Recovery of phospho-ERK activity allows melanoma cells to escape from BRAF inhibitor therapy

K H T Paraiso, I V Fedorenko, L P Cantini, A C Munko, M Hall, V K Sondak, J L Messina, K T Flaherty, K S M Smalley, K H T Paraiso, I V Fedorenko, L P Cantini, A C Munko, M Hall, V K Sondak, J L Messina, K T Flaherty, K S M Smalley

Abstract

Background: Resistance to BRAF inhibitors is an emerging problem in the melanoma field. Strategies to prevent and overcome resistance are urgently required.

Methods: The dynamics of cell signalling, BrdU incorporation and cell-cycle entry after BRAF inhibition was measured using flow cytometry and western blot. The ability of combined BRAF/MEK inhibition to prevent the emergence of resistance was demonstrated by apoptosis and colony formation assays and in 3D organotypic cell culture.

Results: BRAF inhibition led to a rapid recovery of phospho-ERK (pERK) signalling. Although most of the cells remained growth arrested in the presence of drug, a minor population of cells retained their proliferative potential and escaped from BRAF inhibitor therapy. A function for the rebound pERK signalling in therapy escape was demonstrated by the ability of combined BRAF/MEK inhibition to enhance the levels of apoptosis and abrogate the onset of resistance.

Conclusion: Combined BRAF/MEK inhibition may be one strategy to prevent the emergence of drug resistance in BRAF-V600E-mutated melanomas.

Figures

Figure 1
Figure 1
PLX4720 inhibits the growth of melanoma cells harbouring the BRAF-V600E mutation. (A). Increasing concentrations of PLX4720 reduced the growth of melanoma cell lines harbouring the BRAF-V600E mutation (WM35, 1205Lu and WM164), whereas melanoma cell lines that were BRAF wild type were relatively resistant (WM1346, WM1361A and WM1366). Cells were treated with drug (3 nM–30 μM) for 72 h, and cell numbers were quantified using the MTT assay. Bars show s.e. mean. (B) Low doses of PLX4720 are cytostatic in melanoma cells harbouring the BRAF-V600E mutation. 1205Lu cells were treated were either 0.3 or 3 μM PLX4720 for 24 h before being fixed, stained with propidium iodide and analysed by flow cytometry. (C) PLX4720 inhibits MAPK signalling in BRAF-V600E-mutated melanoma cells. Cells were treated with increasing concentrations of PLX4720 (0.03–30 μM, 1 h); proteins were extracted and probed for expression of phospho-ERK (pERK). Blots were stripped once and reprobed for total-ERK to show even protein loading. (D) PLX4720 induces a concentration-dependent reduction in the phosphorylation of the retinoblastoma protein (phospho-RB), induces the cleavage of PARP, stabilises p27 and suppresses the expression of cyclin D1 in WM164 BRAF-V600E-mutated melanoma cells. Cells were treated with increasing concentrations of PLX4720 (3 nM–30 μM) for 24 h, after which time, protein was extracted and resolved by western blotting (C=vehicle control). Blots were stripped once and probed for actin to show equal protein loading.
Figure 2
Figure 2
PLX4720 induces apoptosis in BRAF-V600E-mutated melanoma cell lines. (A) PLX4720 induces apoptosis in three BRAF-mutated melanoma cell lines. Cultures were treated with increasing concentrations of PLX4720 (0.03–30 μM, 48 h), before staining for FITC-annexin-V and flow cytometry. Data show mean of three experiments. (B) PLX4720 reduces viability and invasion of 1205Lu cells grown as 3D collagen-implanted spheroids. Preformed 1205Lu spheroids were implanted into collagen and overlayed with media. Cells were treated with PLX4720 (0.3–30 μM for 72 h) before being treated with calcein-AM and ethidium bromide. Green, viable cells; red, dead cells. Lack of green staining also indicates a loss of cell viability. Magnification × 10. *P<0.05, Significant difference from control. The colour reproduction of this figure is available on the html full text version of the manuscript.
Figure 3
Figure 3
Melanoma cells escape PLX4720 and become resistant. (A) Representative photomicrograph of WM164 and 1205Lu melanoma cells treated with PLX4720 (3 μM) for either 14 or 28 days. (B) Western blot showing levels of pERK expression in PLX4720 naive (N) and resistant (R) (8 weeks, 3 μM) WM164 and 1205Lu cell lines. Note that the resistant cell lines were maintained continuously in the presence of PLX4720 (3 μM). Total-ERK demonstrates even protein loading. (C) Resistant 1205Lu and WM164 cell lines continue to incorporate BrdU in the continual presence of PLX4720 (3 μM). Panel shows either treatment-naive WM164 and 1205Lu cell lines (control) or resistant (chronically treated with PLX4720 for 8 weeks) treated with either PLX4720 (3 μM) or the MEK inhibitor U0126 (3 and 10 μM). Cells were stained for BrdU (20 μM, 1 h) uptake and the cell viability marker 7-AAD and were analysed by flow cytometry. (D) Representative sequencing trace from Exon 3 of MEK1 of 1205Lu cells chronically treated with PLX4720 for 8 weeks, arrow indicates site of P124L mutation identified previously in Emery et al (2009).
Figure 4
Figure 4
pERK signalling recovers after PLX4720 treatment. (A) Naive WM164 melanoma cells were treated with PLX4720 (3 μM, every 24 h) for increasing periods of time (0–48 h) and probed for pERK and total-ERK (tERK). (B) Recovery of pERK is observed in three naive BRAF-V600E-mutated melanoma cell lines. Cells were treated with PLX4720 for 0, 8, 24, 48 h (3 μM) and analysed as in (A). (C) Most cells remain growth arrested even when pERK recovers. WM793 cells were treated with PLX4720 (3 μM) for 0–72 h. Cells were harvested, fixed and stained with propidium iodide before being analysed by flow cytometry. (D) p27 expression levels remain high even when pERK signalling recovers. WM793 cells were treated with PLX4720 as for (C); protein lysates were probed for expression of pERK, total-ERK (tERK), phospho-RB (p-pRB), total retinoblastoma protein (t-RB) and p27. Equal protein loading was confirmed by stripping the blot once and probing for GAPDH expression. (E) PLX4720 treatment leads to a drop in cell numbers followed by stabilisation of the population. WM793, 1205Lu and WM164 melanoma cells were treated with PLX4720 (3 μM) for 0–120 h. At each time point, the cells were removed from the plate and counted. Data show the mean±s.e.mean of three independent experiments.
Figure 5
Figure 5
The function of rebound pERK signalling in the escape from PLX4720 treatment. (A) U0126 blocks the rebound increase in pERK after PLX4720 treatment. Melanoma cells were either treated with vehicle (0), PLX4720 (3 μM) or PLX4720 + U0126 (both 3 μM) for 48 h, protein was then probed for expression of pERK and tERK. (B) Melanoma cells were treated with increasing concentrations of U0126 for 1 h before being probed for pERK and tERK expression. (C) Cells were treated with increasing concentrations of PLX4720 (30 nM–30 μM) for 24 h in the absence or presence of U0126 (3 μM), after which time, protein was extracted and resolved by western blotting and probed for either cleaved PARP (cl-PARP), phospho-ERK (pERK), cyclin D1 (Cyclin D1), p27 or cleaved caspase-3 (cl-casp-3). Blots were stripped once and probed for actin to show equal protein loading. (D) Combined BRAF and MEK inhibition leads to enhanced apoptosis. WM164 cells were treated with either vehicle, U0126 (3 μM, 3U0), PLX4720 (3PLX, 3 μM) or the two inhibitors in combination for 48 h. Levels of apoptosis were measured by annexin-V staining and flow cytometry. Data show the mean of three experiments. *P<0.05.
Figure 6
Figure 6
Dual BRAF/MEK inhibition prevents escape from PLX4720 therapy. (A) WM164, WM793 and 1205Lu melanoma cells were treated with vehicle (1 week), PLX4720 (3 μM), U0126 (3 μM) or the two inhibitors in combination (both 3 μM) for 4 weeks. After this time, colonies were fixed and stained with crystal violet. Photographs are representative of three independent experiments. Photomicrographs show the detail of one colony each on the WM793 plate ( × 4). (B) Combined PLX4720 and U0126 treatment reduce growth of melanoma cells and enhance cell death in a 3D spheroid model. WM164 spheroids were implanted in collagen and treated with PLX4720 (3 μM), U0126 (3 μM) or the two drugs in combination for 72 h. After this time, plates were washed and cells were stained with a cell viability kit. Red=dead cells, green=live cells. The colour reproduction of this figure is available on the html full text version of the manuscript.

References

    1. Bauer S, Duensing A, Demetri GD, Fletcher JA (2007) KIT oncogenic signaling mechanisms in imatinib-resistant gastrointestinal stromal tumor: PI3-kinase/AKT is a crucial survival pathway. Oncogene 26: 7560–7568
    1. Bhatt KV, Spofford LS, Aram G, McMullen M, Pumiglia K, Aplin AE (2005) Adhesion control of cyclin D1 and p27Kip1 levels is deregulated in melanoma cells through BRAF-MEK-ERK signaling. Oncogene 24: 3459–3471
    1. Cartlidge RA, Thomas GR, Cagnol S, Jong KA, Molton SA, Finch AJ, McMahon M (2008) Oncogenic BRAF(V600E) inhibits BIM expression to promote melanoma cell survival. Pigment Cell Melanoma Res 21: 534–544
    1. Christensen C, Guldberg P (2005) Growth factors rescue cutaneous melanoma cells from apoptosis induced by knockdown of mutated (V 600 E) B-RAF. Oncogene 24: 6292–6302
    1. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA (2002) Mutations of the BRAF gene in human cancer. Nature 417: 949–954
    1. Emery CM, Vijayendran KG, Zipser MC, Sawyer AM, Niu L, Kim JJ, Hatton C, Chopra R, Oberholzer PA, Karpova MB, Macconaill LE, Zhang J, Gray NS, Sellers WR, Dummer R, Garraway LA (2009) MEK1 mutations confer resistance to MEK and B-RAF inhibition. Proc Natl Acad Sci USA 106: 20411–20416
    1. Flaherty KT, Puzanov I, Sosman JA, Kim K, Ribas A, McArthur G, Lee RJ, Grippo JF, Nolop K, Chapman PB (2009) Phase I study of PLX4032: proof of concept for V600E BRAF mutation as a therapeutic target in human cancer. J Clin Oncol 27. Abstract 9000
    1. Gray-Schopfer VC, Karasarides M, Hayward R, Marais R (2007) Tumor necrosis factor-alpha blocks apoptosis in melanoma cells when BRAF signaling is inhibited. Cancer Res 67: 122–129
    1. Haass NK, Sproesser K, Nguyen TK, Contractor R, Medina CA, Nathanson KL, Herlyn M, Smalley KS (2008) The mitogen-activated protein/extracellular signal-regulated kinase kinase inhibitor AZD6244 (ARRY-142886) induces growth arrest in melanoma cells and tumor regression when combined with docetaxel. Clin Cancer Res 14: 230–239
    1. Hingorani SR, Jacobetz MA, Robertson GP, Herlyn M, Tuveson DA (2003) Suppression of BRAF(V599E) in human melanoma abrogates transformation. Cancer Res 63: 5198–5202
    1. Karasarides M, Chiloeches A, Hayward R, Niculescu-Duvaz D, Scanlon I, Friedlos F, Ogilvie L, Hedley D, Martin J, Marshall CJ, Springer CJ, Marais R (2004) B-RAF is a therapeutic target in melanoma. Oncogene 23: 6292–6298
    1. King AJ, Patrick DR, Batorsky RS, Ho ML, Do HT, Zhang SY, Kumar R, Rusnak DW, Takle AK, Wilson DM, Hugger E, Wang L, Karreth F, Lougheed JC, Lee J, Chau D, Stout TJ, May EW, Rominger CM, Schaber MD, Luo L, Lakdawala AS, Adams JL, Contractor RG, Smalley KS, Herlyn M, Morrissey MM, Tuveson DA, Huang PS (2006) Demonstration of a genetic therapeutic index for tumors expressing oncogenic BRAF by the kinase inhibitor SB-590885. Cancer Res 66: 11100–11105
    1. Montagut C, Sharma SV, Shioda T, McDermott U, Ulman M, Ulkus LE, Dias-Santagata D, Stubbs H, Lee DY, Singh A, Drew L, Haber DA, Settleman J (2008) Elevated CRAF as a potential mechanism of acquired resistance to BRAF inhibition in melanoma. Cancer Res 68: 4853–4861
    1. Pratilas CA, Taylor BS, Ye Q, Viale A, Sander C, Solit DB, Rosen N (2009) (V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway. Proc Natl Acad Sci USA 106: 4519–4524
    1. Rudin CM, Hann CL, Laterra J, Yauch RL, Callahan CA, Fu L, Holcomb T, Stinson J, Gould SE, Coleman B, LoRusso PM, Von Hoff DD, de Sauvage FJ, Low JA (2009) Treatment of medulloblastoma with Hedgehog pathway inhibitor GDC-0449. N Engl J Med 361: 1173–1178
    1. Sala E, Mologni L, Truffa S, Gaetano C, Bollag GE, Gambacorti-Passerini C (2008) BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells. Mol Cancer Res 6: 751–759
    1. Sawyers C (2004) Targeted cancer therapy. Nature 432: 294–297
    1. Sharma A, Trivedi NR, Zimmerman MA, Tuveson DA, Smith CD, Robertson GP (2005) Mutant V599EB-Raf regulates growth and vascular development of malignant melanoma tumors. Cancer Res 65: 2412–2421
    1. Smalley KS, Brafford P, Haass NK, Brandner JM, Brown E, Herlyn M (2005) Up-regulated expression of zonula occludens protein-1 in human melanoma associates with N-cadherin and contributes to invasion and adhesion. Am J Pathol 166: 1541–1554
    1. Smalley KS, Contractor R, Haass NK, Kulp AN, Atilla-Gokcumen GE, Williams DS, Bregman H, Flaherty KT, Soengas MS, Meggers E, Herlyn M (2007a) An organometallic protein kinase inhibitor pharmacologically activates p53 and induces apoptosis in human melanoma cells. Cancer Res 67: 209–217
    1. Smalley KS, Contractor R, Haass NK, Lee JT, Nathanson KL, Medina CA, Flaherty KT, Herlyn M (2007b) Ki67 expression levels are a better marker of reduced melanoma growth following MEK inhibitor treatment than phospho-ERK levels. Br J Cancer 96: 445–449
    1. Smalley KS, Flaherty KT (2009) Integrating BRAF/MEK inhibitors into combination therapy for melanoma. Br J Cancer 100: 431–435
    1. Smalley KS, Haass NK, Brafford PA, Lioni M, Flaherty KT, Herlyn M (2006) Multiple signaling pathways must be targeted to overcome drug resistance in cell lines derived from melanoma metastases. Mol Cancer Ther 5: 1136–1144
    1. Smalley KSM (2003) A pivotal role for ERK in the oncogenic behaviour of malignant melanoma? Int J Cancer 104: 527–532
    1. Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, Sproesser K, Li L, Smalley KS, Fong D, Zhu YL, Marimuthu A, Nguyen H, Lam B, Liu J, Cheung I, Rice J, Suzuki Y, Luu C, Settachatgul C, Shellooe R, Cantwell J, Kim SH, Schlessinger J, Zhang KY, West BL, Powell B, Habets G, Zhang C, Ibrahim PN, Hirth P, Artis DR, Herlyn M, Bollag G (2008) Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci USA 105(8): 3041–3046
    1. Wang Y, Van Becelaere K, Jiang P, Przybranowski S, Omer C, Sebolt-Leopold J (2005) A role for K-ras in conferring resistance to the MEK inhibitor, CI-1040. Neoplasia 7: 336–347
    1. Wellbrock C, Karasarides M, Marais R (2004a) The RAF proteins take centre stage. Nat Rev Mol Cell Biol 5: 875–885
    1. Wellbrock C, Ogilvie L, Hedley D, Karasarides M, Martin J, Niculescu-Duvaz D, Springer CJ, Marais R (2004b) V599EB-RAF is an oncogene in melanocytes. Cancer Res 64: 2338–2342
    1. Yauch RL, Dijkgraaf GJ, Alicke B, Januario T, Ahn CP, Holcomb T, Pujara K, Stinson J, Callahan CA, Tang T, Bazan JF, Kan Z, Seshagiri S, Hann CL, Gould SE, Low JA, Rudin CM, de Sauvage FJ (2009) Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 326(5952): 572–574

Source: PubMed

3
Abonner