has not been achieved uniformly among racial groups. A large percentage of non-metastatic breast cancer mortality is related to the cardiovascular effects of breast cancer therapies. These effects appear to be more prevalent among patients from historically marginalized racial/ethnic backgrounds, such as African American and Hispanic individuals. Anthracyclines, particularly doxorubicin and daunorubicin, are the first-line treatments for breast cancer patients. However, their use is limited by their dose-dependent and cumulative cardiotoxicity, manifested by cardiomyopathy, ischemic heart disease, arrhythmias, hypertension, thromboembolic disorders, and heart failure. Cardiotoxicity risk factors, such as genetic predisposition and preexisting obesity, diabetes, hypertension, and heart diseases, are more prevalent in racial/ethnic minorities and undoubtedly contribute to the risk. Yet, beyond these risk factors, racial/ethnic minorities also face unique challenges that contribute to disparities in the emerging field of cardio-oncology, including socioeconomic factors, food insecurity, and the inability to access healthcare providers, among others. The current review will address genetic, clinical, and social determinants that potentially contribute to this disparity.
Collapse
Affiliation(s)
- Swetha Balaji
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Antu K. Antony
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Harry Tonchev
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Giorgia Scichilone
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Mohammed Morsy
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Hania Deen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Imaduddin Mirza
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Mohamed M. Ali
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Abeer M. Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
- Department of Kinesiology, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Bagdasaryan AA, Chubarev VN, Smolyarchuk EA, Drozdov VN, Krasnyuk II, Liu J, Fan R, Tse E, Shikh EV, Sukocheva OA. Pharmacogenetics of Drug Metabolism: The Role of Gene Polymorphism in the Regulation of Doxorubicin Safety and Efficacy. Cancers (Basel) 2022; 14:5436. [PMID: 36358854 PMCID: PMC9659104 DOI: 10.3390/cancers14215436] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Breast cancer (BC) is the prevailing malignancy and major cause of cancer-related death in females. Doxorubicin is a part of BC neoadjuvant and adjuvant chemotherapy regimens. The administration of anthracycline derivates, such as doxorubicin, may cause several side effects, including hematological disfunction, gastrointestinal toxicity, hepatotoxicity, nephrotoxicity, and cardiotoxicity. Cardiotoxicity is a major adverse reaction to anthracyclines, and it may vary depending on individual differences in doxorubicin pharmacokinetics. Determination of specific polymorphisms of genes that can alter doxorubicin metabolism was shown to reduce the risk of adverse reactions and improve the safety and efficacy of doxorubicin. Genes which encode cytochrome P450 enzymes (CYP3A4 and CYP2D6), p-glycoproteins (ATP-binding cassette (ABC) family members such as Multi-Drug Resistance 1 (MDR1) protein), and other detoxifying enzymes were shown to control the metabolism and pharmacokinetics of doxorubicin. The effectiveness of doxorubicin is defined by the polymorphism of cytochrome p450 and p-glycoprotein-encoding genes. This study critically discusses the latest data about the role of gene polymorphisms in the regulation of doxorubicin's anti-BC effects. The correlation of genetic differences with the efficacy and safety of doxorubicin may provide insights for the development of personalized medical treatment for BC patients.
Collapse
Affiliation(s)
- Alina A. Bagdasaryan
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Vladimir N. Chubarev
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Elena A. Smolyarchuk
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Vladimir N. Drozdov
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Ivan I. Krasnyuk
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Junqi Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Ruitai Fan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Evgenia V. Shikh
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Healthcare of the Russian Federation (Sechenovskiy University), 8-2 Trubetskaya Str., 119991 Moscow, Russia
| | - Olga A. Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- College of Nursing and Health Sciences, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
12
|
Li MY, Peng LM, Chen XP. Pharmacogenomics in drug-induced cardiotoxicity: Current status and the future. Front Cardiovasc Med 2022; 9:966261. [PMID: 36312261 PMCID: PMC9606405 DOI: 10.3389/fcvm.2022.966261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022] Open
Abstract
Drug-induced cardiotoxicity (DICT) is an important concern of drug safety in both drug development and clinical application. The clinical manifestations of DICT include cardiomyopathy, arrhythmia, myocardial ischemia, heart failure, and a series of cardiac structural and functional changes. The occurrence of DICT has negative impacts on the life quality of the patients, brings additional social and economic burden. It is important to identify the potential factors and explore the mechanisms of DICT. Traditional cardiovascular risk factors can only partially explain the risk of DICT. Pharmacogenomic studies show accumulated evidence of genetics in DICT and suggest the potential to guide precision therapy to reduce risk of cardiotoxicity. The comprehensive application of technologies such as third-generation sequencing, human induced pluripotent stem (iPS) cells and genome editing has promoted the in-depth understanding of the functional role of susceptible genes in DICT. This paper reviewed drugs that cause DICT, the clinical manifestations and laboratory tests, as well as the related content of genetic variations associated with the risk of DICT, and further discussed the implication of new technologies in pharmacogenomics of DICT.
Collapse
Affiliation(s)
- Mo-Yun Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Li-Ming Peng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China,Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Li-Ming Peng
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China,Xiao-Ping Chen
| |
Collapse
|
13
|
Genetic Susceptibility and Mechanisms Underlying the Pathogenesis of Anthracycline-Associated Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5818612. [PMID: 35965684 PMCID: PMC9365594 DOI: 10.1155/2022/5818612] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022]
Abstract
Anthracyclines are chemotherapeutic agents widely used to treat a variety of cancers, and these drugs have revolutionized our management of cancer patients. The dose-dependent cardiotoxicity of anthracyclines, however, remains one of the leading causes of chemotherapy treatment-associated mortality in cancer survivors. Patient threshold doses leading to anthracycline-induced cardiotoxicity (AIC) are highly variable among affected patients. This variability is largely ascribed to genetic variants in individuals' genomes. Here, we briefly discuss the prevailing mechanisms underlying the pathogenesis of AIC, and then, we review the genetic variants, mostly identified through human genetic approaches and identified in cancer survivors. The identification of all genetic susceptibilities and elucidation of underlying mechanisms of AIC can help improve upfront risk prediction assessment for potentially severe cardiotoxicity disease and provide valuable insights into the understanding of AIC pathophysiology, which can be further leveraged to develop targeted pharmacogenetic therapies for those at high risk.
Collapse
|
14
|
Zhang G, Yuan C, Su X, Zhang J, Gokulnath P, Vulugundam G, Li G, Yang X, An N, Liu C, Sun W, Chen H, Wu M, Sun S, Xing Y. Relevance of Ferroptosis to Cardiotoxicity Caused by Anthracyclines: Mechanisms to Target Treatments. Front Cardiovasc Med 2022; 9:896792. [PMID: 35770215 PMCID: PMC9234116 DOI: 10.3389/fcvm.2022.896792] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/24/2022] [Indexed: 12/06/2022] Open
Abstract
Anthracyclines (ANTs) are a class of anticancer drugs widely used in oncology. However, the clinical application of ANTs is limited by their cardiotoxicity. The mechanisms underlying ANTs-induced cardiotoxicity (AIC) are complicated and involve oxidative stress, inflammation, topoisomerase 2β inhibition, pyroptosis, immunometabolism, autophagy, apoptosis, ferroptosis, etc. Ferroptosis is a new form of regulated cell death (RCD) proposed in 2012, characterized by iron-dependent accumulation of reactive oxygen species (ROS) and lipid peroxidation. An increasing number of studies have found that ferroptosis plays a vital role in the development of AIC. Therefore, we aimed to elaborate on ferroptosis in AIC, especially by doxorubicin (DOX). We first summarize the mechanisms of ferroptosis in terms of oxidation and anti-oxidation systems. Then, we discuss the mechanisms related to ferroptosis caused by DOX, particularly from the perspective of iron metabolism of cardiomyocytes. We also present our research on the prevention and treatment of AIC based on ferroptosis. Finally, we enumerate our views on the development of drugs targeting ferroptosis in this emerging field.
Collapse
Affiliation(s)
- Guoxia Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chao Yuan
- Dezhou Second People’s Hospital, Dezhou, China
| | - Xin Su
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianzhen Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Priyanka Gokulnath
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Gururaja Vulugundam
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, Naples, Italy
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Xinyu Yang
- Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na An
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Can Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wanli Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hengwen Chen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shipeng Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Shipeng Sun,
| | - Yanwei Xing
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Yanwei Xing,
| |
Collapse
|
15
|
Kim Y, Seidman JG, Seidman CE. Genetics of cancer therapy-associated cardiotoxicity. J Mol Cell Cardiol 2022; 167:85-91. [PMID: 35358500 PMCID: PMC9107514 DOI: 10.1016/j.yjmcc.2022.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/12/2022] [Accepted: 03/25/2022] [Indexed: 01/03/2023]
Abstract
As the number of cancer survivors has increased significantly over the last decades due to aging of population and development of effective cancer therapies, side effects from cancer therapies have been increasingly recognized. High-dose anthracyclines, immunotherapies, and concurrent radiation, as well as traditional cardiovascular risk factors such as smoking, hypertension, diabetes, hyperlipidemia, and obesity increase risks for unintended cardiovascular toxicity. However, these factors do not fully explain why only a subset of patients develop adverse cardiovascular sequelae from cancer therapies. Recent studies demonstrate that genetics play a substantial role in susceptibility to development of cardiovascular toxicities from cancer therapies. Common single nucleotide polymorphisms in multiple genes involved in various cellular pathways including membrane transport, stress response, and sarcomeres are recognized to increase risks for these toxicities. Pathogenic variants in the genes encoding proteins that comprise sarcomeres also contribute to cardiomyopathy following cancer therapies. Furthermore, genetic manipulations of model systems indicate mechanisms by which cardiotoxicities emerge following cancer immunomodulatory therapies. Continued efforts are needed to enable insights into cardiovascular responsiveness to these multi-targeted therapies, improve risk stratification of patients, and enable therapeutic interventions that limit these unintended adverse consequences from life-saving cancer treatments.
Collapse
Affiliation(s)
- Yuri Kim
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Genetics, Harvard Medical School, Boston, MA, United States of America.
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, United States of America
| | - Christine E Seidman
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, United States of America; Department of Genetics, Harvard Medical School, Boston, MA, United States of America; Howard Hughes Medical Institute, Chevy Chase, MD, United States of America
| |
Collapse
|
16
|
Peña Irun A, Bengochea Botín E, Pariente Rodrigo E. Late anthracycline cardiotoxicity and genetic alteration of iron metabolism. Med Clin (Barc) 2021; 158:499-500. [PMID: 34952712 DOI: 10.1016/j.medcli.2021.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 10/19/2022]
|
17
|
Abstract
The purpose of this review was to systematize data on molecular genetic markers of increased risk of cardiotoxic effects, as well as to search for risk and protective variants of candidate genes. Today, the therapy of malignant neoplasms is based on the use of anthracyclines – drugs of the cytostatic mechanism of action. Along with their effectiveness, these drugs can have a cardiotoxic effect on cardiomyocytes by increasing the amount of reactive oxygen species and disrupting mitochondrial biogenesis. Pathological disorders lead to an increased risk of myocardial dysfunction and a number of other cardiovascular pathologies in patients receiving chemotherapy using anthracyclines. The cardiotoxic effect of anthracyclines leads to cardiomyopathy, heart failure, myocardial infarction, and thrombosis. Early detection of cardiotoxic damage leads to reducing the negative effects of these drugs due to changes in chemotherapy tactics. It is known that the risk of cardiotoxic myocardial damage is genetically determined and controlled by more than 80 genes. In this review, the description of basic molecules such as ATP-binding cassette transporters and solute carrier family (SLC transporters), carbonyl reductase, molecules of antioxidant defense, xenobiotic and iron metabolism was performed. In addition, a special attention is paid to the study of epigenetic and post-translational regulation. The available data are characterized by some inconsistency that may be explained by the ethnic differences of the studied populations. Thus, a more detailed research of various ethnic groups, gene-gene interactions between potential candidate genes and epigenetic regulation is necessary. Thus, understanding the contribution of genetic polymorphism to the development of cardiotoxicity will help to assess the individual risks of cardiovascular pathology in patients with various types of cancer, as well as reduce the risk of myocardial damage by developing individual preventive measures and correcting chemotherapy.
Collapse
|
18
|
Berkman AM, Hildebrandt MA, Landstrom AP. The genetic underpinnings of anthracycline-induced cardiomyopathy predisposition. Clin Genet 2021; 100:132-143. [PMID: 33871046 PMCID: PMC9902211 DOI: 10.1111/cge.13968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Anthracyclines, chemotherapeutic agents that have contributed to significant improvements in cancer survival, also carry risk of both acute and chronic cardiotoxicity. This has led to significantly elevated risks of cardiac morbidity and mortality among cancer survivors treated with these agents. Certain treatment related, demographic, and medical factors increase an individual's risk of anthracycline induced cardiotoxicity; however, significant variability among those affected suggests that there is an underlying genetic predisposition to anthracycline induced cardiotoxicity. The current narrative review seeks to summarize the literature to date that has identified genetic variants associated with anthracycline induced cardiotoxicity. These include variants found in genes that encode proteins associated with anthracycline transportation and metabolism, those that encode proteins associated with the generation of reactive oxygen species, and those known to be associated with cardiac disease. While there is strong evidence that susceptibility to anthracycline induced cardiotoxicity has genetic underpinnings, the majority of work to date has been candidate gene analyses. Future work should focus on genome-wide analyses including genome-wide association and sequencing-based studies to confirm and expand these findings.
Collapse
Affiliation(s)
- Amy M. Berkman
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
| | - Michelle A.T. Hildebrandt
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas, United States
| | - Andrew P. Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
- Department of Cell Biology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
| |
Collapse
|