Skip to main content

Utilization of Molecular Markers in the Diagnosis and Management of Thyroid Nodules

  • Chapter
  • First Online:

Abstract

Maximizing the information obtained through ultrasound examination and fine needle aspiration biopsy of the patient with an indeterminate thyroid nodule by obtaining a molecular profile is increasingly being utilized to guide clinical management. Several molecular tests have recently become available in the last few years. These molecular tests evaluate nodules for the presence of mutations/rearrangements or for the expression of differentially expressed mRNAs or miRNAs. Differences in test characteristics and performance exist for each assay and are discussed here to aid in test selection and clinical interpretation of the results.

This is a preview of subscription content, log in via an institution.

References

  1. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, Mandel SJ, et al. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid. 2009;19(11):1167–214.

    Article  PubMed  Google Scholar 

  2. Gharib H, Papini E, Paschke R, Duick DS, Valcavi R, Hegedus L, et al. American Association of Clinical Endocrinologists, Associazione Medici Endocrinologi, and European Thyroid Association medical guidelines for clinical practice for the diagnosis and management of thyroid nodules: executive summary of recommendations. J Endocrinol Investig. 2010;33(Suppl 5):51–6.

    CAS  Google Scholar 

  3. Ali SZ, Cibas ES. The Bethesda system for reporting thyroid cytopathology. New York: Springer; 2010.

    Book  Google Scholar 

  4. Baloch ZW, LiVolsi VA, Asa SL, Rosai J, Merino MJ, Randolph G, et al. Diagnostic terminology and morphologic criteria for cytologic diagnosis of thyroid lesions: a synopsis of the National Cancer Institute thyroid fine-needle aspiration state of the science conference. Diagn Cytopathol. 2008;36(6):425–37.

    Article  PubMed  Google Scholar 

  5. Baloch ZW, Fleisher S, LiVolsi VA, Gupta PK. Diagnosis of “follicular neoplasm”: a gray zone in thyroid fine-needle aspiration cytology. Diagn Cytopathol. 2002;26(1):41–4.

    Article  PubMed  Google Scholar 

  6. Mazzaferri EL. Management of a solitary thyroid nodule. N Engl J Med. 1993;328(8):553–9.

    Article  CAS  PubMed  Google Scholar 

  7. Burch HB, Burman KD, Cooper DS, Hennessey JV, Vietor NO. A 2015 survey of clinical practice patterns in the management of thyroid nodules. J Clin Endocrinol Metab. 2016;101(7):2853–62.

    Article  CAS  PubMed  Google Scholar 

  8. Alexander EK, Kennedy GC, Baloch ZW, Cibas ES, Chudova D, Diggans J, et al. Preoperative diagnosis of benign thyroid nodules with indeterminate cytology. N Engl J Med. 2012;367(8):705–15.

    Article  CAS  PubMed  Google Scholar 

  9. Lithwick-Yanai G, Dromi N, Shtabsky A, Morgenstern S, Strenov Y, Feinmesser M, et al. Multicentre validation of a microRNA-based assay for diagnosing indeterminate thyroid nodules utilising fine needle aspirate smears. J Clin Pathol. 2017;70(6):500–7.

    Article  CAS  PubMed  Google Scholar 

  10. Bartolazzi A, Orlandi F, Saggiorato E, Volante M, Arecco F, Rossetto R, et al. Galectin-3-expression analysis in the surgical selection of follicular thyroid nodules with indeterminate fine-needle aspiration cytology: a prospective multicentre study. Lancet Oncol. 2008;9(6):543–9.

    Article  CAS  PubMed  Google Scholar 

  11. Keutgen XM, Filicori F, Crowley MJ, Wang Y, Scognamiglio T, Hoda R, et al. A panel of four miRNAs accurately differentiates malignant from benign indeterminate thyroid lesions on fine needle aspiration. Clin Cancer Res. 2012;18(7):2032–8.

    Article  CAS  PubMed  Google Scholar 

  12. Nikiforov YE, Ohori NP, Hodak SP, Carty SE, LeBeau SO, Ferris RL, et al. Impact of mutational testing on the diagnosis and management of patients with cytologically indeterminate thyroid nodules: a prospective analysis of 1056 FNA samples. J Clin Endocrinol Metab. 2011;96(11):3390–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cancer Genome Atlas Research N. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014;159(3):676–90.

    Article  CAS  Google Scholar 

  14. 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(8):625–7.

    Article  CAS  PubMed  Google Scholar 

  15. 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(7):1454–7.

    CAS  PubMed  Google Scholar 

  16. Chiosea S, Nikiforova M, Zuo H, Ogilvie J, Gandhi M, Seethala RR, et al. A novel complex BRAF mutation detected in a solid variant of papillary thyroid carcinoma. Endocr Pathol. 2009;20(2):122–6.

    Article  CAS  PubMed  Google Scholar 

  17. Ciampi R, Nikiforov YE. Alterations of the BRAF gene in thyroid tumors. Endocr Pathol. 2005;16(3):163–72.

    Article  CAS  PubMed  Google Scholar 

  18. Hou P, Liu D, Xing M. Functional characterization of the T1799-1801del and A1799-1816ins BRAF mutations in papillary thyroid cancer. Cell Cycle. 2007;6(3):377–9.

    Article  CAS  PubMed  Google Scholar 

  19. 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(29):4578–80.

    Article  CAS  PubMed  Google Scholar 

  20. Ciampi R, Knauf JA, Kerler R, Gandhi M, Zhu Z, Nikiforova MN, et al. Oncogenic AKAP9-BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J Clin Invest. 2005;115(1):94–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lemoine NR, Mayall ES, Wyllie FS, Williams ED, Goyns M, Stringer B, et al. High frequency of ras oncogene activation in all stages of human thyroid tumorigenesis. Oncogene. 1989;4(2):159–64.

    CAS  PubMed  Google Scholar 

  22. Motoi N, Sakamoto A, Yamochi T, Horiuchi H, Motoi T, Machinami R. Role of ras mutation in the progression of thyroid carcinoma of follicular epithelial origin. Pathol Res Pract. 2000;196(1):1–7.

    Article  CAS  PubMed  Google Scholar 

  23. Namba H, Rubin SA, Fagin JA. Point mutations of ras oncogenes are an early event in thyroid tumorigenesis. Mol Endocrinol. 1990;4(10):1474–9.

    Article  CAS  PubMed  Google Scholar 

  24. Suarez HG, du Villard JA, Severino M, Caillou B, Schlumberger M, Tubiana M, et al. Presence of mutations in all three ras genes in human thyroid tumors. Oncogene. 1990;5(4):565–70.

    CAS  PubMed  Google Scholar 

  25. Nikiforov YE, Seethala RR, Tallini G, Baloch ZW, Basolo F, Thompson LD, et al. Nomenclature revision for encapsulated follicular variant of papillary thyroid carcinoma: a paradigm shift to reduce overtreatment of indolent tumors. JAMA Oncol. 2016;2(8):1098.

    Article  PubMed  Google Scholar 

  26. Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP, Giordano TJ, et al. Correlation between genetic alterations and microscopic features, clinical manifestations, and prognostic characteristics of thyroid papillary carcinomas. Am J Surg Pathol. 2006;30(2):216–22.

    Article  PubMed  Google Scholar 

  27. 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(1):71–7.

    Article  CAS  PubMed  Google Scholar 

  28. Jung CK, Little MP, Lubin JH, Brenner AV, Wells SA Jr, Sigurdson AJ, et al. The increase in thyroid cancer incidence during the last four decades is accompanied by a high frequency of BRAF mutations and a sharp increase in RAS mutations. J Clin Endocrinol Metab. 2013. https://doi.org/10.1210/jc.2013-2503.

  29. Nikiforov YE. RET/PTC rearrangement—a link between Hashimoto’s thyroiditis and thyroid cancer…or not. J Clin Endocrinol Metab. 2006;91(6):2040–2.

    Article  CAS  PubMed  Google Scholar 

  30. Zhu Z, Ciampi R, Nikiforova MN, Gandhi M, Nikiforov YE. Prevalence of RET/PTC rearrangements in thyroid papillary carcinomas: effects of the detection methods and genetic heterogeneity. J Clin Endocrinol Metab. 2006;91(9):3603–10.

    Article  CAS  PubMed  Google Scholar 

  31. Dwight T, Thoppe SR, Foukakis T, Lui WO, Wallin G, Hoog A, et al. Involvement of the PAX8/peroxisome proliferator-activated receptor gamma rearrangement in follicular thyroid tumors. J Clin Endocrinol Metab. 2003;88(9):4440–5.

    Article  CAS  PubMed  Google Scholar 

  32. French CA, Alexander EK, Cibas ES, Nose V, Laguette J, Faquin W, et al. Genetic and biological subgroups of low-stage follicular thyroid cancer. Am J Pathol. 2003;162(4):1053–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Nikiforova MN, Lynch RA, Biddinger PW, Alexander EK, Dorn GW 2nd, Tallini G, et al. 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(5):2318–26.

    Article  CAS  PubMed  Google Scholar 

  34. Marques AR, Espadinha C, Catarino AL, Moniz S, Pereira T, Sobrinho LG, et al. Expression of PAX8-PPAR gamma 1 rearrangements in both follicular thyroid carcinomas and adenomas. J Clin Endocrinol Metab. 2002;87(8):3947–52.

    CAS  PubMed  Google Scholar 

  35. Nikiforova MN, Biddinger PW, Caudill CM, Kroll TG, Nikiforov YE. PAX8-PPARgamma rearrangement in thyroid tumors: RT-PCR and immunohistochemical analyses. Am J Surg Pathol. 2002;26(8):1016–23.

    Article  PubMed  Google Scholar 

  36. Cantara S, Capezzone M, Marchisotta S, Capuano S, Busonero G, Toti P, et al. Impact of proto-oncogene mutation detection in cytological specimens from thyroid nodules improves the diagnostic accuracy of cytology. J Clin Endocrinol Metab. 2010;95(3):1365–9.

    Article  CAS  PubMed  Google Scholar 

  37. Nikiforov YE, Steward DL, Robinson-Smith TM, Haugen BR, Klopper JP, Zhu Z, et al. Molecular testing for mutations in improving the fine-needle aspiration diagnosis of thyroid nodules. J Clin Endocrinol Metab. 2009;94(6):2092–8.

    Article  CAS  PubMed  Google Scholar 

  38. Beaudenon-Huibregtse S, Alexander EK, Guttler RB, Hershman JM, Babu V, Blevins TC, et al. Centralized molecular testing for oncogenic gene mutations complements the local cytopathologic diagnosis of thyroid nodules. Thyroid. 2014;24(10):1479–87.

    Article  CAS  PubMed  Google Scholar 

  39. Eszlinger M, Piana S, Moll A, Bosenberg E, Bisagni A, Ciarrocchi A, et al. Molecular testing of thyroid fine-needle aspirations improves presurgical diagnosis and supports the histologic identification of minimally invasive follicular thyroid carcinomas. Thyroid. 2015;25(4):401–9.

    Article  CAS  PubMed  Google Scholar 

  40. Labourier E, Shifrin A, Busseniers AE, Lupo MA, Manganelli ML, Andruss B, et al. Molecular testing for miRNA, mRNA, and DNA on fine-needle aspiration improves the preoperative diagnosis of thyroid nodules with indeterminate cytology. J Clin Endocrinol Metab. 2015;100(7):2743–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Garcia-Rostan G, Costa AM, Pereira-Castro I, Salvatore G, Hernandez R, Hermsem MJ, et al. Mutation of the PIK3CA gene in anaplastic thyroid cancer. Cancer Res. 2005;65(22):10199–207.

    Article  CAS  PubMed  Google Scholar 

  42. Hou P, Liu D, Shan Y, Hu S, Studeman K, Condouris S, et al. Genetic alterations and their relationship in the phosphatidylinositol 3-kinase/Akt pathway in thyroid cancer. Clin Cancer Res. 2007;13(4):1161–70.

    Article  CAS  PubMed  Google Scholar 

  43. 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(11):4885–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Nikiforov YE, Carty SE, Chiosea SI, Coyne C, Duvvuri U, Ferris RL, et al. Highly accurate diagnosis of cancer in thyroid nodules with follicular neoplasm/suspicious for a follicular neoplasm cytology by ThyroSeq v2 next-generation sequencing assay. Cancer. 2014;120(23):3627–34.

    Article  CAS  PubMed  Google Scholar 

  45. Karunamurthy A, Panebianco F, Hsiao S, Vorhauer J, Nikiforova M, Chiosea SI, et al. Prevalence and phenotypic characteristics of EIF1AX mutations in thyroid nodules. Endocr Relat Cancer. 2016;23(4):295–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kunstman JW, Juhlin CC, Goh G, Brown TC, Stenman A, Healy JM, et al. Characterization of the mutational landscape of anaplastic thyroid cancer via whole-exome sequencing. Hum Mol Genet. 2015;24(8):2318–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah RH, et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J Clin Invest. 2016;126(3):1052–66.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Landa I, Ganly I, Chan TA, Mitsutake N, Matsuse M, Ibrahimpasic T, et al. Frequent somatic TERT promoter mutations in thyroid cancer: higher prevalence in advanced forms of the disease. J Clin Endocrinol Metab. 2013;98(9):E1562–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Liu T, Wang N, Cao J, Sofiadis A, Dinets A, Zedenius J, et al. The age- and shorter telomere-dependent TERT promoter mutation in follicular thyroid cell-derived carcinomas. Oncogene. 2013;33(42):4978–84.

    Article  CAS  PubMed  Google Scholar 

  50. Liu X, Bishop J, Shan Y, Pai S, Liu D, Murugan AK, et al. Highly prevalent TERT promoter mutations in aggressive thyroid cancers. Endocr Relat Cancer. 2013;20(4):603–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Melo M, Rocha AG, Vinagre J, Batista R, Peixoto J, Tavares C, et al. TERT promoter mutations are a major indicator of poor outcome in differentiated thyroid carcinomas. J Clin Endocrinol Metab. 2014. https://doi.org/10.1210/jc.2013-3734.

  52. Fagin JA, Matsuo K, Karmakar A, Chen DL, Tang SH, Koeffler HP. High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas. J Clin Invest. 1993;91(1):179–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Donghi R, Longoni A, Pilotti S, Michieli P, Della Porta G, Pierotti MA. Gene p53 mutations are restricted to poorly differentiated and undifferentiated carcinomas of the thyroid gland. J Clin Invest. 1993;91(4):1753–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dobashi Y, Sugimura H, Sakamoto A, Mernyei M, Mori M, Oyama T, et al. Stepwise participation of p53 gene mutation during dedifferentiation of human thyroid carcinomas. Diagn Mol Pathol. 1994;3(1):9–14.

    Article  CAS  PubMed  Google Scholar 

  55. Ho YS, Tseng SC, Chin TY, Hsieh LL, Lin JD. p53 gene mutation in thyroid carcinoma. Cancer Lett. 1996;103(1):57–63.

    Article  CAS  PubMed  Google Scholar 

  56. Takeuchi Y, Daa T, Kashima K, Yokoyama S, Nakayama I, Noguchi S. Mutations of p53 in thyroid carcinoma with an insular component. Thyroid. 1999;9(4):377–81.

    Article  CAS  PubMed  Google Scholar 

  57. Fuhrer D, Holzapfel HP, Wonerow P, Scherbaum WA, Paschke R. Somatic mutations in the thyrotropin receptor gene and not in the Gs alpha protein gene in 31 toxic thyroid nodules. J Clin Endocrinol Metab. 1997;82(11):3885–91.

    CAS  PubMed  Google Scholar 

  58. Trulzsch B, Krohn K, Wonerow P, Chey S, Holzapfel HP, Ackermann F, et al. Detection of thyroid-stimulating hormone receptor and Gsalpha mutations: in 75 toxic thyroid nodules by denaturing gradient gel electrophoresis. J Mol Med. 2001;78(12):684–91.

    Article  CAS  PubMed  Google Scholar 

  59. Parma J, Duprez L, Van Sande J, Hermans J, Rocmans P, Van Vliet G, et al. Diversity and prevalence of somatic mutations in the thyrotropin receptor and Gs alpha genes as a cause of toxic thyroid adenomas. J Clin Endocrinol Metab. 1997;82(8):2695–701.

    CAS  PubMed  Google Scholar 

  60. Garcia-Jimenez C, Santisteban P. TSH signalling and cancer. Arq Bras Endocrinol Metabol. 2007;51(5):654–71.

    Article  PubMed  Google Scholar 

  61. Nishihara E, Amino N, Maekawa K, Yoshida H, Ito M, Kubota S, et al. Prevalence of TSH receptor and Gsalpha mutations in 45 autonomously functioning thyroid nodules in Japan. Endocr J. 2009;56(6):791–8.

    Article  CAS  PubMed  Google Scholar 

  62. Leeman-Neill RJ, Kelly LM, Liu P, Brenner AV, Little MP, Bogdanova TI, et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer. 2013;120(6):799–807.

    Article  CAS  PubMed  Google Scholar 

  63. Greco A, Pierotti MA, Bongarzone I, Pagliardini S, Lanzi C, Della PG. TRK-T1 is a novel oncogene formed by the fusion of TPR and TRK genes in human papillary thyroid carcinomas. Oncogene. 1992;7(2):237–42.

    CAS  PubMed  Google Scholar 

  64. Greco A, Mariani C, Miranda C, Lupas A, Pagliardini S, Pomati M, et al. The DNA rearrangement that generates the TRK-T3 oncogene involves a novel gene on chromosome 3 whose product has a potential coiled-coil domain. Mol Cell Biol. 1995;15(11):6118–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Martin-Zanca D, Hughes SH, Barbacid M. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature. 1986;319(6056):743–8.

    Article  CAS  PubMed  Google Scholar 

  66. Radice P, Sozzi G, Miozzo M, De Benedetti V, Cariani T, Bongarzone I, et al. The human tropomyosin gene involved in the generation of the TRK oncogene maps to chromosome 1q31. Oncogene. 1991;6(11):2145–8.

    CAS  PubMed  Google Scholar 

  67. Prasad ML, Vyas M, Horne MJ, Virk RK, Morotti R, Liu Z, et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer. 2016;122(7):1097–107.

    Article  CAS  PubMed  Google Scholar 

  68. Kelly LM, Barila G, Liu P, Evdokimova VN, Trivedi S, Panebianco F, et al. Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc Natl Acad Sci U S A. 2014;111(11):4233–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ji JH, Oh YL, Hong M, Yun JW, Lee HW, Kim D, et al. Identification of driving ALK fusion genes and genomic landscape of medullary thyroid cancer. PLoS Genet. 2015;11(8):e1005467.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nikiforov YE, Carty SE, Chiosea SI, Coyne C, Duvvuri U, Ferris RL, et al. Impact of the multi-gene thyroSeq next-generation sequencing assay on cancer diagnosis in thyroid nodules with atypia of undetermined significance/follicular lesion of undetermined significance cytology. Thyroid. 2015;25(11):1217–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Chudova D, Wilde JI, Wang ET, Wang H, Rabbee N, Egidio CM, et al. Molecular classification of thyroid nodules using high-dimensionality genomic data. J Clin Endocrinol Metab. 2010;95(12):5296–304.

    Article  CAS  PubMed  Google Scholar 

  72. Alexander EK, Schorr M, Klopper J, Kim C, Sipos J, Nabhan F, et al. Multicenter clinical experience with the Afirma gene expression classifier. J Clin Endocrinol Metab. 2014;99(1):119–25.

    Article  CAS  PubMed  Google Scholar 

  73. Harrell RM, Bimston DN. Surgical utility of Afirma: effects of high cancer prevalence and oncocytic cell types in patients with indeterminate thyroid cytology. Endocr Pract. 2014;20(4):364–9.

    Article  PubMed  Google Scholar 

  74. Marti JL, Avadhani V, Donatelli LA, Niyogi S, Wang B, Wong RJ, et al. Wide inter-institutional variation in performance of a molecular classifier for indeterminate thyroid nodules. Ann Surg Oncol. 2015;22(12):3996–4001.

    Article  PubMed  PubMed Central  Google Scholar 

  75. McIver B, Castro MR, Morris JC, Bernet V, Smallridge R, Henry M, et al. An independent study of a gene expression classifier (Afirma) in the evaluation of cytologically indeterminate thyroid nodules. J Clin Endocrinol Metab. 2014;99(11):4069–77.

    Article  CAS  PubMed  Google Scholar 

  76. Santhanam P, Khthir R, Gress T, Elkadry A, Olajide O, Yaqub A, et al. Gene expression classifier for the diagnosis of indeterminate thyroid nodules: a meta-analysis. Med Oncol. 2016;33(2):14.

    Article  CAS  PubMed  Google Scholar 

  77. Pankratz DG, Hu Z, Kim SY, Monroe RJ, Wong MG, Traweek ST, et al. Analytical performance of a gene expression classifier for medullary thyroid carcinoma. Thyroid. 2016;26(11):1573–80.

    Article  CAS  PubMed  Google Scholar 

  78. Chou CK, Chen RF, Chou FF, Chang HW, Chen YJ, Lee YF, et al. miR-146b is highly expressed in adult papillary thyroid carcinomas with high risk features including extrathyroidal invasion and the BRAF(V600E) mutation. Thyroid. 2010;20(5):489–94.

    Article  CAS  PubMed  Google Scholar 

  79. Chou CK, Yang KD, Chou FF, Huang CC, Lan YW, Lee YF, et al. Prognostic implications of miR-146b expression and its functional role in papillary thyroid carcinoma. J Clin Endocrinol Metab. 2013;98(2):E196–205.

    Article  CAS  PubMed  Google Scholar 

  80. Benjamin H, Schnitzer-Perlman T, Shtabsky A, VandenBussche CJ, Ali SZ, Kolar Z, et al. Analytical validity of a microRNA-based assay for diagnosing indeterminate thyroid FNA smears from routinely prepared cytology slides. Cancer Cytopathol. 2016;124(10):711–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Ferris RL, Baloch Z, Bernet V, Chen A, Fahey TJ 3rd, Ganly I, et al. American Thyroid Association statement on surgical application of molecular profiling for thyroid nodules: current impact on perioperative decision making. Thyroid. 2015;25(7):760–8.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Yip L, Wharry LI, Armstrong MJ, Silbermann A, McCoy KL, Stang MT, et al. A clinical algorithm for fine-needle aspiration molecular testing effectively guides the appropriate extent of initial thyroidectomy. Ann Surg. 2014;260(1):163–8.

    Article  PubMed  Google Scholar 

  83. Labourier E. Utility and cost-effectiveness of molecular testing in thyroid nodules with indeterminate cytology. Clin Endocrinol. 2016;85(4):624–31.

    Article  Google Scholar 

  84. Yip L, Farris C, Kabaker AS, Hodak SP, Nikiforova MN, McCoy KL, et al. Cost impact of molecular testing for indeterminate thyroid nodule fine-needle aspiration biopsies. J Clin Endocrinol Metab. 2012;97(6):1905–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yuri E. Nikiforov MD, PhD .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG

About this chapter

Cite this chapter

Hsiao, S.J., Nikiforov, Y.E. (2018). Utilization of Molecular Markers in the Diagnosis and Management of Thyroid Nodules. In: Duick, D., Levine, R., Lupo, M. (eds) Thyroid and Parathyroid Ultrasound and Ultrasound-Guided FNA . Springer, Cham. https://doi.org/10.1007/978-3-319-67238-0_15

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-67238-0_15

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-67237-3

  • Online ISBN: 978-3-319-67238-0

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics