Atractyloside inhibits gefitinib‑resistant non‑small‑cell lung cancer cell proliferation
- Authors:
- Published online on: July 30, 2024 https://doi.org/10.3892/ol.2024.14599
- Article Number: 466
-
Copyright: © Yao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Atractyloside, a diterpenoid glycoside, is a naturally occurring active component common in medicines and animal feed, particularly in plants (Asteraceae and Atractylis) native to east Asia. Atractyloside has been used in traditional Chinese medicine to treat nasal congestion and allergic rhinitis (1). Numerous studies (2–4) have demonstrated the biological functions of atractyloside, indicating that it is a strong candidate for the treatment of illnesses, including digestive disorders, liver injury and diabetes. According to a pharmacological clinical study in humans (1), atractyloside has been demonstrated to inhibit the mitochondrial ATP translocase, also known as the adenine nucleotide translocator, and to effectively reduce fat accumulation in the liver (steatosis) and improve insulin sensitivity, which protects the liver. Cho et al (5) revealed that atractyloside is a modest hypoglycemic agent in splenocytes, thus suggesting that it may be used to treat diabetes. Previous research has demonstrated that atractyloside inhibits mitochondrial ATP transporters, leading to cancer cell death (6).
The incidence and mortality rates of cancer are rapidly increasing worldwide. In both sexes, lung cancer is the most commonly diagnosed type of cancer (11.6% of total cases) and the leading cause of cancer-related deaths (18.4% of total cancer deaths) worldwide (7). Lung cancer is histologically classified as small-cell lung cancer (SCLC) and non-SCLC (NSCLC), and ~85% of patients have NSCLC. Of these patients, lung squamous cell carcinoma and lung adenocarcinoma are the most common subtypes, accounting for 40 and 20–25% of global cases, respectively (8,9). Notably, the application of biomarkers for NS CLC is clinically beneficial. Epidermal growth factor receptor (EGFR) is a well-known biomarker for NSCLC management (10). Although small-molecule tyrosine kinase inhibitors (TKIs), such as gefitinib, have curative effects, relapse caused by EGFR mutations usually lead to patients succumbing to the disease 2 years after the first diagnosis (11). First-generation EGFR inhibitors, such as gefitinib and erlotinib, have significantly improved the survival of patients with NSCLC; however, the secondary EGFR-T790M mutation leads to clinical resistance to first-generation EGFR-TKIs (11–13). New biomarkers may improve the diagnosis and treatment of NSCLC.
Brother of the regulator of imprinted sites (BORIS, also known as CTCFL)which is a paralog of CCCTC-binding factor, is commonly expressed in most types of cancer, whereas it is not expressed in the corresponding normal tissues; therefore, it is considered a potential therapeutic target for lung cancer (14–16). In our previous study, it was revealed that BORIS suppressed apoptosis and enhanced 5-fluorouracil resistance in colorectal cancer (17), and BORIS has also been reported to increase resistance to cisplatin treatment in NSCLC (12). Debruyne et al (18) reported that BORIS may be associated with various tumor occurrences, including brain cancer and cervical cancer, drug resistance and the prognosis of patients with cancer. Based on the ubiquitous expression of BORIS and the high incidence of EGFR resistance in NSCLC, it is worth studying whether BORIS influences targeted therapies for lung cancer.
In the present study, the association between BORIS and TKI-resistant NSCLC was assessed. In addition, atractyloside was used to mimic BORIS knockdown to study the therapeutic function of BORIS on the prevention of TKI resistance. The results revealed that atractyloside could facilitate TKIs to suppress NSCLC cell proliferation.
Materials and methods
Cell culture
NSCLC cancer cell lines H1299, PC-9, PC-9-IR and H1975, and the colorectal cancer cell line Caco2 were purchased from The Cell Bank of Type Culture Collection of The Chinese Academy of Sciences. All cells were cultured in RPMI 1640 medium (Nanjing BioChannel Biotechnology Co., Ltd.) containing 10% heat-inactivated fetal bovine serum (GeminiBio) at 37°C in an incubator containing 5% CO2.
Cell transfection and treatment
Lipofectamine® RNAiMAX reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used to transfect small interfering RNA (siRNA) into cells. According to the manufacturer's protocol, H1975, PC-9 and PC-9IR cells at 70% confluence were used for transfection. Briefly, 1 pmol siRNA/well was used for the transfection of cells on a 96-well plate, and 30 pmol siRNA/well was used for the transfection of cells on a 6-well plate. After a 5-min incubation at room temperature, the RNAiMAX-siRNA mixture was added to either 6-well or 96-well plates. The cells were incubated for 48 h without media replacement, after which subsequent experiments were performed. The 96-well plates were used for MTT and cell proliferation assays. The 6-well plates were used for the analysis of transcript or protein expression levels. The siRNA sequences used in the present study are listed in Table I. Negative control siRNA and siBORIS were synthesized by Xiangyin Biotechnology Co., Ltd. The cells were then incubated at 37°C in an incubator containing 5% CO2. After 48 h, the cells were used for subsequent experiments. Following transfection, cells were treated with their respective drug treatments, with the control group receiving an equal volume of DMSO. Atractyloside (cat. no. HY-N1462), the TKI inhibitor gefitinib (cat. no. HY-50895) and erlotinib (cat. no. HY-50896) were purchased from MedChemExpress. Cells were treated with gefitinib (50 µM) 4 h post-transfection at room temperature. Cells were subjected to experiments after 48 or 96 h of gefitinib treatment. In addition, cells were treated with gefitinib (1 µM), erlotinib (5 µM), or atractyloside (1, 2.5 or 5 µM) for 48 h at 37°C prior to performing Cell Counting Kit (CCK)-8 assays, western blotting and reverse transcription-quantitative PCR (RT-qPCR).
Cell viability analysis
A total of 3,000 cells/well were seeded in a 96-well plate for transfection or drug treatment. Subsequently, MTT (500 µg/ml; cat. no. M2128; Sigma-Aldrich; Merck KGaA) was added to the cells and incubated for 4 h at 37°C, and 100 µl dimethyl sulfoxide was added for 15 min at room temperature. Signals were recorded using a BioTek Synergy 2 plate reader at a wavelength of 490 nm (BioTek; Agilent Technologies, Inc.).
CCK-8
A total of 3,000 cells/well were seeded in a 96-well plate and underwent drug treatment. After treatment with drugs (gefitinib, 1 µM; erlotinib, 5 µM; atractyloside, 5 µM) for 48 h at 37°C, the cell culture medium was discarded, and 100 µl medium containing 10 µl CCK-8 (cat. no. K1018; APeXBIO Technology LLC) reagent was added. The cells were then incubated for 1 h at 37°C and signals were recorded using a BioTek Synergy 2 plate reader at a wavelength of 450 nm.
Western blotting
H1299, Caco2, PC-9 and H1975 cells were cultured in a 6-well plate and were lysed using RIPA buffer (cat. no. 20188; MilliporeSigma) containing PMSF (1:100; cat. no. ST506; Beyotime Institute of Biotechnology) and Roche cOmplete™ Protease Inhibitor Cocktail (1:25; cat. no. 04693116001; Sigma-Aldrich; Merck KGaA). After centrifugation at 12,000 × g for 30 min at 4°C, the supernatants were collected, and the total protein was quantified using a detergent-compatible Bradford protein assay kit (cat. no. P0006C; Beyotime Institute of Biotechnology). Samples (30 µg/lane) were separated by SDS-PAGE on a 10% gel and were transferred onto a PVDF membrane (cat. no. ISEQ00010-PVDF; MilliporeSigma). The membrane was blocked with a protein-free rapid blocking buffer (cat. no. PS108P; New Cell & Molecular Biotech Co., Ltd.) for 15 min at room temperature and then incubated at 4°C overnight with the following antibodies: Mouse anti-GAPDH (1:500,000; cat. no. 60004-1-Ig; Proteintech Group, Inc.), rabbit anti-XRCC4 (1:1,000; cat. no. 15817-1-AP; Proteintech Group, Inc.), mouse anti-BORIS (1:1,000; cat. no. sc-377085; Santa Cruz Biotechnology, Inc), rabbit anti-AKT (1:1,000; cat. no. 9272; Cell Signaling Technology, Inc.) and mouse anti-phosphorylated (p)-AKT (1:1,000; cat. no. 66444-1-Ig; Proteintech Group, Inc.). After washing with TBS-1% Tween (TBST) three times (10 min/wash), the membrane was incubated with HRP-conjugated secondary antibodies (anti-rabbit and anti-mouse; 1:5,000; cat. nos. DW-GAR007 and DW0990-100; Hangzhou Dawen Biological Co., Ltd.) for 2 h at room temperature. Signals were detected after washing with TBST three times (10 min/wash) using the Ultrasensitive ECL Kit (cat no. P2300; New Cell & Molecular Biotech Co., Ltd)and ChemiDoc XRS+ system (Bio-Rad Laboratories, Inc.). The relative expression of the protein bands was semi-quantified using ImageJ version 1.53 software (National Institutes of Health).
RT-qPCR
The RNA of treated cells was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) through ethanol precipitation. cDNA was reverse transcribed using the Hifair® II 1st Strand cDNA Synthesis Kit (gDNA digester plus) (cat. no. 11121ES60; Shanghai Yeasen Biotechnology Co., Ltd.), and was used for qPCR analysis. For RT, the temperature settings were as follows: 25°C for 5 min, 42°C for 30 min and 85°C for 5 min. qPCR was performed using the 2X T5 Fast qPCR Mix (SYBR Green; cat. no. 11201ES08; Shanghai Yeasen Biotechnology Co., Ltd.) and a CFX connect real-time PCR detection system (Bio-Rad Laboratories, Inc.). According to the manufacturer's protocol, the thermocycling conditions were as follows: Initial denaturation at 95°C for 5 min, followed by 40 cycles of denaturation at 95°C for 15 sec and annealing/extension at 60°C for 30 sec. GAPDH was used as an internal reference for normalization. The primers used for qPCR are listed in Table II. The qPCR results were analyzed using the 2−ΔΔCq method (19).
Bioinformatics analysis
BORIS expression in NSCLC was determined using the R2 Genomics Analysis and Visualization Platform (http://r2.amc.nl). The Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/) datasets GSE19188 (20) and GSE63074 (21) were utilized for bioinformatics analysis.
Statistical analysis
GraphPad Prism 8 software (Dotmatics) was used for all statistical analyses. All experiments were performed in triplicate. Data are presented as the mean ± standard deviation. Statistical differences were calculated using one-way or two-way ANOVA followed by Tukey's multiple comparisons test, paired Student's t-test or unpaired Student's t-test. P<0.05 was considered to indicate a statistically significant difference.
Results
Bioinformatics analysis
Bioinformatics analysis was performed using the R2 Genomics Analysis and Visualization Platform. Based on the GSE19188 dataset, BORIS expression was elevated in primary NSCLC tissues compared with in normal lung tissues (Fig. 1A). From the GSE63074 dataset, survival rate data were used to perform a risk stratification analysis, categorizing patients into high-risk and low-risk groups according to a previously described method (22); the analysis indicated a significant upregulation of BORIS expression in the high-risk NSCLC group (Fig. 1B). Given that EGFR upregulation or mutation was prevalent in high-risk NSCLC cases, further investigation into the potential association between BORIS and EGFR expression/mutation is warranted.
BORIS knockdown inhibits H1975 cell viability
The H1975 cell line comprises NSCLC cells resistant to gefitinib due to the T790M mutation. The present study knocked down BORIS in H1975 cells and the results demonstrated that the cell viability was significantly decreased in response to successful transfection with siBORIS (Fig. 1D). This finding indicated that the presence of BORIS may maintain the stability of H1975 cells and that its knockdown could be beneficial for treating drug-resistant lung cancer.
BORIS knockdown, alongside gefitinib treatment, inhibits NSCLC cell viability
To further explore the function of BORIS in TKI resistance, PC-9, PC-9IR and H1975 cells underwent BORIS knockdown and gefitinib treatment. PC-9 is an EGFR wild-type NSCLC cell line, whereas PC-9IR and H1975 are EGFR-mutant cells that are resistant to TKIs.
siBORIS effectively reduced the viability of NSCLC cells (Fig. 1D-F). All cell transfections were successful. When siBORIS was combined with gefitinib treatment, cell viability was significantly reduced compared with gefitinib treatment alone, with a more obvious effect observed on the drug-resistant cell lines H1975 and PC9-IR (Fig. 1D and 1F). Verification of the knockdown efficiency of siBORIS is presented in Fig. 1G. These findings suggested that the knockdown of BORIS may be beneficial for lung cancer resistance. Western blot analysis confirmed that the expression of BORIS was decreased in response to siBORIS.
Atractyloside mimics BORIS knockdown to suppress NSCLC viability
In a previous study, a drug that could mimic BORIS knockdown was identified (23). To identify bioactive drugs that might mimic the effects of BORIS knockdown, genes regulated in BORIS-silenced Caco2 cells were analyzed using microarray and a connectivity map database was screened for associated drugs in our previous study (23). Based on gene expression patterns and drug correlation analysis, metronidazole and atractyloside were identified as promising candidates for further study (23). These previous findings using the Caco2 cell line demonstrated that atractyloside (Fig. 1C) inhibited cell viability (23).
The H1299 (wild-type EGFR) and H1975 (EGFR mutation) NSCLC cells were used for assessing the response of siBORIS or atractyloside treatment. The results demonstrated that siBORIS treatment downregulated the expression of DNA repair-related genes, including BRCA-1, MSH6 and c-myc (Fig. 2B). Atractyloside treatment, on the other hand, increased XRCC4 expression while downregulating BRCA-1 (Fig. 2D), which is consistent with our previous observations (23). In H1299 and H1975 cells, atractyloside treatment resulted in DNA damage and upregulation of XRCC4 expression (Fig. 2B and D). These results indicated that atractyloside could mimic the effects of siBORIS to regulate the downstream genes (Fig. 2C and D); however, atractyloside treatment did not influence BORIS expression (Fig. 2A). In the present study atractyloside was used instead of siBORIS in subsequent experiments to regulate BORIS-related downstream genes.
Combination of atractyloside and gefitinib treatment reduces the proliferation of NSCLC cells
To avoid using a high concentration of atractyloside, which would induce mitochondrial permeability transition and cause apoptosis (4), 5 µM atractyloside was selected for application in two lung cancer cell lines, the wild-type cell line PC-9 and the T790M mutant drug-resistant cell line H1975.
In PC-9 cells, it was observed that, after 2 days of administration, gefitinib inhibited cell proliferation, and after 4 days administration, the proliferation of PC-9 cells was significantly inhibited by gefitinib (Fig. 3A). Atractyloside was shown to suppress PC-9 cell proliferation, but was less effective than gefitinib. In addition, the effect of the two-drug combination on cell proliferation was not significant, thus indicating that atractyloside had little effect on wild-type lung cancer cells. In H1975 cells, gefitinib at a concentration of 1 µM did not affect cell proliferation (Fig. 3B). However, when used in combination with atractyloside, cell proliferation was significantly decreased, indicating that the combined administration of atractyloside and gefitinib may affect the proliferation of TKI-resistant cells.
To further verify the inhibitory effect of combination therapy on EGFR-mutant cells, the 2nd-generation EGFR-TKI inhibitor erlotinib was used. After treatment with the drugs for 96 h, a combination of erlotinib and atractyloside suppressed H1975 cell proliferation better than erlotinib alone (Fig. 3C). Atractyloside demonstrated cytotoxic effects on cancer cells, with increasing concentrations inhibiting H1975 cell proliferation (Fig. 3D). When combined with gefitinib, the effect of atractyloside on drug-resistant lung cancer cells was stronger than that on wild-type cells, indicating that the BORIS pathway may be associated with lung cancer resistance. Western blot analysis indicated that atractyloside may suppress NSCLC cell proliferation by inhibiting AKT phosphorylation (Fig. 3E and F). This finding aligns with the results of a previous study demonstrating that AKT phosphorylation promotes lung cancer cell proliferation (24). AKT is a downstream factor of EGFR and can be used to examine the severity of cancer; therefore, the inhibition of AKT phosphorylation indicated that EGFR-related signaling was suppressed by atractyloside.
Atractyloside induces DNA damage in NSCLC
After knockdown of the expression of BORIS in the H1975 cell line, a decrease in the expression levels of the homologous recombination-related genes c-myc, BRCA-1 and the mismatch repair gene MSH6 (25), was detected (Fig. 2C). These findings were consistent with our previous results (12) and indicated the existence of BORIS-stabilized cell DNA. To further verify the effects of atractyloside on H1975 and PC-9 cells, XRCC4 protein expression was detected. The data demonstrated a consistent trend of XRCC4 upregulation, observed in response to both BORIS knockdown and atractyloside treatment (Fig. 4A-D). In PC-9 cells, BORIS knockdown elevated the expression of XRCC4 (Fig. 4A), as did atractyloside (Fig. 4B). In addition, in H1975 cells, BORIS knockdown elevated the expression of XRCC4 (Fig. 4C), as did atractyloside (Fig. 4D). In summary, atractyloside may disrupt DNA stability in H1975 cells and PC-9 cells.
Discussion
Gefitinib is a small-molecule EGFR-TKI that blocks the intracellular receptor binding site of adenosine triphosphate (ATP); this blocks downstream signal transduction, inhibits tumor cell proliferation and promotes apoptosis, all of which have a significant effect on the treatment of advanced NSCLC (13,26). However, the median survival time for patients from 61 centers across 11 European and Asia-Pacific countries with advanced NSCLC was revealed to be only 7–9 months, and drug resistance, frequently arising from secondary mutations, presents a significant obstacle to effective treatment (13). Among mutations, EGFR-T790M is considered the main cause of acquired drug resistance. This mutation competitively reduces binding with EGFR-TKIs to confer drug resistance by increasing the affinity between EGFR and ATP (27). Therefore, attention is required to identify novel ways to deal with this acquired drug resistance.
Through the analysis of BORIS expression in NSCLC in the present study, it was revealed that BORIS expression was increased in tissues from patients with high-risk NSCLC. As EGFR mutations are usually related with high-risk NSCLC, it may be hypothesized that BORIS is associated with EGFR mutations. In addition, BORIS knockdown or atractyloside treatment promoted TKI resistance in H1975 NSCLC cells with T790M mutation. These findings suggested that inhibiting BORIS could be a promising strategy for combination therapy with first-generation EGFR inhibitors in NSCLC. The prognosis of a number of patients with NSCLC is poor because of secondary drug-resistance gene mutations (27). However, the notable effect of BORIS knockdown provides a novel opportunity for treatment. In addition, atractyloside, a BORIS knockdown mimetic, holds promise as a means for treatment of NSCLC. However, the lack of in vivo experiments is a limitation of the present study. In future studies, we plan to conduct in-depth research on atractyloside and verify its medicinal value in xenograft models, since animal experiments may better reflect the occurrence and development of NSCLC. Future research may also construct stable drug-resistant H1975 and PC-9IR NSCLC cell lines with BORIS overexpression, and may assess treatment of drug resistance in lung carcinoma in situ and in a brain metastasis model of lung cancer.
In the present study, the knockdown of BORIS in H1975 cells or the administration of atractyloside decreased the expression of homologous recombination-related genes, such as BRCA-1. In addition, BORIS can influence DNA repair pathways, such as non-homologous end recombination, were compensatively upregulated, indicating that BORIS could stabilize the DNA of NSCLC cells. The H1299 cell line, expressing wild-type EGFR, was used to confirm that atractyloside treatment could induce XRCC4. Notably, the expression of DNA damage repair genes, including BRCA-1, c-myc and MSH6, was detected only in H1795 cells, which harbor an EGFR mutation. EGFR mutations or amplifications in NSCLC cells cause resistance to TKIs and induce downstream constitutive AKT phosphorylation, whereas inhibition of AKT reverses resistance to TKIs. In a study on neuroblastoma, ALK-mutated neuroblastoma cells were resistant to the ALK inhibitor TAE684 (18). Resistant cells exhibited upregulation of BORIS, which could lead to wide-ranging changes in chromatin interactions and transcriptional reprogramming. A 10-fold gain in genome-wide occupancy by BORIS was observed in resistant cells (22,891 vs. 2,211 in the sensitive cells) (18). We observed increased BORIS expression in TKI-resistant NSCLC cells and identified AKT as a potential downstream target of BORIS activation in these cells. The present study observed that atractyloside may inhibit AKT activity and suppress NSCLC cell proliferation, but did not affect the expression of BORIS. Inhibition of AKT by atractyloside suggests crosstalk between BORIS and factors downstream of EGFR.
In summary, BORIS expression was increased in patients with high-risk lung cancer, as determined by comparing groups with different survival rates. Notably, atractyloside is an inhibitor of the BORIS pathway and may be a potential therapeutic drug against TKI resistance.
Acknowledgements
Not applicable.
Funding
This study was supported by grants from the Zhejiang Provincial Natural Science Foundation of China (grant no. HDMY22H318024), the Medical and Health Science and Technology Project of Zhejiang Province (grant no. 2022RC128), and the Foundation of the Zhejiang Academy of Medical Sciences to Yanmei Zhang.
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
Conceptualization of the study was directed by YQ and YZ. The experiments were performed by WY, CL, NZ and YZ. The original draft was written by CL, WY and YZ. The review, editing and revisions were completed by YQ and YZ. The visualization of the data and the generation of the figures were performed by CL and NZ. The funding was provided by YQ and YZ. All authors read and approved the final version of the manuscript. WY and CL confirmed the authenticity of all the raw data.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Glossary
Abbreviations
Abbreviations:
NSCLC |
non-small cell lung cancer |
BORIS |
brother of the regulator of imprinted sites |
ATP |
adenosine triphosphate |
TKI |
tyrosine kinase inhibitor |
References
Chen LY, Hu A and Chang CJ: The degradation mechanism of toxic atractyloside in herbal medicines by decoction. Molecules. 18:2018–2028. 2013. View Article : Google Scholar : PubMed/NCBI | |
Xu J, Liu C, Shi K, Sun X, Song C, Xu K and Liu Y: Atractyloside-A ameliorates spleen deficiency diarrhea by interfering with TLR4/MyD88/NF-κB signaling activation and regulating intestinal flora homeostasis. Int Immunopharmacol. 107:1086792022. View Article : Google Scholar : PubMed/NCBI | |
Zhang P, Cheng X, Sun H, Li Y, Mei W and Zeng C: Atractyloside protect mice against liver steatosis by sctivation of autophagy via ANT-AMPK-mTORC1 Signaling Pathway. Front Pharmacol. 12:7366552021. View Article : Google Scholar : PubMed/NCBI | |
Li H, Shi X, Jiang H, Kang J, Yu M, Li Q, Yu K, Chen Z, Pan H and Chen W: CMap analysis identifies Atractyloside as a potential drug candidate for type 2 diabetes based on integration of metabolomics and transcriptomics. J Cell Mol Med. 24:7417–7426. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cho J, Zhang Y, Park SY, Joseph AM, Han C, Park HJ, Kalavalapalli S, Chun SK, Morgan D, Kim JS, et al: Mitochondrial ATP transporter depletion protects mice against liver steatosis and insulin resistance. Nat Commun. 8:144772017. View Article : Google Scholar : PubMed/NCBI | |
Ling X, Zhou Y, Li SW, Yan B and Wen L: Modulation of mitochondrial permeability transition pore affects multidrug resistance in human hepatocellular carcinoma cells. Int J Biol Sci. 6:773–783. 2010. View Article : Google Scholar : PubMed/NCBI | |
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu WJ, Du Y, Wen R, Yang M and Xu J: Drug resistance to targeted therapeutic strategies in non-small cell lung cancer. Pharmacol Ther. 206:1074382020. View Article : Google Scholar : PubMed/NCBI | |
Herbst RS, Morgensztern D and Boshoff C: The biology and management of non-small cell lung cancer. Nature. 553:446–454. 2018. View Article : Google Scholar : PubMed/NCBI | |
Aran V and Omerovic J: Current approaches in NSCLC targeting K-RAS and EGFR. Int J Mol Sci. 20:57012019. View Article : Google Scholar : PubMed/NCBI | |
Lu X, Yu L, Zhang Z, Ren X, Smaill JB and Ding K: Targeting EGFRL858R/T790M and EGFRL858R/T790M/C797S resistance mutations in NSCLC: Current developments in medicinal chemistry. Med Res Rev. 38:1550–1581. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Song Y, Li C, Ren J, Fang M, Fang J and Wang X: Brother of regulator of imprinted sites inhibits cisplatin-induced DNA damage in non-small cell lung cancer. Oncol Lett. 20:2512020. View Article : Google Scholar : PubMed/NCBI | |
Mok TSK, Kim SW, Wu YL, Nakagawa K, Yang JJ, Ahn MJ, Wang J, Yang JC, Lu Y, Atagi S, et al: Gefitinib plus chemotherapy versus chemotherapy in epidermal growth factor receptor mutation-positive non-small-cell lung cancer resistant to first-line gefitinib (IMPRESS): Overall survival and biomarker analyses. J Clin Oncol. 35:4027–4034. 2017. View Article : Google Scholar : PubMed/NCBI | |
Soltanian S and Dehghani H: BORIS: A key regulator of cancer stemness. Cancer Cell Int. 18:1542018. View Article : Google Scholar : PubMed/NCBI | |
Loukinov D: Targeting CTCFL/BORIS for the immunotherapy of cancer. Cancer Immunol Immunother. 67:1955–1965. 2018. View Article : Google Scholar : PubMed/NCBI | |
Asano T, Hirohashi Y, Torigoe T, Mariya T, Horibe R, Kuroda T, Tabuchi Y, Saijo H, Yasuda K, Mizuuchi M, et al: Brother of the regulator of the imprinted site (BORIS) variant subfamily 6 is involved in cervical cancer stemness and can be a target of immunotherapy. Oncotarget. 7:11223–11237. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Fang M, Song Y, Ren J, Fang J and Wang X: Brother of regulator of imprinted sites (BORIS) suppresses apoptosis in colorectal cancer. Sci Rep. 7:407862017. View Article : Google Scholar : PubMed/NCBI | |
Debruyne DN, Dries R, Sengupta S, Seruggia D, Gao Y, Sharma B, Huang H, Moreau L, McLane M, Day DS, et al: BORIS promotes chromatin regulatory interactions in treatment-resistant cancer cells. Nature. 572:676–680. 2019. View Article : Google Scholar : PubMed/NCBI | |
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI | |
Hou J, Aerts J, den Hamer B, van Ijcken W, den Bakker M, Riegman P, van der Leest C, van der Spek P, Foekens JA, Hoogsteden HC, et al: Gene expression-based classification of non-small cell lung carcinomas and survival prediction. PLoS One. 5:e103122010. View Article : Google Scholar : PubMed/NCBI | |
Huang S, Reitze NJ, Ewing AL, McCreary S, Uihlein AH, Brower SL, Wang D, Wang T, Gabrin MJ, Keating KE, et al: Analytical Performance of a 15-Gene Prognostic Assay for Early-Stage Non-Small-Cell Lung Carcinoma Using RNA-Stabilized Tissue. J Mol Diagn. 17:438–445. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhu CQ, Ding K, Strumpf D, Weir BA, Meyerson M, Pennell N, Thomas RK, Naoki K, Ladd-Acosta C, Liu N, et al: Prognostic and predictive gene signature for adjuvant chemotherapy in resected non-small-cell lung cancer. J Clin Oncol. 28:4417–4424. 2010. View Article : Google Scholar : PubMed/NCBI | |
Fang M, Song Y, Ren J, Yuan H, Fang J, Yan D, Zhang Y and Wang X: Atractyloside mimics BORIS knockdown to induce DNA damage in colorectal cancer cells. Int J Clin Exp Pathol. 11:3286–3293. 2018.PubMed/NCBI | |
Wang R, Wang S, Li Z, Luo Y, Zhao Y, Han Q, Rong XZ, Guo YX and Liu Y: PLEKHH2 binds β-arrestin1 through its FERM domain, activates FAK/PI3K/AKT phosphorylation, and promotes the malignant phenotype of non-small cell lung cancer. Cell Death Dis. 13:8582022. View Article : Google Scholar : PubMed/NCBI | |
Luoto KR, Meng AX, Wasylishen AR, Zhao H, Coackley CL, Penn LZ and Bristow RG: Tumor cell kill by c-MYC depletion: Role of MYC-regulated genes that control DNA double-strand break repair. Cancer Res. 70:8748–8759. 2010. View Article : Google Scholar : PubMed/NCBI | |
Arteaga CL: ErbB-targeted therapeutic approaches in human cancer. Exp Cell Res. 284:122–130. 2003. View Article : Google Scholar : PubMed/NCBI | |
Jin X, Wang J, Shen H, Ran R, Xu K, Zhang W, Tong X and Feng L: Curcumin co-treatment ameliorates resistance to gefitinib in drug- resistant NCI-H1975 lung cancer cells. J Tradit Chin Med. 37:355–360. 2017. View Article : Google Scholar : PubMed/NCBI |