Reduced Proteolytic Shedding of Receptor Tyrosine Kinases Is a Post-Translational Mechanism of Kinase Inhibitor Resistance.

UNLABELLED Kinase inhibitor resistance often involves upregulation of poorly understood "bypass" signaling pathways. Here, we show that extracellular proteomic adaptation is one path to bypass signaling and drug resistance. Proteolytic shedding of surface receptors, which can provide negative feedback on signaling activity, is blocked by kinase inhibitor treatment and enhances bypass signaling. In particular, MEK inhibition broadly decreases shedding of multiple receptor tyrosine kinases (RTK), including HER4, MET, and most prominently AXL, an ADAM10 and ADAM17 substrate, thus increasing surface RTK levels and mitogenic signaling. Progression-free survival of patients with melanoma treated with clinical BRAF/MEK inhibitors inversely correlates with RTK shedding reduction following treatment, as measured noninvasively in blood plasma. Disrupting protease inhibition by neutralizing TIMP1 improves MAPK inhibitor efficacy, and combined MAPK/AXL inhibition synergistically reduces tumor growth and metastasis in xenograft models. Altogether, extracellular proteomic rewiring through reduced RTK shedding represents a surprising mechanism for bypass signaling in cancer drug resistance. SIGNIFICANCE Genetic, epigenetic, and gene expression alterations often fail to explain adaptive drug resistance in cancer. This work presents a novel post-translational mechanism of such resistance: Kinase inhibitors, particularly targeting MAPK signaling, increase tumor cell surface receptor levels due to widely reduced proteolysis, allowing tumor signaling to circumvent intended drug action.

[1]  W. Yang,et al.  Vitamin D-induced ectodomain shedding of TNF receptor 1 as a nongenomic action: D3 vs D2 derivatives , 2016, The Journal of Steroid Biochemistry and Molecular Biology.

[2]  J. Massagué,et al.  Therapy-induced tumour secretomes promote resistance and tumour progression , 2015, Nature.

[3]  Mohammad Fallahi-Sichani,et al.  Systematic analysis of BRAFV600E melanomas reveals a role for JNK/c-Jun pathway in adaptive resistance to drug-induced apoptosis , 2015 .

[4]  A. Giaccia,et al.  An engineered Axl 'decoy receptor' effectively silences the Gas6-Axl signaling axis. , 2014, Nature chemical biology.

[5]  Renhao Li,et al.  Membrane-enabled dimerization of the intrinsically disordered cytoplasmic domain of ADAM10 , 2014, Proceedings of the National Academy of Sciences.

[6]  A. Harris,et al.  ADAM10 mediates trastuzumab resistance and is correlated with survival in HER2 positive breast cancer , 2014, Oncotarget.

[7]  Hakho Lee,et al.  Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor , 2014, Nature Biotechnology.

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

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

[10]  Miles A. Miller,et al.  The Receptor AXL Diversifies EGFR Signaling and Limits the Response to EGFR-Targeted Inhibitors in Triple-Negative Breast Cancer Cells , 2013, Science Signaling.

[11]  Miles A. Miller,et al.  Regulated ADAM17-dependent EGF family ligand release by substrate-selecting signaling pathways , 2013, Proceedings of the National Academy of Sciences.

[12]  Jongyoon Han,et al.  ADAM-10 and -17 regulate endometriotic cell migration via concerted ligand and receptor shedding feedback on kinase signaling , 2013, Proceedings of the National Academy of Sciences.

[13]  R. Sullivan,et al.  Resistance to BRAF-targeted therapy in melanoma. , 2013, European journal of cancer.

[14]  M. Piccart,et al.  Emerging targeted agents in metastatic breast cancer , 2013, Nature Reviews Clinical Oncology.

[15]  Ivan Babic,et al.  De-repression of PDGFRβ transcription promotes acquired resistance to EGFR tyrosine kinase inhibitors in glioblastoma patients. , 2013, Cancer discovery.

[16]  P. Bonate,et al.  Phase II Study Evaluating 2 Dosing Schedules of Oral Foretinib (GSK1363089), cMET/VEGFR2 Inhibitor, in Patients with Metastatic Gastric Cancer , 2013, PloS one.

[17]  E. McDermott,et al.  ADAM-17: a novel therapeutic target for triple negative breast cancer. , 2013, Annals of oncology : official journal of the European Society for Medical Oncology.

[18]  R. Sedláček,et al.  ADAM10/17-dependent release of soluble c-Met correlates with hepatocellular damage. , 2013, Folia biologica.

[19]  Michael Peyton,et al.  An Epithelial–Mesenchymal Transition Gene Signature Predicts Resistance to EGFR and PI3K Inhibitors and Identifies Axl as a Therapeutic Target for Overcoming EGFR Inhibitor Resistance , 2012, Clinical Cancer Research.

[20]  T. Golub,et al.  Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion , 2012, Nature.

[21]  Jane Fridlyand,et al.  Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors , 2012, Nature.

[22]  Carlotta Costa,et al.  MEK inhibition leads to PI3K/AKT activation by relieving a negative feedback on ERBB receptors. , 2012, Cancer research.

[23]  J. Qiao,et al.  Self-control of HGF regulation on human trophoblast cell invasion via enhancing c-Met receptor shedding by ADAM10 and ADAM17. , 2012, The Journal of clinical endocrinology and metabolism.

[24]  Jae Cheol Lee,et al.  Activation of the AXL Kinase Causes Resistance to EGFR-Targeted Therapy in Lung Cancer , 2012, Nature Genetics.

[25]  G. Hortobagyi,et al.  High ERK protein expression levels correlate with shorter survival in triple-negative breast cancer patients. , 2012, The oncologist.

[26]  R. Derynck,et al.  TACE Activation by MAPK-Mediated Regulation of Cell Surface Dimerization and TIMP3 Association , 2012, Science Signaling.

[27]  A. Blackburn,et al.  Abstract 3228: Dichloroacetate reverses the Warburg effect, inhibiting growth and sensitizing breast cancer cells towards apoptosis , 2012 .

[28]  Jian Jin,et al.  Dynamic Reprogramming of the Kinome in Response to Targeted MEK Inhibition in Triple-Negative Breast Cancer , 2012, Cell.

[29]  Gordon B Mills,et al.  Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. , 2012, Cancer cell.

[30]  高橋 良 Early [18F]fluorodeoxyglucose positron emission tomography at two days of gefitinib treatment predicts clinical outcome in patients with adenocarcinoma of the lung , 2012 .

[31]  E. Shimosegawa,et al.  Early [18F]Fluorodeoxyglucose Positron Emission Tomography at Two Days of Gefitinib Treatment Predicts Clinical Outcome in Patients with Adenocarcinoma of the Lung , 2011, Clinical Cancer Research.

[32]  A. Krüger,et al.  Tumor cell-derived Timp-1 is necessary for maintaining metastasis-promoting Met-signaling via inhibition of Adam-10 , 2011, Clinical & Experimental Metastasis.

[33]  Maeve Mullooly,et al.  The ADAMs family of proteases: new biomarkers and therapeutic targets for cancer? , 2011, Clinical Proteomics.

[34]  Douglas A Lauffenburger,et al.  Proteolytic Activity Matrix Analysis (PrAMA) for simultaneous determination of multiple protease activities. , 2011, Integrative biology : quantitative biosciences from nano to macro.

[35]  S. Chandarlapaty,et al.  PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer , 2011, Oncogene.

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

[37]  Luke A. Gilbert,et al.  DNA Damage-Mediated Induction of a Chemoresistant Niche , 2010, Cell.

[38]  A. Krüger,et al.  A Disintegrin and Metalloproteinase-10 (ADAM-10) Mediates DN30 Antibody-induced Shedding of the Met Surface Receptor* , 2010, The Journal of Biological Chemistry.

[39]  D. Doval,et al.  Clinical benefit of INCB7839, a potent and selective ADAM inhibitor, in combination with trastuzumab in patients with metastatic HER2+ breast cancer. , 2010 .

[40]  J. Birt,et al.  Neutralizing TIMP1 Restores Fecundity in a Rat Model of Endometriosis and Treating Control Rats with TIMP1 Causes Anomalies in Ovarian Function and Embryo Development1 , 2010, Biology of reproduction.

[41]  Z. Werb,et al.  Matrix Metalloproteinases: Regulators of the Tumor Microenvironment , 2010, Cell.

[42]  C. Eaves,et al.  Y-box binding protein-1 induces the expression of CD44 and CD49f leading to enhanced self-renewal, mammosphere growth, and drug resistance. , 2010, Cancer research.

[43]  M. Gooz ADAM-17: the enzyme that does it all , 2010, Critical reviews in biochemistry and molecular biology.

[44]  P. Altevogt,et al.  ADAM10 is upregulated in melanoma metastasis compared with primary melanoma. , 2010, The Journal of investigative dermatology.

[45]  R. Derynck,et al.  Direct activation of TACE-mediated ectodomain shedding by p38 MAP kinase regulates EGF receptor-dependent cell proliferation. , 2010, Molecular cell.

[46]  Y. Hitoshi,et al.  R428, a selective small molecule inhibitor of Axl kinase, blocks tumor spread and prolongs survival in models of metastatic breast cancer. , 2010, Cancer research.

[47]  Luca Toschi,et al.  Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. , 2010, Cancer cell.

[48]  P. Scherle,et al.  ADAM‐mediated amphiregulin shedding and EGFR transactivation , 2009, Cell proliferation.

[49]  Joel Greshock,et al.  Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: activation of AXL. , 2009, Cancer research.

[50]  Wei Zhou,et al.  In vivo Antitumor Activity of MEK and Phosphatidylinositol 3-Kinase Inhibitors in Basal-Like Breast Cancer Models , 2009, Clinical Cancer Research.

[51]  Dongsheng Tu,et al.  K-ras mutations and benefit from cetuximab in advanced colorectal cancer. , 2008, The New England journal of medicine.

[52]  J. Fridman,et al.  Synergistic inhibition with a dual epidermal growth factor receptor/HER-2/neu tyrosine kinase inhibitor and a disintegrin and metalloprotease inhibitor. , 2008, Cancer research.

[53]  D. Edwards,et al.  The ADAM metalloproteinases , 2008, Molecular Aspects of Medicine.

[54]  P. Dempsey,et al.  The ADAM10 Prodomain Is a Specific Inhibitor of ADAM10 Proteolytic Activity and Inhibits Cellular Shedding Events* , 2007, Journal of Biological Chemistry.

[55]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[56]  Andy J. Minn,et al.  Genes that mediate breast cancer metastasis to lung , 2005, Nature.

[57]  L. Naldini,et al.  Targeting the tumor and its microenvironment by a dual-function decoy Met receptor. , 2004, Cancer cell.

[58]  B. Szabó,et al.  Putative function of ADAM9, ADAM10, and ADAM17 as APP -secretase , 2003 .

[59]  B. Szabó,et al.  Putative function of ADAM9, ADAM10, and ADAM17 as APP alpha-secretase. , 2003, Biochemical and biophysical research communications.

[60]  Z. Werb,et al.  G protein–coupled receptors , 2002 .

[61]  R. Paxton,et al.  A Proteomic Approach for the Identification of Cell-surface Proteins Shed by Metalloproteases* , 2002, Molecular & Cellular Proteomics.

[62]  C. Basbaum,et al.  Platelet-activating factor receptor and ADAM10 mediate responses to Staphylococcus aureus in epithelial cells , 2002, Nature Medicine.