Cancer Res Treat.  2018 Jan;50(1):239-254. 10.4143/crt.2016.580.

FOXO1 Suppression is a Determinant of Acquired Lapatinib-Resistance in HER2-Positive Gastric Cancer Cells Through MET Upregulation

Affiliations
  • 1Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea. dslanat@snu.ac.kr
  • 2Department of Forensic Medicine, National Forensic Service Busan Institute, Yangsan, Korea.
  • 3Department of Pathology, Seoul National University College of Medicine, Seoul, Korea.
  • 4Department of Pathology, Eulji University Hospital, Eulji University School of Medicine, Daejeon, Korea.
  • 5Department of Pathology, Jeju National University Hospital, Jeju, Korea.
  • 6Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.
  • 7Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea.
  • 8Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea.

Abstract

PURPOSE
Lapatinib is a candidate drug for treatment of trastuzumab-resistant, human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC). Unfortunately, lapatinib resistance renders this drug ineffective. The present study investigated the implication of forkhead box O1 (FOXO1) signaling in the acquired lapatinib resistance in HER2-positive GC cells.
MATERIALS AND METHODS
Lapatinib-resistant GC cell lines (SNU-216 LR2-8) were generated in vitro by chronic exposure of lapatinib-sensitive, HER2-positive SNU-216 cells to lapatinib. SNU-216 LR cells with FOXO1 overexpression were generated by stable transfection of a constitutively active FOXO1 mutant (FOXO1A3). HER2 and MET in SNU-216 LR cells were downregulated using RNA interference. The sensitivity of GC cells to lapatinib and/or cisplatin was determined by crystal violet assay. In addition, Western blot analysis, luciferase reporter assay and reverse transcription-polymerase chain reaction were performed.
RESULTS
SNU-216 LR cells showed upregulations of HER2 and MET, but downregulation of FOXO1 compared to parental SNU-216 cells. FOXO1 overexpression in SNU-216 LR cells significantly suppressed resistance to lapatinib and/or cisplatin. In addition, FOXO1 negatively controlled HER2 and MET at the transcriptional level and was negatively controlled by these molecules at the post-transcriptional level. A positive crosstalk was shown between HER2 and MET, each of which increased resistance to lapatinib and/or cisplatin.
CONCLUSION
FOXO1 serves as an important linker between HER2 and MET signaling pathways through negative crosstalks and is a key regulator of the acquired lapatinib resistance in HER2-positive GC cells. These findings provide a rationale for establishing a novel treatment strategy to overcome lapatinib resistance in a subtype of GC patients.

Keyword

Stomach neoplasms; ErbB-2 receptor; Drug resistance; Lapatinib; Human FOXO1 protein; Human MET protein

MeSH Terms

Blotting, Western
Cell Line
Cisplatin
Down-Regulation
Drug Resistance
Gentian Violet
Humans
In Vitro Techniques
Luciferases
Parents
Receptor, Epidermal Growth Factor
Receptor, ErbB-2
RNA Interference
Stomach Neoplasms*
Transfection
Up-Regulation*
Cisplatin
Gentian Violet
Luciferases
Receptor, Epidermal Growth Factor
Receptor, ErbB-2

Figure

  • Fig. 1. Effect of chronic lapatinib treatment on SNU-216 cells. (A) Lapatinib-resistant (LR), human epidermal growth factor receptor 2 (HER2)–positive gastric cancer (GC) cell lines (SNU-216 LR2-8) were generated from a lapatinib-sensitive, HER2-positive SNU-216 GC cell line by chronic exposure to lapatinib over a period of 8 months. Twenty-four hours after plating, parental and lapatinib-resistant (LR) SNU-216 cells were treated with the indicated concentrations of lapatinib for 3 days, and cell viability was determined using crystal violet assay. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (B) Comparative analysis of total and phosphorylated HER2, MET, and AKT as well as total forkhead box O1 (FOXO1) by Western blot analysis (WB). mRNA expressions of HER2, MET, and FOXO1 were determined by reverse transcription–polymerase chain reaction (RT-PCR). β-Actin protein and mRNA were served as loading controls. (C) FOXO1 transcriptional activity was determined by the luciferase reporter assay and was normalized by β-galactosidase activity. Luciferase activity in parental SNU-216 cells was arbitrarily set to 1. Each bar represents the mean±standard deviation. a)p < 0.05 vs. parental SNU-216 cells.

  • Fig. 2. Effect of forkhead box O1 (FOXO1) overexpression on lapatinib and/or cisplatin sensitivity in lapatinib-resistant (LR) cell lines. SNU-216 LR3 and LR7 cells were transfected with empty pcDNA3 vector (pcDNA3) or FOXO1A3 mutant vector (FOXO1A3). Cell viability was measured by crystal violet assay. (A) FOXO1 overexpression was confirmed by Western blot analysis. (B) FOXO1 transcriptional activity was analyzed by the luciferase reporter assay. (C) Cells were treated with the indicated concentrations of lapatinib, and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (D) Cells were treated with the indicated concentrations of cisplatin, and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (E) Cells were treated with the 1 μmol/L lapatinib alone or combined with 10 μg/mL cisplatin (CDDP), and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). Each bar represents the mean±standard deviation. ns, not significant. a)p < 0.05 vs. pcDNA3 cells, b)p < 0.05 vs. lapatinib-treated pcDNA3 cells, c)p < 0.05 vs. cisplatin-treated pcDNA3 cells, d)p < 0.05 vs. pcDNA3 cells, e)p < 0.05 vs. lapatinib-treated FOXO1A3 cells.

  • Fig. 3. Association between forkhead box O1 (FOXO1) and human epidermal growth factor receptor 2 (HER2)/MET in lapatinib-resistant (LR) cell lines. SNU-216 LR3 and LR7 cells were transfected with empty pcDNA3 vector (pcDNA3) or FOXO1A3 mutant vector (FOXO1A3). (A) The protein expressions of total and phosphorylated HER2, MET, and AKT were determined by Western blot analysis (WB). (B) The mRNA expressions of HER2 and MET were evaluated by reverse transcription-polymerase chain reaction (RT-PCR).

  • Fig. 4. Effect of human epidermal growth factor receptor 2 (HER2) downregulation on lapatinib/cisplatin resistance in lapatinib-resistant (LR) cell lines. SNU-216 LR3 and LR7 cells were infected with a lentivirus containing either control shRNA (shCtrl) or HER2 shRNA (shHER2). Cell viability was measured by crystal violet assay. (A) The protein expressions of HER2, pAKT, and AKT were determined by Western blot analysis. (B) Twenty-four hours after plating, cells were cultured for 3 days and cell growth was determined at the indicated times. (C) Cells were treated with the indicated concentrations of lapatinib, and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (D) Cells were treated with the indicated concentrations of cisplatin, and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (E) Cells were treated with the 1 μmol/L lapatinib alone or combined with 10 μg/mL cisplatin (CDDP), and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). Each bar represents the mean±standard deviation. ns, not significant. a)p < 0.05 vs. shCtrl cells, b)p < 0.05 vs. lapatinib-treated shCtrl cells, c)p < 0.05 vs. cisplatin-treated shCtrl cells, d)p < 0.05 vs. shCtrl cells, e)p < 0.05 vs. lapatinib-treated shHER2 cells.

  • Fig. 5. Effect of MET downregulation on lapatinib/cisplatin resistance in lapatinib-resistant cell lines. SNU-216 LR3 and LR7 cells were transfected with pGFP-v-RS vectors containing either control scrambled shRNA (shCtrl) or MET shRNA (shMET). Cell viability was measured by crystal violet assay. (A) The protein expressions of MET, pAKT, and AKT were termined by Western blot analysis. (B) Cells were cultured for 3 days, and cell growth was determined at the indicated times. (C) Cells were treated with the indicated concentrations of lapatinib and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (D) Cells were treated with the indicated concentrations of cisplatin, and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). (E) Cells were treated with the 1 μmol/L lapatinib alone or combined with 10 μg/mL cisplatin (CDDP), and cell viability was measured after 3 days. The percentage of viable cells is shown relative to untreated cells (considered as 100%). Each bar represents the mean±standard deviation. ns, not significant. a)p < 0.05 vs. shCtrl cells, b)p < 0.05 vs. lapatinib-treated shCtrl cells, c)p < 0.05 vs. cisplatin-treated shCtrl cells, d)p < 0.05 vs. shCtrl cells, e)p < 0.05 vs. lapatinib-treated shMET cells.

  • Fig. 6. The relationships between human epidermal growth factor receptor 2 (HER2), MET, and forkhead box O1 (FOXO1) in lapatinib-resistant cells. (A-C) Cells were infected with a lentivirus containing either control shRNA (shCtrl) or HER2 shRNA (shHER2). (A) The expression and pMET, MET, and FOXO1 protein expression were determined by Western blot analysis. (B) FOXO1 transcriptional activity was determined by the luciferase reporter assay. (C) The mRNA expression of FOXO1 was evaluated by reverse transcription–polymerase chain reaction (RT-PCR). (D-F) Cells were transfected with pGFPv-RS vectors containing either control scrambled shRNA (shCtrl) or MET shRNA (shMET). (D) The expression and pHER2, HER2, and FOXO1 protein expression were determined by Western blot analysis. (E) FOXO1 transcriptional activity was determined by the luciferase reporter assay. (F) mRNA expression of FOXO1 was evaluated by RT-PCR. Each bar represents the mean±standard deviation. a)p < 0.05 vs. shCtrl cells, b)p < 0.05 vs. shCtrl cells.

  • Fig. 7. Model for forkhead box O1 (FOXO1)–dependent acquired lapatinib resistance and the crosstalk among FOXO1, human epidermal growth factor receptor 2 (HER2) and MET in lapatinib-resistant, HER2-positive gastric cancer (GC) cells. Downregulation of FOXO1 leads to coactivation of HER2 and MET, which are essential to lapatinib resistance. Reintroduction of FOXO1 is necessary to reduce the lapatinib resistance in a subpopulation of HER2-positive GC patients showing lapatinib resistance.


Reference

References

1. Gravalos C, Jimeno A. HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 2008; 19:1523–9.
Article
2. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010; 376:687–97.
Article
3. Liu L, Wu N, Li J. Novel targeted agents for gastric cancer. J Hematol Oncol. 2012; 5:31.
Article
4. Spector NL, Xia W, Burris H 3rd, Hurwitz H, Dees EC, Dowlati A, et al. Study of the biologic effects of lapatinib, a reversible inhibitor of ErbB1 and ErbB2 tyrosine kinases, on tumor growth and survival pathways in patients with advanced malignancies. J Clin Oncol. 2005; 23:2502–12.
Article
5. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007; 316:1039–43.
6. Corso S, Ghiso E, Cepero V, Sierra JR, Migliore C, Bertotti A, et al. Activation of HER family members in gastric carcinoma cells mediates resistance to MET inhibition. Mol Cancer. 2010; 9:121.
Article
7. Lee HE, Kim MA, Lee HS, Jung EJ, Yang HK, Lee BL, et al. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer. 2012; 107:325–33.
Article
8. Nakajima M, Sawada H, Yamada Y, Watanabe A, Tatsumi M, Yamashita J, et al. The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer. 1999; 85:1894–902.
Article
9. Ha SY, Lee J, Jang J, Hong JY, Do IG, Park SH, et al. HER2-positive gastric cancer with concomitant MET and/or EGFR overexpression: a distinct subset of patients for dual inhibition therapy. Int J Cancer. 2015; 136:1629–35.
Article
10. Chen CT, Kim H, Liska D, Gao S, Christensen JG, Weiser MR. MET activation mediates resistance to lapatinib inhibition of HER2-amplified gastric cancer cells. Mol Cancer Ther. 2012; 11:660–9.
Article
11. Zhang Z, Wang J, Ji D, Wang C, Liu R, Wu Z, et al. Functional genetic approach identifies MET, HER3, IGF1R, INSR pathways as determinants of lapatinib unresponsiveness in HER2-positive gastric cancer. Clin Cancer Res. 2014; 20:4559–73.
Article
12. Zhao M, Luo R, Liu Y, Gao L, Fu Z, Fu Q, et al. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat Commun. 2016; 7:11309.
Article
13. Goto T, Takano M, Hirata J, Tsuda H. The involvement of FOXO1 in cytotoxic stress and drug-resistance induced by paclitaxel in ovarian cancers. Br J Cancer. 2008; 98:1068–75.
Article
14. Han CY, Cho KB, Choi HS, Han HK, Kang KW. Role of FoxO1 activation in MDR1 expression in adriamycin-resistant breast cancer cells. Carcinogenesis. 2008; 29:1837–44.
Article
15. Park J, Ko YS, Yoon J, Kim MA, Park JW, Kim WH, et al. The forkhead transcription factor FOXO1 mediates cisplatin resistance in gastric cancer cells by activating phosphoinositide 3-kinase/Akt pathway. Gastric Cancer. 2014; 17:423–30.
Article
16. Ko YS, Cho SJ, Park J, Kim Y, Choi YJ, Pyo JS, et al. Loss of FOXO1 promotes gastric tumour growth and metastasis through upregulation of human epidermal growth factor receptor 2/neu expression. Br J Cancer. 2015; 113:1186–96.
Article
17. Kim HP, Han SW, Song SH, Jeong EG, Lee MY, Hwang D, et al. Testican-1-mediated epithelial-mesenchymal transition signaling confers acquired resistance to lapatinib in HER2-positive gastric cancer. Oncogene. 2014; 33:3334–41.
Article
18. Kim WH, Schnaper HW, Nomizu M, Yamada Y, Kleinman HK. Apoptosis in human fibrosarcoma cells is induced by a multimeric synthetic Tyr-Ile-Gly-Ser-Arg (YIGSR)-containing polypeptide from laminin. Cancer Res. 1994; 54:5005–10.
19. Kim JW, Kim HP, Im SA, Kang S, Hur HS, Yoon YK, et al. The growth inhibitory effect of lapatinib, a dual inhibitor of EGFR and HER2 tyrosine kinase, in gastric cancer cell lines. Cancer Lett. 2008; 272:296–306.
Article
20. Satoh T, Xu RH, Chung HC, Sun GP, Doi T, Xu JM, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN: a randomized, phase III study. J Clin Oncol. 2014; 32:2039–49.
21. Sebolt-Leopold JS, English JM. Mechanisms of drug inhibition of signalling molecules. Nature. 2006; 441:457–62.
Article
22. Sharma SV, Settleman J. Oncogene addiction: setting the stage for molecularly targeted cancer therapy. Genes Dev. 2007; 21:3214–31.
Article
23. Wu Y, Shang X, Sarkissyan M, Slamon D, Vadgama JV. FOXO1A is a target for HER2-overexpressing breast tumors. Cancer Res. 2010; 70:5475–85.
Article
24. Chakrabarty A, Bhola NE, Sutton C, Ghosh R, Kuba MG, Dave B, et al. Trastuzumab-resistant cells rely on a HER2-PI3K-FoxO-survivin axis and are sensitive to PI3K inhibitors. Cancer Res. 2013; 73:1190–200.
Article
25. Wang YC, Morrison G, Gillihan R, Guo J, Ward RM, Fu X, et al. Different mechanisms for resistance to trastuzumab versus lapatinib in HER2-positive breast cancers: role of estrogen receptor and HER2 reactivation. Breast Cancer Res. 2011; 13:R121.
Article
26. Rusnak DW, Alligood KJ, Mullin RJ, Spehar GM, Arenas-Elliott C, Martin AM, et al. Assessment of epidermal growth factor receptor (EGFR, ErbB1) and HER2 (ErbB2) protein expression levels and response to lapatinib (Tykerb, GW57-2016) in an expanded panel of human normal and tumour cell lines. Cell Prolif. 2007; 40:580–94.
Article
27. Huang D, Duan H, Huang H, Tong X, Han Y, Ru G, et al. Cisplatin resistance in gastric cancer cells is associated with HER2 upregulation-induced epithelial-mesenchymal transition. Sci Rep. 2016; 6:20502.
Article
28. Karamouzis MV, Konstantinopoulos PA, Papavassiliou AG. Targeting MET as a strategy to overcome crosstalk-related resistance to EGFR inhibitors. Lancet Oncol. 2009; 10:709–17.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr