1
|
Arzuk E, Armağan G. In vitro assessment of the role of endoplasmic reticulum stress in sunitinib-induced liver and kidney toxicity. Toxicol Lett 2025; 403:9-16. [PMID: 39613054 DOI: 10.1016/j.toxlet.2024.11.010] [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: 10/01/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Sunitinib, a multi-targeted tyrosine kinase inhibitor, is prescribed for the treatment of metastatic gastrointestinal stromal tumors, advanced metastatic renal cell carcinoma, and pancreatic neuroendocrine tumors. Hepatotoxicity and nephrotoxicity are significant adverse effects of sunitinib administration; however, there is limited information regarding the molecular mechanisms of these adverse effects. The aim of the present study was to elucidate the role of endoplasmic reticulum stress in hepatotoxicity and nephrotoxicity induced by sunitinib. In addition to endoplasmic reticulum stress, oxidative stress and mitochondrial membrane potential were evaluated to investigate the molecular mechanism more comprehensively. Findings revealed that sunitinib exposure significantly increased the reactive oxygen species levels and decreased the Nrf2 gene expression and GSH/GSSG ratio, suggesting oxidative stress induction in normal hepatocyte (AML12) and normal kidney (HK-2) cell lines. Endoplasmic reticulum stress markers, including ATF4, CHOP, IRE1α, XBP1s and ATF6 mRNA expressions, were upregulated in AML12 cells. Furthermore, enhanced intracellular calcium levels also indicate endoplasmic reticulum stress in hepatocytes. In contrast, sunitinib exposure did not alter endoplasmic reticulum-related gene expression levels and intracellular calcium levels in HK-2 cells. In terms of mitochondrial membrane potential and caspase-3 activity, sunitinib induced mitochondrial membrane damage and increased caspase-3 activation not only in AML12 cells but also in HK-2 cells. The research findings indicate that sunitinib may induce cytotoxic effects in hepatocytes through mechanisms involving oxidative stress, endoplasmic reticulum stress, and mitochondrial damage. However, in the kidney, the toxicity mechanism is different from that of liver, and the endoplasmic reticulum stress does not seem to be involved in this mechanism.
Collapse
Affiliation(s)
- Ege Arzuk
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Ege University, İzmir, Turkey.
| | - Güliz Armağan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, İzmir, Turkey.
| |
Collapse
|
2
|
Zhang S, Zhang X, Ren Y, Huang L, Xu W, Wang H, Lu Q. Regorafenib enhances the efficacy of photodynamic therapy in hepatocellular carcinoma through MAPK signaling pathway suppression. Photodiagnosis Photodyn Ther 2024; 49:104319. [PMID: 39181490 DOI: 10.1016/j.pdpdt.2024.104319] [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: 06/22/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Photodynamic therapy (PDT) is a promising and innovative approach for treating tumors. The synergistic effect of PDT and chemotherapy can enhance the anti-tumor efficacy by leveraging their complementing benefits. In this study, we created lipid vesicles to deliver a photosensitizer (chlorin e6, Ce6) and Regorafenib into tumors for the purpose of examining the effectiveness and mechanism of Lipo-Ce6@Rego-PDT (LCR-P) on Hepatocellular carcinoma (HCC) both in vitro and in vivo. We found that the cytotoxicity on HCC caused by LCR-P was significantly stronger than that caused by Lipo-Ce6-PDT (LC-P). Cellular ROS production in the LCR-P group was approximately higher than that in the LC-P group, and Regorafenib significantly inhibited the phosphorylation of JNK, ERK, and P38 of Lipo-Ce6-PDT group in vitro and in vivo. Furthermore, Regorafenib significantly downregulated the expression of Bcl-2 and upregulated the expression of Bax and cleaved caspase-3 of LC-P group in vitro and in vivo. Compared with LC-P, LCR-P significantly increased cell apoptosis rate. The body weight and HE staining of normal organs primarily indicated the safety of this combined strategy. These results indicate that the combination of Regorafenib and Lipo-Ce6 can significantly enhance the anti-tumor efficiency of PDT for HCC and exhibits good biosafety.
Collapse
Affiliation(s)
- Song Zhang
- Postdoctoral Research Station, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Xiao Zhang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Yali Ren
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Lu Huang
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Weitian Xu
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China; School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430065, China
| | - Haiping Wang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Cancer Institute, School of Medicine, Jianghan University, Wuhan, China.
| | - Qiping Lu
- Postdoctoral Research Station, General Hospital of Central Theater Command, Wuhan, Hubei 430070, China.
| |
Collapse
|
3
|
Boran T, Zengin OS, Seker Z, Gunaydin Akyildiz A, Oztas E, Özhan G. The cyclin-dependent kinase inhibitor abemaciclib-induced hepatotoxicity: Insight on the molecular mechanisms in HepG2/THP-1 co-culture model. Toxicol Lett 2024; 391:1-12. [PMID: 37992977 DOI: 10.1016/j.toxlet.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
Drug-induced liver injury (DILI) is one of the widespread causes of liver injury and immune system plays important role. Abemaciclib (ABE) is a cyclin-dependent kinase inhibitor used as monotherapy or combination therapy in the treatment of breast cancer. Like other kinase inhibitors, the underlying mechanisms of ABE-induced hepatotoxicity are not completely known yet. In the current study, hepatotoxicity of ABE was evaluated with HepG2/THP-1 co-culture model which has been developed in recent years for the evaluation of DILI potential. Following ABE treatment, oxidative stress, mitochondrial damage, cytokine secretion levels, apoptotic/necrotic cell death were determined. According to our results, ROS production along with GSH depletion was observed in HepG2 cells after ABE treatment. ABE promoted secretion of pro-inflammatory mediators (TNF-α and MCP-1) and declined anti-inflammatory cytokine IL-10 release. Besides, NFKβ and JNK1 protein expression levels increased following ABE treatment. ABE enhanced intracellular calcium levels, induced early apoptotic and necrotic cell deaths in HepG2 cells. Furthermore, the changes in some mitochondrial parameters including a reducement in intracellular ATP levels and complex V activity; hyperpolarized mitochondrial membrane potential and enhanced mitochondrial ROS levels were observed, whereas mitochondrial mass did not show any differences after ABE treatments. Therefore, ABE-induced hepatotoxic effects is probably via oxidative stress, inflammatory response and necrotic cell death rather than direct mitochondrial toxicity. In conclusion; the study makes a significant contribution to strengthening the infrastructure we have on in vitro toxicity mechanism evaluations, which are the basis of preclinical toxicity studies.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Istanbul University-Cerrahpasa, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34500 Istanbul, Turkey
| | - Ozge Sultan Zengin
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey
| | - Zehra Seker
- Institute of Graduate Studies in Health Sciences, Istanbul University, 34116 Istanbul, Turkey; Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Aysenur Gunaydin Akyildiz
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34093 Istanbul, Turkey
| | - Ezgi Oztas
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey
| | - Gül Özhan
- Istanbul University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, 34116 Istanbul, Turkey.
| |
Collapse
|
4
|
Parrack PH, Zucker SD, Zhao L. Liver Pathology Related to Onco-Therapeutic Agents. Surg Pathol Clin 2023; 16:499-518. [PMID: 37536885 DOI: 10.1016/j.path.2023.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Oncotherapeutic agents can cause a wide range of liver injuries from elevated liver functions tests to fulminant liver failure. In this review, we emphasize a newer generation of drugs including immune checkpoint inhibitors, protein kinase inhibitors, monoclonal antibodies, and hormonal therapy. A few conventional chemotherapy agents are also discussed.
Collapse
Affiliation(s)
- Paige H Parrack
- Department of Pathology, Brigham and Women's Hospital, 75 Francis street, Boston, MA, 02115, USA; Harvard Medical School
| | - Stephen D Zucker
- Harvard Medical School; Department of Medicine, Brigham and Women's Hospital, 75 Francis street, Boston, MA, 02115, USA
| | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, 75 Francis street, Boston, MA, 02115, USA; Harvard Medical School.
| |
Collapse
|
5
|
Yan M, Li W, Li WB, Huang Q, Li J, Cai HL, Gong H, Zhang BK, Wang YK. Metabolic activation of tyrosine kinase inhibitors: recent advance and further clinical practice. Drug Metab Rev 2023; 55:94-106. [PMID: 36453523 DOI: 10.1080/03602532.2022.2149775] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
At present, receptor tyrosine kinase signaling-related pathways have been successfully mediated to inhibit tumor proliferation and promote anti-angiogenesis effects for cancer therapy. Tyrosine kinase inhibitors (TKIs), a group of novel chemotherapeutic agents, have been applied to treat diverse malignant tumors effectively. However, the latent toxic and side effects of TKIs, such as hepatotoxicity and cardiotoxicity, limit their use in clinical practice. Metabolic activation has the potential to lead to toxic effects. Numerous TKIs have been demonstrated to be transformed into chemically reactive/potentially toxic metabolites following cytochrome P450-catalyzed activation, which causes severe adverse reactions, including hepatotoxicity, cardiotoxicity, skin toxicity, immune injury, mitochondria injury, and cytochrome P450 inactivation. However, the precise mechanisms of how these chemically reactive/potentially toxic species induce toxicity remain poorly understood. In addition, we present our viewpoints that regulating the production of reactive metabolites may decrease the toxicity of TKIs. Exploring this topic will improve understanding of metabolic activation and its underlying mechanisms, promoting the rational use of TKIs. This review summarizes the updated evidence concerning the reactive metabolites of TKIs and the associated toxicities. This paper provides novel insight into the safe use of TKIs and the prevention and treatment of multiple TKIs adverse effects in clinical practice.
Collapse
Affiliation(s)
- Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Wen-Bo Li
- Department of Plastic and Aesthetic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Hua-Lin Cai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Bi-Kui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Yi-Kun Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| |
Collapse
|
6
|
Tang TL, Yang Y, Guo L, Xia S, Zhang B, Yan M. Sunitinib induced hepatotoxicity in L02 cells via ROS-MAPKs signaling pathway. Front Pharmacol 2022; 13:1002142. [PMID: 36386201 PMCID: PMC9643779 DOI: 10.3389/fphar.2022.1002142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/13/2022] [Indexed: 10/19/2024] Open
Abstract
Sunitinib is a multi-targeted tyrosine kinase inhibitor with remarkable anticancer activity, while hepatotoxicity is a potentially fatal adverse effect of its administration. The aim of this study was to elucidate the mechanism of hepatotoxicity induced by Sunitinib and the protective effect of glycyrrhetinic acid (GA). Sunitinib significantly reduced the survival of human normal hepatocytes (L02 cells), induced the increase of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH). Chloroquine (CQ) and Z-VAD-FMK were applied to clarify the cell death patterns induced by Sunitinib. Sunitinib significantly induced L02 cells death by triggering apoptosis and autophagy acted as a self-defense mechanism to promote survival. Sunitinib exposure caused excessive ROS generation which activated mitogen-activated protein kinases (MAPKs) signaling. Mechanistically, SP600125 (JNK inhibitor) and SB203580 (p38 inhibitor) respectively blocked apoptosis and autophagy induced by Sunitinib. And inhibition of ROS by NAC pretreatment ameliorated the effect of Sunitinib on MAPKs phosphorylation. GA alleviated Sunitinib-induced cell damage by inhibiting apoptosis and autophagy. These results suggested ROS/MAPKs signaling pathway was responsible for Sunitinib-induced hepatotoxicity and GA could be a preventive strategy to alleviate liver injury caused by Sunitinib.
Collapse
Affiliation(s)
| | | | | | | | | | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
7
|
Ramos-Casals M, Flores-Chávez A, Brito-Zerón P, Lambotte O, Mariette X. Immune-related adverse events of cancer immunotherapies targeting kinases. Pharmacol Ther 2022; 237:108250. [DOI: 10.1016/j.pharmthera.2022.108250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022]
|
8
|
Claesson A, Parkes K. Non-innocuous Consequences of Metabolic Oxidation of Alkyls on Arenes. J Med Chem 2022; 65:11433-11453. [PMID: 36001003 DOI: 10.1021/acs.jmedchem.2c00833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Reactive metabolite (RM) formation is widely accepted as playing a pivotal role in causing adverse idiosyncratic drug reactions, with most attention paid to drug-induced liver injury. Mechanisms of RM formation are determined by the drug's properties in relation to human enzymes transforming the drug. This Perspective focuses on enzymatic oxidation of alkyl groups on aromatics leading to quinone methides and benzylic alcohol sulfates as RMs, a topic that has not received very much attention. Unlike previous overviews, we will include in our Perspective several fulvene-like methides such as 3-methyleneindole. We also speculate that a few older drugs may form non-reported methides of this class. In addition, we report a few guiding DFT calculations of changes in free energy on going from a benzylic alcohol to the corresponding methide. Particularly facile reactions of 2-aminothiazole-5-methanol and 4-aminobenzyl alcohol are noted.
Collapse
Affiliation(s)
- Alf Claesson
- Awametox AB, Lilldalsvägen 17 A, SE-14461 Rönninge, Sweden
| | - Kevin Parkes
- Consultant, 39 Cashio Lane, Letchworth Garden City, Hertfordshire SG6 1AY, U.K
| |
Collapse
|
9
|
Dong H, You J, Zhao Y, Zheng D, Zhong Y, Li G, Weng Z, Luo H, Jiang S. Study on the Characteristics of Small-Molecule Kinase Inhibitors-Related Drug-Induced Liver Injury. Front Pharmacol 2022; 13:838397. [PMID: 35529445 PMCID: PMC9068902 DOI: 10.3389/fphar.2022.838397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background and Aim: More than half of the small-molecule kinase inhibitors (KIs) induced liver injury clinically. Meanwhile, studies have shown a close relationship between mitochondrial damage and drug-induced liver injury (DILI). We aimed to study KIs and the binding between drugs and mitochondrial proteins to find factors related to DILI occurrence. Methods: A total of 1,223 oral FDA-approved drugs were collected and analyzed, including 44 KIs. Fisher’s exact test was used to analyze DILI potential and risk of different factors. A total of 187 human mitochondrial proteins were further collected, and high-throughput molecular docking was performed between human mitochondrial proteins and drugs in the data set. The molecular dynamics simulation was used to optimize and evaluate the dynamic binding behavior of the selected mitochondrial protein/KI complexes. Results: The possibility of KIs to produce DILI is much higher than that of other types (OR = 46.89, p = 9.28E-13). A few DILI risk factors were identified, including molecular weight (MW) between 400 and 600, the defined daily dose (DDD) ≥ 100 mg/day, the octanol–water partition coefficient (LogP) ≥ 3, and the degree of liver metabolism (LM) more than 50%. Drugs that met this combination of rules were found to have a higher DILI risk than controls (OR = 8.28, p = 4.82E-05) and were more likely to cause severe DILI (OR = 8.26, p = 5.06E-04). The docking results showed that KIs had a significant higher affinity with human mitochondrial proteins (p = 4.19E-11) than other drug types. Furthermore, the five proteins with the lowest docking score were selected for molecular dynamics simulation, and the smallest fluctuation of the backbone RMSD curve was found in the protein 5FS8/KI complexes, which indicated the best stability of the protein 5FS8 bound to KIs. Conclusions: KIs were found to have the highest odds ratio of causing DILI. MW was significantly related to the production of DILI, and the average docking scores of KI drugs were found to be significantly different from other classes. Further analysis identified the top binding mitochondrial proteins for KIs, and specific binding sites were analyzed. The optimization of molecular docking results by molecular dynamics simulation may contribute to further studying the mechanism of DILI.
Collapse
Affiliation(s)
- Huiqun Dong
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Jia You
- Department of Hepatology, Hepatology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yu Zhao
- College of Mathematics and Computer Science, Fuzhou University, Fuzhou, China
| | - Danhua Zheng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Yi Zhong
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
- College of Mathematics and Computer Science, Fuzhou University, Fuzhou, China
| | - Gaozheng Li
- College of Mathematics and Computer Science, Fuzhou University, Fuzhou, China
| | - Zuquan Weng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
- College of Mathematics and Computer Science, Fuzhou University, Fuzhou, China
- *Correspondence: Zuquan Weng, ; Heng Luo, ; Shan Jiang,
| | - Heng Luo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
- College of Mathematics and Computer Science, Fuzhou University, Fuzhou, China
- MetaNovas Biotech Inc., Foster City, CA, United States
- *Correspondence: Zuquan Weng, ; Heng Luo, ; Shan Jiang,
| | - Shan Jiang
- Department of Vascular Thyroid Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou, China
- *Correspondence: Zuquan Weng, ; Heng Luo, ; Shan Jiang,
| |
Collapse
|
10
|
Guo L, Tang T, Fang D, Gong H, Zhang B, Zhou Y, Zhang L, Yan M. An Insight on the Pathways Involved in Crizotinib and Sunitinib Induced Hepatotoxicity in HepG2 Cells and Animal Model. Front Oncol 2022; 12:749954. [PMID: 35155225 PMCID: PMC8832280 DOI: 10.3389/fonc.2022.749954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/13/2022] [Indexed: 11/22/2022] Open
Abstract
Both crizotinib and sunitinib, novel orally-active multikinase inhibitors, exhibit antitumor activity and extend the survival of patients with a malignant tumor. However, some patients may suffer liver injury that can further limit the clinical use of these drugs, however the mechanisms underlying hepatotoxicity are still to be elucidated. Thus, our study was designed to use HepG2 cells in vitro and the ICR mice model in vivo to investigate the mechanisms of hepatotoxicity induced by crizotinib and sunitinib. Male ICR mice were treated orally with crizotinib (70 mg/kg/day) or sunitinib (7.5 mg/kg/day) for four weeks. The results demonstrated that crizotinib and sunitinib caused cytotoxicity in HepG2 cells and chronic liver injury in mice, which were associated with oxidative stress, apoptosis and/or necrosis. Crizotinib- and sunitinib-induced oxidative stress was accompanied by increasing reactive oxygen species and malondialdehyde levels and decreasing the activity of superoxide dismutase and glutathione peroxidase. Notably, the activation of the Kelch-like ECH-associated protein-1/Nuclear factor erythroid-2 related factor 2 signaling pathway was involved in the process of oxidative stress, and partially protected against oxidative stress. Crizotinib and sunitinib induced apoptosis via the mitochondrial pathway, which was characterized by decreasing Bcl2/Bax ratio to dissipate the mitochondrial membrane potential, and increasing apoptotic markers levels. Moreover, the pan-caspase inhibitor Z-VAD-FMK improved the cell viability and alleviated liver damage, which further indicated the presence of apoptosis. Taken together, this study demonstrated that crizotinib- and sunitinib-caused oxidative stress and apoptosis finally impaired hepatic function, which was strongly supported by the histopathological lesions and markedly increased levels of serum alanine aminotransferase, alkaline phosphatase and lactate dehydrogenase.
Collapse
Affiliation(s)
- Lin Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tingli Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dongmei Fang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yueyin Zhou
- Orthodontic Department of Xiangya Stomatology Hospital, Central South University, Changsha, China
| | - Leiyi Zhang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Guan S, Chen X, Chen Y, Wan G, Su Q, Liang H, Yang Y, Fang W, Huang Y, Zhao H, Zhuang W, Liu S, Wang F, Feng W, Zhang X, Huang M, Wang X, Zhang L. FOXO3 mutation predicting gefitinib-induced hepatotoxicity in NSCLC patients through regulation of autophagy. Acta Pharm Sin B 2022; 12:3639-3649. [PMID: 36176901 PMCID: PMC9513443 DOI: 10.1016/j.apsb.2022.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/25/2022] Open
Abstract
Hepatotoxicity is a common side effect for patients treated with gefitinib, but the related pathogenesis is unclear and lacks effective predictor and management strategies. A multi-omics approach integrating pharmacometabolomics, pharmacokinetics and pharmacogenomics was employed in non-small cell lung cancer patients to identify the effective predictor for gefitinib-induced hepatotoxicity and explore optional therapy substitution. Here, we found that patients with rs4946935 AA, located in Forkhead Box O3 (FOXO3) which is a well-known autophagic regulator, had a higher risk of hepatotoxicity than those with the GA or GG variant (OR = 18.020, 95%CI = 2.473 to 459.1784, P = 0.018) in a gefitinib-concentration dependent pattern. Furthermore, functional experiments identified that rs4946935_A impaired the expression of FOXO3 by inhibiting the promotor activity, increasing the threshold of autophagy initiation and inhibiting the autophagic activity which contributed to gefitinib-induced liver injury. In contrast, erlotinib-induced liver injury was independent on the variant and expression levels of FOXO3. This study reveals that FOXO3 mutation, leading to autophagic imbalance, plays important role in gefitinib-induced hepatotoxicity, especially for patients with high concentration of gefitinib. In conclusion, FOXO3 mutation is an effective predictor and erlotinib might be an appropriately and well-tolerated treatment option for patients carrying rs4946935 AA.
Collapse
|
12
|
Du J, Yan H, Xu Z, Yang B, He Q, Wang X, Luo P. Cutaneous toxicity of FDA-approved small-molecule kinase inhibitors. Expert Opin Drug Metab Toxicol 2021; 17:1311-1325. [PMID: 34743659 DOI: 10.1080/17425255.2021.2004116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION By 1 January 2021, the FDA has approved a total of 62 small-molecule kinase inhibitors (SMKIs). The increasing clinical use of small-molecule kinase inhibitors has led to some side effects, the most common of which is cutaneous toxicity, as reflected by approximately 90% (57 of 62) of the FDA-approved SMKIs have reported treatment-related cutaneous toxicities. Since these cutaneous toxicities may have a crucial influence on the emotional, physical and psychosocial health of the patients, it is of great importance for doctors, patients, oncologists and interrelated researchers to be aware of the cutaneous side effects of these drugs in order to make the diagnosis accurate and the treatment appropriate. AREAS COVERED This review aims to summarize the potential cutaneous toxicities and the frequency of occurrence of FDA-approved 62 SMKIs, and provide a succinct overview of the potential mechanisms of certain cutaneous toxicities. The literature review was performed based on PubMed database and FDA official website. EXPERT OPINION It is significant to determine the risk factors for SMKI-induced cutaneous toxicity. The mechanisms underlying SMKI-induced cutaneous toxicities remain unclear at present. Future research should focus on the mechanisms of SMKI-induced cutaneous toxicities to find out mechanistically driven therapies.
Collapse
Affiliation(s)
- Jiangxia Du
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaohong Wang
- Department of Chemotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Jiang H, Jin Y, Yan H, Xu Z, Yang B, He Q, Luo P. Hepatotoxicity of FDA-approved small molecule kinase inhibitors. Expert Opin Drug Saf 2020; 20:335-348. [PMID: 33356646 DOI: 10.1080/14740338.2021.1867104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Given their importance in cellular processes and association with numerous diseases, protein kinases have emerged as promising targets for drugs. The FDA has approved greater than fifty small molecule kinase inhibitors (SMKIs) since 2001. Nevertheless, severe hepatotoxicity and related fatal cases have grown as a potential challenge in the advancement of these drugs, and the identification and diagnosis of drug-induced liver injury (DILI) are thorny problems for clinicians.Areas covered: This article summarizes the progression and analyzes the significant features in the study of SMKI hepatotoxicity, including clinical observations and investigations of the underlying mechanisms.Expert opinion: The understanding of SMKI-associated hepatotoxicity relies on the development of preclinical models and improvement of clinical assessment. With a full understanding of the role of inflammation in DILI and the mediating role of cytokines in inflammation, cytokines are promising candidates as sensitive and specific biomarkers for DILI. The emergence of three-dimensional spheroid models demonstrates potential use in providing clinically relevant data and predicting hepatotoxicity of SMKIs.
Collapse
Affiliation(s)
| | | | - Hao Yan
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou China
| |
Collapse
|