Lysosome-targeting chimaeras for degradation of extracellular proteins

The majority of therapies that target individual proteins rely on specific activity-modulating interactions with the target protein—for example, enzyme inhibition or ligand blocking. However, several major classes of therapeutically relevant proteins have unknown or inaccessible activity profiles and so cannot be targeted by such strategies. Protein-degradation platforms such as proteolysis-targeting chimaeras (PROTACs) 1 , 2 and others (for example, dTAGs 3 , Trim-Away 4 , chaperone-mediated autophagy targeting 5 and SNIPERs 6 ) have been developed for proteins that are typically difficult to target; however, these methods involve the manipulation of intracellular protein degradation machinery and are therefore fundamentally limited to proteins that contain cytosolic domains to which ligands can bind and recruit the requisite cellular components. Extracellular and membrane-associated proteins—the products of 40% of all protein-encoding genes 7 —are key agents in cancer, ageing-related diseases and autoimmune disorders 8 , and so a general strategy to selectively degrade these proteins has the potential to improve human health. Here we establish the targeted degradation of extracellular and membrane-associated proteins using conjugates that bind both a cell-surface lysosome-shuttling receptor and the extracellular domain of a target protein. These initial lysosome-targeting chimaeras, which we term LYTACs, consist of a small molecule or antibody fused to chemically synthesized glycopeptide ligands that are agonists of the cation-independent mannose-6-phosphate receptor (CI-M6PR). We use LYTACs to develop a CRISPR interference screen that reveals the biochemical pathway for CI-M6PR-mediated cargo internalization in cell lines, and uncover the exocyst complex as a previously unidentified—but essential—component of this pathway. We demonstrate the scope of this platform through the degradation of therapeutically relevant proteins, including apolipoprotein E4, epidermal growth factor receptor, CD71 and programmed death-ligand 1. Our results establish a modular strategy for directing secreted and membrane proteins for lysosomal degradation, with broad implications for biochemical research and for therapeutics. Lysosome-targeting chimaeras—in which a small molecule or antibody is connected to a glycopeptide ligand to form a conjugate that can bind a cell-surface lysosome-shuttling receptor and a protein target—are used to achieve the targeted degradation of extracellular and membrane proteins.

[1]  Jun S. Liu,et al.  MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens , 2014, Genome Biology.

[2]  G. Bu,et al.  Apolipoprotein E as a Therapeutic Target in Alzheimer’s Disease: A Review of Basic Research and Clinical Evidence , 2016, CNS Drugs.

[3]  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.

[4]  M. Prata,et al.  A shortcut to the lysosome: the mannose-6-phosphate-independent pathway. , 2012, Molecular genetics and metabolism.

[5]  W. Sly,et al.  Enzyme therapy in mannose receptor-null mucopolysaccharidosis VII mice defines roles for the mannose 6-phosphate and mannose receptors , 2006, Proceedings of the National Academy of Sciences.

[6]  Y. T. Wang,et al.  Rapid and reversible knockdown of endogenous proteins by peptide-directed lysosomal degradation , 2014, Nature Neuroscience.

[7]  Jamie B. Spangler,et al.  Combination antibody treatment down-regulates epidermal growth factor receptor by inhibiting endosomal recycling , 2010, Proceedings of the National Academy of Sciences.

[8]  Nicholas W. Kwiecien,et al.  Improved Precursor Characterization for Data-Dependent Mass Spectrometry. , 2018, Analytical chemistry.

[9]  Soumen Das,et al.  Controlled Synthesis of End-Functionalized Mannose-6-phosphate Glycopolypeptides for Lysosome Targeting. , 2016, ACS macro letters.

[10]  Melina Schuh,et al.  A Method for the Acute and Rapid Degradation of Endogenous Proteins , 2017, Cell.

[11]  J. Weissman,et al.  Functional genomics platform for pooled screening and generation of mammalian genetic interaction maps , 2014, Nature Protocols.

[12]  Anton Arkhipov,et al.  EGFR oligomerization organizes kinase-active dimers into competent signalling platforms , 2016, Nature Communications.

[13]  C. Bustamante,et al.  Chemically tunable mucin chimeras assembled on living cells , 2015, Proceedings of the National Academy of Sciences.

[14]  Marco Y. Hein,et al.  Accurate Proteome-wide Label-free Quantification by Delayed Normalization and Maximal Peptide Ratio Extraction, Termed MaxLFQ * , 2014, Molecular & Cellular Proteomics.

[15]  J. Blenis,et al.  Activation of PI3K/Akt and MAPK pathways regulates Myc-mediated transcription by phosphorylating and promoting the degradation of Mad1 , 2008, Proceedings of the National Academy of Sciences.

[16]  P. Olinga,et al.  Albumin modified with mannose 6‐phosphate: A potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic stellate cells , 1999, Hepatology.

[17]  R. G. Anderson,et al.  Folate receptors targeted to clathrin-coated pits cannot regulate vitamin uptake. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[18]  Max A. Horlbeck,et al.  Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation , 2016, eLife.

[19]  Marco Y. Hein,et al.  The Perseus computational platform for comprehensive analysis of (prote)omics data , 2016, Nature Methods.

[20]  M. Rebelatto,et al.  A Biparatopic HER2-Targeting Antibody-Drug Conjugate Induces Tumor Regression in Primary Models Refractory to or Ineligible for HER2-Targeted Therapy. , 2016, Cancer cell.

[21]  James E. Bradner,et al.  Phthalimide conjugation as a strategy for in vivo target protein degradation , 2015, Science.

[22]  James E. Bradner,et al.  The dTAG system for immediate and target-specific protein degradation , 2018, Nature Chemical Biology.

[23]  E. Isacoff,et al.  Molecular basis for multimerization in the activation of the epidermal growth factor receptor , 2016, eLife.

[24]  M. Naito,et al.  SNIPERs-Hijacking IAP activity to induce protein degradation. , 2019, Drug discovery today. Technologies.

[25]  S. Kornfeld,et al.  Mannose 6-phosphate receptors: new twists in the tale , 2003, Nature Reviews Molecular Cell Biology.

[26]  Israel Steinfeld,et al.  BMC Bioinformatics BioMed Central , 2008 .

[27]  Y. Hathout,et al.  The human secretome atlas initiative: implications in health and disease conditions. , 2013, Biochimica et biophysica acta.

[28]  Margaret R. Heider,et al.  Exorcising the Exocyst Complex , 2012, Traffic.

[29]  Steven P Gygi,et al.  Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry , 2007, Nature Methods.

[30]  Dennis L. Buckley,et al.  Selective Target Protein Degradation via Phthalimide Conjugation , 2015 .

[31]  M. Gary‐Bobo,et al.  Mannose 6-phosphate receptor targeting and its applications in human diseases. , 2007, Current medicinal chemistry.

[32]  Valentin Jaumouillé,et al.  The position of lysosomes within the cell determines their luminal pH , 2016, The Journal of cell biology.

[33]  M. Wilchek,et al.  Affinity cleavage of cell surface antibodies using the avidin-biotin system. , 1993, Journal of immunological methods.

[34]  Jüergen Cox,et al.  The MaxQuant computational platform for mass spectrometry-based shotgun proteomics , 2016, Nature Protocols.

[35]  J. Montero,et al.  A Flexible Route to Mannose 6-Phosphonate Functionalized Derivatives , 2002 .

[36]  R D Klausner,et al.  Exposure of K562 cells to anti-receptor monoclonal antibody OKT9 results in rapid redistribution and enhanced degradation of the transferrin receptor , 1986, The Journal of cell biology.

[37]  D. Berkowitz,et al.  Mono- and bivalent ligands bearing mannose 6-phosphate (M6P) surrogates: targeting the M6P/insulin-like growth factor II receptor. , 2004, Organic letters.

[38]  M. Mann,et al.  Andromeda: a peptide search engine integrated into the MaxQuant environment. , 2011, Journal of proteome research.

[39]  J. Xia,et al.  The role of EGF-EGFR signalling pathway in hepatocellular carcinoma inflammatory microenvironment , 2013, Journal of cellular and molecular medicine.

[40]  Marcel Garcia,et al.  Synthesis of new sulfonate and phosphonate derivatives for cation-independent mannose 6-phosphate receptor targeting. , 2008, Bioorganic & medicinal chemistry letters.

[41]  S. Dawson,et al.  CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity , 2017, Nature.

[42]  J. Montero,et al.  Synthesis and receptor binding affinity of carboxylate analogues of the mannose 6-phosphate recognition marker. , 2006, Bioorganic & medicinal chemistry.

[43]  Qun Zhou,et al.  Conjugation of Mannose 6-Phosphate-containing Oligosaccharides to Acid α-Glucosidase Improves the Clearance of Glycogen in Pompe Mice* , 2004, Journal of Biological Chemistry.

[44]  P. Shang,et al.  Transferrin receptor 1 in cancer: a new sight for cancer therapy. , 2018, American journal of cancer research.

[45]  S. Kornfeld,et al.  Engineering of GlcNAc-1-Phosphotransferase for Production of Highly Phosphorylated Lysosomal Enzymes for Enzyme Replacement Therapy , 2017, Molecular therapy. Methods & clinical development.

[46]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[47]  T. Igawa,et al.  Sweeping antibody as a novel therapeutic antibody modality capable of eliminating soluble antigens from circulation , 2016, Immunological reviews.