Warning: fopen(/home/virtual/enm-kes/journal/upload/ip_log/ip_log_2024-03.txt): failed to open stream: Permission denied in /home/virtual/lib/view_data.php on line 88 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 89 Molecular Pathogenesis and Targeted Therapies in Well-Differentiated Thyroid Carcinoma
Skip Navigation
Skip to contents

Endocrinol Metab : Endocrinology and Metabolism

clarivate
OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > Endocrinol Metab > Volume 29(3); 2014 > Article
Review Article
Molecular Pathogenesis and Targeted Therapies in Well-Differentiated Thyroid Carcinoma
Jung Guk Kim
Endocrinology and Metabolism 2014;29(3):211-216.
DOI: https://doi.org/10.3803/EnM.2014.29.3.211
Published online: September 25, 2014

Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea.

Corresponding author: Jung Guk Kim. Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Jung-gu, Daegu 700-842, Korea. Tel: +82-53-420-5566, Fax: +82-53-426-2046, jugkim@knu.ac.kr

Copyright © 2014 Korean Endocrine Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 3,487 Views
  • 35 Download
  • 12 Web of Science
  • 10 Crossref
  • 12 Scopus
  • Four proto-oncogenes commonly associated with well-differentiated thyroid carcinoma, rearranged during transfection (RET)/papillary thyroid cancer, BRAF, RAS, and PAX8/peroxisome proliferator activated receptor-γ, may carry diagnostic and prognostic significance. These oncogenes can be used to improve the diagnosis and management of well-differentiated thyroid carcinoma. Limited therapeutic options are available for patients with metastatic well-differentiated thyroid cancer, necessitating the development of novel therapies. Vascular endothelial growth factor (VEGF)- and RET-directed therapies such as sorafenib, motesanib, and sunitinib have been shown to be the most effective at inducing clinical responses and stabilizing the disease process. Further clinical trials of these therapeutic agents may soon change the management of thyroid cancer.
Remarkable advances have occurred in recent years in understanding the molecular biology of thyroid cancer. Various somatic mutations associated with the development and progression of thyroid carcinomas have been discovered [1,2,3,4]. Our rapidly expanding knowledge of the molecular genetics of thyroid cancer has shifted into clinical practice, offering better diagnostic accuracy and enabling the development of novel compounds for the treatment of advanced metastatic thyroid carcinoma [5,6,7,8]. As the most common and challenging problem in thyroid carcinoma is related to the diagnosis and prognostication of well-differentiated thyroid cancer (WDTC), this review will focus on the genetic abnormalities found in WDTC and their diagnostic and prognostic use, as well as on therapies targeted to specific mutated sites and kinase pathways.
Chromosomal rearrangements (RET/PTC)
The rearranged during transfection (RET) proto-oncogene is located on chromosome 10q11-12 and encodes a transmembrane receptor with a tyrosine kinase domain. Because it was first found only in papillary thyroid cancer (PTC), it is called RET/PTC [9]. Although more than ten rearrangements have been identified, RET/PTC1 and RET/PTC3 account for most of the rearrangements found in PTC [10]. The RET/PTC chimeric oncogenic protein leads to the activation of several downstream pathways, including mitogen-activated protein kinase (MAPK), extracellular signal-related kinase (ERK), and phosphatidylinositol 3-kinase (PI3K). The prevalence of RET/PTC rearrangements in sporadic papillary carcinoma is approximately 15% to 40% [11]. The RET/PTC rearrangement has also been detected in the follicular variant of PTC (FVPTC) [9]. Clinically, RET/PTC variants are often found in radiation-associated PTC. Following the Chernobyl nuclear plant explosion in 1986, there was an increase in pediatric PTC and an increase in the incidence of an aggressive solid variant of PTC that contained the RET/PTC3 rearrangement. A form of PTC with a later onset, with a more classical phenotype and clinical course, emerged in the Chernobyl survivors and contained RET/PTC1 [12].
RAS mutations
The Rat sarcoma (RAS) oncogene families regulate two important signaling pathways in thyroid cancer, the MAP kinase cascade (Ras-Raf-MEK-ERK) and the PI3K/Akt signaling pathway. Mutations of H-RAS codon 61 and N-RAS codon 61 are the most common RAS mutations associated with thyroid cancer [13]. RAS mutations occur in both benign tumors and thyroid cancers (both PTC and follicular thyroid cancer [FTC]), with variable frequency in anaplastic thyroid cancers. PTC with RAS mutations often display a follicular architecture and are called FVPTC. PTCs carrying a RAS mutation tend to exhibit a lower rate of lymph node metastasis and are more likely to be encapsulated [14].
BRAF and the MAP kinase pathway
The MAP kinase pathway mediates cellular responses to growth signals [4]. The MAPK pathway consists of a three-tiered protein kinase cascade: an extracellular signal (growth factor) interacts with a receptor tyrosine kinase, which then stimulates the activity of a MAPK kinase kinase (a MAPKKK, e.g., Raf) via the G-protein Ras, which activates a MAPKK (e.g., MEK), which in turn activates a MAPK (e.g., ERK). ERK is able to phosphorylate a number of proteins that regulate cytoskeletal organization, metabolism, chromatin remodeling, and numerous transcription factors (Fig. 1). There are three kinds of Raf proteins: A-Raf, B-Raf, and C-Raf (or Raf-1). Activation of B-Raf occurs after binding to Ras alone, whereas for activation of A-Raf and C-Raf, additional signals are required [15]. B-Raf is therefore the most potent activator of downstream effector MEK kinases in the Raf-Raf-MEK-ERK pathway. Mutations in the MAP kinase pathway are found in approximately 30% of all cancers, while around 8% of cancers develop due to BRAF mutations [16,17].
BRAF mutations were first discovered in thyroid cancer in 2003 [1]. A missense mutation in exon 15, which occurs due to the substitution of the amino acid valine for glutamic acid at residue position 600 [16], is present in 36% to 69% of PTC cases and is the most frequent genetic change in PTC [18,19]. The V600E mutation comprises more than 90% of observed BRAF mutations. In subtypes of PTC, the highest rate of BRAF mutation (77%) is found in the tall cell variant of papillary cancer, and the lowest percentage (12%) is found in the FVPTC. In PTC, BRAF mutation is more frequent in older patients, and is associated with extrathyroidal invasion, advanced stage, and worse clinical outcome, including greater rates of recurrence and treatment failure [20,21,22,23]. BRAF mutation is responsible for the suppression of the sodium/iodide symporter (NIS), which is involved in iodine metabolism [24]. By contrast, others have found that the BRAF mutation is not associated with age, gender, multicentricity, recurrence rate, lymphovascular invasion, extranodal extension, central neck involvement, advanced stage (stage III or IV), or distant metastasis [25,26,27].
PAX8/PPAR
The most common genetic abnormalities involved in follicular thyroid carcinoma are deletions, partial deletions, and deletion rearrangements of the short arm of chromosome 3. The PAX8/PPAR fusion oncogene occurs via a balanced translocation between chromosomes 2 and 3 that results in the fusion of the DNA-binding domains of the thyroid transcription factor PAX8 to the peroxisome proliferator activated receptor (PPARγ) [3]. The PAX8/PPAR fusion protein (PPFP) is detected in 36.5% of FTCs (range, 11% to 63%), 9.5% of follicular adenomas, and 13.2% of FVPTCs [28]. This chimeric protein may retard the growth inhibition and follicular differentiation normally induced by PPARγ, resulting in tumorigenesis. PPFP is not the only PPARγ fusion mutation that occurs in FTC. Recently, the CREB3L2-PPARγ fusion, which results from a t(3;7)(p25;q34) chromosomal rearrangement, was identified [29].
The treatment of patients with well-differentiated thyroid carcinoma is based on surgery, high-dose radioactive iodine (RAI), and thyroid hormone therapy. However, for patients with advanced, metastatic thyroid carcinoma, treatment options after radioiodine and thyroid hormone therapy have been limited. In patients with radioiodine-resistant disease, cytotoxic chemotherapy yields low response rates of short duration, is often associated with considerable toxicity, and does not prolong survival.
Recently, antiangiogenic therapy has been investigated for the treatment of advanced thyroid cancer. Investigational targeted therapies for WDTC include agents targeting the VEGF pathway and those targeting multiple kinases such as the MAP kinase and PI3 kinase pathways (Table 1) [6]. These inhibitors are typically administered orally and, because of the targeting similarities, common toxicities exist among these agents, including hypertension, diarrhea, skin rashes, and fatigue.
Sorafenib
Sorafenib is an oral tyrosine kinase inhibitor (TKI) targeting B-Raf and VEGF receptors 2 and 3. The optimal therapeutic dose of sorafenib is 400 mg twice daily [5]. In a phase II trial, 58 WDTC patients with RAI failure were treated with sorafenib over a 10-month period. Of 41 PTC patients, six had a partial response (PR) and 23 had stable disease lasting longer than 6 months. The median duration of the PR was 7.5 months, the median progression-free survival (PFS) was 15 months, and the thyroglobulin of 77% of PTC patients declined more than 25%. No PRs were noted among non-PTC patients [30]. Another phase II study evaluated the effect of 26 weeks of sorafenib therapy on 31 WDTC patients with metastatic or locally advanced, radioiodine-refractory disease. At the study end, 25% of patients had a PR, 34% had stable disease, and the PFS was 58 weeks. Of 21 patients who underwent radioiodine imaging after 26 weeks of treatment, none had induction of uptake in metastatic lesions [7].
Motesanib
Motesanib is also an oral TKI targeting VEGF receptors 1, 2, and 3. In an open-label, phase II study, 57 PTC patients who had progressive, locally advanced or metastatic, radioiodine-resistant WDTC were treated with 125 mg of motesanib administered orally once daily. The objective response rate was 14%, and stable disease was achieved in 67% of the patients and maintained for 24 weeks or longer in 35%. The median duration of the response was 32 weeks and the median PFS was 40 weeks. Serum thyroglobulin concentrations decreased during treatment in 81% of patients [31].
Sunitinib
Sunitinib is approved for the treatment of renal cell carcinoma and is a TKI targeting all three VEGF receptors, RET, and RET/PTC subtypes 1 and 3. Prolonged PRs have been described in three patients (with PTC, FTC, and MTC, respectively) treated with 50 mg of sunitinib daily for 28 days followed by 14 days without treatment per cycle. Fluoro-2-deoxy-D-glucose uptake by positron emission tomography imaging was markedly reduced in the WDTC patients [32]. An open-label phase II trial with sunitinib in 35 patients with progressive WDTC or MTC reported a PR in 13% of 31 WDTC patients, and disease stabilization in 68% of WDTC and 83% of MTC patients [33].
Axitinib and pazopanib
Although the above TKIs targeting the VEGF receptors also inhibit RET, axitinib, and pazopanib do not have any anti-RET activity [34,35]. Axitinib and pazopanib are efficacious in thyroid carcinoma despite the absence of inhibitory activity against RET or other mutated kinases that are oncogenic in thyroid carcinoma, suggesting that VEGF receptor-mediated angiogenesis is likely the primary mechanism by which the other anti-VEGF receptor inhibitory agents function.
Thalidomide
Thalidomide was found to inhibit angiogenesis, but the exact mechanism by which thalidomide exerts its antiangiogenic effects remains unknown. An open-label, phase II trial was initiated to examine the efficacy of thalidomide in 36 patients with progressive, metastatic thyroid carcinoma [36]. Starting at 200 mg daily, the dose of thalidomide was progressively increased as tolerated, with a median maximum daily dose of 600 mg. PRs were achieved in 18% of patients, and 32% had stable disease as their best response. Common adverse events included somnolence, peripheral neuropathy, constipation, dizziness, and infection.
Celecoxib
Cyclooxygenase-2 (COX-2) is overexpressed in many cancers and promotes tumor development and progression. COX-2 mRNA and protein levels are increased in thyroid cancer compared with nonneoplastic and benign thyroid tissues, especially those expressing RET/PTC mutations. One phase II trial was performed in 32 patients with progressive WDTC treated with celecoxib 400 mg orally twice a day for 12 months. One patient had a PR, and one remained stable on therapy for >12 months, but most patients progressed despite treatment. The study was terminated as a result of a lack of efficacy combined with increasing concern about cardiovascular toxicity from COX-2 inhibitors [37].
Rosiglitazone
Rosiglitazone, a PPARγ agonist, has been shown to induce differentiation, cell cycle arrest, and apoptosis in a variety of human cancers, including thyroid carcinoma. Ten patients with radioiodine scan-negative metastatic WDTC were treated with rosiglitazone at 4 mg daily for 1 week, and then 8 mg daily for 7 weeks. Of the ten patients, four had positive radioiodine scans after rosiglitazone therapy, with uptake in the neck in three patients and in the pelvis in one patient. The serum thyroglobulin level decreased in two patients, increased in five patients, and was stable in three patients. No patient developed clinically important toxicity associated with rosiglitazone treatment.
Papillary thyroid carcinomas, follicular thyroid carcinomas, and their subtypes represent the majority of WDTCs. Mutations in RET/PTC, BRAF, RAS, and PAX8/PPARγ are commonly associated with well-differentiated thyroid carcinoma and may carry diagnostic and prognostic significance. These molecular markers are reflected in the Revised Management Guidelines for Patients with Thyroid Nodules and Differentiated Thyroid Cancer, published by the American Thyroid Association [38]. VEGF- and RET-directed therapies such as sorafenib, motesanib, and sunitinib, novel therapeutic agents targeting the above genetic mutations, have been shown to be the most effective at inducing clinical responses and stabilizing the disease process. In the near future, targeted therapy and the use of tumor biomarkers for predicting responses will be the next step in improving the survival of a disease that not long ago was untreatable.

No potential conflict of interest relevant to this article was reported.

  • 1. Cohen Y, Xing M, Mambo E, Guo Z, Wu G, Trink B, Beller U, Westra WH, Ladenson PW, Sidransky D. BRAF mutation in papillary thyroid carcinoma. J Natl Cancer Inst 2003;95:625–627. ArticlePubMedPDF
  • 2. Fusco A, Santoro M, Grieco M, Carlomagno F, Dathan N, Fabien N, Berlingieri MT, Li Z, De Franciscis V, Salvatore D, Melillo RM, Portella G, Cerrato A, Colantuoni V, Vecchio G. RET/PTC activation in human thyroid carcinomas. J Endocrinol Invest 1995;18:127–129. ArticlePubMedPDF
  • 3. Kroll TG, Sarraf P, Pecciarini L, Chen CJ, Mueller E, Spiegelman BM, Fletcher JA. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma [corrected]. Science 2000;289:1357–1360. ArticlePubMed
  • 4. Hilger RA, Scheulen ME, Strumberg D. The Ras-Raf-MEK-ERK pathway in the treatment of cancer. Onkologie 2002;25:511–518. ArticlePubMed
  • 5. Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M, Cao Y, Shujath J, Gawlak S, Eveleigh D, Rowley B, Liu L, Adnane L, Lynch M, Auclair D, Taylor I, Gedrich R, Voznesensky A, Riedl B, Post LE, Bollag G, Trail PA. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 2004;64:7099–7109. ArticlePubMed
  • 6. Gild ML, Bullock M, Robinson BG, Clifton-Bligh R. Multikinase inhibitors: a new option for the treatment of thyroid cancer. Nat Rev Endocrinol 2011;7:617–624. ArticlePubMedPDF
  • 7. Hoftijzer H, Heemstra KA, Morreau H, Stokkel MP, Corssmit EP, Gelderblom H, Weijers K, Pereira AM, Huijberts M, Kapiteijn E, Romijn JA, Smit JW. Beneficial effects of sorafenib on tumor progression, but not on radioiodine uptake, in patients with differentiated thyroid carcinoma. Eur J Endocrinol 2009;161:923–931. ArticlePubMed
  • 8. Kebebew E, Peng M, Reiff E, Treseler P, Woeber KA, Clark OH, Greenspan FS, Lindsay S, Duh QY, Morita E. A phase II trial of rosiglitazone in patients with thyroglobulin-positive and radioiodine-negative differentiated thyroid cancer. Surgery 2006;140:960–966. ArticlePubMed
  • 9. Nikiforov YE. RET/PTC rearrangement in thyroid tumors. Endocr Pathol 2002;13:3–16. ArticlePubMed
  • 10. Vecchio G, Santoro M. Oncogenes and thyroid cancer. Clin Chem Lab Med 2000;38:113–116. ArticlePubMedPDF
  • 11. Musholt TJ, Musholt PB, Khaladj N, Schulz D, Scheumann GF, Klempnauer J. Prognostic significance of RET and NTRK1 rearrangements in sporadic papillary thyroid carcinoma. Surgery 2000;128:984–993. ArticlePubMed
  • 12. Santoro M, Thomas GA, Vecchio G, Williams GH, Fusco A, Chiappetta G, Pozcharskaya V, Bogdanova TI, Demidchik EP, Cherstvoy ED, Voscoboinik L, Tronko ND, Carss A, Bunnell H, Tonnachera M, Parma J, Dumont JE, Keller G, Hofler H, Williams ED. Gene rearrangement and Chernobyl related thyroid cancers. Br J Cancer 2000;82:315–322. ArticlePubMedPMC
  • 13. Vasko V, Ferrand M, Di Cristofaro J, Carayon P, Henry JF, de Micco C. Specific pattern of RAS oncogene mutations in follicular thyroid tumors. J Clin Endocrinol Metab 2003;88:2745–2752. ArticlePubMedPDF
  • 14. Zhu Z, Gandhi M, Nikiforova MN, Fischer AH, Nikiforov YE. Molecular profile and clinical-pathologic features of the follicular variant of papillary thyroid carcinoma. An unusually high prevalence of ras mutations. Am J Clin Pathol 2003;120:71–77. ArticlePubMedPDF
  • 15. Hagemann C, Rapp UR. Isotype-specific functions of Raf kinases. Exp Cell Res 1999;253:34–46. ArticlePubMed
  • 16. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA. Mutations of the BRAF gene in human cancer. Nature 2002;417:949–954. ArticlePubMedPDF
  • 17. Garnett MJ, Marais R. Guilty as charged: B-RAF is a human oncogene. Cancer Cell 2004;6:313–319. ArticlePubMed
  • 18. 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. PubMed
  • 19. Xing M, Vasko V, Tallini G, Larin A, Wu G, Udelsman R, Ringel MD, Ladenson PW, Sidransky D. BRAF T1796A transversion mutation in various thyroid neoplasms. J Clin Endocrinol Metab 2004;89:1365–1368. ArticlePubMed
  • 20. Cantwell-Dorris ER, O'Leary JJ, Sheils OM. BRAFV600E: implications for carcinogenesis and molecular therapy. Mol Cancer Ther 2011;10:385–394. ArticlePubMed
  • 21. Palona I, Namba H, Mitsutake N, Starenki D, Podtcheko A, Sedliarou I, Ohtsuru A, Saenko V, Nagayama Y, Umezawa K, Yamashita S. BRAFV600E promotes invasiveness of thyroid cancer cells through nuclear factor kappaB activation. Endocrinology 2006;147:5699–5707. ArticlePubMedPDF
  • 22. Xing M, Westra WH, Tufano RP, Cohen Y, Rosenbaum E, Rhoden KJ, Carson KA, Vasko V, Larin A, Tallini G, Tolaney S, Holt EH, Hui P, Umbricht CB, Basaria S, Ewertz M, Tufaro AP, Califano JA, Ringel MD, Zeiger MA, Sidransky D, Ladenson PW. BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J Clin Endocrinol Metab 2005;90:6373–6379. ArticlePubMed
  • 23. Caronia LM, Phay JE, Shah MH. Role of BRAF in thyroid oncogenesis. Clin Cancer Res 2011;17:7511–7517. ArticlePubMed
  • 24. Durante C, Puxeddu E, Ferretti E, Morisi R, Moretti S, Bruno R, Barbi F, Avenia N, Scipioni A, Verrienti A, Tosi E, Cavaliere A, Gulino A, Filetti S, Russo D. BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism. J Clin Endocrinol Metab 2007;92:2840–2843. ArticlePubMedPDF
  • 25. Fugazzola L, Mannavola D, Cirello V, Vannucchi G, Muzza M, Vicentini L, Beck-Peccoz P. BRAF mutations in an Italian cohort of thyroid cancers. Clin Endocrinol (Oxf) 2004;61:239–243. ArticlePubMed
  • 26. Gouveia C, Can NT, Bostrom A, Grenert JP, van Zante A, Orloff LA. Lack of association of BRAF mutation with negative prognostic indicators in papillary thyroid carcinoma: the University of California, San Francisco, experience. JAMA Otolaryngol Head Neck Surg 2013;139:1164–1170. ArticlePubMed
  • 27. Puxeddu E, Moretti S, Giannico A, Martinelli M, Marino C, Avenia N, Cristofani R, Farabi R, Reboldi G, Ribacchi R, Pontecorvi A, Santeusanio F. Ret/PTC activation does not influence clinical and pathological features of adult papillary thyroid carcinomas. Eur J Endocrinol 2003;148:505–513. ArticlePubMed
  • 28. Nikiforova MN, Lynch RA, Biddinger PW, Alexander EK, Dorn GW 2nd, Tallini G, Kroll TG, Nikiforov YE. RAS point mutations and PAX8-PPAR gamma rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. J Clin Endocrinol Metab 2003;88:2318–2326. ArticlePubMed
  • 29. Lui WO, Zeng L, Rehrmann V, Deshpande S, Tretiakova M, Kaplan EL, Leibiger I, Leibiger B, Enberg U, Hoog A, Larsson C, Kroll TG. CREB3L2-PPARgamma fusion mutation identifies a thyroid signaling pathway regulated by intramembrane proteolysis. Cancer Res 2008;68:7156–7164. ArticlePubMed
  • 30. Kloos RT, Ringel MD, Knopp MV, Hall NC, King M, Stevens R, Liang J, Wakely PE Jr, Vasko VV, Saji M, Rittenberry J, Wei L, Arbogast D, Collamore M, Wright JJ, Grever M, Shah MH. Phase II trial of sorafenib in metastatic thyroid cancer. J Clin Oncol 2009;27:1675–1684. ArticlePubMedPMC
  • 31. Sherman SI, Wirth LJ, Droz JP, Hofmann M, Bastholt L, Martins RG, Licitra L, Eschenberg MJ, Sun YN, Juan T, Stepan DE, Schlumberger MJ. Motesanib Thyroid Cancer Study Group. Motesanib diphosphate in progressive differentiated thyroid cancer. N Engl J Med 2008;359:31–42. ArticlePubMed
  • 32. Dawson SJ, Conus NM, Toner GC, Raleigh JM, Hicks RJ, McArthur G, Rischin D. Sustained clinical responses to tyrosine kinase inhibitor sunitinib in thyroid carcinoma. Anticancer Drugs 2008;19:547–552. ArticlePubMed
  • 33. Carr LL, Mankoff DA, Goulart BH, Eaton KD, Capell PT, Kell EM, Bauman JE, Martins RG. Phase II study of daily sunitinib in FDG-PET-positive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation. Clin Cancer Res 2010;16:5260–5268. ArticlePubMedPMC
  • 34. Inai T, Mancuso M, Hashizume H, Baffert F, Haskell A, Baluk P, Hu-Lowe DD, Shalinsky DR, Thurston G, Yancopoulos GD, McDonald DM. Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am J Pathol 2004;165:35–52. ArticlePubMedPMC
  • 35. Bible KC, Suman VJ, Menefee ME, Smallridge RC, Molina JR, Maples WJ, Karlin NJ, Traynor AM, Kumar P, Goh BC, Lim WT, Bossou AR, Isham CR, Webster KP, Kukla AK, Bieber C, Burton JK, Harris P, Erlichman C. Mayo Phase 2 Consortium. Mayo Clinic Endocrine Malignances Disease Oriented Group. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J Clin Endocrinol Metab 2012;97:3179–3184. ArticlePubMedPMCPDF
  • 36. Ain KB, Lee C, Williams KD. Phase II trial of thalidomide for therapy of radioiodine-unresponsive and rapidly progressive thyroid carcinomas. Thyroid 2007;17:663–670. ArticlePubMed
  • 37. Mrozek E, Kloos RT, Ringel MD, Kresty L, Snider P, Arbogast D, Kies M, Munden R, Busaidy N, Klein MJ, Sherman SI, Shah MH. Phase II study of celecoxib in metastatic differentiated thyroid carcinoma. J Clin Endocrinol Metab 2006;91:2201–2204. ArticlePubMed
  • 38. American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, Mandel SJ, Mazzaferri EL, McIver B, Pacini F, Schlumberger M, Sherman SI, Steward DL, Tuttle RM. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 2009;19:1167–1214. ArticlePubMed
Fig. 1
MAP kinase pathway and thyroid carcinoma. RTK, receptor tyrosine kinase; TRK, tropomyosin-receptor-kinase; RAS, rat sarcoma; RET/PTC, rearranged during transfection/papillary thyroid cancer; BRAF, v-raf murine sarcoma viral oncogene homolog B1; MEK, mitogen-activated protein kinase; ERK, extracellular signal-related kinase.
enm-29-211-g001.jpg
Table 1
Multi-Kinase Inhibitors and Key Targets
enm-29-211-i001.jpg

Adapted from Gild et al. Nat Rev Endocrinol 2011;7:617-24, with permission from Nature Publishing Group [6].

VEGFR, vascular endothelial growth factor receptor; RET, rearranged during transfection; PDGFR, platelet-derived growth factor receptor; BRAF, v-raf murine sarcoma viral oncogene homolog B1; KIT, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog; FLT3, fms-related tyrosine kinase 3; CSF1R, colony stimulating factor 1 receptor; MET, c-Met, also called hepatocyte growth factor receptor; EGFR, endothelial growth factor receptor; FGFR, fibroblast growth factor receptor.

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Estimating the Growth Rate of Lung Metastases in Differentiated Thyroid Carcinoma: Response Evaluation Criteria in Solid Tumors or Doubling Time?
      Eyun Song, Jonghwa Ahn, Min Ji Jeon, Sang Min Lee, Jeong Hyun Lee, Tae Yong Kim, Jung Hwan Baek, Won Bae Kim, Young Kee Shong, Won Gu Kim
      Thyroid.2020; 30(3): 418.     CrossRef
    • Lenvatinib for Radioactive Iodine-Refractory Differentiated Thyroid Carcinoma and Candidate Biomarkers Associated with Survival: A Multicenter Study in Korea
      Eyun Song, Mijin Kim, Eui Young Kim, Bo Hyun Kim, Dong Yeob Shin, Ho-Cheol Kang, Byeong-Cheol Ahn, Won Bae Kim, Young Kee Shong, Min Ji Jeon, Dong Jun Lim
      Thyroid.2020; 30(5): 732.     CrossRef
    • The role of Slit2 as a tumor suppressor in thyroid cancer
      Min Ji Jeon, Seonhee Lim, Mi-hyeon You, Yangsoon Park, Dong Eun Song, Soyoung Sim, Tae Yong Kim, Young Kee Shong, Won Bae Kim, Won Gu Kim
      Molecular and Cellular Endocrinology.2019; 483: 87.     CrossRef
    • Extended Real-World Observation of Patients Treated with Sorafenib for Radioactive Iodine-Refractory Differentiated Thyroid Carcinoma and Impact of Lenvatinib Salvage Treatment: A Korean Multicenter Study
      Hye-Seon Oh, Dong Yeob Shin, Mijin Kim, So Young Park, Tae Hyuk Kim, Bo Hyun Kim, Eui Young Kim, Won Bae Kim, Jae Hoon Chung, Young Kee Shong, Dong Jun Lim, Won Gu Kim
      Thyroid.2019; 29(12): 1804.     CrossRef
    • Cytological features of warthin‐like papillary thyroid carcinoma: A case report with review of previous cytology cases
      Archana George Vallonthaiel, Shipra Agarwal, Deepali Jain, Rajni Yadav, Nishikant A. Damle
      Diagnostic Cytopathology.2017; 45(9): 837.     CrossRef
    • Low Prevalence of Somatic TERT Promoter Mutations in Classic Papillary Thyroid Carcinoma
      Min Ji Jeon, Won Gu Kim, Soyoung Sim, Seonhee Lim, Hyemi Kwon, Tae Yong Kim, Young Kee Shong, Won Bae Kim
      Endocrinology and Metabolism.2016; 31(1): 100.     CrossRef
    • Molecular Diagnosis Using Residual Liquid-Based Cytology Materials for Patients with Nondiagnostic or Indeterminate Thyroid Nodules
      Hyemi Kwon, Won Gu Kim, Markus Eszlinger, Ralf Paschke, Dong Eun Song, Mijin Kim, Suyeon Park, Min Ji Jeon, Tae Yong Kim, Young Kee Shong, Won Bae Kim
      Endocrinology and Metabolism.2016; 31(4): 586.     CrossRef
    • Alpha lipoic acid inhibits proliferation and epithelial mesenchymal transition of thyroid cancer cells
      Min Ji Jeon, Won Gu Kim, Seonhee Lim, Hyun-Jeung Choi, Soyoung Sim, Tae Yong Kim, Young Kee Shong, Won Bae Kim
      Molecular and Cellular Endocrinology.2016; 419: 113.     CrossRef
    • Upregulation of long noncoding RNA LOC100507661 promotes tumor aggressiveness in thyroid cancer
      Daham Kim, Woo Kyung Lee, Seonhyang Jeong, Mi-Youn Seol, Hyunji Kim, Kyung-Sup Kim, Eun Jig Lee, Jandee Lee, Young Suk Jo
      Molecular and Cellular Endocrinology.2016; 431: 36.     CrossRef
    • Articles in 'Endocrinology and Metabolism' in 2014
      Won-Young Lee
      Endocrinology and Metabolism.2015; 30(1): 47.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
    • XML DownloadXML Download
    Figure

    Endocrinol Metab : Endocrinology and Metabolism