Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells

  • Authors:
    • Kento Kurata
    • Naoyoshi Onoda
    • Satoru Noda
    • Shinichiro Kashiwagi
    • Yuka Asano
    • Kosei Hirakawa
    • Masaichi Ohira
  • View Affiliations

  • Published online on: October 7, 2016     https://doi.org/10.3892/ijo.2016.3723
  • Pages: 2303-2308
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Anaplastic thyroid cancer (ATC) is a rare malignancy that progresses extremely aggressively and often results in dismal prognosis. We investigated the efficacy of inhibiting the activated RAS/RAF/MEK pathway in ATC cells aiming to clarify the mechanism of effect and resistance. Four human ATC cell lines (ACT-1, OCUT-2, OCUT-4 and OCUT-6) were used. OCUT-4 had a BRAF mutation. OCUT-2 had both BRAF and PI3KCA mutations. ACT-1 and OCUT-6 had wild-type BRAF and NRAS mutations. The effects of dabrafenib, a selective inhibitor of the BRAFV600E kinase, and trametinib, a reversible inhibitor of MEK activity, were investigated. Dabrafenib strongly inhibited the viability in BRAF mutated cells by demonstrating G0/G1-arrest via the downregulation of MEK/ERK phosphorylation. Upregulated phosphorylation of MEK was observed in RAS mutated cells after dabrafenib treatment and caused VEGF upregulation, but was not related to the cellular proliferation. Trametinib inhibited the cellular viability to variable degrees in every cell by downregulating ERK phosphorylation. Dual blockade by both inhibitors demonstrated clear cytostatic effect in all the cells. OCUT-4 showed the weakest sensitivity to trametinib, no additional effect of either inhibitor in combination with the other, and an increase of SNAI1 mRNA expression after treatment with inhibitors, suggesting a mechanism for resistance. Our findings demonstrated the efficacy of a mutation-selective BRAF inhibitor and a MEK inhibitor in human ATC cells in a genetic alteration-specific manner.

Introduction

Anaplastic thyroid cancer (ATC) is a rare orphan disease that accounts for 1–3% of thyroid cancers. ATCs progress rapidly and are extremely aggressive toward both adjacent organs by invasion and distant organs by hematological dissemination. Because of the highly malignant potential, ATC cases often become lethal within 6 months from the initial diagnosis, despite intensive therapeutic efforts (13). No standardized therapeutic strategy has been documented to manage ATC, and experimental multimodal therapies with surgery, chemotherapy and/or radiation therapy have been attempted practically. Regrettably, no effective therapeutic method for ATC has been established to date (4,5).

However, several molecular targeted therapies have achieved successful results against ATCs (68). Rosove et al (9) reported an impressive case of an ATC patient successfully treated with a selective BRAFV600E inhibitor, vemurafenib. Possible clinical application of this inhibitor has been demonstrated recently in a phase 2 trial in BRAFV600E mutation-positive ATC patients, demonstrating an overall response rate of 29% (2/7) (10). In a study by Kim et al (11), BRAF mutation in papillary thyroid cancer (PTC) was found more frequently in East Asian countries compared to the Western countries, and the proportion of PTCs among differentiated thyroid cancers (DTCs) was higher in Japan than Western countries. A considerable proportion of ATCs is thought to be derived from long-lasting DTC, and BRAF mutation was found to be maintained during the phenotypical change from DTC to ATC (12). Although the incidence of BRAF mutation was reported to be relatively less common in ATC (15–24%) than that found in PTC (1315), a preliminary finding indicated that the rate of BRAF mutation in a population of Japanese ATC patients was high (6 of 14 patients) [Uchino, et al, Proceedings of the 20th Annual Meeting of Japan Association of Endocrine Surgeons, O-11 65, 2008 (In Japanese)]. In addition, six of seven thyroid cancer cell lines in our series have a BRAF mutation (16). These observations suggest that the frequency of BRAF mutation in ATC is much higher in Japan compared to Western countries.

A previous study of our group demonstrated a possible effect of molecular therapies targeting epidermal growth factor receptor (EGFR), although the effect was limited to the cells with a preserved RAS/RAF/MEK pathway (17). Our more recent study demonstrated that part of this EGFR-targeted therapy resistance could be overcome with an mTOR inhibitor, although we again observed that the efficacy was limited to the cells with an altered PI3K/AKT/mTOR pathway (18). These observations indicated the importance of direct targeting to the RAS/RAF/MEK pathway to manage ATC.

Another research group also demonstrated the importance of BRAF mutation in the aggressive characteristics of thyroid cancer and the efficacy of its inhibition on the management of the disease (19). Several studies described important roles of BRAF gene alteration in genome-wide aberrant methylation (20), vascular endothelial growth factor (VEGF) expression (21), and the induction of epithelial-mesenchymal transformation (EMT) (22).

We conducted a preclinical investigation of the efficacy of inhibiting the RAS/RAF/MEK pathway in a series of authentic ATC cell lines harboring a genetic alteration in either BRAF or NRAS (16). Our specific aims were to determine the efficacies of BRAF/MEK inhibitors in ATC cells and to identify possible differences in the mechanism of blockade between the cell lines according to the differences in the genetic alterations of the cell lines, the levels of VEGF secretion, and/or the expression of EMT markers.

Materials and methods

Cell lines

Four human ATC cell lines, ACT-1, OCUT-2, OCUT-4 and OCUT-6 were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 IU/ml of penicillin and 100 μg/ml of streptomycin at 37°C with 5% CO2 in a humidified condition. The ACT-1 cell line was kindly provided by Dr S. Ohata of Tokushima University. The other three cell lines were established in our institute (16). The OCUT-4 cell line had a BRAF (1799T>A; V600E) gene mutation. The OCUT-2 cell line had both BRAF (1799T>A; V600E) and PI3KCA (3140A>G; H1047R) gene mutations. The ACT-1 line harbored the wild-type BRAF gene and an NRAS (181C>A; Q61K) mutation. The OCUT-6 cells had the wild-type BRAF gene and an NRAS (182A>G; Q61R) mutation.

Inhibitors and drugs

Dabrafenib and trametinib were provided by Novartis (Basel, Switzerland).

Cell viability after exposure to the inhibitors

Cells (1×103) were seeded in each well of a 96-well plastic culture plate and left overnight. They were then treated with the intended doses of inhibitors for 72 h. After the incubation period, MTT reagent (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide, Dojindo Laboratories, Kumamoto, Japan) was added to the final concentration of 0.5 mg/ml, and the cells were incubated again for 2 h under the same condition. The culture plate was centrifuged at 200 g for 5 min, and the supernatant was removed. Dimethyl sulfoxide was added for reaction, and the absorbency at 570 nm was measured with a microplate reader (Infinite F50, Tecan Trading, Männedorf, Switzerland) and calculated using the supplied software. The experiments were carried out three times independently, in triplicate each time, and the average values of the three independent experiments were calculated (17).

Western blotting

Cells were incubated in 10 ml of DMEM containing 1,000 nM dabrafenib or 500 nM trametinib for 1 h. The cells were then rinsed with phosphate-buffered saline (PBS) and lysed with Pro-Prep (iNtRON Biotechnology, Kyungki-Do, Korea). After the protein concentration of each sample was adjusted, the lysates were electrophoretically separated using 4–12% Tris/Gly gels (Novex, Carlsbad, CA, USA) and transferred to a polyvinylidene difluoride membrane (Trans-Blot Turbo Transfer Pack, Bio-Rad, Hercules, CA, USA). Membranes were blocked with skim milk and incubated either with anti-human p44/42 MAPK antibody (#4695S; Cell Signaling Technology, Beverly, MA, USA), anti-human phospho-p44/42 MAPK antibody (T202/Y204) (#9101S; Cell Signaling Technology), anti-human MEK1/2 antibody (#8727S; Cell Signaling Technology), anti-human phospho-MEK1/2 (S217/221) (#9154S; Cell Signaling Technology) and anti-human β-actin antibody (#4963; Cell Signaling Technology) using SNAP i.d. (Merck, Darmstadt, Germany). The bands were detected using an enhanced chemiluminescence system (ImageQuant LAS 4000mini, General Electric, Fairfield, CA, USA).

Cell cycle analysis by flow cytometry

The cells treated with 100 nM of dabrafenib or 5 nM of trametinib for 24 h were collected after brief trypsinization, washed with PBS and fixed with 70% cold ethanol. The samples were then treated with ribonuclease (R6513: Sigma-Aldrich, St. Louis, MO, USA), stained with 10 mg/ml propidium iodine and analyzed by a cell sorter (FACScan, Becton-Dickinson, Mountain View, CA, USA). The cell cycle distributions were quantified using CellQuest software (17).

Measurement of VEGF secretion

Approximately 1×105 cells were seeded on a 10-mm plastic culture plate in 5 ml of culture medium, and treated with either or both 100 nM of dabrafenib or 5 nM of trametinib for 24 h. The conditioned medium was then sampled, and the concentrations of VEGF were measured by an enzyme-linked immunosorbent assay (ELISA; Mitsubishi, Tokyo, Japan). Culture medium without cells was used to measure the baseline concentrations (16).

Reverse transcription-polymerase chain reaction (RT-PCR)

The cells were treated with 1,000 nM of dabrafenib or 500 nM of trametinib for 1 h. After incubation, total cellular RNA was isolated using an RNeasy Mini kit (Qiagen, Hilden, Germany) and was reverse transcribed into cDNA with the use of ReverTra Ace qPCR RT Master Mix (Toyobo, Osaka, Japan) according to the manufacturer’s instructions. Reverse transcription-polymerase chain reaction (RT-PCR) was performed using a StepOnePlus™ Real-Time PCR system (Applied Biosystems, Foster City, CA, USA), with TaqMan® Gene Expression Assays (Thermo Fisher Scientific, Waltham, MA, USA) for GAPDH (Hs02758991), SNAI1 (Hs00195591), SNAI2 (Hs00950344) and TWIST1 (Hs01675818). The threshold cycle (CT) values were used to calculate the relative expression ratios between control and treated cells. We performed the relative quantification of gene expression by the 2−ΔΔCT method (23).

Results

Cell viability after exposure to the inhibitors

The dabrafenib treatment resulted in dose-dependent inhibitions of cell viability. The cellular viability was significantly more strongly inhibited in the OCUT-2 and OCUT-4 cells, which harbor a BRAF V600E mutation, compared to the ACT-1 and OCUT-6 cells, which have the wild-type BRAF gene (Fig. 1, left). The efficacy of trametinib was found in all four cell lines, with no relationship to the gene mutation status. The OCUT-6 line (the NRAS mutant) showed the weakest sensitivity to dabrafenib and the highest sensitivity to trametinib among all of the cell lines. The OCUT-4 line (the BRAF mutant) showed the weakest sensitivity to trametinib (Fig. 1, right). Significant impairment of the cellular viability by trametinib in addition to that by dabrafenib was observed in all cell lines tested (Fig. 2).

Alteration of the phosphorylation status of ERK and MEK after exposure to inhibitors

There was a clear downregulation in the phosphorylation of ERK and MEK after exposure to dabrafenib in the BRAF mutant cell lines, OCUT-2 and -4. The phosphorylation of ERK was also significantly decreased after trametinib exposure in these cell lines, but the phosphorylation of MEK was increased at the same time. The combination treatment with dabrafenib and trametinib resulted in the additional shut-down of ERK phosphorylation. In contrast, an upregulation of the phosphorylation of ERK was observed in the two NRAS mutant cell lines ACT-1 and OCUT-6 after exposure to dabrafenib. Trametinib clearly inhibited the phosphorylation of MEK in these NRAS mutant cells, and significant shutdown of ERK phosphorylation was observed after the dual blockade with dabrafenib and trametinib (Fig. 3).

The effects of the inhibitors on cell cycle progression

Significant increases in the proportion of cells in the G0/G1 phase were observed after exposure to dabrafenib in both the OCUT-2 and -4 lines. The G0/G1 arrest was not observed in the RAS mutant ACT-1 or OCUT-6 cells after dabrafenib treatment. Trametinib induced G0/G1 arrest in all four cell lines. Sub-G1 population cells were scarcely observed (0.7–4.4%) after treatment (Fig. 4).

The effects of the inhibitors on VEGF secretion of the cell lines

The VEGF concentration in the conditioned medium varied among the cell lines. The OCUT-2 cells demonstrated the highest concentration at 13,500 pg/ml and the OCUT-4 cells showed the lowest concentration at 384 pg/ml in the stable condition. The concentration of VEGF decreased after dabrafenib treatment in the OCUT-2 and -4 cells, whereas it increased after dabrafenib treatment in the two cell lines with wild-type BRAF, i.e., the OCUT-6 and ACT-1 cells. A decrease in the VEGF concentration in the conditioned medium was observed in all four cell lines after trametinib treatment (Fig. 5).

The effects of the inhibitors on the expression of EMT markers

A significant decrease in the mRNA expression of the EMT markers snail (SNAI1), slug (SNAI2) and twist (TWIST) was observed in the OCUT-2 cells after exposure to dabrafenib and trametinib, alone and in combination. Increased expression of SNAI1 mRNA was seen in the OCUT-4 cell line after exposure to either and both inhibitors. The changes of SNAI2 and TWIST expressions after treatment were not significant in the OCUT-4 line (Fig. 6).

Discussion

Dabrafenib is a reversible and potent ATP-competitive inhibitor that selectively inhibits the BRAFV600E kinase (24). In the OCUT-2 and -4 cell lines, which harbor a BRAFV600E mutation, dabrafenib clearly inhibited cellular growth by demonstrating G0/G1 arrest. The strongest inhibitory effect was in the OCUT-4 cells, in which the marked activation of a downstream pathway from BRAF gene was observed in the stable culturing condition. The phosphorylations of MEK and ERK were strongly downregulated by exposure to dabrafenib in OCUT-4 cells, causing a significant G0/G1 arrest. In the OCUT-2 line, the mutation in PI3KCA gene in addition to BRAFV600E mutation (16) and signaling through the PI3K/AKT/mTOR pathway can also be expected to contribute to aberrant cell proliferation to some extent. Nevertheless, the dabrafenib treatment resulted in a degree of growth inhibition by G0/G1 arrest in the OCUT-2 cells that was similar to that observed in the OCUT-4 cells. This observation suggested that the activated MAPK/ERK pathway, and not the PI3K/AKT/mTOR pathway, was the main driver for aggressive cell proliferation in OCUT-2 cell line. The results indicate that the inhibition of BRAFV600E by dabrafenib might be effective against cancer cells harboring active alterations in both the MAPK/ERK and PI3K/AKT/mTOR pathways.

We observed an upregulation of phosphorylated ERK after dabrafenib exposure in ACT-1 and OCUT-6 cells, which have an NRAS mutation. The mechanism of the upregulation of p-ERK in RAS mutant cells after treatment with a selective BRAFV600E kinase inhibitor has been investigated. Dimeric complexes with wild-type BRAF, CRAF or kinase-dead BRAF is able to generate excessive downstream signaling under stimulation by mutant RAS enzyme (25). This mechanism resulted in paradoxical phosphorylation in ERK after BRAF inhibition, but did not contribute to the cell cycle progression or cell growth in the present study. However, the VEGF secretion was clearly stimulated in the NRAS mutant cells after dabrafenib treatment in our study. VEGF is well known as a strong inducer of cancer neo-vasculature that contributes to the arrangement of the cancer microenvironment for aggressive growth. Our present findings indicated one of the potential mechanisms of tumor growth in dabrafenib-resistant NRAS mutant cancer cells.

The treatment with trametinib, a reversible allosteric inhibitor of MEK1 and MEK2 activation and kinase activity, resulted in universal growth suppression in all four cell lines independent of the mutational status of BRAF or NRAS. A weak growth-inhibitory effect was observed in the BRAF mutant OCUT-4 cells. After trametinib exposure, inhibition of the phosphorylation of ERK was clear, and cell cycle arrest was obviously identified. Nevertheless, the phosphorylation of MEK was strongly induced in OCUT-4 cells by trametinib treatment, suggesting that resistance to trametinib could be caused by a mechanism other than one downstream of the MAPK/ERK pathway. This hypothesis was also suggested by the result of our dual blockade by dabrafenib and trametinib. There was no additional effect of either inhibitor in combination with the other in the OCUT-4 cell line, suggesting a limited effect of inhibiting the MAPK/MEK pathway. In addition, the OCUT-4 cells showed a different EMT marker expression profile after exposure to the inhibitors. Only this cell line showed an upregulation in the expression of the mRNA of SNAI1.

The expression of EMT markers is thought to have a role in the acquisition of resistance to a cytotoxic drug (26). Our present findings indicated that the phenotypical change through the EMT also contributed to the mechanism of resistance to these inhibitors. Several mechanisms have been confirmed to trigger resistance to BRAF inhibition (2730). Additional investigations are needed to clarify the involvement of the EMT in the effect of BRAF inhibition.

Dabrafenib and trametinib, as monotherapy or in combination, were approved for the treatment of melanomas by the US Food and Drug Administration. Dabrafenib as a treatment for advanced thyroid cancer resulted in durable responses in BRAF-mutant DTC patients (31). A recent report suggested the re-differentiation of iodine-refractory thyroid cancer after dabrafenib treatment (32). These observations clearly indicate the usefulness of BRAF/MEK inhibitors for the management of advanced and inoperable thyroid cancer. The results of the present study suggest the importance of selecting ATC patients in accord with the mutation status of BRAF and RAS when applying inhibitors (33).

Our present findings demonstrated the efficacy of a mutation-selective BRAF inhibitor and a MEK inhibitor in human ATC cell lines. Our observations indicated the existence of a unique driver gene for the aggressive proliferation of ATC cancer cells, and we observed that a cellular growth inhibitory effect can be expected when appropriate inhibitor(s) are selected.

Acknowledgements

This study was supported in part by Grants-in-Aid for Scientific Research (JSPS KAKENHI, #25461992). N. Onoda received honoraria from Bayer and Eisai, research grant from Bayer. The English of this manuscript has been edited and proofread by Ms. Mary Stewart of KN International Inc.

References

1 

Sugitani I, Miyauchi A, Sugino K, Okamoto T, Yoshida A and Suzuki S: Prognostic factors and treatment outcomes for anaplastic thyroid carcinoma: ATC Research Consortium of Japan cohort study of 677 patients. World J Surg. 36:1247–1254. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Kebebew E, Greenspan FS, Clark OH, Woeber KA and McMillan A: Anaplastic thyroid carcinoma. Treatment outcome and prognostic factors. Cancer. 103:1330–1335. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Haymart MR, Banerjee M, Yin H, Worden F and Griggs JJ: Marginal treatment benefit in anaplastic thyroid cancer. Cancer. 119:3133–3139. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Smallridge RC, Ain KB, Asa SL, Bible KC, Brierley JD, Burman KD, Kebebew E, Lee NY, Nikiforov YE, Rosenthal MS, et al; American Thyroid Association Anaplastic Thyroid Cancer Guidelines Taskforce. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid. 22:1104–1139. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Sugino K and Onoda N: Part VII. Anaplastic carcinoma. Treatment of Thyroid cancer. Japanese Clinical Guidelines. Takami H, Ito Y, Noguchi H, Yoshida A and Okamoto T: Springer; Japan, Tokyo: pp. 203–230. 2012

6 

Haugen BR and Sherman SI: Evolving approaches to patients with advanced differentiated thyroid cancer. Endocr Rev. 34:439–455. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Onoda N, Ito Y, Ito K, Sugitani I, Takahashi S, Yamaguchi I, Kawakami Y and Tsukada K: Phase II clinical trial of sorafenib in Japanese patients with anaplastic thyroid carcinoma and locally advanced or metastatic medullary thyroid carcinoma. Thyroid. 25:A1202015.

8 

Takahashi S, Tahara M, Kiyota N, Yamazaki T, Chayahara N, Nakano K, Inagaki R, Toda K, Enokida T, Minami H, et al: Phase II study of lenvatinib, a multi-targeted tyrosine kinase inhibitor, in patients with all histologic subtypes of advanced thyroid cancer. Ann Oncol. 25(Suppl_4): iv340–iv356. 2014.

9 

Rosove MH, Peddi PF and Glaspy JA: BRAF V600E inhibition in anaplastic thyroid cancer. N Engl J Med. 368:684–685. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, Wolf J, Raje NS, Diamond EL, Hollebecque A, et al: Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. 373:726–736. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Kim TH, Park YJ, Lim JA, Ahn HY, Lee EK, Lee YJ, Kim KW, Hahn SK, Youn YK, Kim KH, et al: The association of the BRAF(V600E) mutation with prognostic factors and poor clinical outcome in papillary thyroid cancer: A meta-analysis. Cancer. 118:1764–1773. 2012. View Article : Google Scholar

12 

Smallridge RC and Copland JA: Anaplastic thyroid carcinoma: Pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol). 22:486–497. 2010. View Article : Google Scholar

13 

Xing M: BRAF mutation in thyroid cancer. Endocr Relat Cancer. 12:245–262. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Wang HM, Huang YW, Huang JS, Wang CH, Kok VC, Hung CM, Chen HM and Tzen CY: Anaplastic carcinoma of the thyroid arising more often from follicular carcinoma than papillary carcinoma. Ann Surg Oncol. 14:3011–3018. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Shi X, Liu R, Qu S, Zhu G, Bishop J, Liu X, Sun H, Shan Z, Wang E, Luo Y, et al: Association of TERT promoter mutation 1,295,228 C>T with BRAF V600E mutation, older patient age, and distant metastasis in anaplastic thyroid cancer. J Clin Endocrinol Metab. 100:E632–E637. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Onoda N, Nakamura M, Aomatsu N, Noda S, Kashiwagi S and Hirakawa K: Establishment, characterization and comparison of seven authentic anaplastic thyroid cancer cell lines retaining clinical features of the original tumors. World J Surg. 38:688–695. 2014. View Article : Google Scholar

17 

Nobuhara Y, Onoda N, Yamashita Y, Yamasaki M, Ogisawa K, Takashima T, Ishikawa T and Hirakawa K: Efficacy of epidermal growth factor receptor-targeted molecular therapy in anaplastic thyroid cancer cell lines. Br J Cancer. 92:1110–1116. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Onoda N, Nakamura M, Aomatsu N, Noda S, Kashiwagi S, Kurata K, Uchino S and Hirakawa K: Significant cytostatic effect of everolimus on a gefitinib-resistant anaplastic thyroid cancer cell line harboring PI3KCA gene mutation. Mol Clin Oncol. 3:522–526. 2015.PubMed/NCBI

19 

Salvatore G, De Falco V, Salerno P, Nappi TC, Pepe S, Troncone G, Carlomagno F, Melillo RM, Wilhelm SM and Santoro M: BRAF is a therapeutic target in aggressive thyroid carcinoma. Clin Cancer Res. 12:1623–1629. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Hou P, Liu D and Xing M: Genome-wide alterations in gene methylation by the BRAF V600E mutation in papillary thyroid cancer cells. Endocr Relat Cancer. 18:687–697. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Jo YS, Li S, Song JH, Kwon KH, Lee JC, Rha SY, Lee HJ, Sul JY, Kweon GR, Ro HK, et al: Influence of the BRAF V600E mutation on expression of vascular endothelial growth factor in papillary thyroid cancer. J Clin Endocrinol Metab. 91:3667–3670. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Rusinek D, Szpak-Ulczok S and Jarzab B: Gene expression profile of human thyroid cancer in relation to its mutational status. J Mol Endocrinol. 47:R91–R103. 2011. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Menzies AM and Long GV: Dabrafenib and trametinib, alone and in combination for BRAF-mutant metastatic melanoma. Clin Cancer Res. 20:2035–2043. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Cichowski K and Jänne PA: Drug discovery: Inhibitors that activate. Nature. 464:358–359. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, et al: Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 527:472–476. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Carlino MS, Todd JR, Gowrishankar K, Mijatov B, Pupo GM, Fung C, Snoyman S, Hersey P, Long GV, Kefford RF, et al: Differential activity of MEK and ERK inhibitors in BRAF inhibitor resistant melanoma. Mol Oncol. 8:544–554. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Johannessen CM, Boehm JS, Kim SY, Thomas SR, Wardwell L, Johnson LA, Emery CM, Stransky N, Cogdill AP, Barretina J, et al: COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature. 468:968–972. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar H, et al: Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature. 468:973–977. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, Pochanard P, Kehoe SM, Johannessen CM, Macconaill LE, Hahn WC, et al: Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol. 29:3085–3096. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Falchook GS, Millward M, Hong D, Naing A, Piha-Paul S, Waguespack SG, Cabanillas ME, Sherman SI, Ma B, Curtis M, et al: BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer. Thyroid. 25:71–77. 2015. View Article : Google Scholar :

32 

Rothenberg SM, McFadden DG, Palmer EL, Daniels GH and Wirth LJ: Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clin Cancer Res. 21:1028–1035. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Cabanillas ME, Patel A, Danysh BP, Dadu R, Kopetz S and Falchook G: BRAF inhibitors: Experience in thyroid cancer and general review of toxicity. Horm Cancer. 6:21–36. 2015. View Article : Google Scholar :

Related Articles

Journal Cover

December-2016
Volume 49 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kurata K, Onoda N, Noda S, Kashiwagi S, Asano Y, Hirakawa K and Ohira M: Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells. Int J Oncol 49: 2303-2308, 2016
APA
Kurata, K., Onoda, N., Noda, S., Kashiwagi, S., Asano, Y., Hirakawa, K., & Ohira, M. (2016). Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells. International Journal of Oncology, 49, 2303-2308. https://doi.org/10.3892/ijo.2016.3723
MLA
Kurata, K., Onoda, N., Noda, S., Kashiwagi, S., Asano, Y., Hirakawa, K., Ohira, M."Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells". International Journal of Oncology 49.6 (2016): 2303-2308.
Chicago
Kurata, K., Onoda, N., Noda, S., Kashiwagi, S., Asano, Y., Hirakawa, K., Ohira, M."Growth arrest by activated BRAF and MEK inhibition in human anaplastic thyroid cancer cells". International Journal of Oncology 49, no. 6 (2016): 2303-2308. https://doi.org/10.3892/ijo.2016.3723