The Advantages of Targeted Protein Degradation Over Inhibition: An RTK Case Study.

Proteolysis targeting chimera (PROTAC) technology has emerged over the last two decades as a powerful tool for targeted degradation of endogenous proteins. Herein we describe the development of PROTACs for receptor tyrosine kinases, a protein family yet to be targeted for induced protein degradation. The use of VHL-recruiting PROTACs against this protein family reveals several advantages of degradation over inhibition alone: direct comparisons of fully functional, target-degrading PROTACs with target-inhibiting variants that contain an inactivated E3 ligase-recruiting ligand show that degradation leads to more potent inhibition of cell proliferation and a more durable and sustained downstream signaling response, and thus addresses the kinome rewiring challenge seen with many receptor tyrosine kinase inhibitors. Combined, these findings demonstrate the ability to target receptor tyrosine kinases for degradation using the PROTAC technology and outline the advantages of this degradation-based approach.

[1]  Geoffrey R. Oxnard,et al.  Structural, Biochemical, and Clinical Characterization of Epidermal Growth Factor Receptor (EGFR) Exon 20 Insertion Mutations in Lung Cancer , 2013, Science Translational Medicine.

[2]  A. Scott,et al.  The plasticity of oncogene addiction: implications for targeted therapies directed to receptor tyrosine kinases. , 2009, Neoplasia.

[3]  N. Iqbal,et al.  Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications , 2014, Molecular biology international.

[4]  Ming-Sound Tsao,et al.  An overview of the c-MET signaling pathway , 2011, Therapeutic advances in medical oncology.

[5]  D. Stolz,et al.  Cross-talk between Epidermal Growth Factor Receptor and c-Met Signal Pathways in Transformed Cells* , 2000, The Journal of Biological Chemistry.

[6]  L. Jones,et al.  Selective Downregulation of JAK2 and JAK3 by an ATP-Competitive pan-JAK Inhibitor. , 2017, ACS chemical biology.

[7]  V. Lang,et al.  Efficient protection and isolation of ubiquitylated proteins using tandem ubiquitin‐binding entities , 2009, EMBO reports.

[8]  T. Heightman,et al.  Protein Degradation by In-Cell Self-Assembly of Proteolysis Targeting Chimeras , 2016, ACS central science.

[9]  K. Takeuchi,et al.  Receptor tyrosine kinases and targeted cancer therapeutics. , 2011, Biological & pharmaceutical bulletin.

[10]  Julien Michel,et al.  Targeting the von Hippel–Lindau E3 Ubiquitin Ligase Using Small Molecules To Disrupt the VHL/HIF-1α Interaction , 2012, Journal of the American Chemical Society.

[11]  R. Deshaies,et al.  Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[12]  M. Naujokas,et al.  Met/Hepatocyte Growth Factor Receptor Ubiquitination Suppresses Transformation and Is Required for Hrs Phosphorylation , 2005, Molecular and Cellular Biology.

[13]  A. Misra,et al.  Epidermal growth factor receptor targeting in cancer: a review of trends and strategies. , 2013, Biomaterials.

[14]  J. Ovádi,et al.  Chemically Induced Degradation of Sirtuin 2 (Sirt2) by a Proteolysis Targeting Chimera (PROTAC) Based on Sirtuin Rearranging Ligands (SirReals). , 2017, Journal of medicinal chemistry.

[15]  J. Baselga,et al.  ZD1839, a specific epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, induces the formation of inactive EGFR/HER2 and EGFR/HER3 heterodimers and prevents heregulin signaling in HER2-overexpressing breast cancer cells. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[16]  James S. Duncan,et al.  Resistance to BET Bromodomain Inhibitors Is Mediated by Kinome Reprogramming in Ovarian Cancer. , 2016, Cell reports.

[17]  L. Neckers,et al.  Cancer cells harboring MET gene amplification activate alternative signaling pathways to escape MET inhibition but remain sensitive to Hsp90 inhibitors , 2009, Cell cycle.

[18]  D. Fenyo,et al.  Phosphotyrosine Signaling Networks in Epidermal Growth Factor Receptor Overexpressing Squamous Carcinoma Cells*S , 2005, Molecular & Cellular Proteomics.

[19]  C. Crews,et al.  Chemical biology: Greasy tags for protein removal , 2012, Nature.

[20]  T. Corson,et al.  Small-Molecule Hydrophobic Tagging Induced Degradation of HaloTag Fusion Proteins , 2011, Nature Chemical Biology.

[21]  T. Pellinen,et al.  Small GTPase Rab21 regulates cell adhesion and controls endosomal traffic of β1-integrins , 2006, The Journal of cell biology.

[22]  A. Marcus,et al.  Hepatocyte growth factor is predominantly expressed by the carcinoma cells in non-small-cell lung cancer. , 2001, Human pathology.

[23]  Sun-Mi Park,et al.  Foretinib (GSK1363089), an Orally Available Multikinase Inhibitor of c-Met and VEGFR-2, Blocks Proliferation, Induces Anoikis, and Impairs Ovarian Cancer Metastasis , 2011, Clinical Cancer Research.

[24]  V. Calléja,et al.  A direct role for Met endocytosis in tumorigenesis , 2011, Nature Cell Biology.

[25]  C. Crews,et al.  Development and characterization of proteasome inhibitors. , 2005, Methods in enzymology.

[26]  James S. Duncan,et al.  Inhibition of Lapatinib-Induced Kinome Reprogramming in ERBB2-Positive Breast Cancer by Targeting BET Family Bromodomains. , 2015, Cell reports.

[27]  W. L. Jorgensen,et al.  Small-molecule inhibitors of the interaction between the E3 ligase VHL and HIF1α. , 2012, Angewandte Chemie.

[28]  L. Neckers,et al.  The HSP90 inhibitor ganetespib synergizes with the MET kinase inhibitor crizotinib in both crizotinib-sensitive and -resistant MET-driven tumor models. , 2013, Cancer research.

[29]  A. Ullrich,et al.  The discovery of receptor tyrosine kinases: targets for cancer therapy , 2004, Nature Reviews Cancer.

[30]  C. Klein,et al.  Target Binding Properties and Cellular Activity of Afatinib (BIBW 2992), an Irreversible ErbB Family Blocker , 2012, Journal of Pharmacology and Experimental Therapeutics.

[31]  T. Corson,et al.  Posttranslational protein knockdown coupled to receptor tyrosine kinase activation with phosphoPROTACs , 2013, Proceedings of the National Academy of Sciences.

[32]  I. E. Smith,et al.  HaloPROTACS: Use of Small Molecule PROTACs to Induce Degradation of HaloTag Fusion Proteins. , 2015, ACS chemical biology.

[33]  C. Crews,et al.  Targeted intracellular protein degradation induced by a small molecule: En route to chemical proteomics. , 2008, Bioorganic & medicinal chemistry letters.

[34]  S. Akinaga,et al.  A pharmacodynamic study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response. , 2009, Blood.

[35]  Carmen Birchmeier,et al.  Targeting MET in cancer: rationale and progress , 2012, Nature Reviews Cancer.

[36]  A. Xu,et al.  Receptor tyrosine kinase coactivation networks in cancer. , 2010, Cancer research.

[37]  C. Crews,et al.  Hijacking the E3 Ubiquitin Ligase Cereblon to Efficiently Target BRD4. , 2015, Chemistry & biology.

[38]  A. Ciulli,et al.  Selective Small Molecule Induced Degradation of the BET Bromodomain Protein BRD4 , 2015, ACS chemical biology.

[39]  Eunhwa Ko,et al.  Modular PROTAC Design for the Degradation of Oncogenic BCR-ABL. , 2016, Angewandte Chemie.

[40]  S. Ogawa,et al.  Gastric cancer cell line Hs746T harbors a splice site mutation of c-Met causing juxtamembrane domain deletion. , 2010, Biochemical and biophysical research communications.

[41]  Adriana L. Gonzalez,et al.  Epidermal Growth Factor Receptor (EGFR) Antibody Down-regulates Mutant Receptors and Inhibits Tumors Expressing EGFR Mutations* , 2006, Journal of Biological Chemistry.

[42]  H. Band,et al.  Mutation of the c-Cbl TKB domain binding site on the Met receptor tyrosine kinase converts it into a transforming protein. , 2001, Molecular cell.

[43]  Joon-Oh Park,et al.  MET Amplification Leads to Gefitinib Resistance in Lung Cancer by Activating ERBB3 Signaling , 2007, Science.

[44]  P. Parker,et al.  Protein Kinase C Controls Microtubule-based Traffic but Not Proteasomal Degradation of c-Met* , 2003, Journal of Biological Chemistry.

[45]  Liu Liu,et al.  Discovery of a Small-Molecule Degrader of Bromodomain and Extra-Terminal (BET) Proteins with Picomolar Cellular Potencies and Capable of Achieving Tumor Regression , 2017, Journal of medicinal chemistry.

[46]  P. Jänne,et al.  Pharmacological Targeting of the Pseudokinase Her3 , 2014, Nature chemical biology.

[47]  J. Schlessinger,et al.  Cell Signaling by Receptor Tyrosine Kinases , 2000, Cell.

[48]  D. Lamont,et al.  Structural basis of PROTAC cooperative recognition for selective protein degradation , 2017, Nature chemical biology.

[49]  A. Drilon MET Exon 14 Alterations in Lung Cancer: Exon Skipping Extends Half-Life , 2016, Clinical Cancer Research.

[50]  C. Crews,et al.  Lessons in PROTAC Design from Selective Degradation with a Promiscuous Warhead. , 2017, Cell chemical biology.

[51]  Amanda Doucette,et al.  An inhibitor of NEDD8-activating enzyme as a new approach to treat cancer , 2009, Nature.

[52]  Chad J. Miller,et al.  A comprehensive mathematical model for three-body binding equilibria. , 2013, Journal of the American Chemical Society.

[53]  W. Birchmeier,et al.  Met, metastasis, motility and more , 2003, Nature Reviews Molecular Cell Biology.

[54]  A. Bozec,et al.  EGFR targeting therapies: monoclonal antibodies versus tyrosine kinase inhibitors. Similarities and differences. , 2007, Critical reviews in oncology/hematology.

[55]  M. Wislez,et al.  Mutations at the splice sites of exon 14 of MET gene: a new target for sarcomatoid carcinomas? , 2016, Annals of translational medicine.

[56]  K. Bhalla,et al.  Targeting HSP90 for cancer therapy , 2009, British Journal of Cancer.

[57]  James S. Duncan,et al.  The dynamic nature of the kinome. , 2013, The Biochemical journal.

[58]  C. Crews,et al.  PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer , 2016, Proceedings of the National Academy of Sciences.

[59]  Somasekar Seshagiri,et al.  Somatic mutations lead to an oncogenic deletion of met in lung cancer. , 2006, Cancer research.

[60]  I. E. Smith,et al.  Catalytic in vivo protein knockdown by small-molecule PROTACs. , 2015, Nature chemical biology.

[61]  G. Scagliotti,et al.  Phase III Multinational, Randomized, Double-Blind, Placebo-Controlled Study of Tivantinib (ARQ 197) Plus Erlotinib Versus Erlotinib Alone in Previously Treated Patients With Locally Advanced or Metastatic Nonsquamous Non-Small-Cell Lung Cancer. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[62]  L. Sequist,et al.  Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[63]  L. Neckers,et al.  Low Dose Geldanamycin Inhibits Hepatocyte Growth Factor- and Hypoxia-Stimulated Invasion of Cancer Cells , 2007, Cell cycle.

[64]  M. Berger,et al.  Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. , 2006, Cancer research.