1
|
Mohamad HE, Askar ME, Shaheen MA, Baraka NM, Mahmoud YK. Sacubitril/valsartan alleviates sunitinib-induced cardiac fibrosis and oxidative stress via improving TXNIP/TRX system and downregulation of NF-ĸB/Wnt/β-catenin/SOX9 signaling. Int Immunopharmacol 2024; 132:111963. [PMID: 38560962 DOI: 10.1016/j.intimp.2024.111963] [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: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
We aimed in this study to investigate the possible cardioprotective effects of sacubitril/valsartan against sunitinib-induced cardiac fibrosis (CF) and oxidative stress via targeting thioredoxin-interacting protein/thioredoxin (TXNIP/TRX) system and nuclear factor-kappa B (NF-κB)/Wingless-related MMTV integration site (Wnt)/β-catenin/Sex-determining region Y box 9 (SOX9) signaling. CF was induced in male Wistar albino rats by cumulative dose of sunitinib (300 mg/kg, given over 4 weeks as: 25 mg/kg orally, three times a week), which were co-treated with sacubitril/valsartan (68 mg/kg/day, orally) for four weeks. Significant elevation in blood pressure, cardiac inflammatory and fibrotic markers besides cardiac dysfunction were observed. These alterations were associated with disruption of TXNIP/TRX system, upregulation of NF-κB/Wnt/β-catenin/SOX9 pathway along with marked increase in lysyl oxidase (LOX) and matrix metalloproteinase-1 (MMP-1) expressions and extensive deposition of collagen fibers in cardiac tissues. Luckily, sacubitril/valsartan was able to reverse all of the aforementioned detrimental effects in sunitinib-administered rats. These findings illustrate a potential role of sacubitril/valsartan in alleviating CF and oxidative stress induced by sunitinib via antioxidant, anti-inflammatory and antifibrotic properties. These remarkable effects of sacubitril/valsartan were mediated by its ability to improve TXNIP/TRX system and downregulate NF-κB/Wnt/β-catenin/SOX9 signaling in addition to decreasing LOX and MMP-1 expressions in cardiac tissues. In summary, this study highlights sacubitril/valsartan as a potential therapeutic agent in mitigating CF and oxidative stress especially in cancer cases treated with sunitinib.
Collapse
Affiliation(s)
- Hoda E Mohamad
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mervat E Askar
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Mohamed A Shaheen
- Department of Histology & Cell Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Nourhan M Baraka
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Yasmin K Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
2
|
Sun S, Qin J, Liao W, Gao X, Shang Z, Luo D, Xiong S. Mitochondrial Dysfunction in Cardiotoxicity Induced by BCR-ABL1 Tyrosine Kinase Inhibitors -Underlying Mechanisms, Detection, Potential Therapies. Cardiovasc Toxicol 2023; 23:233-254. [PMID: 37479951 DOI: 10.1007/s12012-023-09800-x] [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] [Received: 05/03/2023] [Accepted: 07/08/2023] [Indexed: 07/23/2023]
Abstract
The advent of BCR-ABL tyrosine kinase inhibitors (TKIs) targeted therapy revolutionized the treatment of chronic myeloid leukemia (CML) patients. Mitochondria are the key organelles for the maintenance of myocardial tissue homeostasis. However, cardiotoxicity associated with BCR-ABL1 TKIs can directly or indirectly cause mitochondrial damage and dysfunction, playing a pivotal role in cardiomyocytes homeostatic system and putting the cancer survivors at higher risk. In this review, we summarize the cardiotoxicity caused by BCR-ABL1 TKIs and the underlying mechanisms, which contribute dominantly to the damage of mitochondrial structure and dysfunction: endoplasmic reticulum (ER) stress, mitochondrial stress, damage of myocardial cell mitochondrial respiratory chain, increased production of mitochondrial reactive oxygen species (ROS), and other kinases and other potential mechanisms of cardiotoxicity induced by BCR-ABL1 TKIs. Furthermore, detection and management of BCR-ABL1 TKIs will promote our rational use, and cardioprotection strategies based on mitochondria will improve our understanding of the cardiotoxicity from a mitochondrial perspective. Ultimately, we hope shed light on clinical decision-making. By integrate and learn from both research and practice, we will endeavor to minimize the mitochondria-mediated cardiotoxicity and reduce the adverse sequelae associated with BCR-ABL1 TKIs.
Collapse
Affiliation(s)
- Sheng Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China
| | - Jiqiu Qin
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhoubiao Shang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dehua Luo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaoquan Xiong
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Medical Oncology, Hospital of Chengdu University of Traditioanal Chinese Medicine, Chengdu, 610075, Sichuan Province, China.
| |
Collapse
|
3
|
Li Y, Yan J, Zhao Q, Zhang Y, Zhang Y. ATF3 promotes ferroptosis in sorafenib-induced cardiotoxicity by suppressing Slc7a11 expression. Front Pharmacol 2022; 13:904314. [PMID: 36210815 PMCID: PMC9537618 DOI: 10.3389/fphar.2022.904314] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Sorafenib is the unique recommended molecular-targeted drug for advanced hepatocellular carcinoma, but its clinical use is limited due to cardiotoxicity. As sorafenib is an efficient ferroptosis inducer, the pathogenesis of this compound to ferroptosis-mediated cardiotoxicity is worth further study. Mice were administered 30 mg/kg sorafenib intraperitoneally for 2 weeks to induce cardiac dysfunction and Ferrostatin-1 (Fer-1) was used to reduce ferroptosis of mice with sorafenib-induced cardiotoxicity. Sorafenib reduced levels of anti-ferroptotic markers involving Slc7a11 and glutathione peroxidase 4 (GPX4), increased malonaldehyde malondialdehyde, apart from causing obvious mitochondria damage, which was alleviated by Fer-1. In vitro experiments showed that Fer-1 inhibited lipid peroxidation and injury of H9c2 cardiomyoblasts induced by sorafenib. Both in vitro and in vivo experiments confirmed that the expression of Slc7a11 was down regulated in sorafenib-induced cardiotoxicity, which can be partially prevented by treatment with Fer-1. Overexpression of Slc7a11 protected cells from ferroptosis, while knock-down of Slc7a11 made cardiomyoblasts sensitive to ferroptosis caused by sorafenib. Finally, by comparing data from the GEO database, we found that the expression of ATF3 was significantly increased in sorafenib treated human cardiomyocytes. In addition, we demonstrated that ATF3 suppressed Slc7a11 expression and promoted ferroptosis. Based on these findings, we concluded that ATF3/Slc7a11 mediated ferroptosis is one of the key mechanisms leading to sorafenib-induced cardiotoxicity. Targeting ferroptosis may be a novel therapeutic approach for preventing sorafenib-induced cardiotoxicity in the future.
Collapse
Affiliation(s)
- Yilan Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Jingru Yan
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Qianqian Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
| | - Yan Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yao Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- *Correspondence: Yao Zhang,
| |
Collapse
|
4
|
Jiang H, Wang C, Zhang A, Li Y, Li J, Li Z, Yang X, Hou Y. ATF4 protects against sorafenib-induced cardiotoxicity by suppressing ferroptosis. Biomed Pharmacother 2022; 153:113280. [PMID: 35724508 DOI: 10.1016/j.biopha.2022.113280] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022] Open
Abstract
Sorafenib (SOR) is an effective chemotherapy drug for hepatocellular carcinoma, renal cell carcinoma, and differentiated thyroid carcinoma. However, a long-standing clinical issue associated with SOR use is an increased risk of cardiotoxicity, but the underlying mechanisms remain obscure. Here we report that ferroptosis of cardiomyocytes is responsible for SOR-induced cardiotoxicity. The specific ferroptosis inhibitor ferrostatin-1 and deferoxamine mesylate, an iron chelator, significantly alleviate SOR-induced cardiac damage. RNA-sequencing revealed that endoplasmic reticulum (ER) stress and the unfolded protein response were predominately activated, which might be attributed to the lipid reactive oxygen species-mediated perturbation of the ER. Activating transcription factor 4 (ATF4) is one of the most significantly up-regulated genes, knockdown of ATF4 exacerbates cardiomyocyte ferroptosis induced by SOR, while overexpression of ATF4 promotes cell survival. Mice with AAV-mediated ATF4 knockdown exhibit lipid peroxidation and more severe cardiomyopathy. Further experiments demonstrated that ATF4 exerts its protective role by elevating SLC7A11 expression, a transport subunit of system Xc-, which promotes cystine uptake and glutathione biosynthesis. The cardioprotective effect of ATF4 was diminished by SLC7A11 knockdown in cardiomyocytes subjected to SOR treatment. Taken together, these findings show that ferroptosis of cardiomyocytes is an important cause of SOR-related cardiotoxicity. ATF4 acts as a key regulator to promote cardiomyocytes survival by up-regulation of SLC7A11 and suppression of ferroptosis.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China; Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Cong Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong 250014, China
| | - An Zhang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Yufeng Li
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Jianping Li
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Zhan Li
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong 250014, China
| | - Xin Yang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Yinglong Hou
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong 250014, China.
| |
Collapse
|
5
|
Morelli MB, Bongiovanni C, Da Pra S, Miano C, Sacchi F, Lauriola M, D’Uva G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front Cardiovasc Med 2022; 9:847012. [PMID: 35497981 PMCID: PMC9051244 DOI: 10.3389/fcvm.2022.847012] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.
Collapse
Affiliation(s)
| | - Chiara Bongiovanni
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Silvia Da Pra
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gabriele D’Uva
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- *Correspondence: Gabriele D’Uva,
| |
Collapse
|
6
|
Dong M, Wang R, Sun P, Zhang D, Zhang Z, Zhang J, Tse G, Zhong L. Clinical significance of hypertension in patients with different types of cancer treated with antiangiogenic drugs. Oncol Lett 2021; 21:315. [PMID: 33692847 PMCID: PMC7933774 DOI: 10.3892/ol.2021.12576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
Hypertension is a common comorbidity in patients receiving antiangiogenic therapy. Prior studies have reported worsening or new-onset hypertension as an adverse event of antiangiogenetic therapy, which can be managed by dose reduction or discontinuation of the culprit medication. By contrast, other studies have found that the occurrence of hypertension is a potential biomarker associated with greater efficacy of antiangiogenic therapy and predicts improved survival. At present, there is no consensus on the effects of hypertension in patients treated with antiangiogenic drugs. The present study reviewed the relationship between antiangiogenic drugs and hypertension in different types of cancer. It was demonstrated that the use of antiangiogenic drugs was associated with an increased risk of hypertension in most types of solid cancers. There was no significant difference in the incidence of hypertension between monoclonal antibody and small-molecule tyrosine kinase inhibitor treatments. Hypertension was more likely to occur in patients younger than 75 years old, female, and those with no history of bevacizumab use. Discontinuation or death caused by hypertension was rare, although previous studies have reported that hypertension was a risk factor for acute and chronic cardiovascular diseases and ischemic stroke. Of note, the early development of hypertension may serve as a potential biomarker associated with greater efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Mei Dong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Rujian Wang
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Ping Sun
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Dongxia Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Zhenzhen Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Jing Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Lin Zhong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
7
|
Abstract
The anticancer drug dasatinib (Sprycel) is a BCR-ABL1-targeted tyrosine kinase inhibitor used in treating chronic myelogenous leukemia that has been shown in clinical trials to display cardiovascular toxicities. While dasatinib potently inhibits BCR-ABL1, it is not a highly selective kinase inhibitor and may have off-target effects. A neonatal rat cardiac myocyte model was used to investigate potential mechanisms by which dasatinib damaged myocytes. The anthracycline cardioprotective drug dexrazoxane was shown to be ineffective in preventing dasatinib-induced myocyte damage. Dasatinib treatment increased doxorubicin accumulation in myocytes and doxorubicin-induced myocyte damage, likely through its ability to bind to one or more ABC-type efflux transporters. Dasatinib induced myocyte damage either after a brief treatment that mimicked the clinical situation, or more potently after continuous treatment. Dasatinib slightly induced apoptosis in myocytes as evidenced by increases in caspase-3/7 activity. Dasatinib treatment reduced pERK levels in myocytes most likely through inhibition of RAF, which dasatinib strongly inhibits. Thus, inhibition of the RAF/MEK/ERK pro-survival pathway in the heart may be, in part, a mechanism by which dasatinib induces cardiovascular toxicity.
Collapse
|
8
|
Jiang L, Ping L, Yan H, Yang X, He Q, Xu Z, Luo P. Cardiovascular toxicity induced by anti-VEGF/VEGFR agents: a special focus on definitions, diagnoses, mechanisms and management. Expert Opin Drug Metab Toxicol 2020; 16:823-835. [PMID: 32597258 DOI: 10.1080/17425255.2020.1787986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Vascular endothelial growth factor (VEGF) is a key target in cancer therapy. However, cardiovascular safety has been one of the most challenging aspects of anti-VEGF/VEGF receptor (VEGFR) agent development and therapy. While accurate diagnostic modalities for assessment of cardiac function have been developed over the past few decades, a lack of an optimal definition and precise mechanism still places a significant limit on the effective management of cardiovascular toxicity. AREAS COVERED Here, we report the cardiovascular toxicity profile associated with anti-VEGF/VEGFR agents and summarize the clinical diagnoses as well as management that are already performed in clinical practice or are currently being investigated. Furthermore, the review discusses the potential molecular toxicological mechanisms, which may provide strategies to prevent toxicity and drive drug discovery. EXPERT OPINION Cardiovascular toxicity associated with anti-VEGF/VEGFR agents has been a substantial risk for cancer treatment. To improve its management, the development of guidelines for prevention, monitoring and treatment of cardiovascular toxicity has become a hot topic. The summary of cardiovascular toxicity profile, mechanisms and management given in this review is not only significant for the optimal use of existing anti-VEGF/VEGFR agents to protect patients predisposed to cardiovascular toxicity but is also beneficial for drug development.
Collapse
Affiliation(s)
- Liyu Jiang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Li Ping
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| |
Collapse
|
9
|
Ma W, Liu M, Liang F, Zhao L, Gao C, Jiang X, Zhang X, Zhan H, Hu H, Zhao Z. Cardiotoxicity of sorafenib is mediated through elevation of ROS level and CaMKII activity and dysregulation of calcium homoeostasis. Basic Clin Pharmacol Toxicol 2019; 126:166-180. [DOI: 10.1111/bcpt.13318] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Wenzhuo Ma
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Mei Liu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Fanfan Liang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Lili Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Chenying Gao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xixi Jiang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xin Zhang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Heqin Zhan
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
- Department of Pharmacology College of Pharmacy Xinxiang Medical University Xinxiang Henan 453003 China
| | - Hao Hu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Zhenghang Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| |
Collapse
|
10
|
Sharma A, McKeithan WL, Serrano R, Kitani T, Burridge PW, Del Álamo JC, Mercola M, Wu JC. Use of human induced pluripotent stem cell-derived cardiomyocytes to assess drug cardiotoxicity. Nat Protoc 2018; 13:3018-3041. [PMID: 30413796 PMCID: PMC6502639 DOI: 10.1038/s41596-018-0076-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cardiotoxicity has historically been a major cause of drug removal from the pharmaceutical market. Several chemotherapeutic compounds have been noted for their propensities to induce dangerous cardiac-specific side effects such as arrhythmias or cardiomyocyte apoptosis. However, improved preclinical screening methodologies have enabled cardiotoxic compounds to be identified earlier in the drug development pipeline. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to screen for drug-induced alterations in cardiac cellular contractility, electrophysiology, and viability. We previously established a novel 'cardiac safety index' (CSI) as a metric that can evaluate potential cardiotoxic drugs via high-throughput screening of hiPSC-CMs. This metric quantitatively examines drug-induced alterations in CM function, using several in vitro readouts, and normalizes the resulting toxicity values to the in vivo maximum drug blood plasma concentration seen in preclinical or clinical pharmacokinetic models. In this ~1-month-long protocol, we describe how to differentiate hiPSCs into hiPSC-CMs and subsequently implement contractility and cytotoxicity assays that can evaluate drug-induced cardiotoxicity in hiPSC-CMs. We also describe how to carry out the calculations needed to generate the CSI metric from these quantitative toxicity measurements.
Collapse
Affiliation(s)
- Arun Sharma
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wesley L McKeithan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ricardo Serrano
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA, USA
| | - Tomoya Kitani
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University School of Medicine, Chicago, IL, USA
| | - Juan C Del Álamo
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, CA, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Chaar M, Kamta J, Ait-Oudhia S. Mechanisms, monitoring, and management of tyrosine kinase inhibitors-associated cardiovascular toxicities. Onco Targets Ther 2018; 11:6227-6237. [PMID: 30288058 PMCID: PMC6163027 DOI: 10.2147/ott.s170138] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The tyrosine kinase inhibitor (TKI) drug class is a prominently used option in the treatment of various cancers. Safety evaluation of these drugs has shown evidence of cardiotoxicity of varying frequency and severity between agents; concern has led to updated labeling, warning prescribers of such. This review seeks to clarify the present dangers and investigate cardiotoxic mechanisms of action for each discussed TKI. Dasatinib was connected primarily with an incidence of fluid retention, edema, QT prolongation, and pulmonary hypertension in clinical studies. It is theorized that this is due to a combination of off-target kinase binding and on-target binding of Bcr-Abl, and less likely, mitochondrial induced apoptosis. Studies showed sorafenib to carry the risk of hypertension, QT prolongation, and myocardial infarction. Proposed mechanisms for these side effects include inhibition of proteins, vascular endothelium growth factor receptor, hERG potassium channels, and the RAF/MERK/ERK pro-survival pathway. Finally, lapatinib showed evidence of decreased left ventricular ejection fraction (LVEF) and QT prolongation in clinical studies. The literature attributes these as side effects of on-target ErbB2 binding leading to mitochondrial induced apoptosis. The concern warranted by these findings is in question. Pooled safety data suggest that the overall risk for cardiotoxicity is minimal in dasatinib and lapatinib. Sorafenib seems to carry a moderate concern. For the discussed agents, recommendations agree that routine monitoring via methods such as electroencephalogram, cardiac biomarkers, and blood pressure is warranted during the course of treatment, in addition to a comprehensive collection of past medical history and risk factors to identify those at heightened risk for cardiovascular events.
Collapse
Affiliation(s)
- Maher Chaar
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA,
| | - Jeff Kamta
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA,
| | - Sihem Ait-Oudhia
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA,
| |
Collapse
|
12
|
Manolis AA, Manolis TA, Mikhailidis DP, Manolis AS. Cardiovascular safety of oncologic agents: a double-edged sword even in the era of targeted therapies - Part 2. Expert Opin Drug Saf 2018; 17:893-915. [PMID: 30126303 DOI: 10.1080/14740338.2018.1513489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Patients with cancer are subject to the cardiotoxic effects of cancer therapy. Improved cancer treatments lead to more cancer-survivors, who though are exposed to various forms of cardiovascular (CV) disease (CVD) as they age. Aging patients are at increased risk of developing both malignancy and CVD or they may have survived some form of CVD as a result of effective CV treatments. Furthermore, patients with CVD may develop cancer and require treatment (and vice versa), all contributing to increased morbidity and mortality. The prevalence of both malignancy and CVD will increase due to the trend toward a longer lifespan. AREAS COVERED In part 2 of this review, the discussion of the CV effects of specific oncology drugs is completed with inclusion of additional immunological agents, current hormonal and other agents. Early detection and monitoring of cardiotoxicity, use of biomarkers and other imaging and diagnostic methods and prevention and treatment options are also discussed. EXPERT OPINION As outlined in part 1 of this review, oncologists need to be aware of the CV adverse-effects of their treatments and make careful and expectant clinical decisions, especially in patients with preexisting CVD or CV risk factors. Similarly, cardiologists should consider a detailed previous history of treatment for malignant disease, including prior chemotherapy exposure, dose(s) received, and/or combined modality therapy with chest radiotherapy. Both specialists should collaborate in order to minimize the impact of these two ubiquitous diseases (cancer and CVD) and mitigate the adverse effects of treatment modalities.
Collapse
Affiliation(s)
| | | | - Dimitri P Mikhailidis
- c Department of Clinical Biochemistry , Royal Free Hospital Campus, University College London Medical School , London , UK
| | - Antonis S Manolis
- d Third Department of Cardiology , Athens University School of Medicine , Athens , Greece
| |
Collapse
|
13
|
Yang X, Huang Q, Bardelang D, Wang C, Lee SMY, Wang R. Supramolecular alleviation of cardiotoxicity of a small-molecule kinase inhibitor. Org Biomol Chem 2018; 15:8046-8053. [PMID: 28795750 DOI: 10.1039/c7ob01505d] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Small-molecule kinase inhibitors (SMKIs) have been widely used in the treatment of a variety of cancers due to their clinically demonstrated efficacy. However, the use of some SMKIs, such as sorafenib (SO), has been plagued by their cardiotoxicity that has been frequently observed in treated patients. Herein we report that the encapsulation of SO by a synthetic receptor cucurbit[7]uril (CB[7]) alleviated the inherent cardiotoxicity of SO, as demonstrated in an in vivo zebrafish model. Moreover, the anti-cancer activity of SO was well preserved, upon its encapsulation by CB[7], as demonstrated by both in vitro and in vivo cancer/angiogenesis models. This discovery may provide new insights into a novel supramolecular formulation of SMKIs for the management of their side-effects.
Collapse
Affiliation(s)
- Xue Yang
- State Key Laboratory of Quality Research in Chinese Medicine, and Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China.
| | | | | | | | | | | |
Collapse
|
14
|
Molecular Mechanisms of the Cardiotoxicity of the Proteasomal-Targeted Drugs Bortezomib and Carfilzomib. Cardiovasc Toxicol 2018; 17:237-250. [PMID: 27388042 DOI: 10.1007/s12012-016-9378-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Bortezomib and carfilzomib are anticancer drugs that target the proteasome. However, these agents have been shown to exhibit some specific cardiac toxicities by as yet unknown mechanisms. Bortezomib and carfilzomib are also being used clinically in combination with doxorubicin, which is also cardiotoxic. A primary neonatal rat myocyte model was used to study these cardiotoxic mechanisms. Exposure to submicromolar concentrations of bortezomib and carfilzomib resulted in significant myocyte damage and induced apoptosis. Both bortezomib and carfilzomib inhibited the chymotrypsin-like proteasomal activity of myocyte lysate in the low nanomolar concentration range and exhibited time-dependent inhibition kinetics. The high sensitivity of myocytes, which were determined to contain high specific levels of chymotrypsin-like proteasomal activity, to the damaging effects of bortezomib and carfilzomib was likely due to the inhibition of proteasomal-dependent ongoing sarcomeric protein turnover. A brief preexposure of myocytes to non-toxic nanomolar concentrations of bortezomib or carfilzomib greatly increased doxorubicin-mediated damage, which suggests that the combination of doxorubicin with either bortezomib or carfilzomib may produce more than additive cardiotoxicity. The doxorubicin cardioprotective agent dexrazoxane partially protected myocytes from doxorubicin plus bortezomib or carfilzomib treatment, in spite of the fact that bortezomib and carfilzomib inhibited the dexrazoxane-induced decreases in topoisomerase IIβ protein levels in myocytes. These latter results suggest that the doxorubicin cardioprotective effects of dexrazoxane and the doxorubicin-mediated cardiotoxicity were not exclusively due to targeting of topoisomerase IIβ.
Collapse
|
15
|
Calistri L, Cordopatri C, Nardi C, Gianni E, Marra F, Colagrande S. Sudden cardiac death in a patient with advanced hepatocellular carcinoma with good response to sorafenib treatment: A case report with literature analysis. Mol Clin Oncol 2017; 6:389-396. [PMID: 28451419 DOI: 10.3892/mco.2017.1132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the principal primary liver tumor, representing the third largest cause of cancer-associated death worldwide. The actual reference standard systemic treatment for advanced HCC is represented by sorafenib, a multi-targeted orally active small-molecule tyrosine kinase inhibitor. Sorafenib has exhibited a good general safety profile in multiple clinical trials. However, adverse drug-associated events are common, occasionally severe, and special attention should be paid to cardiovascular adverse events, particularly in patients with risk factors or known heart disease. In the present study, the case of a patient with no known cardiovascular risk factors affected by highly enhancing advanced HCC in cirrhotic liver, who died during successful sorafenib monotherapy, is reported.
Collapse
Affiliation(s)
- Linda Calistri
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence-Azienda Ospedaliero-Universitaria Careggi, I-50134 Florence, Italy
| | - Cesare Cordopatri
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence-Azienda Ospedaliero-Universitaria Careggi, I-50134 Florence, Italy
| | - Cosimo Nardi
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence-Azienda Ospedaliero-Universitaria Careggi, I-50134 Florence, Italy
| | - Elena Gianni
- Department of Experimental and Clinical Medicine, University of Florence, I-50134 Florence, Italy
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, I-50134 Florence, Italy
| | - Stefano Colagrande
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence-Azienda Ospedaliero-Universitaria Careggi, I-50134 Florence, Italy
| |
Collapse
|
16
|
Bronte E, Bronte G, Novo G, Bronte F, Bavetta MG, Lo Re G, Brancatelli G, Bazan V, Natoli C, Novo S, Russo A. What links BRAF to the heart function? New insights from the cardiotoxicity of BRAF inhibitors in cancer treatment. Oncotarget 2016; 6:35589-601. [PMID: 26431495 PMCID: PMC4742127 DOI: 10.18632/oncotarget.5853] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/22/2015] [Indexed: 12/14/2022] Open
Abstract
The RAS-related signalling cascade has a fundamental role in cell. It activates differentiation and survival. It is particularly important one of its molecules, B-RAF. B-RAF has been a central point for research, especially in melanoma. Indeed, it lacked effective therapeutic weapons since the early years of its study. Molecules targeting B-RAF have been developed. Nowadays, two classes of molecules are approved by FDA. Multi-target molecules, such as Sorafenib and Regorafenib, and selective molecules, such as Vemurafenib and Dabrafenib. Many other molecules are still under investigation. Most of them are studied in phase 1 trials. Clinical studies correlate B-RAF inhibitors and QT prolongation. Though this cardiovascular side effect is not common using these drugs, it must be noticed early and recognize its signals. Indeed, Oncologists and Cardiologists should work in cooperation to prevent lethal events, such as fatal arrhythmias or sudden cardiac death. These events could originate from an uncontrolled QT prolongation.
Collapse
Affiliation(s)
- Enrico Bronte
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Bronte
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppina Novo
- Department of Internal Medicine and Cardiovascular Disease, University of Palermo, Palermo, Italy
| | - Fabrizio Bronte
- DiBiMIS, Section of Gastroenterology, University of Palermo, Palermo, Italy
| | | | - Giuseppe Lo Re
- Department of Radiology, University of Palermo, Palermo, Italy
| | | | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", Chieti, Italy
| | - Salvatore Novo
- Department of Internal Medicine and Cardiovascular Disease, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
17
|
Abdel-Rahman O, ElHalawani H, Ahmed H. Risk of Selected Cardiovascular Toxicities in Patients With Cancer Treated With MEK Inhibitors: A Comparative Systematic Review and Meta-Analysis. J Glob Oncol 2015; 1:73-82. [PMID: 28804776 PMCID: PMC5539872 DOI: 10.1200/jgo.2015.000802] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose We conducted a literature-based meta-analysis of the risk of cardiovascular toxicities associated with MEK inhibitors. Methods Eligible trials included randomized phase II and III trials of patients with cancer who were given a mitogen activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor (trametinib, selumetinib, or cobimetinib) and that described events of hypertension and decreased ejection fraction. Results Our search strategy yielded 300 potentially relevant citations from PubMed/MEDLINE, Google Scholar, and Cochrane Central Register of Controlled Trials. After ineligible studies were excluded, a total of 10 clinical trials were considered eligible for the meta-analysis. The relative risk for all grades of hypertension was 1.54 (95% CI, 1.02 to 2.32; P = .05), 1.85 (95% CI, 1.01 to 3.40; P = .05) for high-grade hypertension, and 4.92 (95% CI, 2.93 to 8.25; P < .001) for decreased ejection fraction. Subgroup analysis revealed no difference between trametinib and selumetinib for risk of hypertension. Conclusion Our meta-analysis demonstrated that MEK inhibitor–based treatment is associated with an increased risk of all-grade and high-grade hypertension and asymptomatic decrease in ejection fraction. Clinicians should be aware of this risk and perform regular assessment.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Omar Abdel-Rahman and Hesham ElHalawani, Ain Shams University; and Hoda Ahmed, Nasser Institute, Cairo, Egypt
| | - Hesham ElHalawani
- Omar Abdel-Rahman and Hesham ElHalawani, Ain Shams University; and Hoda Ahmed, Nasser Institute, Cairo, Egypt
| | - Hoda Ahmed
- Omar Abdel-Rahman and Hesham ElHalawani, Ain Shams University; and Hoda Ahmed, Nasser Institute, Cairo, Egypt
| |
Collapse
|
18
|
Doherty KR, Talbert DR, Trusk PB, Moran DM, Shell SA, Bacus S. Structural and functional screening in human induced-pluripotent stem cell-derived cardiomyocytes accurately identifies cardiotoxicity of multiple drug types. Toxicol Appl Pharmacol 2015; 285:51-60. [PMID: 25841593 DOI: 10.1016/j.taap.2015.03.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/31/2022]
Abstract
Safety pharmacology studies that evaluate new drug entities for potential cardiac liability remain a critical component of drug development. Current studies have shown that in vitro tests utilizing human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CM) may be beneficial for preclinical risk evaluation. We recently demonstrated that an in vitro multi-parameter test panel assessing overall cardiac health and function could accurately reflect the associated clinical cardiotoxicity of 4 FDA-approved targeted oncology agents using hiPS-CM. The present studies expand upon this initial observation to assess whether this in vitro screen could detect cardiotoxicity across multiple drug classes with known clinical cardiac risks. Thus, 24 drugs were examined for their effect on both structural (viability, reactive oxygen species generation, lipid formation, troponin secretion) and functional (beating activity) endpoints in hiPS-CM. Using this screen, the cardiac-safe drugs showed no effects on any of the tests in our panel. However, 16 of 18 compounds with known clinical cardiac risk showed drug-induced changes in hiPS-CM by at least one method. Moreover, when taking into account the Cmax values, these 16 compounds could be further classified depending on whether the effects were structural, functional, or both. Overall, the most sensitive test assessed cardiac beating using the xCELLigence platform (88.9%) while the structural endpoints provided additional insight into the mechanism of cardiotoxicity for several drugs. These studies show that a multi-parameter approach examining both cardiac cell health and function in hiPS-CM provides a comprehensive and robust assessment that can aid in the determination of potential cardiac liability.
Collapse
Affiliation(s)
| | | | | | | | - Scott A Shell
- Quintiles, 777 Oakmont Lane Suite 100, Westmont, IL 60559,USA
| | - Sarah Bacus
- Quintiles, 777 Oakmont Lane Suite 100, Westmont, IL 60559,USA
| |
Collapse
|
19
|
Bronte G, Bronte E, Novo G, Pernice G, Lo Vullo F, Musso E, Bronte F, Gulotta E, Rizzo S, Rolfo C, Silvestris N, Bazan V, Novo S, Russo A. Conquests and perspectives of cardio-oncology in the field of tumor angiogenesis-targeting tyrosine kinase inhibitor-based therapy. Expert Opin Drug Saf 2014; 14:253-67. [PMID: 25494575 DOI: 10.1517/14740338.2015.986092] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Angiogenesis is fundamental for tumor development and progression. Hence, anti-angiogenic drugs have been developed to target VEGF and its receptors (VEGFRs). Several tyrosine kinase inhibitors (TKIs) have been developed over the years and others are still under investigation, each anti-VEGFR TKI showing a different cardiotoxic profile. Knowledge of the cardiac side-effects of each drug and the magnitude of their expression and frequency can lead to a specific approach. AREAS COVERED This work reviews the mechanism of action of anti-VEGFR TKIs and the pathophysiological mechanisms leading to cardiotoxicity, followed by close examination of the most important drugs individually. A literature search was conducted on PubMed selecting review articles, original studies and clinical trials, with a focus on Phase III studies. EXPERT OPINION Side-effects on the cardiovascular system could lead both to the worsening of general health status of cancer patients and to the discontinuation of the cancer treatment affecting its efficacy. Cardiologists often have to face new triggers of heart disease in these patients. They need a specific approach, which must be carried out in cooperation with oncologists. It must start before cancer treatment, continue during it and extend after its completion.
Collapse
Affiliation(s)
- Giuseppe Bronte
- University of Palermo, Department of Surgical, Oncological and Oral Sciences , Palermo , Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kawabata M, Umemoto N, Shimada Y, Nishimura Y, Zhang B, Kuroyanagi J, Miyabe M, Tanaka T. Downregulation of stanniocalcin 1 is responsible for sorafenib-induced cardiotoxicity. Toxicol Sci 2014; 143:374-84. [PMID: 25370841 DOI: 10.1093/toxsci/kfu235] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sorafenib is associated with adverse cardiac effects, including left ventricular dysfunction. However, the precise mechanism remains unclear. Here, we aimed to establish the genes responsible for this cardiotoxicity using zebrafish and human cardiomyocytes. Fluorescent cardiac imaging using pigmentless zebrafish with green fluorescent protein hearts revealed that the ventricular dimensions of the longitudinal axis with sorafenib were significantly shorter than those of the control group. Transcriptome analysis of their hearts revealed that stanniocalcin 1 (stc1) was downregulated by sorafenib. stc1 knockdown in zebrafish revealed that reduction of stc1 decreased the longitudinal dimensions of zebrafish ventricles, similar to that which occurs during sorafenib treatment. STC1 downregulation and cytotoxicity were also seen in human cardiomyocytes exposed to sorafenib. To clarify the molecular function of stc1 in sorafenib-induced cardiotoxicity, we focused on oxidative stress in cardiomyocytes treated with sorafenib. Reactive oxygen species (ROS) production significantly increased in both species of human cardiomyocytes and zebrafish exposed to sorafenib and STC1 knockdown compared with the controls. Finally, we found that forced expression of stc1 normalized impairment, decreasing the longitudinal dimensions in zebrafish treated with sorafenib. Our study demonstrated that STC1 plays a protective role against ventricular dysfunction and ROS overproduction, which are induced by sorafenib treatment. We discovered for the first time that STC1 downregulation is responsible for sorafenib-induced cardiotoxicity through activated ROS generation.
Collapse
Affiliation(s)
- Miko Kawabata
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Noriko Umemoto
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Yasuhito Shimada
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-
| | - Yuhei Nishimura
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-
| | - Beibei Zhang
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Junya Kuroyanagi
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Masayuki Miyabe
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan
| | - Toshio Tanaka
- *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-8507, Japan *Department of Molecular and Cellular Pharmacology, Pharmacogenomics and Pharmacoinformatics, Department of Clinical Anesthesiology, Department of Systems Pharmacology, Mie University Graduate School of Medicine, Mie 514-8507, Japan, Mie University Medical Zebrafish Research Center, Mie 514-8507, Japan, Department of Bioinformatics, Mie University Life Science Research Center, Mie 514-8507, Japan and Department of Omics Medicine, Mie University Industrial Technology Innovation, Mie 514-
| |
Collapse
|
21
|
Daryadel A, Bogdanova A, Gassmann M, Mueller X, Zünd G, Seifert B, Lehalle C, Frossard N, Tavakoli R. Multikinase inhibitor sorafenib prevents pressure overload-induced left ventricular hypertrophy in rats by blocking the c-Raf/ERK1/2 signaling pathway. J Cardiothorac Surg 2014; 9:81. [PMID: 24885948 PMCID: PMC4042218 DOI: 10.1186/1749-8090-9-81] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 05/06/2014] [Indexed: 01/07/2023] Open
Abstract
Background Left ventricular hypertrophy (LVH) is a potent risk factor for sudden death and congestive heart failure. Methods We tested the effect of sorafenib, a multikinase inhibitor (10 mg/kg, given orally, starting 2 days prior to banding, till sacrifice on day 14), on the development of LVH following aortic banding in rats. Results The latter resulted in significant LVH caused by both an increase in cardiomyocyte volume and interstitial collagen deposition. The observed LVH was entirely blocked by sorafenib downregulating both of these components. LVH was associated with PDGF-BB and TGFβ1 overexpression, as well as phosphorylation of c-raf and ERK1/2. Additionally, the transcription factors c-myc and c-fos leading to proliferation as well as the hypertrophy-inducing transcription factor GATA4 and its regulated gene ANP were all upregulated in response to aortic banding. All these overexpressions and upregulations were inhibited upon sorafenib treatment. Conclusion We show that sorafenib exhibits a regulatory role on the occurrence of LVH following AB in rats by blocking the rise in growth factors PDGF-BB and TGFβ1, activation of the corresponding c-Raf-ERK1/2 signaling pathway and effector mechanisms, including GATA4 and ANP. This effect of sorafenib may be of clinical importance in modulating the maladaptive hypertrophic response to pressure overload.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Reza Tavakoli
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
22
|
Molecular basis of cancer-therapy-induced cardiotoxicity: introducing microRNA biomarkers for early assessment of subclinical myocardial injury. Clin Sci (Lond) 2014; 126:377-400. [PMID: 24274966 DOI: 10.1042/cs20120620] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Development of reliable biomarkers for early clinical assessment of drug-induced cardiotoxicity could allow the detection of subclinical cardiac injury risk in vulnerable patients before irreversible damage occurs. Currently, it is difficult to predict who will develop drug-induced cardiotoxicity owing to lack of sensitivity and/or specificity of currently used diagnostics. miRNAs are mRNA regulators and they are currently being extensively profiled for use as biomarkers due to their specific tissue and disease expression signature profiles. Identification of cardiotoxicity-specific miRNA biomarkers could provide clinicians with a valuable tool to allow prognosis of patients at risk of cardiovascular injury, alteration of a treatment regime or the introduction of an adjunct therapy in order to increase the long-term survival rate of patients treated with cardiotoxic drugs.
Collapse
|
23
|
Todaro MC, Oreto L, Qamar R, Paterick TE, Carerj S, Khandheria BK. Cardioncology: State of the heart. Int J Cardiol 2013; 168:680-7. [DOI: 10.1016/j.ijcard.2013.03.133] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 03/19/2013] [Accepted: 03/26/2013] [Indexed: 10/26/2022]
|
24
|
Dy GK, Adjei AA. Understanding, recognizing, and managing toxicities of targeted anticancer therapies. CA Cancer J Clin 2013; 63:249-79. [PMID: 23716430 DOI: 10.3322/caac.21184] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/21/2013] [Accepted: 02/25/2013] [Indexed: 12/11/2022] Open
Abstract
Answer questions and earn CME/CNE Advances in genomics and molecular biology have identified aberrant proteins in cancer cells that are attractive targets for cancer therapy. Because these proteins are overexpressed or dysregulated in cancer cells compared with normal cells, it was assumed that their inhibitors will be narrowly targeted and relatively nontoxic. However, this hope has not been achieved. Current targeted agents exhibit the same frequency and severity of toxicities as traditional cytotoxic agents, with the main difference being the nature of the toxic effects. Thus, the classical chemotherapy toxicities of alopecia, myelosuppression, mucositis, nausea, and vomiting have been generally replaced by vascular, dermatologic, endocrine, coagulation, immunologic, ocular, and pulmonary toxicities. These toxicities need to be recognized, prevented, and optimally managed.
Collapse
Affiliation(s)
- Grace K Dy
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | | |
Collapse
|
25
|
Shah RR, Morganroth J, Shah DR. Cardiovascular Safety of Tyrosine Kinase Inhibitors: With a Special Focus on Cardiac Repolarisation (QT Interval). Drug Saf 2013; 36:295-316. [DOI: 10.1007/s40264-013-0047-5] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
26
|
Hall PS, Harshman LC, Srinivas S, Witteles RM. The frequency and severity of cardiovascular toxicity from targeted therapy in advanced renal cell carcinoma patients. JACC-HEART FAILURE 2013; 1:72-8. [PMID: 24621801 DOI: 10.1016/j.jchf.2012.09.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVES The purpose of this study was to document the incidence and extent of cardiovascular toxicity among advanced renal cell carcinoma patients treated with newer targeted cancer agents. BACKGROUND The potential for targeted cancer agents to induce cardiovascular toxicity has been increasingly recognized, but the overall incidence and extent of toxicity have not been well characterized. Early detection of asymptomatic patients could preempt symptomatic toxicity and reduce treatment-related morbidity and mortality. METHODS The incidence of hypertension, left ventricular dysfunction, and heart failure was assessed for all advanced renal cell carcinoma patients treated with targeted therapies at our institution between 2004 and 2011. Grading was performed according to the Common Terminology Criteria for Adverse Events version 4.0. RESULTS Cardiovascular toxicity developed in 116 of 159 patients (73%), including 52 of 159 patients (33%) when hypertension was excluded. Toxicity varied from occurrences of asymptomatic drops in left ventricular ejection fraction to rises in N-terminal-pro-B-type natriuretic peptide to severe heart failure. The tyrosine kinase inhibitor sunitinib was the agent most frequently used, with 66 of 101 sunitinib-treated patients (65%) developing a form of cardiovascular toxicity, including 32 of 101 patients (32%), excluding hypertension. Other VEGF inhibitors such as bevacizumab, sorafenib, and pazopanib also elicited significant cardiovascular toxicity with incidences ranging from 51% to 68%. CONCLUSIONS The frequency and severity of cardiovascular toxicity in advanced renal cell carcinoma patients treated with targeted cancer therapies are high.
Collapse
Affiliation(s)
- Philip S Hall
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, California
| | - Lauren C Harshman
- Division of Oncology, Stanford University School of Medicine, Stanford, California
| | - Sandy Srinivas
- Division of Oncology, Stanford University School of Medicine, Stanford, California
| | - Ronald M Witteles
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
27
|
Hasinoff BB, Patel D, Wu X. The Dual-Targeted HER1/HER2 Tyrosine Kinase Inhibitor Lapatinib Strongly Potentiates the Cardiac Myocyte-Damaging Effects of Doxorubicin. Cardiovasc Toxicol 2012; 13:33-47. [DOI: 10.1007/s12012-012-9183-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
28
|
Effect of irradiation with red and infrared laser in the treatment of oral mucositis. Lasers Med Sci 2012; 27:1233-40. [DOI: 10.1007/s10103-012-1089-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 03/15/2012] [Indexed: 01/29/2023]
|
29
|
Dasanu CA, Padmanabhan P, Clark BA, Do C. Cardiovascular toxicity associated with small molecule tyrosine kinase inhibitors currently in clinical use. Expert Opin Drug Saf 2012; 11:445-57. [DOI: 10.1517/14740338.2012.672971] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
Rees ML, Khakoo AY. Molecular mechanisms of hypertension and heart failure due to antiangiogenic cancer therapies. Heart Fail Clin 2011; 7:299-311. [PMID: 21749882 DOI: 10.1016/j.hfc.2011.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Targeted antiangiogenic cancer therapies have revolutionized the treatment of highly vascularized cancers such as metastatic renal cell carcinoma and gastrointestinal stromal tumors. Such agents act by inhibiting the actions of proangiogenic growth factors and their receptor tyrosine kinases, which are known to be overexpressed in cancer. However, these factors also play an important role in normal cardiovascular physiology. This article summarizes the incidences of cardiovascular toxicities (namely hypertension and heart failure) associated with the most commonly used antiangiogenic therapies, and then presents data from preclinical and clinical studies to provide some insight into the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Meredith L Rees
- Department of Cardiology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1101, Houston, TX 77030, USA
| | | |
Collapse
|
31
|
van Beuge MM, Poelstra K, Prakash J. Specific delivery of kinase inhibitors in nonmalignant and malignant diseases. Expert Opin Drug Deliv 2011; 9:59-70. [PMID: 22111941 DOI: 10.1517/17425247.2012.638625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Kinase inhibitors have been hailed as a breakthrough in the treatment of cancer. Extensive research is now being devoted to the development of kinase inhibitors as a treatment for many nonmalignant diseases. However, the use of kinase inhibitors in both malignant and nonmalignant diseases is also associated with side effects and the development of resistance. It may be worthwhile to explore whether cell-specific delivery of kinase inhibitors improves therapeutic efficacy and reduces side effects. AREAS COVERED This review aims to provide an overview of the preclinical studies performed to examine the specific targeting of kinase inhibitors in vitro and in vivo. It gives an introduction to kinase signaling pathways induced during disease, along with the possible problems associated with their inhibition. It also discusses the studies on specific delivery and shows that altering the specificity of kinase inhibitors by targeting methods improves their effectivity and safety. EXPERT OPINION Compared with the delivery of cytotoxic compounds, the specific delivery of kinase inhibitors has not yet been studied extensively. The studies discussed in this review provide an insight into methods used to target kinase inhibitors to different organs. The targeting of different kinase inhibitors has improved their therapeutic possibilities, but many questions still remain to be studied.
Collapse
Affiliation(s)
- Marike Marjolijn van Beuge
- University of Groningen, University Centre for Pharmacy, Department of Pharmacokinetics, Toxicology & Targeting, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| | | | | |
Collapse
|
32
|
Bonura F, Di Lisi D, Novo S, D’Alessandro N. Timely Recognition of Cardiovascular Toxicity by Anticancer Agents: A Common Objective of the Pharmacologist, Oncologist and Cardiologist. Cardiovasc Toxicol 2011; 12:93-107. [DOI: 10.1007/s12012-011-9141-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Toubert ME, Vercellino L, Faugeron I, Lussato D, Hindie E, Bousquet G. Fatal heart failure after a 26-month combination of tyrosine kinase inhibitors in a papillary thyroid cancer. Thyroid 2011; 21:451-4. [PMID: 21385075 DOI: 10.1089/thy.2010.0270] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Patients with progressive refractory thyroid cancer are potential candidates for clinical trials using tyrosine kinase inhibitors (TKIs), and a promising proportion of patients in these trials have achieved stable disease. Here we report an unusual adverse experience in a patient receiving a combination of TKIs. SUMMARY The patient was a 62-year-old man with chronic myloid leukemia (CML) and thyroid carcinoma that did not concentrate iodide and had metastases. He was started on imatinib for his CML. About 5 months later he was started on sorafenib for his thyroid cancer. At this time he had no risk factors for cardiac disease except moderate obesity. He had a complete cytogenetic response in his CML, and a partial response in his thyroid cancer. Twenty-one months after starting the combination of TKIs, he manifested signs of coronary artery disease. He received a combination of medications and his TKIs were continued. He died of a sudden myocardial infarction with cardiogenic shock 28 months after starting the combination of TKIs. A retrospective analysis of sequential 18-fludeoxyglucose positron emission tomography scans (18-FDG PET scans) were indicative of cardiac toxicity developing during the period of concomitant administration of TKIs. CONCLUSION We report the first case of apparent lethal cardiotoxicity with imatinib-sorafenib combined therapy. Combination TKI treatment may enhance the risk of adverse effects. Our experience with this patient suggests that cardiac PET scan should be monitored closely in these type of patients.
Collapse
|
34
|
Myositis due to Sorafenib intake in a patient with hepatocellular carcinoma. Dig Liver Dis 2011; 43:333-4. [PMID: 21215716 DOI: 10.1016/j.dld.2010.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 11/23/2010] [Accepted: 11/23/2010] [Indexed: 12/11/2022]
|
35
|
Abernethy AP. Critical Summaries of Innovations in Palliative Care Pharmacotherapy. J Pain Palliat Care Pharmacother 2010. [DOI: 10.3109/15360288.2010.526686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
36
|
Hasinoff BB, Patel D. The lack of target specificity of small molecule anticancer kinase inhibitors is correlated with their ability to damage myocytes in vitro. Toxicol Appl Pharmacol 2010; 249:132-9. [PMID: 20832415 DOI: 10.1016/j.taap.2010.08.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 08/24/2010] [Accepted: 08/26/2010] [Indexed: 10/19/2022]
Abstract
Many new targeted small molecule anticancer kinase inhibitors are actively being developed. However, the clinical use of some kinase inhibitors has been shown to result in cardiotoxicity. In most cases the mechanisms by which they exert their cardiotoxicity are not well understood. We have used large scale profiling data on 8 FDA-approved tyrosine kinase inhibitors and 10 other kinase inhibitors to a panel of 317 kinases in order to correlate binding constants and kinase inhibitor binding selectivity scores with kinase inhibitor-induced damage to neonatal rat cardiac myocytes. The 18 kinase inhibitors that were the subject of this study were: canertinib, dasatinib, dovitinib, erlotinib, flavopiridol, gefitinib, imatinib, lapatinib, midostaurin, motesanib, pazopanib, sorafenib, staurosporine, sunitinib, tandutinib, tozasertib, vandetanib and vatalanib. The combined tyrosine kinase and serine-threonine kinase selectivity scores were highly correlated with the myocyte-damaging effects of the kinase inhibitors. This result suggests that myocyte damage was due to a lack of target selectivity to binding of both tyrosine kinases and serine-threonine kinases, and was not due to binding to either group specifically. Finally, the strength of kinase inhibitor binding for 290 kinases was examined for correlations with myocyte damage. Kinase inhibitor binding was significantly correlated with myocyte damage for 12 kinases. Thus, myocyte damage may be multifactorial in nature with the inhibition of a number of kinases involved in producing kinase inhibitor-induced myocyte damage.
Collapse
Affiliation(s)
- Brian B Hasinoff
- Faculty of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Avenue, Winnipeg, Manitoba R3E0T5, Canada.
| | | |
Collapse
|