RT Journal Article SR Electronic T1 Pharmacologically targeting a novel pathway of sodium iodide symporter trafficking to enhance radioiodine uptake JF bioRxiv FD Cold Spring Harbor Laboratory SP 622241 DO 10.1101/622241 A1 Alice Fletcher A1 Martin L. Read A1 Caitlin E.M. Thornton A1 Dean P. Larner A1 Vikki L. Poole A1 Katie Brookes A1 Hannah R. Nieto A1 Mohammed Alshahrani A1 Rebecca J. Thompson A1 Gareth G. Lavery A1 Moray J. Campbell A1 Kristien Boelaert A1 Andrew S. Turnell A1 Vicki E. Smith A1 Christopher J. McCabe YR 2019 UL http://biorxiv.org/content/early/2019/05/07/622241.abstract AB Radioiodine treatment fails ≥25% of patients with thyroid cancer and has been proposed as a potential treatment for breast cancer. Cellular iodide uptake is governed by the sodium iodide symporter (NIS), which is frequently mislocalized in thyroid and breast tumours. However, the trafficking of NIS to the plasma membrane (PM) is ill-defined. Through mass spectrometry, co-immunoprecipitation, cell surface biotinylation and proximity ligation assays we identify two proteins which control NIS subcellular trafficking: ADP-ribosylation factor 4 (ARF4) and valosin-containing protein (VCP). HiLo microscopy revealed ARF4 enhanced NIS trafficking in co-incident PM vesicles, governed by a C-terminal VXPX motif, whilst papillary thyroid cancers (PTC) demonstrate repressed ARF4 expression. In contrast, VCP, the central protein in ER-associated degradation, specifically bound NIS and decreased its PM localization. Five chemically distinct allosteric VCP inhibitors all overcame VCP-mediated repression of NIS function. In mice, two re-purposed FDA-approved VCP inhibitors significantly enhanced radioiodine uptake into thyrocytes, whilst human primary thyrocytes showed similar increases. Critically, PTC patients with high tumoural VCP expression who received radioiodine had strikingly worse disease-free survival. These studies now delineate the mechanisms of NIS trafficking, and for the first time open the therapeutic possibility of systemically enhancing radioiodine uptake in patients via FDA-approved drugs.One Sentence Summary Novel NIS interactors ARF4 and VCP alter NIS trafficking in vitro, and FDA-approved VCP inhibitors can significantly enhance radioiodine uptake.