2023 Impact Factor
Gefitinib, an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor developed by AstraZeneca, is widely used for treating advanced non-small cell lung cancer (NSCLC) and is clinically approved as a standard first-line therapeutic agent for patients with somatic EGFR mutation-positive advanced NSCLC (Hida
Antipyretic analgesics may be used to alleviate cancer pain and cancer-related low-grade fever or even high fever. Recently, acetaminophen (APAP) or ibuprofen was often used clinically to relieve fever and pain associated with the coronavirus disease 2019 (COVID-19) infection in patients susceptible to COVID-19 (Rinott
In this study, we simulated the combination strategy in an animal model at clinical doses and confirmed that gefitinib in combination with APAP exhibits additional hepatotoxicity, suggested that this exacerbating effect is through the large-scale production of ROS, damaging hepatocytes, thereby mediating apoptosis. When the two drugs are taken together, the upregulation of p62 expression brought on by gefitinib is inhibited by APAP intervention, which causes Keap1 to accumulate. In the cytoplasm, free Nrf2 combines with Keap1 and then is degraded. The synthesis of antioxidant factors decreases, and ROS levels rise, ultimately resulting in ROS-mediated cell apoptosis. This study provides a feasible theoretical guidance for the combined use of gefitinib and APAP in clinical practice, and emphasizes the risk of aggravating hepatotoxicity.
We purchased 6-8 weeks old male Institute of Cancer Research (ICR) mice from Shanghai Experimental Animal Center (Shanghai, China). All experimental procedures and methods were approved by the Center for Drug Safety Evaluation and Research of Zhejiang University and were bred according to the Institutional Animal Care and Use Committee (IACUC) protocols of Zhejiang University (Approval No: IACUC-23-350). The mice were housed in the Zhejiang University animal facility under a 12 h light/dark cycle and were provided food and water
Human hepatocyte cell line, HL-7702, was purchased from Jennio Biological Technology (JNO-048, Guangzhou, China). The human hepatoma cell line, HepG2, was obtained from the Cell Bank of China Science Academy (TCHu 72, Shanghai, China). HL-7702 cells were maintained in Roswell Park Memorial Institute (RPMI)-1640 (31800, Gibco, CA, USA), while HepG2 cells were maintained in Dulbecco’s Modified Eagle’s Medium (DMEM, 10569010, Gibco, NY, USA) supplemented with 10% fetal bovine serum (SH30396.03, Hyclone, Logan, UT, USA), 100 U/mL penicillin (Invitrogen, Carlsbad, CA, USA) and 100 μg/mL streptomycin (Invitrogen) in a humidified atmosphere with 5% CO2 at 37°C.
Gefitinib (SML1657), N-acetyl-L-cysteine (NAC, A7250), and 3-Methyladenine (3-MA, 5142-23-4) were purchased from Sigma-Aldrich. APAP (T0065), necrostatin-1 (T1847) and ferrostatin-1 (T6500) were purchased from Topscience (Shanghai, China). Z-VAD-FMK (C1202) was purchased from Beyotime (Shanghai, China). Z-YVAD-FMK (HY-P1009) was purchased from MedChem Express (Shanghai, China). Throughout the experiments (except otherwise stated), HL-7702 cells and HepG2 cells were treated with 10 μM gefitinib and/or 1.25 mM APAP for 24 h. In selected samples, 20 μM Z-VAD-FMK, 10 μM Z-YVAD-FMK, 1 μM ferrostatin-1, 20 μM necrostatin-1, 1 mM 3-MA and 1 mM NAC were used.
Blood samples were collected and left for more than 2 h, and then centrifuged at 4000 rpm for 15 min to obtain serum for the determining alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels using an automated chemical analyzer (XN-1000V, Sysmex, Kobe, Japan).
Liver tissues were fixed in formalin (F8775, Sigma-Aldrich), embedded in paraffin, and cut into 3 μm sections. After dewaxing and rehydration, the sections were stained in hematoxylin (C0105, Beyotime) for 8 min and rinsed with tap water for 5 min and thereafter, stained in eosin (C0105, Beyotime) for 30 s. Finally, the sections were dehydrated and sealed with neutral resin to observe the morphology of the liver using a fluorescent microscope (IX81-FV1000, Olympus, Tokyo, Japan).
A colorimetric assay using sulforhodamine B (SRB, S1402, Sigma-Aldrich) was used to assess cell viability as previously described (Rubinstein
Apoptosis rates were determined using the Pharmingen™ FITC Annexin V Apoptosis Detection Kit I (556547, BD Biosciences, NJ, USA). The procedure was performed according to the manufacturer’s instructions. Briefly, the cells were processed for the indicated time, harvested, and washed with precooled phosphate buffer saline (PBS) for binding and Annexin V-PI staining. For each sample, 1×104 cells were obtained and analyzed using BD FACSCalibur™ flow cytometry (342973, BD Biosciences).
Intracellular reactive oxygen species (ROS) levels were determined using a ROS Assay Kit (S0033S, Beyotime) according to the manufacturer’s instructions. Cells were harvested and incubated with 10 μM dichlorofluorescin diacetate (DCFH-DA) probes for 20 min at 37°C. The cells were washed with serum-free culture medium and resuspended. The cells were obtained and analyzed using BD FACSCalibur™ flow cytometry (342973, BD Biosciences).
Total protein was extracted from cells or liver tissues using a lysis buffer (150 mM NaCl, 50 mM Tris-HCl, 2 mM EGTA, 2 mM EDTA, 25 mM β-sodium glycerophosphate, 25 mM NaF, 0.3% Triton X-100, 0.3% NP-40, 0.3% leupeptin, 0.1% NaVO3 and 0.1% PMSF). Protein lysates (20-50 μg per sample) were separated on 10% sodium dodecyl-sulfate polyacrylamide gel electrophoresis gels, transferred to polyvinylidene difluoride membranes (IPVH00010, Millipore Corporation, Boston, MA, USA) and blocked with a blocking buffer. Blots were cropped according to their molecular weights before probing with primary antibodies. Incubation with primary antibodies, secondary antibodies and the Western Lightning Plus-ECL reagent (NEL105001EA, PerkinElmer, Waltham, MA, USA) was performed for signal detection.
The following antibodies were used: Primary antibody against cleaved PARP (ET1608-10) was purchased from Huabio (Hangzhou, China). Primary antibodies against Nrf2 (12721S) γH2AX (97148SF) and p62 (5114s) were obtained from Cell Signaling Technology (Beverly, MA, USA). Primary antibody against β-actin (ACTB) (db7283) was obtained from Diagbio (Hangzhou, China). Primary antibody against p53 (sc-126) was obtained from Santa Cruz Biotechnology (TX, USA). Primary antibody against Keap1 (10503-2-AP) was obtained from Proteintech (IL, USA).
A one-step TUNEL Apoptosis Detection Kit (C1088, Beyotime) was used to detect apoptosis in mouse liver sections according to the manufacturer’s instructions. Briefly, tissue sections were pretreated with proteinase K (ST532, Beyotime) working solution after dewaxing and rehydration. Then, TUNEL detection solution was added to the tissue samples and incubated for 60 min at 37°C in a light-proof humidified chamber. Finally, the nuclei were stained with 4’,6-diamidino-2-phenylindole (DAPI, D212, Dojindo, Kumamoto, Japan) and the TUNEL signals were observed and captured with a fluorescent microscope (IX81-FV1000, Olympus).
The comet assay was performed as described previously (Olive and Banáth, 2006). Firstly, a single-cell suspension was prepared in PBS at a density of approximately 2×104 cells/mL. Next, pre-warmed 0.5% standard-gelling-temperature agarose, containing approximately 2000 cells, was placed on the microscope slides and spread sequentially (placed sequentially for more than 10 min) on the slides. The slides were immersed in an alkaline solution (pH >13) at 4°C for >1 h to lyse the samples. The slides were then submerged in a horizontal electrophoresis chamber containing a fresh cold alkaline solution (pH 12.3) for 20 min to unwind the DNA, and electrophoresis was performed at 300 mA for 20 min. Subsequently, the samples were neutralized with Tris-HCl (pH 7.5) for 15 min and dehydrated. Finally, the samples were stained with DAPI for 5 min, and images were captured using a fluorescent microscope (IX81-FV1000, Olympus). Using the Comet Assay Software Project, each sample was statistically analyzed using approximately 30 comet images.
After drug treatment, the cells were collected with the Trizol reagent (15596–026, Invitrogen) for total RNA extraction. Equal amounts of RNA were reverse transcribed into complementary DNA using a cDNA reverse transcription kit (AT311; Transgene, Beijing, China). qPCR was performed using the TB Green Premix Ex Taq™ (Tli RNaseH Plus) (RR420A, Takara, Tokyo, Japan) on a QuantStudio™ 3 Real-Time PCR Instrument (A28132, Thermo Fisher Scientific, Waltham, MA, USA). Samples were amplified in two steps: the first step at 95°C (3 s), followed by 95°C (5 s) and 60°C (31 s) for 40 cycles. Relative quantification was determined by the ΔΔCt method.
The primer sequences were as follows:
The ARE-driven reporter gene construct pGL4.27-ARE-NRF2-SPE was purchased from MIAOLING BIOLOGY (P40937, Hubei, China). HL-7702 cells were transfected with pGL4.37 plasmid using jetPRIME (101000046, Polyplus-transfection, Illkirch, France). Briefly, HL-7702 cells were seeded and grown to approximately 70% confluence. The related plasmid was transfected into cells with the transfection reagent for 4-6 h and then replaced with a fresh culture medium. Cells stably transfected with the ARE reporter were exposed to 10 μM gefitinib and/or 1.25 mM APAP for 24 h. Luciferase activity in cell lysates was measured using the Duo-Lite Luciferase Assay System (DD1205-01; Vazyme, Jiangsu, China).
After specific treatments, HL-7702 cells grown in 96-well plates were washed twice with PBS and fixed with fresh 4% paraformaldehyde (P6148, Sigma-Aldrich) for 20 min at 25°C. Then, the cells were permeabilized with 0.1% Triton X-100 in PBS for 10 min at 4°C and blocked with 4% bovine serum albumin (B2064, Sigma-Aldrich) in PBS for 30 min at 37°C. Thereafter, the cells were incubated with primary antibody against Nrf2 (ab62352; Abcam, Cambridge, UK) at 4°C overnight. After washing with PBS, the cells were incubated with Alexa Fluor 488-conjugated secondary antibodies (A11008; 1:100, Thermo Fisher Scientific, Waltham, MA, USA) at room temperature for 1 h, stained with DAPI for 5 min, and imaged using a fluorescence microscope (IX81-FV1000, Olympus).
Statistical comparisons of multiple groups were performed using one-way analysis of variance. All experiments were conducted in triplicate. Data were analyzed using GraphPad Prism 6.0 and expressed as mean ± standard error of mean (SEM).
To observe hepatotoxicity, we designed the administration of gefitinib and APAP in ICR mice (Fig. 1A). Gefitinib and APAP doses were chosen based on clinical recommendations (Peacock
Cell death involves a signaling cascade comprising several effector molecules and has unique biochemical characteristics, including various forms of apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. Given that the combination of gefitinib and APAP can directly inhibit hepatocyte survival, we investigated the specific mechanisms underlying hepatocyte death. We used a variety of cell death inhibitors, such as apoptosis inhibitor Z-VAD-FMK, necroptosis inhibitor necrostatin-1, pyroptosis inhibitor Z-YVAD-FMK, ferroptosis inhibitor ferrostatin-1 and autophagy inhibitor 3-MA to investigate the cell death. The appropriate drug concentration was determined based on relevant studies (Liao
There is growing evidence that ROS accumulation is involved in hepatotoxicity and that ROS plays a considerable role in cell apoptosis (Yan
Oxidative DNA damage caused by amplified ROS generation is one of the most common types of DNA damage in cells, in which the double strand of the cellular DNA breaks. Phosphorylation of histone H2AX (γH2AX) is considered a biomarker that can reflect the extent of DNA damage and repair (Mah
High levels of ROS are associated with oxidative stress and disease progression. Nrf2 is a key upstream transcription factor for cellular resistance to oxidative stress and plays an antioxidant and anti-mortality role by upregulating heme oxygenase-1 (HO-1), superoxide dismutase 1 (SOD1), and other antioxidant proteins to scavenge ROS under oxidative stress (Zhang, 2006; Done and Traustadóttir, 2016; Jayasuriya
The antioxidant genes, HO-1 and SOD1, are induced by Nrf2 nuclear translocation. HO-1 and SOD-1 transcriptional levels (Fig. 4D) detected in cells treated with gefitinib plus APAP were significantly downregulated compared with those in the gefitinib-only group, which may be responsible for the weakened resistance to oxidative stress. ARE signaling pathway is a major defense mechanism against oxidative stress (Motahari
Moreover, the p62/Keap1/Nrf2 antioxidative signaling pathway is involved in protection against ferroptosis in hepatocellular carcinoma cells cells. p62 prevents Nrf2 degradation and enhances Nrf2 nuclear accumulation via Keap1 (Sun
Overall, our preliminary study suggests that Nrf2 levels might be associated with the exacerbation of liver injury. This could be a way to reduce the risk of hepatotoxicity by precisely regulating the level of Nrf2 or using ROS scavengers.
In this study, we revealed the critical role of apoptosis in the exacerbation of gefitinib plus APAP-induced hepatotoxicity and provided mechanistic insight into ROS-promoting pathways. The liver injury occurred after the co-administration of clinically appropriate doses of gefitinib and APAP in ICR mice. Mechanistically, the combination of gefitinib and APAP downregulated gefitinib-induced increase in the p62/Keap1/Nrf2 signaling pathway in hepatocytes, thus increasing intracellular ROS production, leading to DNA damage and enhanced apoptosis.
Gefitinib at a dose of 250 mg once daily (Ramalingam
Previous studies have reported that APAP overdose induces predominantly necrotic or programmed hepatocyte necrosis and rarely involves apoptosis, but the effects of safe and regular low-dose APAP administration in the liver remain unclear. Our results show that low-dose APAP had little effect on hepatocyte survival and caused only a slight increase in apoptosis. When APAP was combined with gefitinib, we speculated that the gefitinib-induced liver injury was exacerbated by an increase in ROS produced by gefitinib and the promotion of hepatocyte apoptosis.
p53 protein is a central biomarker of cellular responses to various types of damage and regulates apoptosis and autophagy in response to oxidative stress (Shi and Dansen, 2020). We detected the expression level of the P53 protein and found no obvious increase after administration (Supplementary Fig. 4). Therefore, we focused on Nrf2 as a key regulatory factor of toxicity.
To explore the possible mechanisms underlying gefitinib plus APAP-induced oxidative stress, we evaluated Nrf2 protein expression, an antioxidant transcription factor involved in redox homeostasis, which plays a vital role in drug-induced liver injury (Chao
Several studies have revealed that p62 is instrumental in regulating the Keap1/Nrf2 pathway. p62 autophagy-dependent degradation of Keap1 can result in the attenuation of Nrf2 ubiquitination and increased protein stability (Komatsu
Our study indicates that the p62/Keap1/Nrf2 antioxidative signaling pathway is involved in the combination with gefitinib and APAP-induced apoptosis. Preventing the expression of p62 promotes the degradation of Nrf2 and attenuates subsequent Nrf2 nuclear accumulation through the accumulation of Keap1. To the best of our knowledge, this is the first study to demonstrate that the combination of gefitinib and APAP enhances oxidative stress via the p62/Keap1/Nrf2 signaling pathway.
This work was supported by National Natural Science Foundation of China (No.82104315), Natural Science Foundation of Zhejiang Province of China (No. LQ22H310002), Special Pharmacy Project of Zhejiang Pharmaceutical Association (No.2023ZYY31), Yangtze River Delta Health Scientific Research Project of Zhejiang Province (No.2023CSJ-3-A002) and Youth Fund Project of Hangzhou Red Cross Hospital (No.HHQN2023007).
All authors declare that they have no conflict of interest.
Conceptualization and study design: P. L., H. Y. and J. J.; Experimental work: J. X. and X. H.; Animal models: X. H. and Y. Z.; Analysis and interpretation of data: J. X., X. H., Y. Z. and Z. X., X. C.; Writing the draft manuscript: J. X. and X. H.; Review and editing the manuscript: Z. X., X. C., B. Y., Q. H., P. L., H. Y. and J. J.; Funding acquisition: H. Y. All authors have read and agreed to the published version of the manuscript.