Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Cancer drug addiction is relayed by an ERK2-dependent phenotype switch

Abstract

Observations from cultured cells1,2,3, animal models4 and patients5,6,7 raise the possibility that the dependency of tumours on the therapeutic drugs to which they have acquired resistance represents a vulnerability with potential applications in cancer treatment. However, for this drug addiction trait to become of clinical interest, we must first define the mechanism that underlies it. We performed an unbiased CRISPR–Cas9 knockout screen on melanoma cells that were both resistant and addicted to inhibition of the serine/threonine-protein kinase BRAF, in order to functionally mine their genome for ‘addiction genes’. Here we describe a signalling pathway comprising ERK2 kinase and JUNB and FRA1 transcription factors, disruption of which allowed addicted tumour cells to survive on treatment discontinuation. This occurred in both cultured cells and mice and was irrespective of the acquired drug resistance mechanism. In melanoma and lung cancer cells, death induced by drug withdrawal was preceded by a specific ERK2-dependent phenotype switch, alongside transcriptional reprogramming reminiscent of the epithelial–mesenchymal transition. In melanoma cells, this reprogramming caused the shutdown of microphthalmia-associated transcription factor (MITF), a lineage survival oncoprotein; restoring this protein reversed phenotype switching and prevented the lethality associated with drug addiction. In patients with melanoma that had progressed during treatment with a BRAF inhibitor, treatment cessation was followed by increased expression of the receptor tyrosine kinase AXL, which is associated with the phenotype switch. Drug discontinuation synergized with the melanoma chemotherapeutic agent dacarbazine by further suppressing MITF and its prosurvival target, B-cell lymphoma 2 (BCL-2), and by inducing DNA damage in cancer cells. Our results uncover a pathway that underpins drug addiction in cancer cells, which may help to guide the use of alternating therapeutic strategies for enhanced clinical responses in drug-resistant cancers.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Genome-wide CRISPR–Cas9 knockout screen to break cancer drug addiction identifies several signalling pathway components.
Figure 2: Conserved drug addiction pathway in multiple cell lines with distinct therapy resistance mechanisms.
Figure 3: ERK2 and JUNB act in an MITF-dependent genetic phenotype switch that controls drug addiction.
Figure 4: Conservation of drug addiction mechanism in human lung cancer cells, and drug withdrawal synergizing with dacarbazine in tumour cell elimination.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Suda, K. et al. Conversion from the “oncogene addiction” to “drug addiction” by intensive inhibition of the EGFR and MET in lung cancer with activating EGFR mutation. Lung Cancer 76, 292–299 (2012)

    Article  Google Scholar 

  2. Sun, C. et al. Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature 508, 118–122 (2014)

    Article  ADS  CAS  Google Scholar 

  3. Moriceau, G. et al. Tunable-combinatorial mechanisms of acquired resistance limit the efficacy of BRAF/MEK cotargeting but result in melanoma drug addiction. Cancer Cell 27, 240–256 (2015)

    Article  CAS  Google Scholar 

  4. Thakur, M. D. et al. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature 494, 251–255 (2013)

    Article  ADS  Google Scholar 

  5. Seifert, H. et al. Prognostic markers and tumour growth kinetics in melanoma patients progressing on vemurafenib. Melanoma Res. 26, 138–144 (2016)

    Article  CAS  Google Scholar 

  6. Dietrich, S. et al. Continued response off treatment after BRAF inhibition in refractory hairy cell leukemia. J. Clin. Oncol. 31, e300–e303 (2013)

    Article  CAS  Google Scholar 

  7. Dooley, A. J., Gupta, A. & Middleton, M. R. Ongoing response in BRAF V600E-mutant melanoma after cessation of intermittent vemurafenib therapy: a case report. Target. Oncol. 11, 557–563 (2016)

    Google Scholar 

  8. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014)

    Article  ADS  CAS  Google Scholar 

  9. Kemper, K. et al. BRAF(V600E) kinase domain duplication identified in therapy-refractory melanoma patient-derived xenografts. Cell Rep. 16, 263–277 (2016)

    Article  CAS  Google Scholar 

  10. Hoek, K. S. & Goding, C. R. Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res. 23, 746–759 (2010)

    Article  CAS  Google Scholar 

  11. Hoek, K. S. et al. In vivo switching of human melanoma cells between proliferative and invasive states. Cancer Res. 68, 650–656 (2008)

    Article  CAS  Google Scholar 

  12. Wellbrock, C. & Arozarena, I. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy. Pigment Cell Melanoma Res. 28, 390–406 (2015)

    Article  CAS  Google Scholar 

  13. Garraway, L. A. et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436, 117–122 (2005)

    Article  ADS  CAS  Google Scholar 

  14. Verfaillie, A. et al. Decoding the regulatory landscape of melanoma reveals TEADS as regulators of the invasive cell state. Nat. Commun. 6, 6683 (2015)

    Article  ADS  CAS  Google Scholar 

  15. Konieczkowski, D. J. et al. A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. Cancer Discov. 4, 816–827 (2014)

    Article  CAS  Google Scholar 

  16. Müller, J. et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 5, 5712 (2014)

    Article  ADS  Google Scholar 

  17. Ramsdale, R. et al. The transcription cofactor c-JUN mediates phenotype switching and BRAF inhibitor resistance in melanoma. Sci. Signal. 8, ra82 (2015)

    Article  Google Scholar 

  18. Riesenberg, S. et al. MITF and c-Jun antagonism interconnects melanoma dedifferentiation with pro-inflammatory cytokine responsiveness and myeloid cell recruitment. Nat. Commun. 6, 8755 (2015)

    Article  ADS  CAS  Google Scholar 

  19. Shin, S., Dimitri, C. A., Yoon, S.-O., Dowdle, W. & Blenis, J. ERK2 but not ERK1 induces epithelial-to-mesenchymal transformation via DEF motif-dependent signaling events. Mol. Cell 38, 114–127 (2010)

    Article  CAS  Google Scholar 

  20. Kemper, K. et al. Intra- and inter-tumor heterogeneity in a vemurafenib-resistant melanoma patient and derived xenografts. EMBO Mol. Med. 7, 1104–1118 (2015)

    Article  CAS  Google Scholar 

  21. Seghers, A. C., Wilgenhof, S., Lebbé, C. & Neyns, B. Successful rechallenge in two patients with BRAF-V600-mutant melanoma who experienced previous progression during treatment with a selective BRAF inhibitor. Melanoma Res. 22, 466–472 (2012)

    Article  Google Scholar 

  22. Schreuer, M. et al. Combination of dabrafenib plus trametinib for BRAF and MEK inhibitor pretreated patients with advanced BRAF(V600)-mutant melanoma: an open-label, single arm, dual-centre, phase 2 clinical trial. Lancet Oncol. 18, 464–472 (2017)

    Article  CAS  Google Scholar 

  23. Harries, M. et al. Treatment patterns of advanced malignant melanoma (stage III-IV) - A review of current standards in Europe. Eur. J. Cancer 60, 179–189 (2016)

    Article  Google Scholar 

  24. Jain, T. & Bryce, A. Intermittent BRAF inhibition can achieve prolonged disease control in BRAF mutant melanoma. Cureus 7, e410 (2015)

    PubMed  PubMed Central  Google Scholar 

  25. Abdel-Wahab, O. et al. Efficacy of intermittent combined RAF and MEK inhibition in a patient with concurrent BRAF- and NRAS-mutant malignancies. Cancer Discov. 4, 538–545 (2014)

    Article  Google Scholar 

  26. Dooley, A. J., Gupta, A., Bhattacharyya, M. & Middleton, M. R. Intermittent dosing with vemurafenib in BRAF V600E-mutant melanoma: review of a case series. Ther. Adv. Med. Oncol. 6, 262–266 (2014)

    Article  CAS  Google Scholar 

  27. van Steensel, B., Smogorzewska, A. & de Lange, T. TRF2 protects human telomeres from end-to-end fusions. Cell 92, 401–413 (1998)

    Article  CAS  Google Scholar 

  28. Vredeveld, L. C. W. et al. Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis. Genes Dev. 26, 1055–1069 (2012)

    Article  CAS  Google Scholar 

  29. Fontana, M. F. et al. JUNB is a key transcriptional modulator of macrophage activation. J. Immunol. 194, 177–186 (2015)

    Article  CAS  Google Scholar 

  30. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005)

    Article  ADS  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to K. Suda and T. Mitsudomi for HCC827 and HCC827CLR cells, J. Villanueva for 451Lu cells, I. de Krijger for technical advice on FISH, A. Broeks and the NKI Biobank for help with patient samples, R. Beijersbergen for providing the GeCKO library and A. Huitema for providing DTIC. We thank all members of the Peeper and Blank labs for advice. This work was financially supported by the European Research Council under the European Union’s Seventh Framework Programme (FP7/2007–2013) /ERC synergy grant agreement number 319661 COMBATCANCER (X.K., D.S.P.), and a Fellowship (T.K.), a Queen Wilhelmina Award (D.S.P.) and a research grant (nr. 10304; X.K., D.S.P.), all by the Dutch Cancer Society/Koningin Wilhelmina Fonds.

Author information

Authors and Affiliations

Authors

Contributions

X.K., T.K. and D.S.P. designed all experiments. All in vitro experiments were carried out by X.K., with the exception of a reverse transcription PCR analysis done by K.K. Xenograft experiments were done by A.S. T.K. performed all bioinformatic analyses. J.-Y.S. analysed mouse tumours. J.B., E.A.R., M.H.G.F. and H.W.M.N. analysed human melanoma samples in collaboration with C.U.B. X.K. and D.S.P. wrote the manuscript. All authors revised and approved the manuscript. The project was supervised by D.S.P.

Corresponding author

Correspondence to Daniel S. Peeper.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks R. Marais, D. Solit and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 Drug addiction phenotype in acquired-drug-resistant melanoma cells.

Acquired BRAFi-resistant 451LuBR cells were cultured in the presence or absence of 1 μM BRAFi; acquired BRAFi + MEKi-resistant A375BMR, A101DBMR and Mel888BMR cells were cultured in the presence or absence of 0.5 μM BRAFi + 0.05 μM MEKi. Photographs were taken after seven days. The images are representative of three independent biological experiments.

Extended Data Figure 2 Generation of a diverse melanoma cell line panel with distinct drug resistance mechanisms.

a, 451Lu and 451LuBR cells were treated with increasing concentrations of dabrafenib (0, 0.01, 0.1, 1 and 10 μM) for six hours. Total cell lysates were subjected to immunoblotting with indicated antibodies. ERK1/2 served as a loading control. b, A375 and A375BMR cells were treated with increasing concentrations of the BRAFi dabrafenib + MEKi trametinib (0 + 0 μM, 0.01 + 0.001 μM, 0.1 + 0.01 μM and 1 + 0.1 μM) for six hours. Total cell lysates were subjected to immunoblotting with indicated antibodies. ERK1/2 served as a loading control. c, Quantification of BRAF amplification by quantitative PCR on genomic DNA of A375 and A375BMR cells. CRAF was included as a negative control. Cycle threshold values were normalized to LINE. P value calculated by unpaired two-sided Student’s t-test. Data in graphs are mean ± s.d. from three technical replicates; confidence interval 95%. d, Mel888 and Mel888BMR cells were treated with vehicle or 0.5 μM BRAFi + 0.05 μM MEKi for six hours. Total cell lysates were subjected to immunoblotting with indicated antibodies. ERK1/2 served as a loading control. For gel source images, see Supplementary Fig. 1. Data in ad are representative of three independent biological experiments.

Extended Data Figure 3 Disruption of MEK1 prevents mortality in drug-addicted 451LuBR cells after drug withdrawal.

a, 451LuBR, A375BMR, A101DBMR and Mel888BMR cells were infected with lentivirus that expressed sgRNAs targeting genes MAP2K1 (encoding MEK1), MAP2K2 (encoding MEK2) or a scrambled sequence (sgCtrl). Cells were seeded in the presence or absence of MAPK pathway inhibitors, and fixed, stained and photographed after 14 days (BRAFi or BRAFi + MEKi group) or 21 days (no drug group). b, Cell lysates from a were subjected to immunoblotting using the indicated antibodies. HSP90 served as a loading control. For gel source images, see Supplementary Fig. 1. Data in a and b are representative of three independent biological experiments.

Extended Data Figure 4 ERK rebound contributes to drug withdrawal lethality irrespective of drug resistance mechanism.

a, Four parental drug-sensitive cell lines were cultured without MAPK pathway inhibitors, and their drug-resistant counterparts were cultured in the presence or absence of MAPK pathway inhibitors, for the indicated number of days. Total cell lysates were subjected to immunoblotting with indicated antibodies. HSP90 served as a loading control. b, 451LuBR cells were cultured in the presence of 1 μM BRAFi, a high concentration (5 μM) of ERKi, a low concentration (0.05 μM) of ERKi, or vehicle control (no drug). A375BMR, A101DBMR and Mel888BMR cells were cultured with 0.5 μM BRAFi + 0.05 μM MEKi, 5 μM ERKi, 0.05 μM ERKi or vehicle control (no drug). Cells were fixed, stained and photographed after 14 days. For gel source images, see Supplementary Fig. 1. Data in a and b are representative of two independent biological experiments.

Extended Data Figure 5 Drug addiction is relayed by an MITF-dependent phenotype switch.

a, Drug-addicted A375BMR cells were cultured in the absence of 0.5 μM BRAFi + 0.05 μM MEKi for 0, 6, 24 and 48 h, and 451LuBR cells were cultured in the absence of 1 μM BRAFi for 0, 1, 3 and 5 days. Total RNA was isolated and subjected to sequencing analysis. Unsupervised clustering of the 200 most variably expressed genes is represented for the first (either 0 h or 0 days) and last time points (either 48 h or 5 days) in a heat map. b, Sequence data in a (all time points) were subjected to GSEA to determine the correlation of the indicated gene sets with the drug addiction effect in A375BMR and 451LuBR cells. For P value calculation, see Methods. c, 451LuBR and A101DBMR cells were infected with lentivirus that expressed sgRNAs targeting gene encoding cJUN (sgJUN) or a scrambled sequence (sgCtrl). Cells were seeded in the presence or absence of MAPK pathway inhibitors, and fixed, stained and photographed after 14 days (BRAFi or BRAFi + MEKi group) or 21 days (no drug group). d, Total cell lysates of the samples in c were subjected to immunoblotting analysis with indicated antibodies. HSP90 served as a loading control. e, 451LuBR and A101DBMR cells were cultured in the presence or absence of the MAPK pathway inhibitors for 24 h and then subjected to a transwell migration assay. The cells that migrated through the membrane were fixed, stained and photographed after 8 (A101DBMR) or 24 (451LuBR) hours. f, Quantification of the migration capacity (e) from three representative images from each sample; P value calculated by unpaired two-sided Student’s t-test. Data in graphs are mean ± s.d. For gel source images, see Supplementary Fig. 1. Data in ce are representative of two independent biological experiments.

Extended Data Figure 6 Phenotype switch is not observed in non drug-addicted cells upon drug withdrawal.

a, D10BMR cells were cultured with 1 μM BRAFi + 0.1 μM MEKi, 0.5 μM BRAFi + 0.05 μM MEKi or vehicle control (no drug); SK-Mel-28BR cells were cultured with either 3 μM BRAFi dabrafenib, 1 μM dabrafenib or vehicle control (no drug); Mel888PLXR and A375PLXR cells were cultured with 3 μM BRAFi PLX4720, 1 μM PLX4720 or vehicle control (no drug); Cells were fixed, stained and photographed after ten days. b, On the third day, total cell lysates of the samples in a were subjected to immunoblotting analysis with indicated antibodies. HSP90 served as a loading control. For gel source images, see Supplementary Fig. 1. Data in a and b are representative of two independent biological experiments.

Extended Data Figure 7 ERK2 and JUNB act in an MITF-dependent genetic phenotype switching program controlling drug addiction.

a, Mel888BMR cells were infected with lentivirus expressing sgRNAs targeting genes encoding JUNB (sgJUNB), ERK2 (sgERK2) or a scrambled sequence (sgCtrl). Cells were cultured in the presence or absence of 0.5 μM BRAFi + 0.05 μM MEKi for five days. Total RNA was isolated and subjected to sequencing analysis. Unsupervised clustering of the 200 most variably expressed genes is represented in a heat map. b, Genes involved in phenotype switching were selected from the sequence data in a and are represented in a heat map. c, Sequence data in a were subjected to GSEA to determine the correlation of the indicated gene sets with the extent of rescue of the drug addiction phenotype by sgJUNB and sgERK2 in Mel888BMR cells. dw, drug withdrawal. For P value calculation, see Methods.

Extended Data Figure 8 Pharmacological ERK inhibition blocks phenotype switch in drug-addicted cells upon drug withdrawal.

a, Mel888BMR, A101DBMR and A375BMR cells were cultured with 0.5 μM BRAFi + 0.05 μM MEKi, vehicle or 0.05 μM ERKi, 451LuBR cells were cultured with 1 μM BRAFi, vehicle or 0.05 μM ERKi for three days. Total cell lysates were subjected to immunoblotting with indicated antibodies. HSP90 served as a loading control. b, Photographs taken of cells in a, on the third day. c, Mel888 MEK1T55delinsRT and A375 MEK1T55delinsRT cells were cultured in the presence of 0.3 μM BRAFi, low concentration (0.05 μM) of ERKi or vehicle control (no drug). Cells were fixed, stained and photographed after ten days. d, On the fifth day, total cell lysates from cells in c were subjected to immunoblotting with indicated antibodies. HSP90 served as a loading control. e, HCC827CLR cells were cultured in the presence of 1 μM EGFR TKI CL-387785, a high concentration (5 μM) of ERKi, a low concentration (0.5 μM) of ERKi, or with vehicle control (no drug). Cells were fixed, stained and photographed after 14 days. For gel source images, see Supplementary Fig. 1. Data in ae are representative of three independent biological experiments.

Extended Data Figure 9 ERK2 and ERK1 have distinct functions in drug addiction.

a, 451LuBR control cells (sgCtrl), ERK2 knockout cells (sgERK2), ERK1 (v5-tagged)-overexpressing sgERK2 cells (sgERK2, ERK1 OE) and ERK1 knockout cells (sgERK1) were cultured in the presence or absence of 1 μM BRAFi for up to six days. Total cell lysates were subjected to immunoblotting analysis with indicated antibodies. HSP90 served as a loading control. b, Cells described in a were cultured in the presence or absence of BRAFi, and fixed, stained and photographed after 14 days (BRAFi group) or 21 days (no drug group). For gel source images, see Supplementary Fig. 1. Data in a and b are representative of two independent biological experiments.

Extended Data Figure 10 Dynamic phenotype of drug resistance and drug addiction in melanoma.

a, Schematic flow chart indicating drug withdrawal resistant cells were generated from drug-addicted cells with long-term drug withdrawal. b, Parental (A375, 451Lu and A101D) cells, acquired-drug-resistant (A375BMR, 451LuBR and A101DBMR) cells and spontaneously developed drug withdrawal-resistant (A375WR, 451LuWR and A101DWR) cells were cultured with MAPK pathway inhibitors or vehicle. Cells were fixed, stained and photographed after 14 days. c, Total cell lysates from cells in b were subjected to immunoblotting with pERK and ERK antibodies after six hours treatment. For gel source images, see Supplementary Fig. 1. Data in b and c are representative of three independent biological experiments.

Supplementary information

Reporting Summary (PDF 67 kb)

Supplementary Figures

This file contains the uncropped blots from Figures 1-4, Extended Data Figures 2-6 and 8-10. (PDF 28527 kb)

Supplementary Tables

This file contains 3 tables. Table 1 contains full sequencing data of screen clones, table 2 contains primer and oligo sequences for PCR and sgRNA plasmid construction. Table 3 contains xenograft tumor volume measurements. (PDF 295 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kong, X., Kuilman, T., Shahrabi, A. et al. Cancer drug addiction is relayed by an ERK2-dependent phenotype switch. Nature 550, 270–274 (2017). https://doi.org/10.1038/nature24037

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature24037

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing