Skip to main content
Erschienen in: International Journal of Clinical Oncology 5/2023

Open Access 01.03.2023 | Original Article

Regulation of MEK inhibitor selumetinib sensitivity by AKT phosphorylation in the novel BRAF L525R mutant

verfasst von: Chikako Nakai, Sachiyo Mimaki, Koutatsu Matsushima, Eiji Shinozaki, Kentaro Yamazaki, Kei Muro, Kensei Yamaguchi, Tomohiro Nishina, Satoshi Yuki, Kohei Shitara, Hideaki Bando, Yutaka Suzuki, Kiwamu Akagi, Shogo Nomura, Satoshi Fujii, Masaya Sugiyama, Nao Nishida, Masashi Mizokami, Yasuhiro Koh, Takuya Koshizaka, Hideki Okada, Yukiko Abe, Atsushi Ohtsu, Takayuki Yoshino, Katsuya Tsuchihara

Erschienen in: International Journal of Clinical Oncology | Ausgabe 5/2023

Abstract

Background

Oncogenic mutations in BRAF genes are found in approximately 5–10% of colorectal cancers. The majority of BRAF mutations are located within exons 11–15 of the catalytic kinase domains, with BRAF V600E accounting for more than 80% of the observed BRAF mutations. Sensitivity to BRAF- and mitogen-activated protein kinase (MEK) inhibitors varies depending on BRAF mutations and tumor cell types. Previously, we newly identified, BRAF L525R-mutation, in the activation segment of the kinase in colorectal cancer patient. Here, we characterized the function of the BRAF L525R mutation.

Methods

HEK293 cells harboring a BRAF mutation (V600E or L525R) were first characterized and then treated with cetuximab, dabrafenib, and selumetinib. Cell viability was measured using WST-1 assay and the expression of proteins involved in the extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) signaling pathways was evaluated using western blot analysis.

Results

The MEK inhibitor selumetinib effectively inhibited cell proliferation and ERK phosphorylation in BRAF L525R cells but not in BRAF V600E cells. Further studies revealed that AKT phosphorylation was reduced by selumetinib in BRAF L525R cells but not in BRAF V600E cells or selumetinib-resistant BRAF L525R cells. Moreover, the AKT inhibitor overcame the selumetinib resistance.

Conclusions

We established a model system harboring BRAF L525R using HEK293 cells. BRAF L525R constitutively activated ERK. AKT phosphorylation caused sensitivity and resistance to selumetinib. Our results suggest that a comprehensive network analysis may provide insights to identify effective therapies.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10147-023-02318-w.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AKT
Protein kinase B
COSMIC
Catalog of somatic mutations in cancer
CRC
Colorectal cancer
EGFR
Epidermal growth factor receptor
ERK
Extracellular signal-regulated kinase
MEK
Mitogen-activated protein kinase

Introduction

BRAF is a serine-threonine kinase that acts downstream of the epidermal growth factor (EGF) receptor (EGFR) in the rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) kinase pathway [1]. The RAF/MEK/ERK pathway has attracted much attention in the search for novel chemotherapeutic agents since mutations in BRAF are found in approximately 60% of melanoma [2, 3], 35–50% of papillary thyroid cancers [4, 5], 35% of low-grade ovarian serous tumors [6, 7], 5–10% of colorectal cancers (CRC) [810], and 5% of non-small cell lung cancers [11]. Genomic analyses have revealed various BRAF mutations in cancers, most of which occur in the kinase domain of the enzyme [12]. More than 80% of BRAF mutations include T to A transversions at nucleotide 1799, resulting in a substitution of valine (V) with glutamic acid (E) at codon 600 (V600E) in exon 15. Subtypes of BRAF mutations are classified as high, intermediate, and impaired depending on their effect on the kinase activity [13]. V600E causes constitutive activation of the downstream effectors MEK and ERK [14]. Another activation segment mutant, L597V, and two glycine-rich loop mutants, G464V and G469A, also enhance MEK/ERK signaling [2, 13]. In contrast, the BRAF G469E mutation significantly decreases MEK/ERK signaling. Meanwhile, other BRAF mutations, such as R462I, I463S, and G464E, do not increase MEK/ERK signaling [13, 15].
Although several BRAF mutations have been identified, the biological effects of these mutations are not fully understood [2, 13, 16, 17]. In a previous study using comprehensive genome-wide sequencing, we identified some non-V600E BRAF mutations in specimens from metastatic CRC (mCRC) patients with anti-EGFR antibody resistance [18]. Among the newly identified BRAF mutations, Q524L and L525R in the activation segment and L525R lead to enhanced kinase activity and increased ERK phosphorylation, but Q524L demonstrates similar activity as wild-type (WT) BRAF in transiently expressed cells [18]. We hypothesized that L525R may contribute to primary resistance to cetuximab, consistent with the lack of response to anti-EGFR antibody treatment.
Several preclinical and clinical studies have demonstrated that MAPK pathway inhibitors, such as inhibitors of BRAF and MEK, exhibit antitumor activity in cancers with BRAF mutations [1921]. However, the use of these inhibitors as monotherapy frequently results in the development of resistance. Combination therapy using both BRAF and MEK inhibitors can improve overall survival for patients with BRAF-mutant melanoma [2224]. Moreover, accumulating evidence suggests that the combination of an MEK inhibitor, such as selumetinib, and a phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) inhibitor can be effective in many cancer therapies [25]. In the current study, we functionally analyzed the BRAF L525R mutation in a stable cell line. We also evaluated the PI3K/mechanistic target of rapamycin (mTOR) inhibitor for inhibiting the proliferation of selumetinib-resistant BRAF L525R cells. We hypothesize that the PI3K/AKT signaling pathway underlies the drug resistance observed in these cells and propose the use of combination therapies.

Materials and methods

Chemicals and antibodies

Cetuximab (ERBITUX®) was purchased from Merck Serono Co., Inc. (Darmstadt, Germany) and dabrafenib was purchased from Adoq Bioscience (Irvine, CA, USA). Selumetinib and BEZ235 were purchased from Selleck Chemicals (Houston, TX, USA). Anti-EGFR (D38B1) XP, anti-phospho-EGFR (Tyr1068), anti-p42/44, anti-Pp42/44 (T202/Y204), anti-AKT (pan), anti-pAKT (S437), anti-Flag, and anti-GAPDH antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA).

Cell culture and transfection

Human embryonic kidney 293 (HEK293) cells expressing WT EGFR [18] were cultured at 37 °C under 5% CO2 in Dulbecco’s Modified Eagle’s Medium supplemented with 10% fetal bovine serum. Flag-tagged BRAF V600E and BRAF L525R constructs were generated by performing site-directed mutagenesis of flag-tagged WT clones using the Prime STAR Mutagenesis Basal Kit (Takara Bio Inc., Shiga. Japan) by following the manufacturer’s instructions. Stable cell clones co-transfected with WT EGFR and BRAF were obtained by transfection of cells using FuGENE HD (Promega Corporation, Madison, WI, USA) followed by puromycin selection. All clones were confirmed by sequencing. The selumetinib-resistant L525R (L525R-R) cells were established by growing the L525R clone in the presence of 10 µM selumetinib for 2 months. The surviving cells at that point were considered selumetinib-resistant and maintained in the presence of 20 µM of selumetinib.

Cell viability

Cells were seeded in 96-well culture plates at approximately 1 × 104 cells/well and treated with cetuximab (0.05–50 µg/mL), dabrafenib (0.01–10 µM), selumetinib (0.01–10 µM), or BEZ235 (0.01–1 µM) for 72 h. Cell viability was assessed using Cell Counting Kit-8 (CCK8; Dojindo Laboratories, Kumamoto, Japan). The optical density of the cell culture medium in each well was measured at 450 nm using a microplate reader (Molecular Devices, San Jose, CA, USA). Three independent experiments were performed in triplicate for each of the drug concentrations.

Western blotting

Cells were lysed in a buffer containing 50 mM Tris (pH 7.5), 150 mM NaCl, 1% NP-40, 0.25% sodium deoxycholate, 5 mM EDTA (pH 8.0), 50 mM NaF, 1 mM Na3VO4, and a commercial protease inhibitor cocktail (P8340) (Sigma-Aldrich Co. LLC, St. Louis, MO, USA). Protein concentration of the cell lysates was determined using a bicinchoninic acid (BCA) protein assay (Thermo Fisher Scientific, Waltham, MA, USA). Whole cell lysates were separated on a 4–20% gradient sodium dodecyl sulfate–polyacrylamide gel (FUJIFILM Wako Pure Chemicals, Osaka, Japan) and transferred onto polyvinylidene fluoride membranes (Bio-Rad Laboratories, Hercules, CA, USA). The membranes were incubated with primary antibodies for EGFR (1:1000 dilution, Cell Signaling), EGFR (Tyr 1068) (1:1000 dilution, Cell Signaling), p42/44 (1:1000 dilution, Cell Signaling), Pp42/44 (1:1000 dilution, Cell Signaling), AKT (1:1000 dilution, Cell Signaling), pAKT (S437) (1:1000 dilution, Cell Signaling), Flag (1:1000 dilution, Cell Signaling), and GAPDH (1:1000 dilution, Cell Signaling). The membranes were incubated with horseradish peroxidase-labeled secondary antibodies (1:10,000 dilution, Cell Signaling). The signals were visualized using electrochemiluminescence detection reagents. Images of the protein bands were acquired using an ImageQuant LAS 4000 mini Biomolecular Imager and Image Quant TL software (GE Healthcare, Chicago, IL, USA).

Statistical analysis

Data are presented as the mean ± SD of at least three independent experiments. Differences between two groups were analyzed using a paired two-tailed Student’s t-test. Results with P < 0.05 were considered statistically significant.

Results

Cell line characteristics

To characterize the biological properties of the BRAF L525R mutant, we established a culture model system. Two independent cell clones overexpressing BRAF L525R cDNA were isolated, confirmed by sequencing, and selected for individual screening. All experiments were performed using both cell clones identified, but only one set of results are shown since the reactivity of both the clones to the drugs was comparable. We first determined the growth inhibitory effect of cetuximab against the L525R-expressing cells. The cells were treated with cetuximab for 72 h at concentrations ranging from 0.05 to 50 µg/mL and cell viability was measured. Cetuximab reduced cell growth of WT cells in a dose-dependent manner but did not affect the growth of BRAF L525R or BRAF V600E cells (Fig. 1a and Fig. S1(a), P < 0.01). To identify the effect of L525R, basal and phosphorylated ERK protein level was evaluated by western blot analysis. ERK phosphorylation was enhanced in BRAF L525R cells to the equivalent level as in BRAF V600E cells (Fig. 1b and Fig. S1(b)), suggesting that L525R consistently activated endogenous ERK similar to V600E. Then, we evaluated the effect of the L525R on the EGFR signaling pathway. EGFR phosphorylation was observed after stimulation with 10 ng/mL exogenous EGF in Vector control, WT, V600E and L525R cells, and cetuximab treatment inhibited the EGF-induced EGFR phosphorylation. However, ERK phosphorylation caused by L525R and V600E was not inhibited by cetuximab (Fig. 1c).

Selumetinib effectively inhibited cell proliferation and ERK phosphorylation in BRAF L525R cells

Next, we evaluated the effect of BRAF and MEK inhibitors on L525R cells. First, we determined the proliferation of BRAF L525R and BRAF V600E cells after treatment with dabrafenib, a specific inhibitor of BRAF V600E [24]. Dabrafenib effectively inhibited cell proliferation of BRAF V600E cells in a dose-dependent manner; however, no significant difference was observed in BRAF L525R cells (Fig. 2a and Fig. S2(a), P < 0.01). We also evaluated proliferation of BRAF L525R and BRAF V600E cells after treatment with selumetinib, which is a highly specific inhibitor of MEK [25]. Selumetinib effectively inhibited cell proliferation of BRAF L525R cells in a dose-dependent manner, but no significant difference was observed in BRAF V600E cells as well as WT and vector control cells (Fig. 2b and Fig. S2(b), P < 0.01).

L525R decreased AKT phosphorylation

To determine whether dabrafenib and selumetinib could prevent ERK phosphorylation, expression levels of phosphorylated ERK were determined by western blot analysis. Dabrafenib treatment exerted a minor inhibitory effect on ERK phosphorylation, even when BRAF L525R cells were treated with 10 µM dabrafenib; however, it inhibited ERK phosphorylation in BRAF V600E cells in a time-dependent and dose-dependent manner (Fig. 3a, b). The inhibition was the most significant after a 30-min treatment (P < 0.01). In comparison, treatment of BRAF L525R cells with selumetinib remarkably inhibited ERK phosphorylation in a dose-dependent manner, and to a much greater extent than observed in BRAF V600E cells (Fig. 3a, c,  P< 0.01). These results suggest that L525R cells are more sensitive to selumetinib than V600E cells, though both BRAF mutants equally activate ERK. Further, to characterize L525R, the phosphorylation status of another EGFR downstream target AKT was evaluated. Western blot analysis showed that basal levels of AKT phosphorylation were substantially reduced in BRAF L525R (Fig. 3d). Furthermore, whether selumetinib treatment upregulated AKT phosphorylation was analyzed. The level of AKT phosphorylation in BRAF V600E cells under selumetinib treatment was the same as that in vector control and WT cells (Fig. S3). However, the level of AKT phosphorylation in BRAF L525R cells remained low.

AKT phosphorylation correlated with selumetinib resistance

To confirm the association between selumetinib resistance and AKT phosphorylation, selumetinib-resistant L525R-R cells were selected by culturing BRAF L525R cells in presence of sub-lethal concentrations of selumetinib. After 2 months of selumetinib selection, two independent selumetinib-resistant clones were isolated. Results from one of the two clones are shown as the reactivity of both the clones to the drugs was comparable. The L525R-R cells were characterized with respect to cell growth and the AKT signaling pathway. Selumetinib (10 µM) strongly inhibited the proliferation of BRAF L525R cells but no such effect was observed in L525R-R cells (Fig. 4a). The basal level of ERK phosphorylation was diminished in L525R-R cells compared to that in WT cells and BRAF L525 cells, whereas AKT phosphorylation was restored to levels comparable with WT cells (Fig. 4b). The expression and phosphorylation of EGFR were also upregulated in BRAF L525R-R cells compared with those in BRAF L525R cells (Fig. S4).

Selumetinib resistance is impaired by AKT/mTOR inhibition

To explore whether a combination of selumetinib and the dual AKT/mTOR inhibitor BEZ235 [26, 27] could achieve greater growth inhibition, we evaluated cell growth and ERK phosphorylation in L525R-R cells. First, we determined the sensitivity of L525R-R cells to BEZ235. We observed that treatment with BEZ235 alone inhibited cell proliferation in a dose-dependent manner in vector control, WT, the parental BRAF L525R, and BRAF L525R-R cells (Fig. 5a). The growth inhibition in L525R-R cells was achieved at a lower concentration of BEZ235 compared with that in vector control, WT, and the parental BRAF L525R cells. Moreover, cell viability was significantly lower for L525R-R cells than for BRAF L525R cells at each concentration (P < 0.01) (Fig. 5a and Fig. S5(a)). This indicated that L525R-R cells were more sensitive to BEZ235 compared to BRAF L525R cells. To confirm this observation, ERK and AKT signals were evaluated by western blot analysis. BEZ235 reduced AKT phosphorylation corresponding with cell growth inhibition (Fig. 5b).
The effect of combining selumetinib with BEZ235 was also evaluated using an in vitro resistant model. The combination of selumetinib and BEZ235 improved the growth inhibition, but the improving outcomes were only additive (Fig. S5(b)).

Discussion

BRAF mutations are found in a wide range of cancers. Most BRAF mutations are activating mutations that are likely involved in oncogenesis and may therefore be attractive targets for molecular therapy. In the current study, we analyzed the function of the BRAF L525R mutation, which was not registered in the Catalog of Somatic Mutations in Cancer (COSMIC). This mutation was identified during our previous clinical observation study, a biomarker research for anti-EGFR monoclonal antibodies in a comprehensive cancer genomics study of patients with CRC (BREAC) [18]. A cell line harboring the BRAF L525R mutation had not been established before; therefore, we established a model system using recombinant EGFR-expressing HEK293 cells to analyze the in vitro response to BRAF and MEK inhibitors through the ERK and AKT signaling pathways. We demonstrated that the L525R mutation resulted in a significant increase in endogenous ERK phosphorylation, which was similar to that of the V600E mutation and constitutively stimulated the MEK/ERK signaling pathway in the absence of extracellular stimuli. We believe that this culture model system will prove useful for analyzing drug responses related to the MEK/ERK signaling pathway, even though the model did not fully represent the pathophysiological function of BRAF mutants.
Selumetinib is a potent and highly selective MEK inhibitor [2628]. Selumetinib inhibited cell proliferation and ERK phosphorylation in cells harboring BRAF L525R mutation, while those with the BRAF V600E mutation were only partially inhibited, even though ERK phosphorylation was impaired. We also observed the inhibition of basal levels of AKT phosphorylation in BRAF L525R cells. Furthermore, AKT phosphorylation level in BRAF V600E cells was the same as that in vector control and WT cells, whereas the level in BRAF L525R cells remained low. Therefore, the ERK signaling pathway may be required instead of the AKT signaling pathway for the proliferation of these cells. Both ERK and AKT signaling pathways are involved in numerous biological processes. The ERK signaling pathway negatively regulates the PI3K/AKT signaling pathway in response to growth factor stimulation [29, 30]. The level of AKT phosphorylation is one of the causes of the difference in sensitivity of BRAF L525R and BRAF V600E cells to selumetinib; but, the underlying molecular mechanism remains unknown. Previous studies using melanoma cell lines reported that lower levels of phosphorylated AKT were associated with enhanced sensitivity to selumetinib [31]. In addition, the accumulating evidence suggests that compensatory activation of the PI3K/Akt signaling pathway contributes to acquired resistance to MEK inhibitors [3234].
In BRAF L525R-R cells, the level of MEK/ERK phosphorylation decreased, whereas that of AKT phosphorylation was restored. Further, the inhibition of PI3K/mTOR with BEZ235 could overcome the acquired resistance of BRAF L525R-R cells to selumetinib. However, the expression of Flag-tagged BRAF L525R decreased in BRAF L525R cells during the acquisition of resistance to selumetinib. During prolonged selumetinib exposure, clonal selection owing to epigenetic dysregulation or genetic alteration may lead to dysregulation of BRAF L525R. The transcriptional upregulation and activation of receptor tyrosine kinases (RTK), including EGFR, as well as the activation of EGFR-mediated AKT signaling pathway via epigenetic deregulation results in the development of resistance to MEK and BRAF inhibitors [3537]. These previous findings are consistent with our observations of EGFR and AKT activation in L525R-R cells.
Though the precise molecular mechanisms underlying the suppression of AKT phosphorylation observed in BRAF L525R cells were not elucidated, BRAF L525R and L525R-R cells can reversibly switch between ERK and AKT signaling pathways (Fig. 6). It seems to be a strategy to maintain the homeostasis of the balance of oncogenic signals. Interestingly, this balancing was not observed in the cells expressing BRAF V600E, whose enzymatic activity was as high as BRAF L525R. The compensatory loops between ERK and AKT signaling pathways have been described in cancer therapies [38]. Further investigations on whether individual BRAF kinase mutants differently affect MEK/ERK and AKT signaling pathways are warranted. This is also clinically important to distinguish these characters for selecting the most appropriate molecular targeted drugs for patients with BRAF mutation-harboring tumors. It would be helpful to routinely analyze the activity of BRAF and downstream signaling pathways to evaluate the vulnerability of tumor cells.
BRAF is believed to be a critical cancer driver gene. The discovery of specific activating mutations in genes controlling regulatory pathways has advanced the development of mutation-specific drugs. Unfortunately, targeted therapies have led to unexpected responses in signaling networks [39]. For instance, recent analysis identified thousands of phosphorylation events orchestrated in response to MAPK blockade in BRAF mutated cells [40] and in mutated EGFR signaling in response to EGFR tyrosine kinase inhibitors [41, 42]. A clinical study demonstrated that a three-drug combination improves survival for patients with BRAF mutated CRC [43]. However, little is currently known regarding gene expression, methylation, and post-translational control of protein signaling in tumor cells of patients. Overall, the consequence of the current proteomic approach described here provides novel insights into potential anticancer therapy.

Acknowledgements

This work was supported by the Adaptable and Seamless Technology Transfer Program (ASTEP) through targeted-driven R&D, Practical Application Development by SME Start-up, Japan Science and Technology Agency (JST), National Cancer Center Research and Development Fund (23-A-2), G&G Science Co., Ltd., and Medical & Biological Laboratories Co., Ltd. We would like to acknowledge our funding agencies and all the authors involved in this study. We also thank Editage (www.​editage.​com) for English language editing.

Declarations

Conflict of interest

The authors declare that there is no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Pessonnaux C, Eychène A (2001) The Raf/MEK/ERK pathway: new concepts of activation. Biol Cell 93:53–62CrossRef Pessonnaux C, Eychène A (2001) The Raf/MEK/ERK pathway: new concepts of activation. Biol Cell 93:53–62CrossRef
2.
Zurück zum Zitat Davies H, Bignell GR, Cox C et al (2002) Mutations of the BRAF gene in human cancer. Nature 417:949–954PubMedCrossRef Davies H, Bignell GR, Cox C et al (2002) Mutations of the BRAF gene in human cancer. Nature 417:949–954PubMedCrossRef
3.
Zurück zum Zitat Long GV, Menzies AM, Nagrial AM et al (2011) Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 29:1239–1246PubMedCrossRef Long GV, Menzies AM, Nagrial AM et al (2011) Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 29:1239–1246PubMedCrossRef
4.
Zurück zum Zitat Puxeddu E, Filetti S (2014) BRAF mutation assessment in papillary thyroid cancer: are we ready to use it in clinical practice? Endocrine 45:341–343PubMedCrossRef Puxeddu E, Filetti S (2014) BRAF mutation assessment in papillary thyroid cancer: are we ready to use it in clinical practice? Endocrine 45:341–343PubMedCrossRef
5.
Zurück zum Zitat Kimura ET, Nikoiforova MN, Zhu Z et al (2003) High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res 63:1454–1457PubMed Kimura ET, Nikoiforova MN, Zhu Z et al (2003) High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res 63:1454–1457PubMed
6.
Zurück zum Zitat Vereczkey I, Serester O, Dobos J et al (2011) Molecular characterization of 103 ovarian serous and mucinous tumors. Pathol Oncol Res 17:551–559PubMedCrossRef Vereczkey I, Serester O, Dobos J et al (2011) Molecular characterization of 103 ovarian serous and mucinous tumors. Pathol Oncol Res 17:551–559PubMedCrossRef
7.
Zurück zum Zitat Grisham RN, Iyer G, Garg K et al (2013) BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer 119:548–554PubMedCrossRef Grisham RN, Iyer G, Garg K et al (2013) BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer 119:548–554PubMedCrossRef
8.
Zurück zum Zitat Souglakos J, Phillips J, Wang R et al (2009) Prognostic and predictive value of common mutations for treatment response and survival in patients with metastatic colorectal cancer. Br J Cancer 101:465–472PubMedPubMedCentralCrossRef Souglakos J, Phillips J, Wang R et al (2009) Prognostic and predictive value of common mutations for treatment response and survival in patients with metastatic colorectal cancer. Br J Cancer 101:465–472PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Clancy C, Burke JP, Kalady MF et al (2013) BRAF mutation is associated with distinct clinicopathological characteristics in colorectal cancer: a systematic review and meta-analysis. Colorectal Dis 15:e711–e718PubMedCrossRef Clancy C, Burke JP, Kalady MF et al (2013) BRAF mutation is associated with distinct clinicopathological characteristics in colorectal cancer: a systematic review and meta-analysis. Colorectal Dis 15:e711–e718PubMedCrossRef
10.
Zurück zum Zitat Yokota T, Ura T, Shibata N et al (2011) BRAF mutation is a powerful prognostic factor in advanced and recurrent colorectal cancer. Br J Cancer 104:856–862PubMedPubMedCentralCrossRef Yokota T, Ura T, Shibata N et al (2011) BRAF mutation is a powerful prognostic factor in advanced and recurrent colorectal cancer. Br J Cancer 104:856–862PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Marchetti A, Felicioni L, Malatesta S et al (2011) Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol 29:3574–3579PubMedCrossRef Marchetti A, Felicioni L, Malatesta S et al (2011) Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol 29:3574–3579PubMedCrossRef
12.
Zurück zum Zitat Forbes SA, Beare D, Boutselakis H et al (2017) COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 45:D777–D783PubMedCrossRef Forbes SA, Beare D, Boutselakis H et al (2017) COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 45:D777–D783PubMedCrossRef
13.
Zurück zum Zitat Dankner M (2018) Targeted therapy for colorectal cancers with non-V600 BRAF mutations: perspectives for precision oncology. JCO Precis Oncol 2:1–12PubMed Dankner M (2018) Targeted therapy for colorectal cancers with non-V600 BRAF mutations: perspectives for precision oncology. JCO Precis Oncol 2:1–12PubMed
14.
15.
Zurück zum Zitat Ikenoue T, Hikiba Y, Kanai F et al (2004) Different effects of point mutations within the B-Raf glycine-rich loop in colorectal tumors on mitogen-activated protein/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase and nuclear factor κB pathway and cellular transformation. Cancer Res 64:3428–3435PubMedCrossRef Ikenoue T, Hikiba Y, Kanai F et al (2004) Different effects of point mutations within the B-Raf glycine-rich loop in colorectal tumors on mitogen-activated protein/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase and nuclear factor κB pathway and cellular transformation. Cancer Res 64:3428–3435PubMedCrossRef
16.
Zurück zum Zitat Osumi H, Shinozaki E, Wakatsuki T et al (2019) Non-V600 BRAF mutations and EGFR signaling pathway in colorectal cancer. Int J Cancer 145:2488–2495PubMedCrossRef Osumi H, Shinozaki E, Wakatsuki T et al (2019) Non-V600 BRAF mutations and EGFR signaling pathway in colorectal cancer. Int J Cancer 145:2488–2495PubMedCrossRef
17.
Zurück zum Zitat Jones JC, Renfro LA, Al-Shamsi HO et al (2017) Non-V600 BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancer. J Clin Oncol 35:2624–2626PubMedPubMedCentralCrossRef Jones JC, Renfro LA, Al-Shamsi HO et al (2017) Non-V600 BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancer. J Clin Oncol 35:2624–2626PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Shinozaki E, Yoshino T, Yamazaki K et al (2017) Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment in patients with pretreated metastatic colorectal cancer: the biomarker research for anti-EGFR monoclonal antibodies by comprehensive cancer genomics (BREAC) study. Br J Cancer 117:1450–1458PubMedPubMedCentralCrossRef Shinozaki E, Yoshino T, Yamazaki K et al (2017) Clinical significance of BRAF non-V600E mutations on the therapeutic effects of anti-EGFR monoclonal antibody treatment in patients with pretreated metastatic colorectal cancer: the biomarker research for anti-EGFR monoclonal antibodies by comprehensive cancer genomics (BREAC) study. Br J Cancer 117:1450–1458PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Spagnolo F, Ghiorzo P, Orgiano L et al (2015) BRAF-mutant melanoma: treatment approaches, resistance mechanisms, and diagnostic strategies. Onco Targets Ther 8:157–168PubMedPubMedCentralCrossRef Spagnolo F, Ghiorzo P, Orgiano L et al (2015) BRAF-mutant melanoma: treatment approaches, resistance mechanisms, and diagnostic strategies. Onco Targets Ther 8:157–168PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Subbiah V, Puzanov I, Blay JY et al (2020) Pan-cancer efficacy of vemurafenib in BRAFV600-mutant non-melanoma cancers. Cancer Discov 10:657–663PubMedPubMedCentralCrossRef Subbiah V, Puzanov I, Blay JY et al (2020) Pan-cancer efficacy of vemurafenib in BRAFV600-mutant non-melanoma cancers. Cancer Discov 10:657–663PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Dummer R, Ascierto PA, Gogas HJ et al (2018) Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicenter, open-label, randomized phase 3 trial. Lancet Oncol 19:603–615PubMedCrossRef Dummer R, Ascierto PA, Gogas HJ et al (2018) Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicenter, open-label, randomized phase 3 trial. Lancet Oncol 19:603–615PubMedCrossRef
23.
Zurück zum Zitat Eroglu Z, Ribas A (2016) Combination therapy with BRAF and MEK inhibitor melanoma: latest evidence and place in therapy. Ther Adv Med Oncol 8:48–56PubMedPubMedCentralCrossRef Eroglu Z, Ribas A (2016) Combination therapy with BRAF and MEK inhibitor melanoma: latest evidence and place in therapy. Ther Adv Med Oncol 8:48–56PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Li Y, Dong Q, Cui Y (2019) Synergistic inhibition of MEK and reciprocal feedback networks for targeted intervention in malignancy. Cancer Biol Med 16:415–434PubMedPubMedCentralCrossRef Li Y, Dong Q, Cui Y (2019) Synergistic inhibition of MEK and reciprocal feedback networks for targeted intervention in malignancy. Cancer Biol Med 16:415–434PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Krishnamurthy A, Dasari A, Noonan AM et al (2018) Phase Ib results of the rational combination of selumetinib and cyclosporin A in advanced solid tumors with an expansion cohort in metastatic colorectal cancer. Cancer Res 78:5398–5407PubMedPubMedCentralCrossRef Krishnamurthy A, Dasari A, Noonan AM et al (2018) Phase Ib results of the rational combination of selumetinib and cyclosporin A in advanced solid tumors with an expansion cohort in metastatic colorectal cancer. Cancer Res 78:5398–5407PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Greystoke A, Steele N, Arkenau HT et al (2017) SELECT-3: A phase I study of selumetinib in combination with platinum-doublet chemotherapy for advanced NSCLC in the first-line setting. Br J Cancer 117:938–946PubMedPubMedCentralCrossRef Greystoke A, Steele N, Arkenau HT et al (2017) SELECT-3: A phase I study of selumetinib in combination with platinum-doublet chemotherapy for advanced NSCLC in the first-line setting. Br J Cancer 117:938–946PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Carvajal RD, Sosman JA, Quevedo JF et al (2014) Effect of selumetinib versus chemotherapy on progression-free survival in uveal melanoma: a randomized clinical trial. JAMA 311:2397–2405PubMedPubMedCentralCrossRef Carvajal RD, Sosman JA, Quevedo JF et al (2014) Effect of selumetinib versus chemotherapy on progression-free survival in uveal melanoma: a randomized clinical trial. JAMA 311:2397–2405PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Hideki H, Yoshiki T, Kei N et al (2008) Down-regulation of the PI3K-kinase/Akt pathway by ERK MAP kinase in growth factor signaling. Genes Cells 13:941–947CrossRef Hideki H, Yoshiki T, Kei N et al (2008) Down-regulation of the PI3K-kinase/Akt pathway by ERK MAP kinase in growth factor signaling. Genes Cells 13:941–947CrossRef
30.
Zurück zum Zitat Jeremiah NW, Leonard SJ, Scot RK (2011) ERK and Akt signaling pathways function through parallel mechanisms to promote mTORC1 signaling. Am J Physiol 300:C1172–C1180CrossRef Jeremiah NW, Leonard SJ, Scot RK (2011) ERK and Akt signaling pathways function through parallel mechanisms to promote mTORC1 signaling. Am J Physiol 300:C1172–C1180CrossRef
31.
Zurück zum Zitat Sweetlove M, Wrightson E, Kolekarn S et al (2015) Inhibitors of pan-PI3K signaling synergizes with BRAF or MEK inhibitors to prevent BRAF-mutant melanoma cell growth. Front Oncol 5:135PubMedPubMedCentralCrossRef Sweetlove M, Wrightson E, Kolekarn S et al (2015) Inhibitors of pan-PI3K signaling synergizes with BRAF or MEK inhibitors to prevent BRAF-mutant melanoma cell growth. Front Oncol 5:135PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Griffin M, Scotto D, Josephs DH et al (2017) BRAF inhibitors: resistance and the promise of combination treatments for melanoma. Oncotarget 8:78174–78192PubMedPubMedCentralCrossRef Griffin M, Scotto D, Josephs DH et al (2017) BRAF inhibitors: resistance and the promise of combination treatments for melanoma. Oncotarget 8:78174–78192PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Van der Noord VE, McLaughlin RP, Smid M et al (2019) An increased cell cycle gene network determines MEK and Akt inhibitor double resistance in triple-negative breast cancer. Sci Rep 9:13308PubMedPubMedCentralCrossRef Van der Noord VE, McLaughlin RP, Smid M et al (2019) An increased cell cycle gene network determines MEK and Akt inhibitor double resistance in triple-negative breast cancer. Sci Rep 9:13308PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Melotti L, Meco D, Battagtia A et al (2020) Targeting the mitogen-activated protein kinase and protein kinase A pathways overcomes a acquired resistance to Selumetinib in low-grade glioma cells. Oncol Rep 45:752–763PubMed Melotti L, Meco D, Battagtia A et al (2020) Targeting the mitogen-activated protein kinase and protein kinase A pathways overcomes a acquired resistance to Selumetinib in low-grade glioma cells. Oncol Rep 45:752–763PubMed
35.
Zurück zum Zitat Jinhua W, Sharon KH, Diego MM et al (2015) Epigenetic changes of EGFR play an important role in BRAF inhibitor resistant cutaneous melanomas. J Invest Dermatol 135:532–541CrossRef Jinhua W, Sharon KH, Diego MM et al (2015) Epigenetic changes of EGFR play an important role in BRAF inhibitor resistant cutaneous melanomas. J Invest Dermatol 135:532–541CrossRef
36.
Zurück zum Zitat Mehwish K, Mohammad FS (2019) Epigenetic mechanisms of escape from BRAF oncogene dependency. Cancers 11:1480CrossRef Mehwish K, Mohammad FS (2019) Epigenetic mechanisms of escape from BRAF oncogene dependency. Cancers 11:1480CrossRef
37.
Zurück zum Zitat Vikrant R, Veena P, Swati A (2021) Molecular pathways and epigenetic factors regulating chemoresistance in cancer. Chemonicles Oncol 4:1010 Vikrant R, Veena P, Swati A (2021) Molecular pathways and epigenetic factors regulating chemoresistance in cancer. Chemonicles Oncol 4:1010
39.
Zurück zum Zitat Parker R, Bligh RC, Molloy MP (2014) Phosphoproteomics of MAPK inhibition in BRAF-mutated cells and a role for the lethal synergism of dual BRAF and CK2 inhibition. Mol Cancer Ther 13:1894–1906PubMedCrossRef Parker R, Bligh RC, Molloy MP (2014) Phosphoproteomics of MAPK inhibition in BRAF-mutated cells and a role for the lethal synergism of dual BRAF and CK2 inhibition. Mol Cancer Ther 13:1894–1906PubMedCrossRef
40.
Zurück zum Zitat Zhang X, Maity T, Kashyap MK et al (2017) Quantitative tyrosine phosphoproteomics of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor-treated lung adenocarcinoma cells reveals potential novel biomarkers of therapeutic response. Mol Cell Proteomics 16:891–910PubMedPubMedCentralCrossRef Zhang X, Maity T, Kashyap MK et al (2017) Quantitative tyrosine phosphoproteomics of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor-treated lung adenocarcinoma cells reveals potential novel biomarkers of therapeutic response. Mol Cell Proteomics 16:891–910PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Awasthi S, Maity T, Oyler BL et al (2018) Dataset describing the development, optimization, and application of SRM/MRM-based targeted proteomics strategy for quantification of potential biomarkers of EGFR TKI sensitivity. Data Brief 19:424–436PubMedPubMedCentralCrossRef Awasthi S, Maity T, Oyler BL et al (2018) Dataset describing the development, optimization, and application of SRM/MRM-based targeted proteomics strategy for quantification of potential biomarkers of EGFR TKI sensitivity. Data Brief 19:424–436PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Kopetz S, Grothey A, Yaeger R et al (2019) Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Eng J Med 381:1632–1643CrossRef Kopetz S, Grothey A, Yaeger R et al (2019) Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N Eng J Med 381:1632–1643CrossRef
43.
Zurück zum Zitat Chen L, Liu S, Tao Y (2020) Regulating tumor suppressor genes: post-translational modifications. Signal Trans Target Ther 5:90CrossRef Chen L, Liu S, Tao Y (2020) Regulating tumor suppressor genes: post-translational modifications. Signal Trans Target Ther 5:90CrossRef
Metadaten
Titel
Regulation of MEK inhibitor selumetinib sensitivity by AKT phosphorylation in the novel BRAF L525R mutant
verfasst von
Chikako Nakai
Sachiyo Mimaki
Koutatsu Matsushima
Eiji Shinozaki
Kentaro Yamazaki
Kei Muro
Kensei Yamaguchi
Tomohiro Nishina
Satoshi Yuki
Kohei Shitara
Hideaki Bando
Yutaka Suzuki
Kiwamu Akagi
Shogo Nomura
Satoshi Fujii
Masaya Sugiyama
Nao Nishida
Masashi Mizokami
Yasuhiro Koh
Takuya Koshizaka
Hideki Okada
Yukiko Abe
Atsushi Ohtsu
Takayuki Yoshino
Katsuya Tsuchihara
Publikationsdatum
01.03.2023
Verlag
Springer Nature Singapore
Erschienen in
International Journal of Clinical Oncology / Ausgabe 5/2023
Print ISSN: 1341-9625
Elektronische ISSN: 1437-7772
DOI
https://doi.org/10.1007/s10147-023-02318-w

Weitere Artikel der Ausgabe 5/2023

International Journal of Clinical Oncology 5/2023 Zur Ausgabe

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.