Rationale for therapeutic decision-making in locally advanced and metastatic radioactive iodine (RAI)-refractory differentiated thyroid cancer, starting from a clinical case

Cristina Alina Silaghi, Oana Stãnoiu-Pînzariu, Horaţiu Silaghi, Doina Piciu, Carmen Emanuela Georgescu

Abstract


Iodine uptake and organification are the hallmarks of thyroid cells differentiation. The loss of these characteristics in thyroid cancer leads to radioactive iodine refractoriness, a rare condition that bears a low survival rate and poor prognosis. We present a 52-year-old patient presenting dry cough and dyspnea in the supine position. Imaging examinations revealed a thyroid nodule with a high suspicion of malignancy in the right thyroid lobe, multiple laterocervical and mediastinal lymph nodes, lung, bone, and brain metastases. Fine needle aspiration cytologic features have advocated for papillary thyroid cancer (PTC). The patient underwent total thyroidectomy and selective lymphadenectomy. Subsequently, the patient received suppressive treatment with levothyroxine and four courses of radioactive iodine therapy. In addition, to treat bone and brain metastases, the patient experienced external radiotherapy and glucocorticoid therapy. Despite this rigorous therapeutic management, the patient obtained an incomplete structural and functional response. Although the last two posttherapeutic 131I whole-body scans were negative, the patient had elevated stimulated thyroglobulin levels and loco-regional recurrence by thyroid ultrasound. This aspect would suggest that thyroid cells become unable to uptake 131I, most likely through the emergence of new genetic mutations in the cancer cells. In conclusion, our patient's case suggests a 131I-refractory PTC, requiring the initiation of novel targeted systemic agents such as tyrosine kinase inhibitors, in order to improve structural and functional outcomes of radioactive iodine therapy and to afford prolonged progression-free survival advantage.

Keywords


papillary thyroid cancer; radioactive iodine-refractory; differentiated thyroid cancer; sodium/iodide symporter; tyrosine kinase inhibitor; brain metastasis

Full Text:

PDF

References


Haugen BR, Alexander EK, Bible KC, et al. 2015 American Thyroid Association Management Guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: The American Thyroid Association Guidelines Task Force on thyroid nodules and differentiated thyroid cancer. Thyroid. 2016; 26(1):1-133. https://doi.org/10.1089/thy.2015.0020

Brenner H. Long-term survival rates of cancer patients achieved by the end of the 20th century: a period analysis. Lancet. 2002; 360(9340):1131-1135. https://doi.org/10.1016/S0140-6736(02)11199-8

Durante C, Haddy N, Baudin E, et al. Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy. J Clin Endocrinol Metab. 2006; 91(8):2892-2899. https://doi.org/10.1210/jc.2005-2838

Henriques de Figueiredo B, Godbert Y, Soubeyran I, et al. Brain metastases from thyroid carcinoma: a retrospective study of 21 patients. Thyroid. 2014; 24(2):270-276. https://doi.org/10.1089/thy.2013.0061

Jayarangaiah A, Sidhu G, Brown J, et al. Therapeutic options for advanced thyroid cancer. Int J Clin Endocrinol Metab. 2019; 5(1):26-34. https://doi.org/10.17352/ijcem.000040

Stewart KE, Strachan MWJ, Srinivasan D, MacNeill M, Wall L, Nixon IJ. Tyrosine kinase inhibitor therapy in locally advanced differentiated thyroid cancer: a case report. Eur Thyroid J. 2019; 8(2):102-107. https://doi.org/10.1159/000494880

Szumowski P, Abdelrazek S, Iwanicka D, et al. Dosimetry during adjuvant 131I therapy in patients with differentiated thyroid cancer-clinical implications. Sci Rep. 2021; 11(1):13930. https://doi.org/10.1038/s41598-021-93431-1

Iñiguez-Ariza NM, Bible KC, Clarke BL. Bone metastases in thyroid cancer. J Bone Oncol. 2020; 21:100282. https://doi.org/10.1016/j.jbo.2020.100282

Dinneen SF, Valimaki MJ, Bergstralh EJ, Goellner JR, Gorman CA, Hay ID. Distant metastases in papillary thyroid carcinoma: 100 cases observed at one institution during 5 decades. J Clin Endocrinol Metab. 1995; 80(7):2041-2045. https://doi.org/10.1210/jcem.80.7.7608252

Ikekubo K, Hino M, Ito H, et al. Seven cases of brain metastasis from papillary thyroid carcinoma. Kaku Igaku. 2000; 37(4):349-357.

Tahmasebi FC, Farmer P, Powell SZ, et al. Brain metastases from papillary thyroid carcinomas. Virchows Arch. 2013; 462(4):473-480. https://doi.org/10.1007/s00428-013-1394-4

Osborne JR, Kondraciuk JD, Rice SL, et al. Thyroid cancer brain metastasis: survival and genomic characteristics of a large tertiary care cohort. Clin Nucl Med. 2019; 44(7):544-549. https://doi.org/10.1097/RLU.0000000000002618

Saito F, Uruno T, Shibuya H, et al. Prognosis after brain metastasis from differentiated thyroid carcinoma. World J Surg. 2016; 40(3):574-81. https://doi.org/10.1007/s00268-016-3405-5

Hirsch D, Gorshtein A, Robenshtok E, et al. Second radioiodine treatment: limited benefit for differentiated thyroid cancer with locoregional persistent disease. J Clin Endocrinol Metab. 2018; 103(2):469-476. https://doi.org/10.1210/jc.2017-01790

Fullmer T, Cabanillas ME, Zafereo M. Novel therapeutics in radioactive iodine-resistant thyroid cancer. Front Endocrinol (Lausanne). 2021; 12:720723. https://doi.org/10.3389/fendo.2021.720723

Borrelli N, Panebianco F, Condello V, et al. Characterization of activating mutations of the MEK1 gene in papillary thyroid carcinomas. Thyroid. 2019; 29(9):1279-1285. https://doi.org/10.1089/thy.2019.0065

Seo GH, Cho YY, Chung JH, Kim SW. Increased risk of leukemia after radioactive iodine therapy in patients with thyroid cancer: a nationwide, population-based study in Korea. Thyroid. 2015; 25(8):927-934. https://doi.org/10.1089/thy.2014.0557

Alsaud A, Mohamed S, Yassin MA, Ashour A, Obeidat K, Azrieh B. Acute myeloid leukemia after low-dose radioiodine therapy for papillary thyroid carcinoma. Case Rep Oncol. 2020; 13(1):207-211. https://doi.org/10.1159/000505686

Chakravarty D, Santos E, Ryder M, et al. Small- molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. J Clin Invest. 2011; 121(12):4700-4711. https://doi.org/10.1172/JCI46382

Zhang Z, Liu D, Murugan AK, Liu Z, Xing M. Histone deacetylation of NIS promoter underlies BRAF V600E-promoted NIS silencing in thyroid cancer. Endocr Relat Cancer. 2014; 21(2):161–173. https://doi.org/10.1530/ERC-13- 0399

Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet. 2014; 384(9940):319-328. https://doi.org/10.1016/S0140-6736(14)60421-9

Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med. 2015; 372:621-630 https://doi.org/10.1056/NEJMoa1406470

Pitoia F, Jerkovich F. Selective use of Sorafenib in the treatment of thyroid cancer. Drug Des Devel Ther. 2016; 10:1119–1131. https://doi.org/10.2147/DDDT. S82972

Cabanillas ME, de Souza JA, Geyer S, et al. Cabozantinib as salvage therapy for patients with tyrosine kinase inhibitor-refractory differentiated thyroid cancer: results of a multicenter phase II international thyroid oncology group trial. J Clin Oncol. 2017; 35(29):3315–3321. https://doi.org/10.1200/JCO.2017.73.0226

Cohen EE, Tortorici M, Kim S, Ingrosso A, Pithavala YK, Bycott P. A Phase II trial of axitinib in patients with various histologic subtypes of advanced thyroid cancer: long-term outcomes and pharmacokinetic/ pharmacodynamic analyses. Cancer Chemother Pharmacol. 2014; 74(6):1261–1270. https://doi.org/10.1007/s00280-014-2604-8

Bible KC, Suman VJ, Molina JR, et al. Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study. Lancet Oncol. 2010; 11(10):962–972. https://doi.org/10.1016/S1470-2045(10)70203-5

Yansong L, Yang H, Ding Y, et al. Donafenib in progressive locally advanced or metastatic radioactive iodine-refractory differentiated thyroid cancer: results of a randomized, multicenter, phase II trial. Thyroid. 2020; 31(4):607-615. https://doi.org/10.1089/thy.2020.0235

Jin N, Jiang T, Rosen DM, Nelkin BD, Ball DW. Synergistic action of a RAF inhibitor and a dual PI3K/mTOR inhibitor in thyroid cancer. Clin Cancer Res. 2011; 17(20):6482–6489. https://doi.org/10.1158/1078-0432.ccr-11-0933

Tsumagari K, Abd Elmageed ZY, Sholl AB, et al. Bortezomib sensitizes thyroid cancer to BRAF inhibitor in vitro and in vivo. Endocr Relat Cancer. 2017; 25(1):99–109. https://doi.org/10.1530/erc-17-0182

Kebebew E, Ghosh C, Kumar S, et al. A combinatorial strategy for targeting BRAF V600E mutant cancers with BRAF V600E inhibitor (PLX4720) and tyrosine kinase inhibitor (ponatinib). Clin Cancer Res. 2020; 26:2022-2036. https://doi.org/10.1158/1078-0432.ccr-19-1606

Ho AL, Grewal RK, Leboeuf R, et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. N Engl J Med. 2013; 368(7):623–632. https://doi.org/10.1056/nejmoa1209288

Rothenberg SM, McFadden DG, Palmer EL, Daniels GH, Wirth LJ. Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clin Cancer Res. 2014; 21(5):1028–1035. https://doi.org/10.1158/1078-0432.ccr-14-2915

Dunn LA, Sherman EJ. Baxi SS, et al. J Clin Endocrinol Metab. 2019; 104(5):1417-1428. https://doi.org/10.1210/jc.2018-01478

Jaber T, Waguespack SG, Cabanillas ME, et al. Targeted therapy in advanced thyroid cancer to resensitize tumors to radioactive iodine. J Clin Endocrinol Metab. 2018; 103(10): 3698–3705. https://doi.org/10.1210/jc.2018-00612




DOI: http://dx.doi.org/10.22551/2021.33.0804.10190

Copyright (c) 2021 Cristina Alina Silaghi, Oana Stãnoiu-Pînzariu, Horaţiu Silaghi, Doina Piciu, Carmen Emanuela Georgescu

Creative Commons License
Archive of Clinical Cases is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

 

ISSN: 2360-6975