Skip to main content

New insights into the diagnosis of nodular goiter

Abstract

Preoperative diagnostic investigations of nodular goiter are based on two main examinations: ultrasonography of the thyroid gland and ultrasound-guided fine-needle aspiration biopsy. So far, FNAB has been the best method for the differentiation of nodules, but in some cases it fails to produce a conclusive diagnosis. Some of the biopsies do not provide enough material to establish the diagnosis, in some other biopsies cytological picture is inconclusive.

Determining the eligibility of thyroid focal lesions for surgery has been more and more often done with molecular methods. The most common genetic changes leading to the development of thyroid cancer include mutations, translocations and amplifications of genes, disturbances in gene methylation and dysregulation of microRNA. The mutations of Ras proto-oncogenes and BRAF gene as well as disturbances of DNA methylation in promoter regions of genes regulating cell cycle (e.g. hypermethylation of RASSF1A gene and TIMP-3 gene) play an important role in the process of neoplastic transformation of thyreocyte. The advances in molecular biology made it possible to investigate these genetic disturbances in DNA and/or RNA from peripheral blood, postoperative thyroid tissue material and cytology specimens obtained through fine-needle aspiration biopsy of focal lesions in the thyroid gland.

As it became possible to analyze the mutations and methylation of genes from cell material obtained through fine-needle aspiration biopsy, it would be beneficial to introduce the techniques of molecular biology in the pre-operative diagnosis of nodular goiter as a valuable method, complementary to ultrasonography and FNAB. The knowledge obtained from molecular studies might help to determine the frequency of follow-up investigations in patients with nodular goiter and to select patients potentially at risk of developing thyroid cancer, which would facilitate their qualification for earlier strumectomy.

Introduction

Nodular goiter is the most common pathology of the thyroid gland. Palpable thyroid nodules are found in 3-7% of adult population and are more frequent in women. Ultrasonography which has been introduced in the diagnosis of thyroid gland has confirmed earlier autopsy reports indicating that focal lesions are found in as many as 50% of clinically normal thyroid glands [1, 2]. Preoperative diagnostic investigations of nodular goiter are currently based on two main examinations: ultrasonography of the thyroid gland and ultrasound-guided fine-needle aspiration biopsy. Determining the eligibility of thyroid focal lesions for surgery has been more and more often done with molecular methods providing the information on possible presence of mutations and epigenetic changes which play an important role in malignant transformation [3–12]. Most focal lesions in the thyroid gland are of benign nature. The incidence of thyroid cancer in multinodular goiter is estimated at approx. 5-10%. Thyroid cancer is more common in solitary thyroid nodules (approx. 10-20%) than in multinodular goiter [1, 13]. Clinical examination of patients with thyroid nodules remains important component of cancer risk assessment. The risk factors include positive family history (this is especially true for medullary carcinoma and some papillary carcinomas), age (under 20 years and over 60 years), sex (males are at greater risk) and history of head and neck irradiation, particularly in the childhood. Other very important symptoms include dysphonia, raucity and neck pain in patients with hard, not easily movable lump [14, 15].

Ultrasonographic examination of focal thyroid lesions, particularly those in multinodular goiter is helpful in selecting one or more foci for fine-needle aspiration biopsy. It is currently believed that the most important criteria for the evaluation of malignant potential of a thyroid nodule is not its size but rather its vascularization, the presence of microcalcifications, height/width ratio, structure (solid or solid-fluid), echogenicity and border margins as well as the presence of so called halo. Hence, thyroid nodules most suspected of malignancy are those with the following ultrasonographic features: enhanced central vascularization or no flow in power doppler; microcalcifications; nodule height exceeding its width; solid lesions are more suspicious than solid-fluid ones; hypoechogenic lesions raise more concerns than isoechogenic ones; lesions with rough margins and those without a halo or lesions with irregular, thick halo are more suspicious [16]. Nowadays, ultrasound-guided fine-needle aspiration biopsy is a gold standard in the diagnosis of nodular goiter. It is technically simple, safe and inexpensive. Cytology assessment of the specimens obtained through fine-needle aspiration biopsy is based on international classification known as the Bethesda System of Reporting Thyroid Cytopathology [17]. According to this classification, the findings of fine-needle aspiration biopsy of thyroid nodule can be divided into 6 groups of diagnostic cytopathology categories: I-non-diagnostic or unsatisfactory, II- benign, III- atypia of undetermined significance or follicular lesion of undetermined significance, IV- follicular neoplasm or suspicious for a follicular neoplasm, V- suspicious for malignancy and VI- malignant. Cytopathology diagnoses falling within groups IV, V and VI are indications for surgery. Diagnoses classified as group III and those classified as group I mean that fine-needle aspiration biopsy should be repeated. It is also worth emphasis that even the diagnosis of a benign lesion (group II ) in fine-needle aspiration biopsy carries 3% risk of false negative result [17]. Irrespective of the chosen classification of cytopathology findings, one should assume that approx. 20% of thyroid nodule biopsies produce results that require final diagnosis based on postoperative histopathology report [13].

The scintigraphy of the thyroid gland is currently considered less useful in the diagnosis of possible malignancy in the nodular goiter since it has been demonstrated that malignant lesions may be found not only in cold nodules but also in approx. 3% of solitary hot thyroid nodules [18].

Thyroid cancer is the most common malignancy of endocrine system accounting for 2.5% of all cancers diagnosed in humans [19]. It has been diagnosed more frequently since 1990s, particularly in women [20]. In Poland, there are 1500 to 1800 new cases of thyroid cancer per year [21]. The most common histological type of thyroid cancer is papillary thyroid carcinoma (PTC) and the second most common differentiated thyroid cancer is follicular thyroid carcinoma (FTC); they account for 80% and 15% of all thyroid cancers, respectively [22, 23]. PTC and FTC are both classified as differentiated cancers of the thyroid gland, deriving from highly differentiated follicular cell which is dedifferentiated in the presence of various factors, fails to undergo apoptosis and becomes capable of uncontrolled proliferation and forming metastases [24, 25]. Neogenesis is a multistage process, involving multiple genes as well as endo- and exogenous factors. At the initial stage of neogenesis called initiation, the cell is damaged. The second stage called promotion is the clonal expansion of abnormal cell triggered by mitogenic signal, which produces a group of dividing cells with still benign genotype. The third stage is neoplastic transformation of some clonal cells caused by further genetic changes – oncogenesis is part of progression stage.

Review

The most common genetic changes resulting in the development of thyroid cancer are mutations, translocations and amplifications of genes, disturbances in gene methylation and dysregulation of microRNA [3, 4, 6, 26]. The advances in molecular biology made it possible to investigate these genetic disturbances in DNA and/or RNA from peripheral blood, postoperative thyroid tissue material and cytology specimens obtained through fine-needle aspiration biopsy of focal lesions of the thyroid gland. They cast new light on the genesis of benign and malignant lesions in the thyroid gland and also opened new perspectives for preoperative diagnosis of focal thyroid lesions. The first Polish center to conduct molecular tests of cytology material obtained from fine-needle aspiration biopsy of the thyroid gland was in Szczecin and these tests were aimed at the detection of somatic mutations of TSH receptor gene and G protein alpha chain [27, 28]. New molecular diagnostic tools applied to fine-needle aspiration biopsy allowed for more precise qualification of patients for total or partial strumectomy. The key role in the neoplastic transformation of thyroid follicular cell is played by the inactivation of suppression genes and activation of oncogenes [3, 4, 24, 25, 29]. Mutations observed in thyroid cancers usually affect RAS, BRAF, PTEN, CTNNB1, TP53, IDH1, ALK and EGFR genes [9, 12, 25, 30].

BRAF gene plays a very important role in the etiopathogenesis of papillary thyroid carcinoma [7, 8, 31–33]. The predominant BRAF gene mutation reported in PTC, observed in 36-80% of PTC cases is the thymine-to-adenine transversion at position 1799 (T1799A) in exon 15, resulting in the substitution of valine (V) by glutamic acid (E) at codon 600 (V600E) [22, 33–40]. This specific V600E BRAF mutation represents 99% of all BRAF mutations found in thyroid cancer [6]. Many studies prove that this mutation is found only in papillary thyroid carcinoma and in few cases of anaplastic cancer [34, 36, 41]. The presence of BRAF T1799A oncogene is an unfavorable prognostic factor in PTC as it increases the aggressive nature of cancer through raising its invasiveness, accelerating relapses and the occurrence of metastases [23, 33, 34, 36, 37, 42–44] BRAF gene encodes BRAF protein. BRAF protein belongs to a class of serine/threonine kinases and subfamily of RAF proteins [8, 45, 46]. Cytoplasmic RAF proteins make up RAS-RAF-MEK-ERK pathway which is involved in the transduction of mitogenic signal from the cell surface to cell nucleus [47–49]. This pathway using tyrosine kinase receptor is a mitogen-activated kinase cascade called MAPK (Mitogen-Activated Protein Kinase) [32, 47–49]. BRAF gene mutation activating MAPK pathway is most likely the main contributor to the development and progression of PTC. T1799A BRAF oncogene is present at all stages of PTC progression, it may even be there at early stages of the development of micropapillary cancer [6, 31, 34, 50]. The presence of BRAF mutation in cytology material obtained from FNAB of thyroid nodule indicates the necessity of surgical treatment. It should be remembered, however, that false positive results indicating the presence of BRAF in the nodule are reported in 0.2-5.7% of cases, while false negative results are found in 1.9-5.8% of cases [51–54]. Since in some cases of thyroid nodule biopsy, the specimens for cytology assessment do not contain enough cells with mutated BRAF and the result obtained is negative despite the mutation in thyroid nodule, currently there are indications to re-evaluate BRAF mutation in follow-up biopsy performed a few months later, especially when the nodule presents ultrasonography features of malignancy and cytology assessment of focal thyroid lesion gives non-diagnostic results, there are signs of atypia or the nodule is of benign nature [11].

Following BRAF mutation, the second most common mutation observed in thyroid cancer are the mutations of RAS proto-oncogenes, which play an important role in the initiation of thyreocyte neoplastic transformation [12, 55, 56]. Proto-oncogenes of RAS family (N-RAS, H-RAS, K-RAS) are located on 1, 11 and 12 chromosome, respectively. They are involved in the control of growth and differentiation of cells. They encode G membrane proteins showing intrinsic GTP-ase activity and participate in signal transmission from membrane tyrosine kinase receptor to cell nucleus using both MAPK and PI3K-AKT pathways. Mutations at codons 12, 13 and 61 transform these proto-oncogenes into active oncogenes [6, 57]. Mutations of RAS proto-oncogene make the protein encoded by this proto-oncogene lose its intrinsic GTP-ase activity and there is a constitutive activation of signal transduction pathway. In thyroid tumorigenesis, PI3K-AKT is a preferable pathway [55]. RAS gene mutations are observed both in benign and malignant thyroid neoplasms. These mutations can be found in 40-50% of FTC cases, in 5-20% of PTC cases, in 20-40% of poorly differentiated and anaplastic cases as well as in approx. 30% of follicular adenomas [9, 32, 35, 56, 58, 59]. RAS-positive follicular adenoma may be a precursor of both follicular cancer and follicular variant of papillary carcinoma [6]. Recent studies have emphasized an important role of RAS mutation as a valuable diagnostic marker in tumors with very difficult or impossible diagnosis based on cytology assessment of fine-needle aspiration biopsy material, which is true for follicular variant of papillary cancer and follicular adenoma [60, 61]. Diagnostic difficulties with follicular variant of papillary cancer result from the absence of papillary proliferation and limited nuclear features typical for papillary cancer. To differentiate between adenoma and follicular cancer, the assessment of vascular invasion and capsule infiltration is necessary, which cannot be based on cytology material obtained from fine-needle aspiration biopsy [39, 56]. Most RAS-positive thyroid nodules with indeterminate cytology and without suspicious ultrasonography features turn out to be a follicular form of papillary cancer with low level of malignancy in post-operative histopathology examination [12]. RAS mutations are found in a large percentage of poorly differentiated and anaplastic thyroid cancers, therefore, it seems advisable to consider surgical treatment of all RAS-positive thyroid nodules to prevent cancer progression [12].

Yet another kind of genetic changes found in thyroid cancer are oncogenic rearrangements resulting from gene translocations, with RET/PTC and PAX8/PPARγ being the most common [62–64]. More than 10 types of RET/PTC translocation have been described but the two most frequently occurring are RET/PTC1 and RET/PTC3 [62, 65]. RET is a proto-oncogene encoding RTK. RET/PTC is formed as a consequence of genetic recombination between the 3’ portion of RET tyrosine kinase and the 5’ portion of a partner gene. In the case of RET/PTC1 rearrangement, the partner gene is CCDC6 known as H4 (coiled-coil domain-containing gene 6), while in RET/PTC3 rearrangement, the partner gene is NCOA4 known as ELE1 (nuclear receptor co-activator 4). The structural basis for RET/PTC transformation is close vicinity of RET and the partner gene in cell nucleus [66, 67]. The consequence of this rearrangement is ligand-independent dimerization and constitutive activation of RET tyrosine kinase [25]. RET/PTC1 is the most frequent type accounting for 60-70% of re-arrangements and RET/PTC3 is observed in 20-30% of PTC cases [68]. RET/PTC1 re-arrangements are more common in classic forms of papillary cancer and in papillary microcarcinoma [3, 69]. On the other hand, RET/PTC3 re-arrangements are more common in solid papillary cancers, which was observed especially in the Ukraine and Belarus after Chernobyl disaster [70].

Another important re-arrangement observed in thyroid cancer is PAX8/PPARγ as a consequence of the translocation of genetic material between chromosomes 2 and 3. Then, PAX8 gene which encodes thyroid-specific transcription factor domain is combined with PPARγ (peroxisome proliferator-activated receptor-γ). PAX8/PPARγ rearrangement is observed mainly in follicular cancers, but also in the follicular form of papillary cancer and follicular adenomas [64, 71].

It is not only the translocation of genetic material but also the amplification of oncogenes that may play a very important role in the thyroid tumorigenesis. This is especially the case in the genes encoding MAPK pathway kinases using tyrosine kinase receptor but also in the genes encoding PI3K-AKT pathway. Elevated number of oncogene copies are more common in anaplastic cancer than in differentiated cancers of the thyroid gland, which suggests that it may be of considerable relevance for cancer aggressiveness and the rate of its progression [72].

It has been demonstrated that neoplastic transformation of the thyroid gland is also affected by epigenetic mechanisms, i.e. the mechanisms influencing the regulation and modification of genetic material, not affecting the nucleotide sequence [4, 5, 25, 29, 73–75]. These mechanisms include DNA methylation and histone modification. DNA methylation takes place through covalent modification of cytosines and it is catalyzed by DNA methyltransferases, which attach methyl group at the carbon 5’ position of cytosine ring. This modification applies only to cytosines (C) which are followed by guanine (G) in the sequence. CG sequences are grouped in the genome sites known as CpG islands, where CG dinucleotide repetitions extend over 1,000 - 2,000 base pairs [76]. In the case of genes with vital significance for fundamental cellular processes, with widespread expression in the tissues, CpG islands associated with them are almost always found on 5’ side of encoding sequences, typically in the promoters of these genes. The general mechanism of silencing expression of hypermethylation-dependent DNA genes has several aspects. The most important of them is to prevent the binding of transcription factors to promoters and sequences regulating transcription, on the basis of spatial conflict. These data are consistent with observations which imply that CpG islands of suppressor genes in healthy somatic cells are usually characterized by low levels of methylation or no methylation at all. During oncogenesis, hypermethylation of these sites often occurs, which causes silencing of their expression. Thus, in the selective strategy of a neoplasm, hypermethylation of genes is aimed at marking those genome areas which are to undergo deletion processes, leading to irreversible loss of growth control. The result of this situation is not only accelerated growth of cells but also the beginning of particularly dangerous genetic instability [77, 78]. The genes controlling the proliferation of cells, which undergo hypermethylation in the papillary carcinoma include TIMP-3 (tissue inhibitor of metalloproteinase-3, inhibitor of extracellular metalloproteinases), DAPK (calcium-dependent protein kinase), taking part in programmed cell death, SLC5A8 (sodium symporter), DNA repair genes (hMLH1, PCNA) and thyroid-specific genes (NIS-sodium-iodine symporter, TSHR- thyroid stimulating hormone receptor). The genes encoding the suppressors of neoplasia undergoing hypermethylation in follicular cancer include PTEN (phosphatase inhibiting one of mitogen signal transduction pathway), RASSF1A (signal protein of mitogen RAS pathway), thyroid-specific genes (NIS, TSHR) and TRbeta (receptor beta for thyroid hormones) [1, 4, 5, 75, 79, 80]. Special attention should be paid to TIMP-3 and RASSF1A genes taking part in the tumorigenesis of the thyroid gland [4, 5, 29]. TIMP-3 inhibits the growth, angiogenesis and invasion of many cancers. Hypermethylation of this gene is particularly important in the onset of papillary thyroid cancer. It has been demonstrated that there is a correlation between loss of TIMP-3 gene function as metalloproteinase inhibitor associated with hypermethylation and extrathyroid invasion of papillary carcinoma, lymph node metastases and multifocal nature of this cancer [4, 75, 81, 82]. Protein products of RASSF1A suppressor gene participate in controlling cell cycle, controlling the differentiation and proliferation of cells through direct regulation of transcription and regulation of proapoptic signal pathways. Epigenetic silencing of this gene expression through promoter hypermethylation may lead to unauthorized divisions of mutated cells. Decreased expression of RASSF1A gene and/or reduced activity of its protein products are also affected by changes in DNA sequence related to the acquisition of genome instability by neoplastic cells, resulting from loss of heterozygosity as well as from the instability of microsatellite sequences [1, 4, 5, 29]. Hypermethylation of RASSF1A gene is observed both in benign thyroid neoplasms and in thyroid cancers, particularly in FTC. Methylation levels above 50% of alleles was only observed in follicular thyroid cancer, while it was not observed in benign neoplasms of the thyroid gland, suggesting that methylation through silencing both RASSF1A gene alleles may play an important role in the pathogenesis and development of follicular thyroid cancer [1, 4, 5, 29].

Conclusion

New opportunities for the analysis of mutation and methylation of genes obtained from fine-needle aspiration biopsy presented in this article confirm clinical benefits from introducing molecular studies into pre-operative diagnostic investigations of the thyroid gland as a valuable method complementary to ultrasonography and cytology evaluation of thyroid bioptates, particularly when qualifying patients with follicular adenomas and follicular lesions with undetermined significance for surgical treatment.

Abbreviations

FNAB:

Fine-needle aspiration biopsy

Approx:

Approximately

PTC:

Papillary thyroid carcinoma

FTC:

Follicular thyroid carcinoma

RNA:

Ribonucleic acid

DNA:

Deoxyribonucleic acid

V:

Valine

E:

Glutamic acid

RET:

a proto-oncogene encoding RTK

TSHR:

Thyroid stimulating hormone receptor

CCDC6:

Coiled-coil domain-containing gene 6

MAPK:

Mitogen-activated protein kinase

C:

Cytosine

G:

Guanine

NCOA4:

Nuclear receptor co-activator 4

ELE1:

Nuclear receptor co-activator

4TIMP-3:

Tissue inhibitor of metalloproteinase-3

DAPK:

Calcium-dependent protein kinase

SLC5A8:

Sodium symporter

NIS:

Sodium-iodine symporter

TRbeta:

Receptor beta for thyroid hormones

RASSF1A:

Signal protein of mitogen RAS pathway

PPARγ:

Peroxisome proliferator-activated receptor-γ.

References

  1. Lesnik DJ, O’Leary MA, Noordzij JP, Randolph GW: Surgical management of benign diseases of the thyroid gland. In Diseases of the thyroid and parathyroid glands. Practical Guide. Edited by: Arora A, Tolley NS, Tuttle RM, Lewiński A. red. polskiego wydania, Warszawa: MediPage; 2013:83. in Polish

    Google Scholar 

  2. Mazzaferri EL: Management of a solitary thyroid nodule. N Engl J Med 1993,328(8):553–559.

    Article  CAS  PubMed  Google Scholar 

  3. Nikiforov YE: Molecular analysis of thyroid tumors. Mod Pathol 2011,24(Suppl 2):S34–43.

    Article  CAS  PubMed  Google Scholar 

  4. Xing M: Minireview: Gene methylation in thyroid tumorigenesis. Endocrinology 2007,148(3):948–953.

    Article  CAS  PubMed  Google Scholar 

  5. Hoque MO, Rosenbaum E, Westra WH, Xing M, Ladenson P, Zeiger MA, Sidransky D, Umbricht CB: Quantitative assessment of promoter methylation profiles in thyroid neoplasms. J Clin Endocrinol Metab 2005,90(7):4011–4018.

    Article  CAS  PubMed  Google Scholar 

  6. Witt RL, Ferris RL, Pribitkin EA, Sherman SI, Steward DL, Nikiforov YE: Diagnosis and management of differentiated thyroid cancer using molecular biology. Laryngoscope 2013,123(4):1059–1064.

    Article  PubMed  Google Scholar 

  7. Cappola AR, Mandel SJ: Molecular testing in thyroid cancer: BRAF mutation status and mortality. JAMA 2013,309(14):1529–1530.

    Article  CAS  PubMed  Google Scholar 

  8. Derdas SP, Soulitzis N, Balis V, Sakorafas GH, Spandidos DA: Expression analysis of B-Raf oncogene in V600E-negative benign and malignant tumors of the thyroid gland: correlation with late disease onset. Med Oncol 2013,30(1):336.

    Article  PubMed  Google Scholar 

  9. Puxeddu E, Moretti S, Elisei R, Romei C, Pascucci R, Martinelli M, Marino C, Avenia N, Rossi ED, Fadda G, Cavaliere A, Ribacchi R, Falorni A, Pontecorvi A, Pacini F, Pinchera A, Santeusanio F: BRAF(V599E) mutation is the leading genetic event in adult sporadic papillary thyroid carcinomas. J Clin Endocrinol Metab 2004,89(5):2414–2420.

    Article  CAS  PubMed  Google Scholar 

  10. Khan MS, Pandith AA, Ul Hussain M, Iqbal M, Khan NP, Wani KA, Masoodi SR, Mudassar S: Lack of mutational events of RAS genes in sporadic thyroid cancer but high risk associated with HRAS T81C single nucleotide polymorphism (case–control study). Tumour Biol 2013,34(1):521–529.

    Article  CAS  PubMed  Google Scholar 

  11. Yoon JH, Kim EK, Moon HJ, Kwak JY: Is follow-up BRAF(V600E) mutation analysis helpful in the differential diagnosis of thyroid nodules with negative results on initial analysis? PLoS One 2013,8(3):e58592.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  12. Gupta N, Dasyam AK, Carty SE, Nikiforova MN, Ohori NP, Armstrong M, Yip L, LeBeau SO, McCoy KL, Coyne C, Stang MT, Johnson J, Ferris RL, Seethala R, Nikiforov YE, Hodak SP: RAS mutations in thyroid FNA specimens are highly predictive of predominantly low-risk follicular-pattern cancers. J Clin Endocrinol Metab 2013,98(5):914–22.

    Article  Google Scholar 

  13. Fadda G, Basolo F, Bondi A, Bussolati G, Crescenzi A, Nappi O, Nardi F, Papotti M, Taddei G, Palombini L: SIAPEC-IAP Italian Consensus Working Group. Cytological classification of thyroid nodules. Proposal of the SIAPEC-IAP Italian Consensus Working Group. Pathologica. 2010,102(5):405–408.

    CAS  PubMed  Google Scholar 

  14. 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, American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer: Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 2009,19(11):1167–1214.

    Article  PubMed  Google Scholar 

  15. Bahn RS, Castro MR: Approach to the patient with nontoxic multinodular goiter. J Clin Endocrinol Metab 2011,96(5):1202–1212.

    Article  CAS  PubMed  Google Scholar 

  16. Adamczewski Z, Lewiński A: Proposed algorithm for management of patients with thyroid nodules/focal lesions, based on ultrasound (US) and fine-needle aspiration biopsy (FNAB); our own experience. Thyroid Res 2013 Apr 20,6(1):6.

    Article  PubMed Central  PubMed  Google Scholar 

  17. Cibas ES, Ali SZ: The Bethesda System for Reporting Thyroid Cytopathology. Thyroid 2009,19(11):1159–1165.

    Article  PubMed  Google Scholar 

  18. Mirfakhraee S, Mathews D, Peng L, Woodruff S, Zigman JM: A solitary hyperfunctioning thyroid nodule harboring thyroid carcinoma: review of the literature. Thyroid Res 2013,6(1):7.

    Article  PubMed Central  PubMed  Google Scholar 

  19. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ: Cancer statistics. CA Cancer J Clin 2010, 59: 225–249.

    Article  Google Scholar 

  20. Holt EH: Care of the pregnant thyroid cancer patient. Curr Opin Oncol 2010,22(1):1–5.

    Article  PubMed  Google Scholar 

  21. Jarząb B, Szpak-Ulczok S: Thyroid cancer. Epidemiology, risk factors and prognosis. In Clinical Endocrinology. Edited by: Milewicz A. Wrocław: Polish Society of Endocrinology; 2012:307.

    Google Scholar 

  22. Xing M, Vasko V, Tallini G, Larin A, Wu G, Udelsman R, Ringel MD, Ladenson PW, Sidransky D: BRAFT1796A transversion mutatin in various thyroid neoplasms. J Clin Endocrinol Metab 2004, 89: 1365–1368.

    Article  CAS  PubMed  Google Scholar 

  23. Kopczyńska E, Junik R, Tyrakowski T: Mutacja genu BRAF w raku tarczycy. Pol Merk Lek 2006, 116: 210–213. XX

    Google Scholar 

  24. Kuźnicka-Puzianowska M, Pietrzak M: Genetic factors predisposing to the formation of papillary thyroid cancer. Endokr Pol 2005,3(56):339–345.

    Google Scholar 

  25. Xing M: Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer 2013,13(3):184–199.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Bhaijee F, Nikiforov YE: Molecular analysis of thyroid tumors. Endocr Pathol 2011,22(3):126–133.

    Article  CAS  PubMed  Google Scholar 

  27. Syrenicz A: A search for somatic mutations in the thyrotropin receptor gene and G protein alpha-subunit 201 and 227 codons using cytological material from thyroid nodules an attempt to link the mutation with clinical results of conservative treatment of nodular goiter with thyroxine or thyroxine and iodine. Habilitation thesis. Pomorska Akad Med 2000, 57: s94.

    Google Scholar 

  28. Syrenicz A, Kurzawski G, Ciechanowicz A: The dedection of somatic mutations of thyrotropin receptor gene in fine needle biopsy samples from thyroid nodules. Endocr Regul 1999,33(3):95–110.

    CAS  PubMed  Google Scholar 

  29. Pfeifer GP, Dammann R: Methylation of the tumor suppressor gene RASSF1A in human tumors. Biochemistry (Mosc) 2005,70(5):576–583.

    Article  CAS  Google Scholar 

  30. Gustafson S, Zbuk KM, Scacheri C, Eng C: Cowden syndrome. Semin Oncol 2007,34(5):428–434.

    Article  CAS  PubMed  Google Scholar 

  31. Xing M: BRAF mutation in thyroid cancer. Endocr Relat Cancer 2005,12(2):245–262.

    Article  CAS  PubMed  Google Scholar 

  32. 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(8):625–627.

    Article  CAS  PubMed  Google Scholar 

  33. Xing M, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, Robenshtok E, Fagin JA, Puxeddu E, Fugazzola L, Czarniecka A, Jarzab B, O'Neill CJ, Sywak MS, Lam AK, Riesco-Eizaguirre G, Santisteban P, Nakayama H, Tufano RP, Pai SI, Zeiger MA, Westra WH, Clark DP, Clifton-Bligh R, Sidransky D, Ladenson PW, Sykorova V: Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA 2013,309(14):1493–1501.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW, Knauf JA, Basolo F, Zhu Z, Giannini R, Salvatore G, Fusco A, Santoro M, Fagin JA, Nikiforov YE: BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J Clin Endocrinol Metab 2003,88(11):5399–5404.

    Article  CAS  PubMed  Google Scholar 

  35. 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.

    CAS  PubMed  Google Scholar 

  36. Namba H, Nakashima M, Hayashi T, Hayashida N, Maeda S, Rogounovitch TI, Ohtsuru A, Saenko VA, Kanematsu T, Yamashita S: Clinical implication of hot spot BRAF mutation, V599E, in papillary thyroid cancers. J Clin Endocrinol Metab 2003,88(9):4393–4397.

    Article  CAS  PubMed  Google Scholar 

  37. Xing M, Tufano RP, Tufaro AP, Basaria S, Ewertz M, Rosenbaum E, Byrne PJ, Wang J, Sidransky D, Ladenson PW: Detection of BRAF mutation on fine needle aspiration biopsy specimens: a new diagnostic tool for papillary thyroid cancer. J Clin Endocrinol Metab 2004, 89: 2867–2872.

    Article  CAS  PubMed  Google Scholar 

  38. 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, et al.: Mutations of the BRAF gene in human cancer. Nature 2002,417(6892):949–954.

    Article  CAS  PubMed  Google Scholar 

  39. Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP, Giordano TJ, Biddinger PW, Nikiforov YE: Correlation between genetic alterations and microscopic features, clinical manifestations, and prognostic characteristics of thyroid papillary carcinomas. Am J Surg Pathol 2006,30(2):216–222.

    Article  PubMed  Google Scholar 

  40. Santoro A, Pannone G, Carosi MA, Francesconi A, Pescarmona E, Russo GM, Feola A, Losito S, Franco R, Nappi L, Aquino G, De Rosa G, Di Domenico M, Bufo P: BRAF mutation and RASSF1A expression in thyroid carcinoma of southern Italy. J Cell Biochem 2013,114(5):1174–82.

    Article  CAS  PubMed  Google Scholar 

  41. Xing M: BRAFV600E mutation and papillary thyroid cancer: chicken or egg? J Clin Endocrinol Metab 2012,97(7):2295–2298.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Howell GM, Carty SE, Armstrong MJ, Lebeau SO, Hodak SP, Coyne C, Stang MT, McCoy KL, Nikiforova MN, Nikiforov YE, Yip L: Both BRAF V600E mutation and older age (≥65 years) are associated with recurrent papillary thyroid cancer. Ann Surg Oncol 2011,18(13):3566–3571.

    Article  PubMed  Google Scholar 

  43. Xing M: BRAF mutation in papillary thyroid cancer: pathogenic role, molecular bases, and clinical implications. Endocr Rev 2007 Dec,28(7):742–762.

    Article  CAS  PubMed  Google Scholar 

  44. Lee JI, Jang HW, Kim SW, Kim JW, Oh YL, Chung JH: BRAFV600E mutation in fine-needle aspiration aspirates: Association with poorer prognostic factors in larger papillary thyroid carcinomas. Head Neck 2013,35(4):548–53.

    Article  PubMed  Google Scholar 

  45. Dhillon AS, Kolch W: Oncogenic B-Raf mutations: crystal clear at last. Cancer Cell 2004, 5: 303–330.

    Article  CAS  PubMed  Google Scholar 

  46. Chong H, Vikis HG, Guan KL: Mechanisms of regulating the Raf kinase family. Cell Signal 2003,15(5):463–469.

    Article  CAS  PubMed  Google Scholar 

  47. Peyssonnaux C, Eychene A: The Raf/MEK/ERK pathway: new concepts of activation. Biol. Cell. 2001, 93: 53–62.

    Article  CAS  PubMed  Google Scholar 

  48. Xing M: Recent advances in molecular biology of thyroid cancer and their clinical implications. Otolaryngol Clin North Am 2008,41(6):1135–1146.

    Article  PubMed Central  PubMed  Google Scholar 

  49. Saji M, Ringel MD: The PI3K-Akt-mTOR pathway in initiation and progression of thyroid tumors. Mol Cell Endocrinol 2010,28(321(1)):20–28.

    Article  Google Scholar 

  50. Sedliarou I, Saenko V, Lantsov D, Rogounovitch T, Namba H, Abrosimov A, Lushnikov E, Kumagai A, Nakashima M, Meirmanov S, Mine M, Hayashi T, Yamashita S: The BRAFT1796A transversion is a prevalent mutational event in human thyroid microcarcinoma. Int J Oncol 2004,25(6):1729–35.

    CAS  PubMed  Google Scholar 

  51. Kwak JY, Kim EK, Kim HJ, Kim MJ, Son EJ, Moon HJ: How to combine ultrasound and cytological information in decision making about thyroid nodules. Eur Radiol 2009,19(8):1923–1931.

    Article  PubMed  Google Scholar 

  52. Kwak JY, Koo H, Youk JH, Kim MJ, Moon HJ, Son EJ, Kim EK: Value of US correlation of a thyroid nodule with initially benign cytologic results. Radiology 2010,254(1):292–300.

    Article  PubMed  Google Scholar 

  53. Haberal AN, Toru S, Ozen O, Arat Z, Bilezikçi B: Diagnostic pitfalls in the evaluation of fine needle aspiration cytology of the thyroid: correlation with histopathology in 260 cases. Cytopathology 2009,20(2):103–108.

    Article  CAS  PubMed  Google Scholar 

  54. Ylagan LR, Farkas T, Dehner LP: Fine needle aspiration of the thyroid: a cytohistologic correlation and study of discrepant cases. Thyroid 2004,14(1):35–41.

    Article  PubMed  Google Scholar 

  55. Abubaker J, Jehan Z, Bavi P, Sultana M, Al-Harbi S, Ibrahim M, Al-Nuaim A, Ahmed M, Amin T, Al-Fehaily M, Al-Sanea O, Al-Dayel F, Uddin S, Al-Kuraya KS: Clinicopathological analysis of papillary thyroid cancer with PIK3CA alterations in a Middle Eastern population. J Clin Endocrinol Metab 2008,93(2):611–618.

    Article  CAS  PubMed  Google Scholar 

  56. 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):71–77.

    Article  CAS  PubMed  Google Scholar 

  57. Garcia-Rostan G, Zhao H, Camp RL, Pollan M, Herrero A, Pardo J, Wu R, Carcangiu ML, Costa J, Tallini G: ras mutations are associated with aggressive tumor phenotypes and poor prognosis in thyroid cancer. J Clin Oncol 2003,21(17):3226–3235.

    Article  CAS  PubMed  Google Scholar 

  58. Puxeddu E, Moretti S, Elisei R, Romei C, Pascucci R, Martinelli M, Marino C, Avenia N, Rossi ED, Fadda G, Cavaliere A, Ribacchi R, Falorni A, Pontecorvi A, Pacini F, Pinchera A, Santeusanio F: BRAF(V599E) mutation in the leading genetic event in adult sporadic papillary thyroid carcinomas. J Clin Endocrinol Metab 2004, 89: 2414–2420.

    Article  CAS  PubMed  Google Scholar 

  59. Ezzat S, Zheng L, Kolenda J, Safarian A, Freeman JL, Asa SL: Prevalence of activating ras mutations in morphologically characterized thyroid nodules. Thyroid 1996,6(5):409–416.

    Article  CAS  PubMed  Google Scholar 

  60. Elsheikh TM, Asa SL, Chan JK, DeLellis RA, Heffess CS, LiVolsi VA, Wenig BM: Interobserver and intraobserver variation among experts in the diagnosis of thyroid follicular lesions with borderline nuclear features of papillary carcinoma. Am J Clin Pathol 2008,130(5):736–744.

    Article  PubMed  Google Scholar 

  61. Lloyd RV, Erickson LA, Casey MB, Lam KY, Lohse CM, Asa SL, Chan JK, DeLellis RA, Harach HR, Kakudo K, LiVolsi VA, Rosai J, Sebo TJ, Sobrinho-Simoes M, Wenig BM, Lae ME: Observer variation in the diagnosis of follicular variant of papillary thyroid carcinoma. Am J Surg Pathol 2004,28(10):1336–1340.

    Article  PubMed  Google Scholar 

  62. Rabes HM, Demidchik EP, Sidorow JD, Lengfelder E, Beimfohr C, Hoelzel D, Klugbauer S: Pattern of radiation-induced RET and NTRK1 rearrangements in 191 post-chernobyl papillary thyroid carcinomas: biological, phenotypic, and clinical implications. Clin Cancer Res 2000,6(3):1093–1103.

    CAS  PubMed  Google Scholar 

  63. Ciampi R, Nikiforov YE: RET/PTC rearrangements and BRAF mutations in thyroid tumorigenesis. Endocrinology 2007,148(3):936–941.

    Article  CAS  PubMed  Google Scholar 

  64. Eberhardt NL, Grebe SK, McIver B, Reddi HV: The role of the PAX8/PPARgamma fusion oncogene in the pathogenesis of follicular thyroid cancer. Mol Cell Endocrinol 2010,321(1):50–56.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  65. 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, Höfler H, Williams ED: Gene rearrangement and Chernobyl related thyroid cancers. Br J Cancer 2000,82(2):315–322.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  66. Santoro M, Melillo RM, Fusco A: RET/PTC activation in papillary thyroid carcinoma: European Journal of Endocrinology Prize Lecture. Eur J Endocrinol 2006,155(5):645–653.

    Article  CAS  PubMed  Google Scholar 

  67. Nikiforova MN, Stringer JR, Blough R, Medvedovic M, Fagin JA, Nikiforov YE: Proximity of chromosomal loci that participate in radiation-induced rearrangements in human cells. Science 2000,290(5489):138–141.

    Article  CAS  PubMed  Google Scholar 

  68. 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):3603–3610.

    Article  CAS  PubMed  Google Scholar 

  69. Tallini G, Santoro M, Helie M, Carlomagno F, Salvatore G, Chiappetta G, Carcangiu ML, Fusco A: RET/PTC oncogene activation defines a subset of papillary thyroid carcinomas lacking evidence of progression to poorly differentiated or undifferentiated tumor phenotypes. Clin Cancer Res 1998,4(2):287–294.

    CAS  PubMed  Google Scholar 

  70. Thomas GA, Bunnell H, Cook HA, Williams ED, Nerovnya A, Cherstvoy ED, Tronko ND, Bogdanova TI, Chiappetta G, Viglietto G, Pentimalli F, Salvatore G, Fusco A, Santoro M, Vecchio G: High prevalence of RET/PTC rearrangements in Ukrainian and Belarussian post-Chernobyl thyroid papillary carcinomas: a strong correlation between RET/PTC3 and the solid-follicular variant. J Clin Endocrinol Metab 1999,84(11):4232–4238.

    CAS  PubMed  Google Scholar 

  71. 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(5):2318–2326.

    Article  CAS  PubMed  Google Scholar 

  72. Liu Z, Hou P, Ji M, Guan H, Studeman K, Jensen K, Vasko V, El-Naggar AK, Xing M: Highly prevalent genetic alterations in receptor tyrosine kinases and phosphatidylinositol 3-kinase/akt and mitogen-activated protein kinase pathways in anaplastic and follicular thyroid cancers. J Clin Endocrinol Metab 2008,93(8):3106–3116.

    Article  CAS  PubMed  Google Scholar 

  73. Lee JJ, Geli J, Larsson C, Wallin G, Karimi M, Zedenius J, Höög A, Foukakis T: Gene-specific promoter hypermethylation without global hypomethylation in follicular thyroid cancer. Int J Oncol 2008,33(4):861–869.

    PubMed  Google Scholar 

  74. Yoshikawa H: DNA methylation and cancer. Gan To Kagaku Ryoho 2007,34(2):145–149.

    CAS  PubMed  Google Scholar 

  75. Hu S, Liu D, Tufano RP, Carson KA, Rosenbaum E, Cohen Y, Holt EH, Kiseljak-Vassiliades K, Rhoden KJ, Tolaney S, Condouris S, Tallini G, Westra WH, Umbricht CB, Zeiger MA, Califano JA, Vasko V, Xing M: Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. Int J Cancer 2006,119(10):2322–2329.

    Article  CAS  PubMed  Google Scholar 

  76. Russo VE, Martienssen RA, Riggs AD: Epigenetic mechanisms of gene regulation. New York: Cold Spring Harbor Laboratory Press; 1996.

    Google Scholar 

  77. Meehan RR, Lewis JD, Bird AP: Characterizatin of MeCP2, a vertebrate DNA binding protein with affinity for methylated DNA. Nucleic Acids Res 1992, 20: 5085–5092.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  78. Lewis JD, Meehan RR, Henzel WJ, Maurer-Fogy I, Jeppesen P, Klein F, Bird A: Purification, sequence and cellular localization of a novel chromosomal protein that binds to methylated DNA. Cell 1992, 69: 905–914.

    Article  CAS  PubMed  Google Scholar 

  79. Smith JA, Fan CY, Zou C, Bodenner D, Kokoska MS: Methylation Status of Genes in Papillary Thyroid Carcinoma. Arch Otolaryngol Head Neck Surg 2007,133(10):1006–1011.

    Article  PubMed  Google Scholar 

  80. Schagdarsurengin U, Gimm O, Hoang-Vu C, Dralle H, Pfeifer GP, Dammann R: Frequent Epigenetic Silencing of the CpG Island Promoter of RASSF1A in Thyroid Carcinoma. Cancer Res 2002, 62: 3698–3701.

    CAS  PubMed  Google Scholar 

  81. Qi JH, Ebrahem Q, Moore N, Murphy G, Claesson-Welsh L, Bond M, Baker A, Anand-Apte B: A novel function for tissue inhibitor of metalloproteinases-3 (TIMP-3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med. 2003, 9: 407–415.

    Article  CAS  PubMed  Google Scholar 

  82. Baylin SB, Ohm JE: Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 2006,6(2):107–116.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from the Polish Ministry of Science and Higher Education (no. N N402 466739).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Anhelli Syrenicz.

Additional information

Competing interests

The authors have non-financial competing interests (political, personal, religious, ideological, academic, intellectual, commercial or any other) to declare in relation to this manuscript.

Authors’ contributions

Prof AS have made substantial contributions to conception and design, acquisition of data and analysis and interpretation of data; have been also involved in drafting the manuscript, have given final approval of the version to be published; MK - have made substantial contributions to conception and design, acquisition of data and analysis and interpretation of data; have been also involved in drafting the manuscript, AC - have been also involved in drafting the manuscript, AS - participated in the sequence alignment and drafted the manuscript, ABK - carried out the molecular genetic studies, MP - carried out the molecular genetic studies. All authors read and approved the final manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Syrenicz, A., Koziołek, M., Ciechanowicz, A. et al. New insights into the diagnosis of nodular goiter. Thyroid Res 7, 6 (2014). https://doi.org/10.1186/1756-6614-7-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1756-6614-7-6

Keywords