American Association for Cancer Research
Browse
ccr-23-2043_supplementary_figure_s6_suppfs6.pdf (17.03 MB)

Supplementary Figure S6 from Combined Vorinostat and Chloroquine Inhibit Sodium–Iodide Symporter Endocytosis and Enhance Radionuclide Uptake In Vivo

Download (17.03 MB)
journal contribution
posted on 2024-04-01, 07:21 authored by Martin L. Read, Katie Brookes, Ling Zha, Selvambigai Manivannan, Jana Kim, Merve Kocbiyik, Alice Fletcher, Caroline M. Gorvin, George Firth, Gilbert O. Fruhwirth, Juan P. Nicola, Sissy Jhiang, Matthew D. Ringel, Moray J. Campbell, Kavitha Sunassee, Philip J. Blower, Kristien Boelaert, Hannah R. Nieto, Vicki E. Smith, Christopher J. McCabe

Supplementary Figure S6

Funding

U.S. Department of Defense (DOD)

Wellcome Trust (WT)

Medical Research Council (MRC)

British Thyroid Foundation (BTF)

History

ARTICLE ABSTRACT

Patients with aggressive thyroid cancer are frequently failed by the central therapy of ablative radioiodide (RAI) uptake, due to reduced plasma membrane (PM) localization of the sodium/iodide symporter (NIS). We aimed to understand how NIS is endocytosed away from the PM of human thyroid cancer cells, and whether this was druggable in vivo. Informed by analysis of endocytic gene expression in patients with aggressive thyroid cancer, we used mutagenesis, NanoBiT interaction assays, cell surface biotinylation assays, RAI uptake, and NanoBRET to understand the mechanisms of NIS endocytosis in transformed cell lines and patient-derived human primary thyroid cells. Systemic drug responses were monitored via 99mTc pertechnetate gamma counting and gene expression in BALB/c mice. We identified an acidic dipeptide within the NIS C-terminus that mediates binding to the σ2 subunit of the Adaptor Protein 2 (AP2) heterotetramer. We discovered that the FDA-approved drug chloroquine (CQ) modulates NIS accumulation at the PM in a functional manner that is AP2 dependent. In vivo, CQ treatment of BALB/c mice significantly enhanced thyroidal uptake of 99mTc pertechnetate in combination with the histone deacetylase (HDAC) inhibitor vorinostat/SAHA, accompanied by increased thyroidal NIS mRNA. Bioinformatic analyses validated the clinical relevance of AP2 genes with disease-free survival in RAI-treated DTC, enabling construction of an AP2 gene–related risk score classifier for predicting recurrence. NIS internalization is specifically druggable in vivo. Our data, therefore, provide new translatable potential for improving RAI therapy using FDA-approved drugs in patients with aggressive thyroid cancer.See related commentary by Lechner and Brent, p. 1220