Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2011 Dec;13(12):1132–1142. doi: 10.1593/neo.111102

Identification of MET and SRC Activation in Melanoma Cell Lines Showing Primary Resistance to PLX40321,2

Elisabetta Vergani *, Viviana Vallacchi *, Simona Frigerio *, Paola Deho *, Piera Mondellini , Paola Perego , Giuliana Cassinelli , Cinzia Lanzi , Maria Adele Testi §, Licia Rivoltini *, Italia Bongarzone , Monica Rodolfo *
PMCID: PMC3257188  PMID: 22241959

Abstract

PLX4032/vemurafenib is a first-in-class small-molecule BRAFV600E inhibitor with clinical activity in patients with BRAF mutant melanoma. Nevertheless, drug resistance develops in treated patients, and strategies to overcome primary and acquired resistance are required. To explore the molecular mechanisms involved in primary resistance to PLX4032, we investigated its effects on cell proliferation and signaling in a panel of 27 genetically characterized patient-derived melanoma cell lines. Cell sensitivity to PLX4032 was dependent on BRAFV600E and independent from other gene alterations that commonly occur in melanoma such as PTEN loss, BRAF, and MITF gene amplification. Two cell lines lacking sensitivity to PLX4032 and harboring a different set of genetic alterations were studied as models of primary resistance. Treatment with the MEK inhibitor UO126 but not with PLX4032 inhibited cell growth and ERK activation. Resistance to PLX4032 was maintained after CRAF down-regulation by siRNA indicating alternative activation of MEK-ERK signaling. Genetic characterization by multiplex ligation-dependent probe amplification and analysis of phosphotyrosine signaling by MALDI-TOF mass spectrometry analysis revealed the activation of MET and SRC signaling, associated with the amplification of MET and of CTNNB1 and CCND1 genes, respectively. The combination of PLX4032 with drugs or siRNA targeting MET was effective in inhibiting cell growth and reducing cell invasion and migration in melanoma cells with MET amplification; similar effects were observed after targeting SRC in the other cell line, indicating a role for MET and SRC signaling in primary resistance to PLX4032. Our results support the development of classification of melanoma in molecular subtypes for more effective therapies.

Introduction

Among the common gene alterations occurring in melanoma pathogenesis, the most frequent is the T1799A transversion in the v-raf murine sarcoma viral oncogene homolog B1 (BRAF) gene that causes a glutamic acid substitution for valine at position 600 in the encoded kinase, which is detectable in approximately 50% of tumor lesions. BRAF is a serine/threonine-specific protein kinase that is activated by RAS G protein, which is activated downstream of growth factor receptors, cytokines, and hormones in the RAS/MEK/extracellular signal-regulated kinase (ERK) signaling cascade. The V600E change activates the RAF kinase function to constitutively activate the mitogen-activated protein kinase (MAPK) pathway through the hyperactivation of ERK, which promotes cell survival, proliferation, invasion, and angiogenesis. BRAF mutation acts as a driver determining a state of “oncogene addiction,” unresponsive to inhibition by MAPK/ERK kinase (MEK)-dependent feedback but displaying increased sensitivity to the direct inhibition of BRAF and MEK [1]. MAPK signaling determines the cascade activation of other pathways that interact at different levels. This network signals also to the phosphoinositide 3-kinase/v-akt murine thymoma viral oncogene homolog (AKT)/mammalian target of rapamycin pathway, which is constitutively activated in melanoma and may offer compensatory routes to promote cell proliferation and survival [2].

In view of the relevance of RAS/BRAF/MAPK-activated signaling in melanoma, several inhibitors have been produced targeting the RAF kinases, some showing selectivity for mutant BRAF, or targeting the downstream kinase MEK. Several of these inhibitors are currently being evaluated in clinical trials [3]. PLX4032 is an azaindole derivative ATP-competitive inhibitor specific for V600E mutant BRAF which displayed promising efficacy in preclinical studies [4–7]. Phase 1 to 2 clinical trials have shown response rates of more than 50% in patients with melanoma carrying the BRAFV600E mutation, a result confirmed in a phase 3 trial reporting improved rates of overall and progression-free survival [8–10]. Despite this encouraging evidence, the clinical results pointed at secondary resistance as a common feature of kinase-targeted drugs and a major issue for investigations. Studies investigating the mechanisms associated to the acquisition of resistance have reported different genetic and epigenetic alterations, which promote ERK activation by MEK-dependent mechanisms bypassing BRAF inhibition, detectable in tumor biopsies from patients who developed resistance to PLX4032 treatment after clinical response. These alterations included de novo somatic mutations in MEK1, neuroblastoma RAS viral (v-ras) oncogene homolog (NRAS), or phosphatase and tensin homolog (PTEN) genes, but not in the targeted BRAF gene, as well as hyperactivation of platelet-derived growth factor receptor β, insulin-like growth factor 1 receptor (IGF1R), and MAP3K8 kinases [11–14].

In the current report, we focused on melanoma showing primary resistance that were identified by screening a panel of patient-derived genetically characterized BRAFV600E-mutated melanoma cell lines to identify alterations that are associated with the cellular response to PLX4032. We investigated at the genetic and molecular levels two melanoma cell lines that displayed poor sensitivity to PLX4032 as models of primary resistance. By genetic characterization and by using a phosphoproteomic approach, we identified and validated further targets for pharmacological intervention and examined the effects of the combination of PLX4032 with other kinase inhibitors as an approach to overcome resistance.

Materials and Methods

Cells and Cellular Assays

The short-term melanoma cell lines LM4–LM41 have previously been described [15]; LM42 and LM43 were derived from visceral metastases and were similarly generated and characterized. The cell line LM17R was generated by treating the parental cell line LM17 with PLX4032 (3.2 µM) for 96 hours, allowing the few surviving cells to regrow, and repeating treatment for 11 times. MTT assays were used to evaluate the inhibition of cell growth at 72 hours, adding drugs 24 hours after cell plating. The bioluminescent ToxiLight bioassay kit (Lonza, Valais, Switzerland) was used to measure the release of adenylate kinase (AK) from dying cells. Caspase 3 activation was measured using the Active Caspase 3 Apoptosis Kit (Becton Dickinson, Franklin Lakers, NJ). The analysis of the cell cycle was performed by determining the DNA content distribution after propidium iodide staining using a FACSCalibur and ModFit LT v3.1 software. Silencing of v-raf-1 murine leukemia viral oncogene homolog 1 (CRAF) and met proto-oncogene (MET) was obtained using SMART pool small interfering RNA (siRNA; L-003601 and L-003156; Dharmacon, Lafayette, CO) and Lipofectamine 2000 (Gibco, Grand Island, NY). A scrambled control was used (D-001810-10). Invasion assays were performed as previously described [16] on cells exposed for 24 hours to the inhibitors. Scratch wound assays were set on confluent cell monolayer in six-well plates. The monolayer was scratched using a sterile pipette tip, rinsed to remove detached cells, and treated with inhibitors for 72 hours. Matrix metalloproteinase 2 and 9 (MMP-2/-9) activity was assessed using 10% SDS-PAGE gelatin substrate zymography (Invitrogen, Carlsbad, CA) in serum-free conditioned medium after concentration with Amicon Ultra 10K (Millipore, Billerica, MA). Anti-human β1-integrin antibody (552828; Becton Dickinson) was used with APC-conjugated anti-rat immunoglobulin G ( Jackson ImmunoResearch, Plymouth, PA) and analyzing staining by FACS analysis. Fluorescent in situ hybridization (FISH) analysis was performed using the probe kit D7S522/CEP7 according to the manufacturer's protocol (Abbott Vysis, Abbott Park, IL).

Genetic Analysis

Copy numbers of BRAF, microphthalmia-associated transcription factor (MITF), MET, cyclin D1 (CCND1), and β-catenin (CTNNB1) genes in melanoma samples were determined by quantitative real-time polymerase chain reaction (PCR) analysis using TaqMan Copy Number Assays from Applied Biosystems (Branchburg, NJ). In particular, the copy number of BRAF gene was evaluated by targeting intron 13 (Hs04958893_cn) and intron 16 (Hs05004157_cn), whereas a single assay was used for MITF (Hs02258756_cn), MET (Hs00305306_cn), CCND1 (Hs01425024_cn), and CTNNB1 (Hs02393264_cn). TaqMan copy number reference assay RNase P was used as endogenous reference gene. DNA isolated from blood samples of healthy donors was used as control. PCRs were performed in quadruplicate and run on the ABI Prism 7900HT machine. Results were analyzed using the Copy Caller software version 1.1 and copy numbers 4 or higher were considered gene amplifications. The methylation status of the PTEN promoter was determined after bisulfite conversion using the EZ DNA Methylation-Gold Kit (Zymo Research, Irvine, CA) by performing PCR analysis using previously reported primers and protocols with minor modifications [17]. Multiplex ligation-dependent probe amplification (MLPA) SALSA kits P005, P006, and P007 were used to profile changes in chromosomal regions as detailed by the manufacturer (MRC-Holland, Amsterdam, the Netherlands). Results were analyzed by Coffalyser v 9.4 software by normalizing to three samples of normal DNA. The resulting values were categorized as homozygous loss (≤0.3), loss of heterozygosity (≤0.6), gain (≥1.3), and amplification (≥2).

Materials

The following antibodies were used: anti-pERK1/2 (M8159), anti-ERK (M5670), and anti-vinculin (V9131) from Sigma (St. Louis, MO); anti-AKT (610861) from Becton Dickinson; anti-pAKT (4051), anti-pSRC (2105), anti-pMET (3077), anti-phosphorylated signal transducer and activator of transcription 3 (STAT3; 9131), anti-pPaxillin (2541), and anti-pp130CAS (4011S) from Cell Signaling Technology (Danvers, MA); anti-Src (05-184), anti-p70 S6 kinase (p70S6K; 05-781), anti-pp70 S6 kinase (04-392), and anti-Src homology 2 domain-containing transforming protein (SHC; 06-203) from Upstate Biotechnology (Lake Placid, NY); anti-CCND1 (M7155) from Dako (Glostrup, Denmark); anti-MET (sc-10), anti-STAT3 (sc-483), anti-CRAF (sc-133), anti-phosphorylated focal adhesion kinase (pFAK; sc-101679), anti-FAK (sc-932), anti-pSHC (sc-18074-R), and anti-actin (sc-166) from Santa Cruz Biotechnology (Santa Cruz, CA); anti-paxillin (P13520) from Transduction Laboratories (Lexington, KY); anti-p130CAS (ab33539) from Abcam (Cambridge, UK); anti-breast cancer resistance protein (BCRP; MON9041) and anti-multidrug resistance protein 4 (MRP4; MON9069) from Monosan (Valter Occhiena, Torino, Italy); anti-KIT (v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog; 566) from MBL (Woburn, MA); and peroxidase-conjugated secondary antibodies anti-mouse immunoglobulin (Becton Dickinson) and anti-rabbit immunoglobulin G (Amersham, Piscataway, NJ) were used. For anti-phosphorylated tyrosine (pTyr) immunoprecipitation and MALDI-TOF mass spectrometry analysis, samples were processed as previously described [18,19]. Only proteins identified in at least three separate experiments were considered.

PLX4032 was obtained by agreement with Plexxikon, Inc (Berkeley, CA). SU11274/Sugen, UO126, PHA-665752 (Sigma), BMS-354825/Dasatinib (LC Laboratories, Woburn, MA), JNJ-38877605, SGX-523 (Selleck, Houston, TX), and E804/Indirubin (Calbiochem, Gibbstown, NJ) were purchased. After dose-response tests, the drugs were used at the concentrations indicated.

Data Analysis

Fitted lines were generated using the four-parameter nonlinear regression with a sigmoidal dose response (variable slope), and the IC50 values (growth-adjusted inhibitory concentration of 50%) for inhibition of cell growth at 72 hours of PLX4032 treatment were calculated using Prism v 5.0 software. Student's t test and one-way analysis of variance (ANOVA) followed by the Bonferroni correction were used to evaluate statistical significance. Drug interaction was evaluated as described elsewhere [20] with interaction index values greater than 1 indicating synergism. The reported data are representative of three independent experiments.

Results

PLX4032 Growth Inhibitory Effects in BRAFV600E-Mutated Melanoma Cells Are Not Associated with Other Common Melanoma Gene Alterations Including PTEN Loss

The growth inhibitory effect of PLX4032 was tested in a panel of 27 genetically characterized melanoma cell lines, including 20 lines that were heterozygous for the V600E BRAF mutation and 7 lines carrying wild-type BRAF gene. The effect of other genetic alterations, including mutations in CDKN2A, PTEN, and tumor protein p53 (TP53) and amplification of BRAF and MITF, on melanoma cell sensitivity to PLX4032 was considered. We found that PLX4032 inhibition of cell growth was strictly dependent on the presence of BRAFV600E and independent of other gene alterations. In fact, 18 of 20 BRAFV600E-mutated melanoma cell lines were sensitive to the compound, with IC50 values ranging between 0.01 and 1 µM, whereas 2 cell lines displayed a poor sensitivity and showed IC50 values that were approximately 10 µM. The different IC50 values were not associated with the mutational profiles of the cell lines, including the amplification of the BRAF or MITF genes, or to the expression of KIT protein (Table 1).

Table 1.

Genetic Characterization and PLX4032 Sensitivity of the Melanoma Cell Lines.

Melanoma* IC50 PLX4032 (µM) BRAF nBRAF 7q34§ nMITF 3p14§ NRAS PTEN TP53 p16 p14/ARF cKIT
LM4 0.065 V600E 4 2 wt wt/+ Y236H del del +
LM14 0.128 V600E 2 2 wt wt/+ wt del del -
LM15 0.088 V600E 3 4 wt wt/+ wt del del -
LM16 0.023 V600E 4 10 wt wt/+ wt L65P wt/+ L862L/+
LM17 0.238 V600E 3 3 wt P38S/+ S127F A148T/+ wt/+ -
LM17R 4.044 V600E 3 3 wt +
LM20 8.907 V600E 2 4 wt wt/+ Y234C wt/- wt/+ -
LM21 0.027 V600E 3 3 wt P246S/+ S127F del del +
LM25 0.122 V600E 2 6 wt wt/+ E258K R80stop/- P94L/- -
LM26 1.195 V600E 3 3 wt C105fsX112/- R175H del del -
LM27 0.031 V600E 7 4 wt wt/+ wt IVS2-2A>G/- wt/- +
LM28 0.398 V600E 3 3 wt wt/+ wt del del +
LM30 0.031 V600E 2 2 wt wt/+ wt del del -
LM33 0.057 V600E 2 5 wt wt/+ wt del del -
LM34 0.097 V600E 5 3 wt wt/+ wt A148T/- wt/- -
LM36 1.778 V600E 4 2 wt wt/+ wt del del I798I/-
LM38 8.871 V600E 11 3 wt M/- wt del del L862L/-
LM39 0.153 V600E 2 2 wt wt/+ wt del del -
LM41 0.166 V600E 3 3 wt wt/+ wt wt/- wt/+ -
LM42 0.032 V600E 2 3 wt M/- c993+1 G>A del del -
LM43 0.017 V600E 8 4 wt M/- wt del del L862L/-
LM3 >6.400 wt 3 4 Q61R wt/- wt wt/- wt/- +
LM18 >6.400 wt 2 3 Q61R wt/+ wt del del +
LM19 >6.400 wt 3 9 wt wt/+ wt del del +
LM23 >6.400 wt 2 3 wt wt/+ wt del del -
LM24 >6.400 wt 2 3 Q61R wt/+ wt del wt/+ +
LM32 >6.400 wt 8 3 wt wt/- wt del del -
LM35 >6.400 wt 4 6 wt M/- wt del del +

+, - indicates presence or absence of protein expression; del, homozygous deletion; M, gene methylation; wt, wild-type gene.

Polymorphisms are shown in italics. PLX4032-resistant cell lines are shown in bold.

*

Cell lines are numbered according to the list reported in Daniotti et al. [15]. All lines are shown wild-type for PI3KCA exons 9 and 20, GNAQ exon 5, and CDK4 exon 2, except LM17 cells that carry a K22R mutation in CDK4 exon 2.

IC50 values were calculated by modeling results of growth inhibition assays using a nonlinear regression curve fit with a sigmoidal dose-response (variable slope).

Heterozygous mutations; exons 11 and 15 for BRAF and exons 1 and 2 for NRAS gene were sequenced.

§

Gene copy number as evaluated by quantitative PCR as detailed in Materials and Methods.

KIT gene exons 9, 11, 13, 14, 17, and 18 were sequenced. Polymorphism L862L (rs3733542) in exon 18 and the silent mutation I798I in exon 17 were detected. KIT protein expression was evaluated by Western blot.

Melanoma cell lines LM20 and LM38 showed primary resistance to PLX4032 lacked p16 and KIT protein expression but showed different gene alterations because LM20 cells harbored MITF amplification and mutated TP53, whereas LM38 lacked p14/ARF gene and PTEN expression because of gene methylation. PTEN deficiency has been hypothesized to promote melanoma cell proliferation and survival through AKT activation, which may decrease the dependency on ERK signaling. Moreover, PTEN loss has been detected in a melanoma tissue biopsy obtained from a patient relapsing on treatment with PLX4032 [13]. When response of melanoma cell lines to PLX4032 concentrations inhibiting cell growth was examined, we found that the drug produced an accumulation in the G1 phase of cell cycle regardless of PTEN status (Figures 1 and W1). Growth inhibition was associated with apoptotic cell death, as documented by AK release and activation of caspase 3, at higher levels in PTEN-positive samples, indicating a role for PTEN in the induction of cell death in response to PLX4032 (Figure 1, A and B).

Figure 1.

Figure 1

Effects of PLX4032 on BRAFV600E-mutated melanoma cells expressing or lacking PTEN. (A) Inhibition of cell growth (72 hours), AK release (72 hours), and activated caspase 3 (48 hours) after treatment with PLX4032 (3.2 µM). The percentage of growth was calculated as: (OD570 of wells that contained the drug/OD570 of the drug-free wells) x 100. Error bars, SD. (B) Percentages of cells in sub G1, G1, S, and G2/M phases of cell cycle after 24 hours of treatment with PLX4032 (3.2 µM). (C) Modulation of phospho-signaling after 24 hours of PLX4032 treatment (3.2 µM).

Modulation of MAPK and AKT Signaling by PLX4032 Treatment

To define the cellular response that was associated with PLX4032 sensitivity, we examined the effect of treatment on downstream signaling pathways that regulate cell growth and survival. PLX4032 treatment strongly reduced the levels of pERK and pAKT in most drug-sensitive cell lines, independently of PTEN status. In addition, down-regulation of p70S6K, which is activated downstream of the mammalian target of rapamycin signaling, was detectable in most lines, and CCND1 expression was downregulated in all drug-sensitive cell lines, consistently with an accumulation in the G1 phase of the cell cycle. In contrast, pAKT, pERK, pp70S6K, and cyclin D1 levels were not affected by the treatment in the resistant LM20 and LM38 cells, in keeping with the poor antiproliferative and cytotoxic effects (Figure 1C).

A resistant cell line (LM17R) was generated by repeated drug exposure from the cell line LM17, which showed extensive cell death after PLX4032 treatment. LM17R showed reduced sensitivity to the antiproliferative effect of PLX4032, diminished AK release, caspase 3 activation, and G1 block of the cell cycle, as well as unresponsiveness of pERK, pAKT, and CCND1 (Figure 2). Sequence analysis confirmed the presence of the heterozygous V600E BRAF mutation and excluded the presence of secondary mutations in exons 11 and 15 and in RAS gene; in addition, the same number of copies of the BRAF gene as the parental LM17 cells was detected.

Figure 2.

Figure 2

Selection of PLX4032-resistant variant LM17R by long-term exposure of LM17 cells to PLX4032. (A) Growth curves showing decreased sensitivity to PLX4032 in LM17R cells. Calculated IC50 values were 0.2 and 4 µM for LM17 and LM17R, respectively. (B) Enhanced ERK and AKT signaling in the resistant variant LM17R. Expression levels as evaluated by Western blot analysis in parental and resistant cells treated with PLX4032 (3.2 µM for 24 hours). (C) Cell growth, cell death, caspase 3 staining, and cell cycle analysis were tested at 72, 48, and 24 hours of treatment with PLX4032 (3.2 µM). *P < .0001 by Student's t test.

To assess whether the MAPK pathway can be modulated downstream of mutated BRAF in resistant cells, we tested whether MEK inhibition affected pERK levels and cell proliferation. Treatment with the MEK1/2 inhibitor UO126 reduced pERK signal and inhibited proliferation in LM20 and LM38 as well as in LM17R cells compared with that in LM17 (Figure 3, A and B), indicating that these cell lines retained the susceptibility to MEK inhibition.

Figure 3.

Figure 3

Resistance to PLX4032 is independent of MEK and CRAF. (A) The proliferation of melanoma LM20, LM38, LM17R, and LM17 cells was similarly inhibited by 72 hours of treatment with UO126 (IC50 values: 9.7, 4, 6.3, and 16.1 µM, respectively). (B) Levels of pERK are downregulated after 24 hours of incubation with UO126 (25 µM). (C) Western blot analysis of LM38 and LM17R cells that were transfected with CRAF or control siRNA for 24 hours before being treated for 24 hours with PLX4032 (3.2 µM), showing CRAF protein levels downregulated to 14% and to 20%, respectively, as determined by quantification of the signal by Image Quant v5.2 software. (D) LM38 and LM17R cells were treated for 24 hours with CRAF or control siRNA before 72 hours of treatment with PLX4032 (3.2 µM). Absorbance at 570 nm after MTT staining is shown, indicating that CRAF inhibition does not increase sensitivity to PLX4032.

A shift in signaling from BRAF to CRAF after BRAF inhibition has been described in melanoma cells, with CRAF mediating ERK activation [21]. Therefore, we silenced CRAF in LM38 cells using specific siRNA to test whether the sensitivity to PLX4032 increased by reducing CRAF levels. The CRAF siRNA downregulated CRAF protein levels without affecting pERK levels and cell sensitivity to PLX4032. Similar results were obtained also in LM17R cells (Figure 3, C and D).

Molecular Characterization of Melanoma Cell Lines Showing Resistance to PLX4032

To identify new potential markers that are associated with PLX4032 resistance and candidate genes, the MLPA analysis was used to genetically characterize the resistant melanoma cell lines. Several probes showed values indicating gene gain or loss (Figure W2). Amplification of CCND1 at 11q13 and of CTNNB1 at 3p21 was detected in LM20 cells, whereas the LM38 line showed a different pattern of alterations, including MET amplification at 7q31. MET, CCND1, and CTNNB1 gene amplifications in LM38 and in LM20 were confirmed by FISH analysis (Figure 4A and data not shown) and by using quantitative PCR assessing gene copy number (Table W1). MLPA analysis showed no difference in the pattern of alterations between LM17R and LM17, indicating that the acquisition of resistance to PLX4032 was not associated to gain or loss of the tested genes.

Figure 4.

Figure 4

Molecular characteristics of the PLX4032-resistant LM20 and LM38 cell lines. (A) FISH analysis with orange labeled probe targeting MET gene and green control probe in LM38 cells. Four green and more than eight orange signals are shown in cells indicated by the arrows. (B) Detection of pMET, pSRC, and pSTAT3 signals after 24 hours of treatment with PLX4032 (3.2 µM) in LM38 and LM20 cells by Western blot analysis. (C) Identification of some relevant immunoaffinity-purified proteins from LM38 and LM20 cell lines. Top: Coomassie blue staining of anti-pTyr affinity-purified proteins from LM38 cells. Bottom: Silver staining of anti-pTyr affinity-purified proteins from LM20 cells. Protein extracts were incubated with anti-pTyr agarose-conjugated antibody. Bound proteins were washed, eluted, and resolved by 4% to 12% SDS-PAGE. Mw indicates molecular weight markers. Swiss Prot ID indicates accession number.

To further explore the mechanisms of PLX4032 resistance, a proteomic-multiplexed analysis of pTyr signaling and antibody validation was used to screen pTyr proteins that were modulated by treatment in PLX4032-sensitive and -resistant melanoma cells. We observed a high degree of heterogeneity in the pTyr profiles in the different cell lines (Figure W3). To identify the most abundant phosphorylated proteins in LM20 and LM38 cell lines, protein bands from anti-pTyr immunoprecipitates of cell lysates were resolved in SDS-PAGE, excised from preparative silver-stained gel, and processed for MALDI-TOF mass spectrometry analysis. The identified proteins indicated that pTyr-based cell signaling was activated in the v-src sarcoma viral oncogene homolog (SRC)/FAK axis in LM20 cells, whereas it was prevalently activated in the MET axis in LM38 cells (Figure 4C). These data were consistent with MET gene amplification in LM38 cells and CTNNB1 amplification in LM20 cells for the role of SRC activity in regulating CTNNB1 signaling. Immunoblot analysis confirmed the presence of the phosphorylated MET receptor in LM38 cells, whereas the phosphorylated form of STAT3, which is activated downstream of SRC, was detectable in LM20 cells. The MET and STAT3 proteins were present but not phosphorylated in the other cell line. In particular, high levels of non-tyrosine-phosphorylated STAT3 were detected in LM38 cells, and both lines showed high pSRC levels, which were not reduced by PLX4032 treatment (Figure 4B).

To define whether PLX4032 resistance was mediated by the increased expression of ABC transporters, we assessed protein expression of ABCB1/Gp170, ABCC1/MRP1, ABCC2/MRP2, ABCC4/MRP4, and ABCG2/BCRP in the resistant melanoma cell lines. Differential expression was observed for BCRP and MRP4 (Figure W4). However, BCRP overexpression did not result in resistance to PLX4032 as shown by using a mutant BRAF isogenic model system [22]. In addition, topotecan, a well-known MRP4 substrate, displayed a similar effect in LM17 and LM17R cells despite increased MRP4 levels (data not shown). Thus, PLX4032 resistance is not determined by ABC transporters.

MET and SRC as Additional Targets for Combined Treatment with PLX4032

On the basis of the results of molecular profiling, MET and SRC represented new candidate targets expressed at high levels and activated in LM38 and LM20 melanoma cells intrinsically resistant to PLX4032. We thus tested the effect of combining PLX4032 with drugs that inhibited MET and SRC kinases.

The MET inhibitor SU11274 significantly inhibited the proliferation of most of the melanoma cell lines that were examined, including PLX4032-resistant lines, with IC50 values of approximately 10 µM (data not shown). The combined treatment with SU11274 and PLX4032 produced a synergistic interaction when tested in LM38 cells (interaction index = 2.5), and growth inhibition was associated with an accumulation of cells in G1 and AK release in the absence of caspase 3 activation (Figure 5A and not shown). The potentiating effect that was obtained by the concomitant inhibition was evident also when other MET inhibitors were tested (Figure 5B). After the cotreatment with SU11274 and PLX4032, pERK and pAKT were not downregulated; in contrast, we found a strong down-regulation of MET signaling through pFAK and pSHC (Figure 5C).

Figure 5.

Figure 5

Cotreatment with MET inhibitor and PLX4032 inhibits growth, invasion, and migration of LM38-resistant melanoma. (A) Growth inhibition (72 hours), AK release (72 hours), and cell cycle (24 hours) in LM38 cells treated with PLX4032 and/or SU11274. *P < .0001 by one-way ANOVA followed by the Bonferroni correction. ⋆: interaction index = 2.5. (B) The inhibitory effect of PLX4032 combined with JNJ-38877605 (J), PHA-665752 (PHA), and SGX-523 (SGX) on proliferation is shown. ⋆: interaction index = 2.2, 1.22, and 1.33, respectively. P < .0001 by one-way ANOVA followed by the Bonferroni correction. (C) Western blot analysis showing the regulation of downstream MET targets in LM38 cells after 24 hours of treatment with PLX4032 and/or SU11274. (D) The Matrigel cell invasion assay showing the effect of exposure to PLX4032, SU11274 or both in LM38 cells. The percent inhibition of migration at 24 hours with or without HGF compared with that of untreated cells is shown. *P < .0001 compared with treatment with PLX4032 by Student's t test. (E) FACS analysis of β1-integrin expression after 24 hours of exposure to PLX4032 and/or SU11274 in LM38 cells. Mean fluorescence intensity after treatment is indicated. (F) Scratch wound assay showing closure of a scratch wound in cultured LM38 cells under control conditions or in the presence of PLX4032, SU11274, or both for 72 hours. Medium was replaced every day to remove detached dead cells. Magnification, x2.5. - indicates untreated control; P, PLX4032 (3.2 µM); S, SU11274 (10 µM).

Because MET is involved in tumor invasion, we evaluated the effects of the combined treatment on the ability of melanoma cells to invade Matrigel and migrate in vitro. LM38 melanoma cells were highly responsive to the MET ligand hepatocyte growth factor (HGF), as the addiction of HGF determined a significant increase in the number of cells that migrated through the Matrigel layer (not shown), further confirming the role of MET signaling in mediating the invasive capacity in these cells. Indeed, blocking MET signaling by treatment with SU11274 alone or in combination with PLX4032 strongly inhibited Matrigel invasion. Notably, a moderate effect was observed after treatment with PLX4032, indicating that BRAF inhibition, although not affecting cell growth, may alter the invasive activity of melanoma cells, even in the presence of exogenous HGF (Figure 5D). Moreover, LM38 cells produced HGF (data not shown), thus suggesting that an autocrine loop contribute to MET pathway constitutive activation. In addition, the combined drugs downregulated the expression of β1-integrin, the receptor for extracellular matrix laminin that is involved in adhesive and invasive cellular processes (Figure 5E). Scratch wound assays showed that the combination of PLX4032 with SU11274 prevented wound closure, whereas the single drugs impaired wound healing to a limited extent, confirming the effect of the combination on cell migration (Figure 5F).

To confirm that MET inhibition can cooperate with BRAF inhibition siRNA silencing of MET was tested. A synergic effect on cell proliferation was detected (interaction index = 1.36), and down-regulation of MET and SHC signal was shown, whereas pERK and pAKT levels were maintained (Figure 6, A and B).

Figure 6.

Figure 6

MET silencing increases sensitivity to PLX4032 in LM38 PLX4032 resistant cell line. (A) LM38 cells were treated for 96 hours with MET or control siRNA and with PLX4032 (3.2 µM). After MTT staining, the percentage of cell growth was calculated compared with the untreated control. *P < .0001 by one-way ANOVA followed by the Bonferroni correction. (B) Western blot analyses of LM38 cells showing modulation of MET signaling after 96 hours of the indicated treatments. MET protein levels were downregulated to 20%, as determined by quantification of the signal by Image Quant v5.2 software.

To assess the functional relevance of the SRC pathway in LM20 cells, the BMS-354825 multikinase inhibitor targeting SRC family kinases was used. When tested in the panel of melanoma cell lines, BMS-354825 displayed a poor inhibitory effect on cell growth, and its antiproliferative effect was not related to the expression of KIT protein, which is one of the kinases targeted by the compound (not shown). BMS-354825 showed a weak inhibitory effect on cell growth in LM20 cells, whereas the combination of BMS-354825 with PLX4032 displayed significant antiproliferative and cytotoxic effects (interaction index = 2.1). Another SRC inhibitor, E804, exerted an additive effect with PLX4032, further corroborating the role of SRC signaling in LM20 cells (Figure 7A). Treatment with BMS-354825 downregulated the levels of phosphorylated SRC protein and of the downstream targets paxillin and p130CAS; in addition, BMS-354825 reduced pFAK levels. In contrast, no effect was detectable on pERK and pAKT levels also with this drug combination, suggesting that it is not a necessary requirement to impair cell proliferation (Figure 7B and data not shown). The combined treatment with PLX4032 and BMS-354825 decreased MMP-2 production by LM20 melanoma cells, which was measured using gelatin-gel zymography (Figure 7C), and reduced the expression of β1-integrin (Figure 7D).

Figure 7.

Figure 7

Cotreatment with SRC inhibitors and PLX4032 inhibits LM20 melanoma cell growth and downregulates MMP-2 and β1-integrin. (A) Growth inhibition, AK release (72 hours), and activated caspase 3 (48 hours) in LM20 cells that were treated with PLX4032 and/or BMS-354825. *P < .0001 by one-way ANOVA followed by the Bonferroni correction. ⋆: interaction index = 2.1. Bottom right, growth inhibitory effect of PLX4032 combined with the SRC inhibitor E804. *P < .0001 compared with single treatments by Student's t test. (B) Western blot analysis showing regulation of downstream SRC targets in LM20 cells after 24 hours of treatment with PLX4032 and/or BMS-354825. (C) Gelatin zymography detecting MMP-2 in supernatants from LM20 cells collected after 24 hours of exposure to PLX4032, BMS-354825, or their combination. MMP-2 band was detectable at 72 kDa. (D) FACS analysis of β1-integrin expression after 24 hours of exposure to PLX4032 and/or BMS-354825 in LM20 cells. Mean fluorescence intensity after treatment is indicated. - indicates untreated control; B, BMS-354825 (100 nM); E, E804; P, PLX4032 (3.2 µM); pos, positive control.

Discussion

It is not yet known how other concurrent genetic alterations in addition to BRAF mutations may affect the clinical efficacy of the BRAF inhibitor PLX4032 in metastatic melanoma and whether a classification level can be defined for the molecular profiles that are associated with primary resistance. Although BRAF, NRAS, and KIT mutations are mutually exclusive, mutated BRAF melanoma may carry common alterations in CDKN2A, PTEN, and TP53 genes, as well as alterations of CDK4, CTNNB1, FGFR2, MITF, ERBB4, MMP, and GRIN2A genes [3,15,23], and other potential driver mutations still poorly characterized [24]. Here, we show that, apart from BRAF mutation, the gene alterations that are common in melanoma, such as PTEN and TP53 mutations, and BRAF and MITF amplification, are not associated with PLX4032 sensitivity in a large panel of genetically characterized short-term melanoma cell lines.

Studies performed on melanoma tissue from few patients relapsing on treatment with PLX4032 have ruled out the occurrence of additional secondary mutations in the BRAF gene and have reported the overgrowth of NRAS mutated [11], PTEN deleted [13], and C121S MEK1 mutated [14] metastases in different individual cases. These results suggest that the mechanisms that mediate acquired resistance rely on different genetic alterations that may include the overgrowth of preexisting genetic variants selected by the treatment as well as de novo mutations.

The in vitro studies on primary resistance to BRAF inhibitors have detected CCND1 gene amplification in cell lines that were resistant to the BRAF inhibitor SB590885 [25]. Other studies have identified different changes in MEK1 and BRAF T529N causing resistance to PLX4720 [26,27]. Melanoma cell lines carrying homozygous BRAFV600E mutation were shown to be more sensitive to PLX4032 than those carrying heterozygous BRAFV600E mutation [28–30]. Although homozygosity is rare, the 7q34 chromosomal region where the BRAF gene is located is frequently amplified in melanoma lesions and especially in BRAFV600E-mutated melanomas [31]. Amplification of the mutated BRAF allele was detected in association with acquired resistance to MEK inhibitors in a melanoma cell line in a previous study [32]. In our panel of melanoma cell lines, BRAF gene amplification was detected in 30% of the cell lines, including the resistant LM38 melanoma model, whereas in the resistant variant LM17R, which was obtained by long-term exposure to PLX4032 in vitro, the BRAF gene was not amplified compared with the parental cell line.

In addition to BRAF gene amplification, LM38 melanoma cells resistant to PLX4032 lacked PTEN. We detected lower levels of cytotoxicity in PTEN-negative melanoma cells after exposure to PLX4032 compared with melanomas with intact PTEN, but a similar block of cell cycle, suggesting a role for PTEN in the cytotoxic effect of PLX4032. This finding is in agreement with studies reporting that PTEN loss contributes to PLX4720 resistance by suppressing BIM-mediated apoptosis [33].

The PLX4032-resistant line LM20 harbored amplified MITF gene. MITF gene amplification was detected in 30% of our BRAFV600E-mutated cell lines. Unexpectedly, however, melanomas with amplified MITF (≥4 copies) showed lower IC50 values than melanomas without MITF amplification when only cell lines carrying two gene copies were considered (0.05 vs 0.4 µM, P = .0013), suggesting that MITF amplification does not contribute to PLX4032 resistance.

Because it has been shown that kinase inhibitors are able to interact with members of the ABC family of transporters and that ABC transporters can mediate resistance to kinase inhibitors [34,35], we tested whether BCRP and MRP4 showing overexpression in resistant cells play a role in PLX4032 resistance. The results of these experiments do not indicate a role for BCRP or MRP4 in resistance to PLX4032.

By expanding the genetic characterization to the analysis of altered chromosomal regions by MLPA, the amplification of MET gene in LM38 cells and of CCND1 and CTNNB1 genes in LM20 cells was detected. This pattern was consistent with the pTyr profiling analysis as detected by MALDI-TOF indicating activated MET and SRC signaling. The amplification of the MET gene has been reported in melanoma [36] along with chromosome 7 polysomy [31]. The amplification of CCND1 was detected in approximately 25% melanoma bearing mutated BRAF [37]. Although CTNNB1 mutations have been reported in melanoma, gene amplification was not formerly shown, although it was detected by MLPA in melanoma lesions [38].

Epigenetic changes providing compensatory signaling to bypass BRAF blockade and activate ERK are associated with acquired resistance to BRAF inhibitors. Several different mechanisms have been described, including the activation of a platelet-derived growth factor receptor β, IGF1R/phosphoinositide 3-kinase and MAP3K8/COT signaling [11–13]. Moreover, increased CRAF protein levels and switching from BRAF to CRAF dependency has been associated with the in vitro acquired resistance to AZ628 BRAF inhibitor [21]. Although our data do not support a role for CRAF in resistance to PLX4032, in the current study, LM17R cells with acquired resistance to PLX4032 showed increased IGFR1 signaling and consistently higher levels of pAKT compared with that of the parental LM17 cell line (data not shown). Up-regulation of IGF1R signaling was reported to occur in two of four melanoma cell variants that were selected in vitro for resistance to the 885 BRAF inhibitor [13], therefore appearing as a rather common mechanism by which melanoma cells compensate BRAF inhibition.

Targeting other signaling molecules in crucial pathways may represent an approach to enhance the clinical impact of treatment with PLX4032. Preclinical studies showed that MEK inhibitors in combination with PLX4720 reduced cell growth and pERK expression [12] and may prevent the emergence of resistant clones [26].

We show that simultaneously targeting multiple pathways may represent a promising option for treating PLX4032-resistant melanomas. Treatment with the MET inhibitor SU11274 inhibited the growth of LM38 cells harboring constitutively activated MET and the combination with PLX4032 increased this effect. The treatment specifically inhibited MET kinase activity and downstream signaling. It is possible that the effects of SU11274 resulted from the inhibition of additional kinases involved in MET-dependent downstream responses or reduced because of off-target effects. SU11274 was reported to reduce proliferation in some melanoma cell lines [39,40] and HGF-induced motility and invasion in cell models of other tumor types. MET inhibition with other drugs or by specific siRNA confirmed the role of MET signaling in LM38 cells resistant to PLX4032. MET overexpression has been shown to contribute to resistance to cytotoxic drugs in ovarian cancer [41]. Although MET gene mutations are very rare [39,40,42], MET gene amplification [36] and autocrine production of HGF [43] occur frequently in melanoma. MET activation has been associated to NRAS mutation in melanoma [44]. In addition, MET signaling is upregulated by MITF [45].

BMS-354825, which is a multikinase inhibitor targeting the SRC family kinases, induced apoptosis in LM20 cells when combined with PLX4032. BMS-354825 was reported to downregulate activated SRC, FAK, and EphA2 in melanoma cells and to inhibit proliferation in some melanoma cell lines [46,47]. However, BMS-354825 alone did not significantly affect the growth of LM20 cells. Likely, STAT3 activation regulated an oncogenic signaling in LM20 cells. Moreover, the combination of PLX4032 with SU11274 or with BMS-354825 reduced the invasive and migratory capacities, consistently with inhibition of MMP-2 activity and the expression of β1-integrin, suggesting that the drug combination may result in an inhibitory effect on melanoma growth and dissemination. These results are consistent with a regulatory role of MAPK signaling on the expression of MMPs [48] and β1-integrin [49]. Furthermore, these data revealed that cell functions other than proliferation and survival are reduced by exposure to PLX4032, suggesting that they are governed by signaling molecules affected by PLX4032 treatment. Because of these effects, we can hypothesize that synergic inhibition of cell proliferation of PLX4032 with MET or SRC inhibitors results from some inhibitory effects on MAPK signaling exerted by PLX4032, which are overridden by compensatory routes exerted by other MEK activators when used as a single treatment.

SRC and MET have been implicated in the development and progression of several types of tumors as a result of the interaction with receptor tyrosine kinases and their downstream effectors leading to proliferation, cell growth, survival, motility, migration, and angiogenesis. In particular, aberrant MET activation, due to overexpression, mutations, or gene amplification, has been associated with poor clinical outcome and drug resistance in lung, hepatic, renal, and colorectal carcinoma [50]. The nonreceptor protein tyrosine kinase SRC acts as a signal transducer from the cell surface receptors by sequential phosphorylation of tyrosine residues on different substrates. SRC is a key molecule in tumor progression providing oncogenic signals for cell survival, epithelial-mesenchymal transition, mitogenesis, invasion, angiogenesis, and metastasis. Aberrant expression and activation of SRC occur in breast, prostate, lung, and colorectal carcinomas, in association with poor clinical outcome, and have stimulated interest in using SRC kinase inhibitors as therapeutic cancer agents, some of which have entered clinical experimentation [51].

Our results highlight the complexity of signaling in melanoma and support the relevance of genetic and proteomic profiling to build rational combination treatments with targeted agents.

Supplementary Material

Supplementary Figures and Tables
neo1312_1132SD1.pdf (716.3KB, pdf)

Acknowledgments

The authors thank Donata Penso, Enrica Favini, Elisabetta Corna, and Andrea Lampis for their contribution to the study. The authors thank Plexxikon Inc. (Berkeley, CA) and Gideon Bollag for providing us PLX4032.

Abbreviations

AK

adenylate kinase

AKT

v-akt murine thymoma viral oncogene homolog

ANOVA

analysis of variance

BCRP

breast cancer resistance protein

BRAF

v-raf murine sarcoma viral oncogene homolog B1

CCND1

cyclin D1

CRAF

v-raf-1 murine leukemia viral oncogene homolog 1

CTNNB1

β-catenin

ERK

extracellular signal-regulated kinase

FAK

focal adhesion kinase

FISH

fluorescent in situ hybridization

HGF

hepatocyte growth factor

IC50

growth-adjusted inhibitory concentration of 50%

IGF1R

insulin-like growth factor 1 receptor

KIT

v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog

MALDI-TOF

matrix-assisted laser desorption/ionization time-of-flight

MAPK

mitogen-activated protein kinase

MEK

MAPK/ERK kinases

MET

met proto-oncogene

MITF

microphthalmia-associated transcription factor

MLPA

multiplex ligation-dependent probe amplification

MMP-2

matrix metalloproteinase 2

MRP4

multidrug resistance protein 4

NRAS

neuroblastoma RAS viral (v-ras) oncogene homolog

p70S6K

70-kDa ribosomal protein S6 kinase

PTEN

phosphatase and tensin homolog

pTyr

phosphorylated tyrosine

SHC

Src homology 2 domain-containing transforming protein

SRC

v-src sarcoma viral oncogene homolog

STAT3

signal transducer and activator of transcription 3

TP53

tumor protein p53

Footnotes

1

This work was supported by EU grant CHEMORES (no. 037665) and by Associazione Italiana Ricerca sul Cancro. E.V. and V.V. were supported by the Italian Health Ministry (RFPS project 2006 no. 4-341458). The authors declare no conflict of interest.

2

This article refers to supplementary materials, which are designated by Table W1 and Figures W1 to W4 and are available online at www.neoplasia.com.

References

  • 1.Dhomen N, Marais R. New insight into BRAF mutations in cancer. Curr Opin Genet Dev. 2007;17:31–39. doi: 10.1016/j.gde.2006.12.005. [DOI] [PubMed] [Google Scholar]
  • 2.Zheng B, Jeong JH, Asara JM, Yuan YY, Granter SR, Chin L, Cantley LC. Oncogenic B-RAF negatively regulates the tumor suppressor LKB1 to promote melanoma cell proliferation. Mol Cell. 2009;33:237–247. doi: 10.1016/j.molcel.2008.12.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Romano E, Schwartz GK, Chapman PB, Wolchock JD, Carvajal RD. Treatment implications of the emerging molecular classification system for melanoma. Lancet Oncol. 2011;12:913–922. doi: 10.1016/S1470-2045(10)70274-6. [DOI] [PubMed] [Google Scholar]
  • 4.Sala E, Mologni L, Truffa S, Gaetano C, Bollag GE, Gambacorti-Passerini C. BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells. Mol Cancer Res. 2008;6:751–759. doi: 10.1158/1541-7786.MCR-07-2001. [DOI] [PubMed] [Google Scholar]
  • 5.Yang H, Higgins B, Kolinsky K, Packman K, Go Z, Iyer R, Kolis S, Zhao S, Lee R, Grippo JF, et al. RG7204 (PLX4032), a selective BRAFV600E inhibitor, displays potent antitumor activity in preclinical melanoma models. Cancer Res. 2010;70:5518–5527. doi: 10.1158/0008-5472.CAN-10-0646. [DOI] [PubMed] [Google Scholar]
  • 6.Joseph EW, Pratilas CA, Poulikakos PI, Tadi M, Wang W, Taylor BS, Halilovic E, Persaud Y, Xing F, Viale A, et al. The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner. Proc Natl Acad Sci USA. 2010;107:14903–14908. doi: 10.1073/pnas.1008990107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lee JT, Li L, Brafford PA, van den Eijnden M, Halloran MB, Sproesser K, Haass NK, Smalley KS, Tsai J, Bollag G, et al. PLX4032, a potent inhibitor of the B-raf V600E oncogene, selectively inhibits V600E-positive melanomas. Pigment Cell Melanoma Res. 2010;23:820–827. doi: 10.1111/j.1755-148X.2010.00763.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363:809–819. doi: 10.1056/NEJMoa1002011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bollag G, Hirth P, Tsai J, Zhang J, Ibrahim PN, Cho H, Spevak W, Chang C, Zhang Y, Habets G. Clinical efficacy of a RAF inhibitor needs broad target blockade in BRAF-mutant melanoma. Nature. 2010;467:596–599. doi: 10.1038/nature09454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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. 2010;468:973–977. doi: 10.1038/nature09626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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. 2010;468:968–972. doi: 10.1038/nature09627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, et al. Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell. 2010;18:683–695. doi: 10.1016/j.ccr.2010.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.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. 2011;29:3085–3096. doi: 10.1200/JCO.2010.33.2312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Daniotti M, Oggionni M, Ranzani T, Vallacchi V, Campi V, Di Stasi D, Torre GD, Perrone F, Luoni C, Suardi S, et al. BRAF alterations are associated with complex mutational profiles in malignant melanoma. Oncogene. 2004;23:5968–5977. doi: 10.1038/sj.onc.1207780. [DOI] [PubMed] [Google Scholar]
  • 16.Cassinelli G, Lanzi C, Petrangolini G, Tortoreto M, Pratesi G, Cuccuru G, Laccabue D, Supino R, Belluco S, Favini E, et al. Inhibition of c-met and prevention of spontaneous metastatic spreading by the 2-indolinone RPI-1. Mol Cancer Ther. 2006;5:2388–2397. doi: 10.1158/1535-7163.MCT-06-0245. [DOI] [PubMed] [Google Scholar]
  • 17.Soria JC, Lee HY, Lee JI, Wang L, Issa JP, Kemp BL, Liu DD, Kurie JM, Mao L, Khuri FR. Lack of PTEN expression in non-small cell lung cancer could be related to promoter methylation. Clin Cancer Res. 2002;8:1178–1184. [PubMed] [Google Scholar]
  • 18.Gorla L, Cantu M, Micciche F, Patelli C, Mondellini P, Pierotti MA, Bongarzone I. RET oncoproteins induce tyrosine phosphorylation changes of proteins involved in RNA metabolism. Cell Signal. 2006;18:2272–2282. doi: 10.1016/j.cellsig.2006.05.016. [DOI] [PubMed] [Google Scholar]
  • 19.Caccia D, Micciche F, Cassinelli G, Mondellini P, Casalini P, Bongarzone I. Dasatinib reduces FAK phosphorylation increasing the effects of RPI-1 inhibition in a RET/PTC1-expressing cell line. Mol Cancer. 2010;9:278. doi: 10.1186/1476-4598-9-278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zanchi C, Zuco V, Lanzi C, Supino R, Zunino F. Modulation of survival signaling pathways and persistence of the genotoxic stress as a basis for the synergistic interaction between the atypical retinoid ST1926 and the epidermal growth factor receptor inhibitor ZD1839. Cancer Res. 2005;65:2364–2372. doi: 10.1158/0008-5472.CAN-04-2495. [DOI] [PubMed] [Google Scholar]
  • 21.Montagut C, Sharma SV, Shioda T, McDermott U, Ulman M, Ulkus LE, Dias-Santagata D, Stubbs H, Lee DY, Singh A, et al. Elevated CRAF as a potential mechanism of acquired resistance to BRAF inhibition in melanoma. Cancer Res. 2008;68:4853–4861. doi: 10.1158/0008-5472.CAN-07-6787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Perego P, De Cesare M, De Isabella P, Carenini N, Beggiolin G, Pezzoni G, Palumbo M, Tartaglia L, Pratesi G, Pisano C, et al. A novel 7-modified camptothecin analog overcomes breast cancer resistance protein-associated resistance in a mitoxantrone-selected colon carcinoma cell line. Cancer Res. 2001;61:6034–6037. [PubMed] [Google Scholar]
  • 23.Wei X, Walia V, Lin JC, Teer JK, Prickett TD, Gartner J, Davis S, Stemke-Hale K, Davies MA, et al. NISC Comparative Sequencing Program, author. Exome sequencing identifies GRIN2A as frequently mutated in melanoma. Nat Genet. 2011;43:442–446. doi: 10.1038/ng.810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Pleasance ED, Cheetham RK, Stephens PJ, McBride DJ, Humphray SJ, Greenman CD, Varela I, Lin ML, Ordonez GR, Bignell GR, et al. A comprehensive catalogue of somatic mutations from a human cancer genome. Nature. 2010;463:191–196. doi: 10.1038/nature08658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Smalley KS, Lioni M, Dalla Palma M, Xiao M, Desai B, Egyhazi S, Hansson J, Wu H, King AJ, Van Belle P, et al. Increased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAFV600E-mutated melanomas. Mol Cancer Ther. 2008;7:2876–2883. doi: 10.1158/1535-7163.MCT-08-0431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Emery CM, Vijayendran KG, Zipser MC, Sawyer AM, Niu L, Kim JJ, Hatton C, Chopra R, Oberholzer PA, Karpova MB, et al. MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci USA. 2009;106:20411–20416. doi: 10.1073/pnas.0905833106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Whittaker S, Kirk R, Hayward R, Zambon A, Viros A, Cantarino N, Affolter A, Nourry A, Niculescu-Duvaz D, Springer C, et al. Gatekeeper mutations mediate resistance to BRAF-targeted therapies. Sci Transl Med. 2010;2:35ra41. doi: 10.1126/scitranslmed.3000758. [DOI] [PubMed] [Google Scholar]
  • 28.Halaban R, Zhang W, Bacchiocchi A, Cheng E, Parisi F, Ariyan S, Krauthammer M, McCusker JP, Kluger Y, Sznol M. PLX4032, a selective BRAF (V600E) kinase inhibitor, activates the ERK pathway and enhances cell migration and proliferation of BRAF melanoma cells. Pigment Cell Melanoma Res. 2010;23:190–200. doi: 10.1111/j.1755-148X.2010.00685.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sondergaard JN, Nazarian R, Wang Q, Guo D, Hsueh T, Mok S, Sazegar H, MacConaill LE, Barretina JG, Kehoe SM, et al. Differential sensitivity of melanoma cell lines with BRAFV600E mutation to the specific raf inhibitor PLX4032. J Transl Med. 2010;8:39. doi: 10.1186/1479-5876-8-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tap WD, Gong KW, Dering J, Tseng Y, Ginther C, Pauletti G, Glaspy JA, Essner R, Bollag G, Hirth P, et al. Pharmacodynamic characterization of the efficacy signals due to selective BRAF inhibition with PLX4032 in malignant melanoma. Neoplasia. 2010;12:637–649. doi: 10.1593/neo.10414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Willmore-Payne C, Holden JA, Hirschowitz S, Layfield LJ. BRAF and c-kit gene copy number in mutation-positive malignant melanoma. Hum Pathol. 2006;37:520–527. doi: 10.1016/j.humpath.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • 32.Corcoran RB, Dias-Santagata D, Bergethon K, Iafrate AJ, Settleman J, Engelman JA. BRAF gene amplification can promote acquired resistance to MEK inhibitors in cancer cells harboring the BRAF V600E mutation. Sci Signal. 2010;3:ra84. doi: 10.1126/scisignal.2001148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Paraiso KH, Xiang Y, Rebecca VW, Abel EV, Chen YA, Munko AC, Wood E, Fedorenko IV, Sondak VK, Anderson AR, et al. PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression. Cancer Res. 2011;71:2750–2760. doi: 10.1158/0008-5472.CAN-10-2954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen YJ, Huang WC, Wei YL, Hsu SC, Yuan P, Lin HY, Wistuba II, Lee JJ, Yen CJ, Su WC, et al. Elevated BCRP/ABCG2 expression confers acquired resistance to gefitinib in wild-type EGFR-expressing cells. PLoS One. 2011;6:e21428. doi: 10.1371/journal.pone.0021428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lemos C, Kathmann I, Giovannetti E, Calhau C, Jansen G, Peters GJ. Impact of cellular folate status and epidermal growth factor receptor expression on BCRP/ABCG2-mediated resistance to gefitinib and erlotinib. Br J Cancer. 2009;100:1120–1127. doi: 10.1038/sj.bjc.6604980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Moore SR, Persons DL, Sosman JA, Bobadilla D, Bedell V, Smith DD, Wolman SR, Tuthill RJ, Moon J, Sondak VK, et al. Detection of copy number alterations in metastatic melanoma by a DNA fluorescence in situ hybridization probe panel and array comparative genomic hybridization: a Southwest Oncology Group study (S9431) Clin Cancer Res. 2008;14:2927–2935. doi: 10.1158/1078-0432.CCR-07-4068. [DOI] [PubMed] [Google Scholar]
  • 37.Utikal J, Udart M, Leiter U, Peter RU, Krahn G. Additional cyclin D(1) gene copies associated with chromosome 11 aberrations in cutaneous malignant melanoma. Int J Oncol. 2005;26:597–605. [PubMed] [Google Scholar]
  • 38.Takata M, Suzuki T, Ansai S, Kimura T, Shirasaki F, Hatta N, Saida T. Genome profiling of melanocytic tumors using multiplex ligationdependent probe amplification (MLPA): its usefulness as an adjunctive diagnostic tool for melanocytic tumors. J Dermatol Sci. 2005;40:51–57. doi: 10.1016/j.jdermsci.2005.06.009. [DOI] [PubMed] [Google Scholar]
  • 39.Puri N, Ahmed S, Janamanchi V, Tretiakova M, Zumba O, Krausz T, Jagadeeswaran R, Salgia R. c-Met is a potentially new therapeutic target for treatment of human melanoma. Clin Cancer Res. 2007;13:2246–2253. doi: 10.1158/1078-0432.CCR-06-0776. [DOI] [PubMed] [Google Scholar]
  • 40.Kenessey I, Keszthelyi M, Kramer Z, Berta J, Adam A, Dobos J, Mildner M, Flachner B, Cseh S, Barna G, et al. Inhibition of c-met with the specific small molecule tyrosine kinase inhibitor SU11274 decreases growth and metastasis formation of experimental human melanoma. Curr Cancer Drug Targets. 2010;10:332–342. doi: 10.2174/156800910791190184. [DOI] [PubMed] [Google Scholar]
  • 41.Tang MK, Zhou HY, Yam JW, Wong AS. c-Met overexpression contributes to the acquired apoptotic resistance of nonadherent ovarian cancer cells through a cross talk mediated by phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1/2. Neoplasia. 2010;12:128–138. doi: 10.1593/neo.91438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Seidl H, Weger W, Wolf P, Kerl H, Schaider H. Lack of oncogenic mutations in the c-met catalytic tyrosine kinase domain in acral lentiginous melanoma. Int J Dermatol. 2008;47:1327–1329. doi: 10.1111/j.1365-4632.2008.03818.x. [DOI] [PubMed] [Google Scholar]
  • 43.Li G, Schaider H, Satyamoorthy K, Hanakawa Y, Hashimoto K, Herlyn M. Downregulation of E-cadherin and desmoglein 1 by autocrine hepatocyte growth factor during melanoma development. Oncogene. 2001;20:8125–8135. doi: 10.1038/sj.onc.1205034. [DOI] [PubMed] [Google Scholar]
  • 44.Chattopadhyay C, Ellerhorst JA, Ekmekcioglu S, Greene VR, Davies MA, Grimm EA. Association of activated c-met with NRAS-mutated human melanomas: a possible avenue for targeting. Int J Cancer. doi: 10.1002/ijc.26487. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Beuret L, Flori E, Denoyelle C, Bille K, Busca R, Picardo M, Bertolotto C, Ballotti R. Up-regulation of MET expression by α-melanocyte-stimulating hormone and MITF allows hepatocyte growth factor to protect melanocytes and melanoma cells from apoptosis. J Biol Chem. 2007;282:14140–14147. doi: 10.1074/jbc.M611563200. [DOI] [PubMed] [Google Scholar]
  • 46.Eustace AJ, Crown J, Clynes M, O'Donovan N. Preclinical evaluation of dasatinib, a potent src kinase inhibitor, in melanoma cell lines. J Transl Med. 2008;6:53. doi: 10.1186/1479-5876-6-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jilaveanu LB, Zito CR, Aziz SA, Chakraborty A, Davies MA, Camp RL, Rimm DL, Dudek A, Sznol M, Kluger HM. In vitro studies of dasatinib, its targets and predictors of sensitivity. Pigment Cell Melanoma Res. 2011;24:386–389. doi: 10.1111/j.1755-148X.2011.00835.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Genersch E, Hayess K, Neuenfeld Y, Haller H. Sustained ERK phosphorylation is necessary but not sufficient for MMP-9 regulation in endothelial cells: involvement of ras-dependent and -independent pathways. J Cell Sci. 2000;23(113 pt):4319–4330. doi: 10.1242/jcs.113.23.4319. [DOI] [PubMed] [Google Scholar]
  • 49.Sumimoto H, Miyagishi M, Miyoshi H, Yamagata S, Shimizu A, Taira K, Kawakami Y. Inhibition of growth and invasive ability of melanoma by inactivation of mutated BRAF with lentivirus-mediated RNA interference. Oncogene. 2004;23:6031–6039. doi: 10.1038/sj.onc.1207812. [DOI] [PubMed] [Google Scholar]
  • 50.Stella GM, Benvenuti S, Comoglio PM. Targeting the MET oncogene in cancer and metastases. Expert Opin Investig Drugs. 2010;19:1381–1394. doi: 10.1517/13543784.2010.522988. [DOI] [PubMed] [Google Scholar]
  • 51.Sen B, Johnson FM. Regulation of SRC family kinases in human cancers. J Signal Transduct. 2011;2011:865819. doi: 10.1155/2011/865819. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Figures and Tables
neo1312_1132SD1.pdf (716.3KB, pdf)

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES