Endocrinol Metab.  2019 Sep;34(3):215-225. 10.3803/EnM.2019.34.3.215.

Radioactive Iodine-Refractory Differentiated Thyroid Cancer and Redifferentiation Therapy

Affiliations
  • 1Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China. linys@pumch.cn
  • 2Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, China.
  • 3Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
  • 4Department of Oncology, Peking University International Hospital, Beijing, China. liangjun1959@aliyun.com

Abstract

The retained functionality of the sodium iodide symporter (NIS) expressed in differentiated thyroid cancer (DTC) cells allows the further utilization of post-surgical radioactive iodine (RAI) therapy, which is an effective treatment for reducing the risk of recurrence, and even the mortality, of DTC. Whereas, the dedifferentiation of DTC could influence the expression of functional NIS, thereby reducing the efficacy of RAI therapy in advanced DTC. Genetic alternations (such as BRAF and the rearranged during transfection [RET]/papillary thyroid cancer [PTC] rearrangement) have been widely reported to be prominently responsible for the onset, progression, and dedifferentiation of PTC, mainly through activating the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling cascades. These genetic alternations have been suggested to associate with the reduced expression of iodide-handling genes in thyroid cancer, especially the NIS gene, disabling iodine uptake and causing resistance to RAI therapy. Recently, novel and promising approaches aiming at various targets have been attempted to restore the expression of these iodine-metabolizing genes and enhance iodine uptake through in vitro studies and studies of RAI-refractory (RAIR)-DTC patients. In this review, we discuss the regulation of NIS, known mechanisms of dedifferentiation including the MAPK and PI3K pathways, and the current status of redifferentiation therapy for RAIR-DTC patients.

Keyword

Thyroid neoplasms; Sodium-iodide symporter; Isotopes

MeSH Terms

Humans
In Vitro Techniques
Iodine
Ion Transport
Isotopes
Mortality
Protein Kinases
Recurrence
Sodium Iodide
Thyroid Gland*
Thyroid Neoplasms*
Transfection
Iodine
Isotopes
Protein Kinases
Sodium Iodide

Figure

  • Fig. 1 Regulation of the sodium iodide symporter (NIS) upstream enhancer (NUE) at the transcriptional level in thyroid cells. TSHR, thyroid stimulating hormone receptor; AC, adenylyl cyclase; cAMP, cyclic adenosine monophosphate; PKA, protein kinase A; CRE, cAMP-response element; CREM, CRE-modulator; Ref1, apurinic apyrimidinic endonuclease redox effector factor-1; Pax8, paired box gene-8; TGFβ, transforming growth factor β; TLR4, Toll-like receptor 4; NF-κB, p65, a member of the class II nuclear factor κ-light-chain-enhancer of activated B cells, p65; PTTG1, pituitary tumor-transforming gene-1; PBF, PTTG1-binding factor.

  • Fig. 2 Known pathways involved in the regulation of sodium iodide symporter (NIS) in thyroid cancer. RTK, receptor tyrosine kinase; IGF-1, insulin-like growth factor-1; TGFβ, transforming growth factor β; PTC, papillary thyroid cancer; PI3K, phosphoinositide 3-kinase; RasGRP3, Ras guanyl releasing protein 3; PAX8, paired box gene-8; MEK, mitogen-activated extracellular signal-regulated kinase; ERK, extracellular regulated protein kinase; mTOR, mechanistic target of rapamycin; VEGFA, vascular endothelial growth factor A; MET, mesenchymal to epithelial transition factor; TSP1, thrombospondin 1; TIMP3, tissue inhibitor of metalloproteinases 3.


Cited by  1 articles

Treatment Effect of Combining Lenvatinib and Vemurafenib for BRAF Mutated Anaplastic Thyroid Cancer
Chae Moon Hong, Ji Min Oh, Prakash Gangadaran, Ramya Lakshmi Rajendran, Byeong-Cheol Ahn
Int J Thyroidol. 2021;14(2):127-134.    doi: 10.11106/ijt.2021.14.2.127.


Reference

1. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019; 144:1941–1953.
Article
2. Mao Y, Xing M. Recent incidences and differential trends of thyroid cancer in the USA. Endocr Relat Cancer. 2016; 23:313–322.
Article
3. Lim H, Devesa SS, Sosa JA, Check D, Kitahara CM. Trends in thyroid cancer incidence and mortality in the United States, 1974-2013. JAMA. 2017; 317:1338–1348.
Article
4. Durante C, Haddy N, Baudin E, Leboulleux S, Hartl D, Travagli JP, 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; 91:2892–2899.
Article
5. Xing M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer. 2013; 13:184–199.
Article
6. Zaballos MA, Santisteban P. Key signaling pathways in thyroid cancer. J Endocrinol. 2017; 235:R43–R61.
Article
7. Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer. 2005; 12:245–262.
Article
8. Venkateswaran A, Marsee DK, Green SH, Jhiang SM. Forskolin, 8-Br-3′,5′-cyclic adenosine 5′-monophosphate, and catalytic protein kinase A expression in the nucleus increase radioiodide uptake and sodium/iodide symporter protein levels in RET/PTC1-expressing cells. J Clin Endocrinol Metab. 2004; 89:6168–6172.
Article
9. Riesco-Eizaguirre G, Rodriguez I, De la Vieja A, Costamagna E, Carrasco N, Nistal M, et al. The BRAFV600E oncogene induces transforming growth factor beta secretion leading to sodium iodide symporter repression and increased malignancy in thyroid cancer. Cancer Res. 2009; 69:8317–8325.
10. Riesco-Eizaguirre G, Santisteban P. A perspective view of sodium iodide symporter research and its clinical implications. Eur J Endocrinol. 2006; 155:495–512.
Article
11. Castro MR, Bergert ER, Goellner JR, Hay ID, Morris JC. Immunohistochemical analysis of sodium iodide symporter expression in metastatic differentiated thyroid cancer: correlation with radioiodine uptake. J Clin Endocrinol Metab. 2001; 86:5627–5632.
Article
12. Wapnir IL, van de Rijn M, Nowels K, Amenta PS, Walton K, Montgomery K, et al. Immunohistochemical profile of the sodium/iodide symporter in thyroid, breast, and other carcinomas using high density tissue microarrays and conventional sections. J Clin Endocrinol Metab. 2003; 88:1880–1888.
Article
13. American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, et al. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid. 2009; 19:1167–1214.
Article
14. Tuttle RM, Ahuja S, Avram AM, Bernet VJ, Bourguet P, Daniels GH, et al. Controversies, consensus, and collaboration in the use of (131)I therapy in differentiated thyroid cancer: a joint statement from the American Thyroid Association, the European Association of Nuclear Medicine, the Society of Nuclear Medicine and Molecular Imaging, and the European Thyroid Association. Thyroid. 2019; 29:461–470.
Article
15. Taki K, Kogai T, Kanamoto Y, Hershman JM, Brent GA. A thyroid-specific far-upstream enhancer in the human sodium/iodide symporter gene requires Pax-8 binding and cyclic adenosine 3′,5′-monophosphate response element-like sequence binding proteins for full activity and is differentially regulated in normal and thyroid cancer cells. Mol Endocrinol. 2002; 16:2266–2282.
Article
16. Costamagna E, Garcia B, Santisteban P. The functional interaction between the paired domain transcription factor Pax8 and Smad3 is involved in transforming growth factor-beta repression of the sodium/iodide symporter gene. J Biol Chem. 2004; 279:3439–3446.
17. Kambe F, Nomura Y, Okamoto T, Seo H. Redox regulation of thyroid-transcription factors, Pax-8 and TTF-1, is involved in their increased DNA-binding activities by thyrotropin in rat thyroid FRTL-5 cells. Mol Endocrinol. 1996; 10:801–812.
Article
18. Tell G, Pellizzari L, Cimarosti D, Pucillo C, Damante G. Ref-1 controls pax-8 DNA-binding activity. Biochem Biophys Res Commun. 1998; 252:178–183.
Article
19. Mansouri A, Chowdhury K, Gruss P. Follicular cells of the thyroid gland require Pax8 gene function. Nat Genet. 1998; 19:87–90.
Article
20. Chun JT, Di Dato V, D'Andrea B, Zannini M, Di Lauro R. The CRE-like element inside the 5′-upstream region of the rat sodium/iodide symporter gene interacts with diverse classes of b-Zip molecules that regulate transcriptional activities through strong synergy with Pax-8. Mol Endocrinol. 2004; 18:2817–2829.
Article
21. Fenton MS, Marion KM, Hershman JM. Identification of cyclic adenosine 3′,5′-monophosphate response element modulator as an activator of the human sodium/iodide symporter upstream enhancer. Endocrinology. 2008; 149:2592–2606.
Article
22. Nicola JP, Velez ML, Lucero AM, Fozzatti L, Pellizas CG, Masini-Repiso AM. Functional toll-like receptor 4 conferring lipopolysaccharide responsiveness is expressed in thyroid cells. Endocrinology. 2009; 150:500–508.
Article
23. Nicola JP, Nazar M, Mascanfroni ID, Pellizas CG, Masini-Repiso AM. NF-kappaB p65 subunit mediates lipopolysaccharide-induced Na(+)/I(−) symporter gene expression by involving functional interaction with the paired domain transcription factor Pax8. Mol Endocrinol. 2010; 24:1846–1862.
24. Boelaert K, Smith VE, Stratford AL, Kogai T, Tannahill LA, Watkinson JC, et al. PTTG and PBF repress the human sodium iodide symporter. Oncogene. 2007; 26:4344–4356.
Article
25. Stratford AL, Boelaert K, Tannahill LA, Kim DS, Warfield A, Eggo MC, et al. Pituitary tumor transforming gene binding factor: a novel transforming gene in thyroid tumorigenesis. J Clin Endocrinol Metab. 2005; 90:4341–4349.
Article
26. Saez C, Martinez-Brocca MA, Castilla C, Soto A, Navarro E, Tortolero M, et al. Prognostic significance of human pituitary tumor-transforming gene immunohistochemical expression in differentiated thyroid cancer. J Clin Endocrinol Metab. 2006; 91:1404–1409.
27. Kogai T, Brent GA. The sodium iodide symporter (NIS): regulation and approaches to targeting for cancer therapeutics. Pharmacol Ther. 2012; 135:355–370.
Article
28. Xing M. BRAF mutation in papillary thyroid cancer: pathogenic role, molecular bases, and clinical implications. Endocr Rev. 2007; 28:742–762.
Article
29. Cohen Y, Xing M, Mambo E, Guo Z, Wu G, Trink B, et al. BRAF mutation in papillary thyroid carcinoma. J Natl Cancer Inst. 2003; 95:625–627.
Article
30. Liu D, Liu Z, Condouris S, Xing M. BRAF V600E maintains proliferation, transformation, and tumorigenicity of BRAF-mutant papillary thyroid cancer cells. J Clin Endocrinol Metab. 2007; 92:2264–2271.
Article
31. Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA. High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 2003; 63:1454–1457.
32. Soares P, Trovisco V, Rocha AS, Lima J, Castro P, Preto A, et al. BRAF mutations and RET/PTC rearrangements are alternative events in the etiopathogenesis of PTC. Oncogene. 2003; 22:4578–4580.
Article
33. Kim TH, Park YJ, Lim JA, Ahn HY, Lee EK, Lee YJ, et al. The association of the BRAF(V600E) mutation with prognostic factors and poor clinical outcome in papillary thyroid cancer: a meta-analysis. Cancer. 2012; 118:1764–1773.
34. Giordano TJ, Kuick R, Thomas DG, Misek DE, Vinco M, Sanders D, et al. Molecular classification of papillary thyroid carcinoma: distinct BRAF, RAS, and RET/PTC mutation-specific gene expression profiles discovered by DNA microarray analysis. Oncogene. 2005; 24:6646–6656.
Article
35. Riesco-Eizaguirre G, Gutierrez-Martinez P, Garcia-Cabezas MA, Nistal M, Santisteban P. The oncogene BRAF V600E is associated with a high risk of recurrence and less differentiated papillary thyroid carcinoma due to the impairment of Na+/I− targeting to the membrane. Endocr Relat Cancer. 2006; 13:257–269.
36. Mian C, Barollo S, Pennelli G, Pavan N, Rugge M, Pelizzo MR, et al. Molecular characteristics in papillary thyroid cancers (PTCs) with no 131I uptake. Clin Endocrinol (Oxf). 2008; 68:108–116.
37. Xing M, Westra WH, Tufano RP, Cohen Y, Rosenbaum E, Rhoden KJ, et al. BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J Clin Endocrinol Metab. 2005; 90:6373–6379.
Article
38. Ricarte-Filho JC, Ryder M, Chitale DA, Rivera M, Heguy A, Ladanyi M, et al. Mutational profile of advanced primary and metastatic radioactive iodine-refractory thyroid cancers reveals distinct pathogenetic roles for BRAF, PIK3CA, and AKT1. Cancer Res. 2009; 69:4885–4893.
Article
39. Barollo S, Pennelli G, Vianello F, Watutantrige Fernando S, Negro I, Merante Boschin I, et al. BRAF in primary and recurrent papillary thyroid cancers: the relationship with (131)I and 2-[(18)F]fluoro-2-deoxy-D-glucose uptake ability. Eur J Endocrinol. 2010; 163:659–663.
Article
40. Yang K, Wang H, Liang Z, Liang J, Li F, Lin Y. BRAFV600E mutation associated with non-radioiodine-avid status in distant metastatic papillary thyroid carcinoma. Clin Nucl Med. 2014; 39:675–679.
Article
41. Barneda-Zahonero B, Parra M. Histone deacetylases and cancer. Mol Oncol. 2012; 6:579–589.
Article
42. Horikoshi M. Histone acetylation: from code to web and router via intrinsically disordered regions. Curr Pharm Des. 2013; 19:5019–5042.
Article
43. Hou P, Bojdani E, Xing M. Induction of thyroid gene expression and radioiodine uptake in thyroid cancer cells by targeting major signaling pathways. J Clin Endocrinol Metab. 2010; 95:820–828.
Article
44. Durante C, Puxeddu E, Ferretti E, Morisi R, Moretti S, Bruno R, et al. BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism. J Clin Endocrinol Metab. 2007; 92:2840–2843.
Article
45. Di Cristofaro J, Silvy M, Lanteaume A, Marcy M, Carayon P, De Micco C. Expression of tpo mRNA in thyroid tumors: quantitative PCR analysis and correlation with alterations of ret, Braf, ras and pax8 genes. Endocr Relat Cancer. 2006; 13:485–495.
46. 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; 21:161–173.
Article
47. Jo YS, Li S, Song JH, Kwon KH, Lee JC, Rha SY, et al. Influence of the BRAF V600E mutation on expression of vascular endothelial growth factor in papillary thyroid cancer. J Clin Endocrinol Metab. 2006; 91:3667–3670.
Article
48. Nucera C, Porrello A, Antonello ZA, Mekel M, Nehs MA, Giordano TJ, et al. B-Raf(V600E) and thrombospondin-1 promote thyroid cancer progression. Proc Natl Acad Sci U S A. 2010; 107:10649–10654.
Article
49. Knauf JA, Sartor MA, Medvedovic M, Lundsmith E, Ryder M, Salzano M, et al. Progression of BRAF-induced thyroid cancer is associated with epithelial-mesenchymal transition requiring concomitant MAP kinase and TGFβ signaling. Oncogene. 2011; 30:3153–3162.
Article
50. Hu S, Liu D, Tufano RP, Carson KA, Rosenbaum E, Cohen Y, et al. Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. Int J Cancer. 2006; 119:2322–2329.
Article
51. Liu R, Xing M. TERT promoter mutations in thyroid cancer. Endocr Relat Cancer. 2016; 23:R143–R155.
Article
52. Alzahrani AS, Alsaadi R, Murugan AK, Sadiq BB. TERT promoter mutations in thyroid cancer. Horm Cancer. 2016; 7:165–177.
Article
53. Sohn SY, Park WY, Shin HT, Bae JS, Ki CS, Oh YL, et al. Highly concordant key genetic alterations in primary tumors and matched distant metastases in differentiated thyroid cancer. Thyroid. 2016; 26:672–682.
Article
54. Bu R, Siraj AK, Divya SP, Kong Y, Parvathareddy SK, Al-Rasheed M, et al. Telomerase reverse transcriptase mutations are independent predictor of disease-free survival in Middle Eastern papillary thyroid cancer. Int J Cancer. 2018; 142:2028–2039.
Article
55. Liu R, Bishop J, Zhu G, Zhang T, Ladenson PW, Xing M. Mortality risk stratification by combining BRAF V600E and TERT promoter mutations in papillary thyroid cancer: genetic duet of BRAF and TERT promoter mutations in thyroid cancer mortality. JAMA Oncol. 2017; 3:202–208.
56. Yang X, Li J, Li X, Liang Z, Gao W, Liang J, et al. TERT promoter mutation predicts radioiodine-refractory character in distant metastatic differentiated thyroid cancer. J Nucl Med. 2017; 58:258–265.
Article
57. Liu R, Zhang T, Zhu G, Xing M. Regulation of mutant TERT by BRAF V600E/MAP kinase pathway through FOS/GABP in human cancer. Nat Commun. 2018; 9:579.
Article
58. Trapasso F, Iuliano R, Chiefari E, Arturi F, Stella A, Filetti S, et al. Iodide symporter gene expression in normal and transformed rat thyroid cells. Eur J Endocrinol. 1999; 140:447–451.
Article
59. Wang J, Knauf JA, Basu S, Puxeddu E, Kuroda H, Santoro M, et al. Conditional expression of RET/PTC induces a weak oncogenic drive in thyroid PCCL3 cells and inhibits thyrotropin action at multiple levels. Mol Endocrinol. 2003; 17:1425–1436.
Article
60. Xing M. Genetic alterations in the phosphatidylinositol-3 kinase/Akt pathway in thyroid cancer. Thyroid. 2010; 20:697–706.
Article
61. Song J, Qiu W, Deng X, Qiu Z, Fan Y, Yang Z. A somatic mutation of RasGRP3 decreases Na(+)/I(−) symporter expression in metastases of radioactive iodine-refractory thyroid cancer by stimulating the Akt signaling pathway. Am J Cancer Res. 2018; 8:1847–1855.
62. Kimura T, Van Keymeulen A, Golstein J, Fusco A, Dumont JE, Roger PP. Regulation of thyroid cell proliferation by TSH and other factors: a critical evaluation of in vitro models. Endocr Rev. 2001; 22:631–656.
Article
63. Garcia B, Santisteban P. PI3K is involved in the IGF-I inhibition of TSH-induced sodium/iodide symporter gene expression. Mol Endocrinol. 2002; 16:342–352.
Article
64. Kogai T, Sajid-Crockett S, Newmarch LS, Liu YY, Brent GA. Phosphoinositide-3-kinase inhibition induces sodium/iodide symporter expression in rat thyroid cells and human papillary thyroid cancer cells. J Endocrinol. 2008; 199:243–252.
Article
65. de Souza EC, Padron AS, Braga WM, de Andrade BM, Vaisman M, Nasciutti LE, et al. MTOR downregulates iodide uptake in thyrocytes. J Endocrinol. 2010; 206:113–120.
66. Short SC, Suovuori A, Cook G, Vivian G, Harmer C. A phase II study using retinoids as redifferentiation agents to increase iodine uptake in metastatic thyroid cancer. Clin Oncol (R Coll Radiol). 2004; 16:569–574.
Article
67. Kebebew E, Lindsay S, Clark OH, Woeber KA, Hawkins R, Greenspan FS. Results of rosiglitazone therapy in patients with thyroglobulin-positive and radioiodine-negative advanced differentiated thyroid cancer. Thyroid. 2009; 19:953–956.
Article
68. Sherman EJ, Su YB, Lyall A, Schoder H, Fury MG, Ghossein RA, et al. Evaluation of romidepsin for clinical activity and radioactive iodine reuptake in radioactive iodine-refractory thyroid carcinoma. Thyroid. 2013; 23:593–599.
Article
69. Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. N Engl J Med. 2013; 368:623–632.
Article
70. Dunn LA, Sherman EJ, Baxi SS, Tchekmedyian V, Grewal RK, Larson SM, et al. Vemurafenib redifferentiation of BRAF mutant, RAI-refractory thyroid cancers. J Clin Endocrinol Metab. 2019; 104:1417–1428.
Article
71. Cheng L, Jin Y, Liu M, Ruan M, Chen L. HER inhibitor promotes BRAF/MEK inhibitor-induced redifferentiation in papillary thyroid cancer harboring BRAFV600E. Oncotarget. 2017; 8:19843–19854.
Article
72. Schmutzler C, Schmitt TL, Glaser F, Loos U, Kohrle J. The promoter of the human sodium/iodide-symporter gene responds to retinoic acid. Mol Cell Endocrinol. 2002; 189:145–155.
Article
73. Grunwald F, Menzel C, Bender H, Palmedo H, Otte R, Fimmers R, et al. Redifferentiation therapy-induced radioiodine uptake in thyroid cancer. J Nucl Med. 1998; 39:1903–1906.
74. Simon D, Koehrle J, Reiners C, Boerner AR, Schmutzler C, Mainz K, et al. Redifferentiation therapy with retinoids: therapeutic option for advanced follicular and papillary thyroid carcinoma. World J Surg. 1998; 22:569–574.
Article
75. Simon D, Korber C, Krausch M, Segering J, Groth P, Gorges R, et al. Clinical impact of retinoids in redifferentiation therapy of advanced thyroid cancer: final results of a pilot study. Eur J Nucl Med Mol Imaging. 2002; 29:775–782.
Article
76. Kim WG, Kim EY, Kim TY, Ryu JS, Hong SJ, Kim WB, et al. Redifferentiation therapy with 13-cis retinoic acids in radioiodine-resistant thyroid cancer. Endocr J. 2009; 56:105–112.
Article
77. Courbon F, Zerdoud S, Bastie D, Archambaud F, Hoff M, Eche N, et al. Defective efficacy of retinoic acid treatment in patients with metastatic thyroid carcinoma. Thyroid. 2006; 16:1025–1031.
Article
78. Corton JC, Anderson SP, Stauber A. Central role of peroxisome proliferator-activated receptors in the actions of peroxisome proliferators. Annu Rev Pharmacol Toxicol. 2000; 40:491–518.
Article
79. Roberts-Thomson SJ. Peroxisome proliferator-activated receptors in tumorigenesis: targets of tumour promotion and treatment. Immunol Cell Biol. 2000; 78:436–441.
Article
80. Frohlich E, Machicao F, Wahl R. Action of thiazolidinediones on differentiation, proliferation and apoptosis of normal and transformed thyrocytes in culture. Endocr Relat Cancer. 2005; 12:291–303.
Article
81. Park JW, Zarnegar R, Kanauchi H, Wong MG, Hyun WC, Ginzinger DG, et al. Troglitazone, the peroxisome proliferator-activated receptor-gamma agonist, induces antiproliferation and redifferentiation in human thyroid cancer cell lines. Thyroid. 2005; 15:222–231.
82. Kitazono M, Robey R, Zhan Z, Sarlis NJ, Skarulis MC, Aikou T, et al. Low concentrations of the histone deacetylase inhibitor, depsipeptide (FR901228), increase expression of the Na(+)/I(−) symporter and iodine accumulation in poorly differentiated thyroid carcinoma cells. J Clin Endocrinol Metab. 2001; 86:3430–3435.
Article
83. Kelly WK, O'Connor OA, Krug LM, Chiao JH, Heaney M, Curley T, et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol. 2005; 23:3923–3931.
Article
84. Amiri-Kordestani L, Luchenko V, Peer CJ, Ghafourian K, Reynolds J, Draper D, et al. Phase I trial of a new schedule of romidepsin in patients with advanced cancers. Clin Cancer Res. 2013; 19:4499–4507.
Article
85. Nilubol N, Merkel R, Yang L, Patel D, Reynolds JC, Sadowski SM, et al. A phase II trial of valproic acid in patients with advanced, radioiodine-resistant thyroid cancers of follicular cell origin. Clin Endocrinol (Oxf). 2017; 86:128–133.
Article
86. Liu D, Hu S, Hou P, Jiang D, Condouris S, Xing M. Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant. Clin Cancer Res. 2007; 13:1341–1349.
Article
87. Hayes DN, Lucas AS, Tanvetyanon T, Krzyzanowska MK, Chung CH, Murphy BA, et al. Phase II efficacy and pharmacogenomic study of Selumetinib (AZD6244; ARRY-142886) in iodine-131 refractory papillary thyroid carcinoma with or without follicular elements. Clin Cancer Res. 2012; 18:2056–2065.
Article
88. 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. 2015; 21:1028–1035.
Article
89. Furuya F, Shimura H, Suzuki H, Taki K, Ohta K, Haraguchi K, et al. Histone deacetylase inhibitors restore radioiodide uptake and retention in poorly differentiated and anaplastic thyroid cancer cells by expression of the sodium/iodide symporter thyroperoxidase and thyroglobulin. Endocrinology. 2004; 145:2865–2875.
Article
90. Pugliese M, Fortunati N, Germano A, Asioli S, Marano F, Palestini N, et al. Histone deacetylase inhibition affects sodium iodide symporter expression and induces 131I cytotoxicity in anaplastic thyroid cancer cells. Thyroid. 2013; 23:838–846.
Article
91. Fu H, Cheng L, Jin Y, Cheng L, Liu M, Chen L. MAPK inhibitors enhance HDAC inhibitor-induced redifferentiation in papillary thyroid cancer cells harboring BRAF (V600E): an in vitro study. Mol Ther Oncolytics. 2019; 12:235–245.
92. Nagarajah J, Le M, Knauf JA, Ferrandino G, Montero-Conde C, Pillarsetty N, et al. Sustained ERK inhibition maximizes responses of BrafV600E thyroid cancers to radioiodine. J Clin Invest. 2016; 126:4119–4124.
Article
93. Liu YY, Zhang X, Ringel MD, Jhiang SM. Modulation of sodium iodide symporter expression and function by LY294002, Akti-1/2 and rapamycin in thyroid cells. Endocr Relat Cancer. 2012; 19:291–304.
Article
94. Plantinga TS, Heinhuis B, Gerrits D, Netea MG, Joosten LA, Hermus AR, et al. mTOR inhibition promotes TTF1-dependent redifferentiation and restores iodine uptake in thyroid carcinoma cell lines. J Clin Endocrinol Metab. 2014; 99:E1368–E1375.
Article
95. Montero-Conde C, Ruiz-Llorente S, Dominguez JM, Knauf JA, Viale A, Sherman EJ, et al. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov. 2013; 3:520–533.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr