ZEB1‐mediated melanoma cell plasticity enhances resistance to MAPK inhibitors

Targeted therapies with MAPK inhibitors (MAPKi) are faced with severe problems of resistance in BRAF‐mutant melanoma. In parallel to the acquisition of genetic mutations, melanoma cells may also adapt to the drugs through phenotype switching. The ZEB1 transcription factor, a known inducer of EMT and invasiveness, is now considered as a genuine oncogenic factor required for tumor initiation, cancer cell plasticity, and drug resistance in carcinomas. Here, we show that high levels of ZEB1 expression are associated with inherent resistance to MAPKi in BRAFV600‐mutated cell lines and tumors. ZEB1 levels are also elevated in melanoma cells with acquired resistance and in biopsies from patients relapsing while under treatment. ZEB1 overexpression is sufficient to drive the emergence of resistance to MAPKi by promoting a reversible transition toward a MITFlow/p75high stem‐like and tumorigenic phenotype. ZEB1 inhibition promotes cell differentiation, prevents tumorigenic growth in vivo, sensitizes naive melanoma cells to MAPKi, and induces cell death in resistant cells. Overall, our results demonstrate that ZEB1 is a major driver of melanoma cell plasticity, driving drug adaptation and phenotypic resistance to MAPKi.

[1]  Annette Meeson,et al.  Epithelial-to-mesenchymal transition: what is the impact on breast cancer stem cells and drug resistance. , 2014, Cancer treatment reviews.

[2]  T. Keck,et al.  ZEB1-associated drug resistance in cancer cells is reversed by the class I HDAC inhibitor mocetinostat , 2015, EMBO molecular medicine.

[3]  Charis Eng,et al.  Direct evidence for epithelial-mesenchymal transitions in breast cancer. , 2008, Cancer research.

[4]  Corbin E. Meacham,et al.  Tumour heterogeneity and cancer cell plasticity , 2013, Nature.

[5]  Irving L. Weissman,et al.  Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271 , 2011, Nature.

[6]  D. Dean,et al.  Different thresholds of ZEB1 are required for Ras-mediated tumour initiation and metastasis , 2014, Nature Communications.

[7]  S. Morrison,et al.  Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. , 2010, Cancer cell.

[8]  A. Dhillon,et al.  Fra-1 controls motility of bladder cancer cells via transcriptional upregulation of the receptor tyrosine kinase AXL , 2012, Oncogene.

[9]  T. Tan,et al.  Epithelial-mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients , 2014, EMBO molecular medicine.

[10]  Irving L. Weissman,et al.  Human Melanoma Initiating Cells Express Neural Crest Nerve Growth Factor Receptor CD271 , 2010, Nature.

[11]  Robert A. Weinberg,et al.  Poised Chromatin at the ZEB1 Promoter Enables Breast Cancer Cell Plasticity and Enhances Tumorigenicity , 2013, Cell.

[12]  Tatyana Chernova,et al.  Direct repression of cyclin D1 by SIP1 attenuates cell cycle progression in cells undergoing an epithelial mesenchymal transition. , 2007, Molecular biology of the cell.

[13]  Mohamed H. Sayegh,et al.  Identification of cells initiating human melanomas , 2008, Nature.

[14]  J. Dou,et al.  Effect of downregulation of ZEB1 on vimentin expression, tumour migration and tumourigenicity of melanoma B16F10 cells and CSCs , 2014, Cell biology international.

[15]  A. Hauschild,et al.  Improved overall survival in melanoma with combined dabrafenib and trametinib. , 2015, The New England journal of medicine.

[16]  A. Puisieux,et al.  Oncogenic roles of EMT-inducing transcription factors , 2014, Nature Cell Biology.

[17]  Frank McCormick,et al.  Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond , 2014, Nature Reviews Cancer.

[18]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[19]  H. Moch,et al.  Human CD271-positive melanoma stem cells associated with metastasis establish tumor heterogeneity and long-term growth. , 2011, Cancer research.

[20]  Steven J. M. Jones,et al.  Genomic Classification of Cutaneous Melanoma , 2015, Cell.

[21]  R. Weinberg,et al.  Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits , 2009, Nature Reviews Cancer.

[22]  J. Larkin,et al.  Tunable-combinatorial mechanisms of acquired resistance limit the efficacy of BRAF/MEK cotargeting but result in melanoma drug addiction. , 2015, Cancer cell.

[23]  R. Moon,et al.  WNT5A enhances resistance of melanoma cells to targeted BRAF inhibitors. , 2014, The Journal of clinical investigation.

[24]  W. Huber,et al.  which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. MAnorm: a robust model for quantitative comparison of ChIP-Seq data sets , 2011 .

[25]  N. Rosen,et al.  Tumor adaptation and resistance to RAF inhibitors , 2013, Nature Medicine.

[26]  J. Lachuer,et al.  A switch in the expression of embryonic EMT-inducers drives the development of malignant melanoma. , 2013, Cancer cell.

[27]  P. Bahadoran,et al.  CD271 is an imperfect marker for melanoma initiating cells , 2014, Oncotarget.

[28]  T. Graeber,et al.  Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma , 2014, Nature Communications.

[29]  R. Huang,et al.  Epithelial-Mesenchymal Transitions in Development and Disease , 2009, Cell.

[30]  C. Wellbrock,et al.  Microphthalmia‐associated transcription factor in melanoma development and MAP‐kinase pathway targeted therapy , 2015, Pigment cell & melanoma research.

[31]  S. Nelson,et al.  Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation , 2010, Nature.

[32]  H. Lehrach,et al.  The Nerve Growth Factor Receptor CD271 Is Crucial to Maintain Tumorigenicity and Stem-Like Properties of Melanoma Cells , 2014, PloS one.

[33]  G. Berx,et al.  Identification of a ZEB2-MITF-ZEB1 transcriptional network that controls melanogenesis and melanoma progression , 2014, Cell Death and Differentiation.

[34]  N. Hayward,et al.  Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma , 2014, Nature Communications.

[35]  J. Mesirov,et al.  A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. , 2014, Cancer discovery.

[36]  Benjamin E. Gross,et al.  The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. , 2012, Cancer discovery.

[37]  A. Hauschild,et al.  Improved survival with vemurafenib in melanoma with BRAF V600E mutation. , 2011, The New England journal of medicine.

[38]  S. Rorive,et al.  Different levels of Twist1 regulate skin tumor initiation, stemness, and progression. , 2015, Cell stem cell.

[39]  K. Hoek,et al.  Cancer stem cells versus phenotype‐switching in melanoma , 2010, Pigment cell & melanoma research.

[40]  J. Lachuer,et al.  EMT Inducers Catalyze Malignant Transformation of Mammary Epithelial Cells and Drive Tumorigenesis towards Claudin-Low Tumors in Transgenic Mice , 2012, PLoS genetics.

[41]  Tuan Zea Tan,et al.  Epithelial–mesenchymal status renders differential responses to cisplatin in ovarian cancer , 2014, Oncogene.

[42]  S. Morrison,et al.  Efficient tumor formation by single human melanoma cells , 2008, Nature.

[43]  Benjamin E. Gross,et al.  Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal , 2013, Science Signaling.

[44]  A. Roesch Tumor heterogeneity and plasticity as elusive drivers for resistance to MAPK pathway inhibition in melanoma , 2014, Oncogene.

[45]  Li Ma,et al.  ZEB1: At the crossroads of epithelial-mesenchymal transition, metastasis and therapy resistance , 2015, Cell cycle.

[46]  J. Pringle,et al.  Plasticity of melanoma and EMT-TF reprogramming , 2013, Oncotarget.

[47]  Wenjun Guo,et al.  The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells , 2008, Cell.

[48]  W. Woodward,et al.  ATM-mediated stabilization of ZEB1 promotes DNA damage response and radioresistance through CHK1 , 2014, Nature Cell Biology.

[49]  Alexander Roesch,et al.  A Temporarily Distinct Subpopulation of Slow-Cycling Melanoma Cells Is Required for Continuous Tumor Growth , 2010, Cell.

[50]  Rajiv Narayan,et al.  A melanocyte lineage program confers resistance to MAP kinase pathway inhibition , 2013, Nature.

[51]  A. McKenna,et al.  The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma. , 2014, Cancer discovery.

[52]  D. Kusewitt,et al.  Slug expression during melanoma progression. , 2012, The American journal of pathology.

[53]  A. Puisieux,et al.  Generation of Breast Cancer Stem Cells through Epithelial-Mesenchymal Transition , 2008, PloS one.

[54]  S. Aerts,et al.  Decoding the regulatory landscape of melanoma reveals TEADS as regulators of the invasive cell state , 2015, Nature Communications.

[55]  J. Settleman,et al.  EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer , 2010, Oncogene.

[56]  Antoni Ribas,et al.  Non-genomic and Immune Evolution of Melanoma Acquiring MAPKi Resistance , 2015, Cell.

[57]  K. Flaherty,et al.  Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. , 2012, The New England journal of medicine.