ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Oncogenes and tumor suppressor genes in thyroid nodules and nonmedullary thyroid cancer

Oncogenes and tumor suppressor genes in thyroid nodules and nonmedullary thyroid cancer
Literature review current through: Jan 2024.
This topic last updated: Dec 06, 2023.

INTRODUCTION — A thyroid tumor develops when the growth of a single thyroid epithelial cell escapes from the normal mechanisms regulating cell division and gains a selective growth advantage. Continued growth leads, in time, to a clinically evident tumor mass. Unregulated cell division can result from mutations in both oncogenes and tumor suppressor genes.

Although thyroid epithelial tumors arise from the same cell type, they have diverse clinical characteristics. Most produce less thyroid hormone than normal thyroid tissue. Most are benign, but some are slow-growing cancers, and a few are highly aggressive cancers. Most thyroid tumors are sporadic and not familial.

An understanding of the mutations of the proto-oncogenes and tumor suppressor genes that occur in these tumors may explain the diverse clinical characteristics of thyroid tumors, provide diagnostic information, and direct therapy. The molecular profiling of thyroid neoplasms is commercially available to help distinguish benign from malignant thyroid nodules using DNA, mRNA, and microRNA from fine-needle aspiration (FNA) biopsy samples. The accuracy of this molecular testing continues to improve. Molecular profiling is now clinically indicated to direct the initial therapy of biopsy proven anaplastic thyroid cancers. Furthermore, identification of RET and TRK rearrangements can now direct therapy in aggressive differentiated thyroid cancers.

This topic review will discuss the tumorigenic gene changes found in thyroid epithelial (nonmedullary) tumors. A summary of these changes is found in the table (table 1). Medullary thyroid cancer is included in this table for comparison but is discussed elsewhere, as are the characteristics and treatment of the specific tumors. (See "Medullary thyroid cancer: Clinical manifestations, diagnosis, and staging" and "Medullary thyroid cancer: Surgical treatment and prognosis" and "Papillary thyroid cancer: Clinical features and prognosis" and "Follicular thyroid cancer (including oncocytic carcinoma of the thyroid)" and "Anaplastic thyroid cancer".)

AUTONOMOUSLY FUNCTIONING THYROID ADENOMAS — Autonomously functioning thyroid adenomas (or nodules) are benign tumors that produce thyroid hormone. Clinically, they present as a single nodule that is hyperfunctioning ("hot") on thyroid radionuclide scan, sometimes causing hyperthyroidism. (See "Diagnostic approach to and treatment of thyroid nodules".)

Many of these tumors are caused by somatic mutations in genes that code for two proteins of the thyroid-stimulating hormone (TSH) stimulation cascade (figure 1):

The TSH receptor

The alpha subunit of the guanyl nucleotide stimulatory protein (Gs)

Normally, TSH stimulates thyroid epithelial cells by binding to receptors on the plasma membrane of the cells. The signal is transduced by complexes formed between the alpha subunit of Gs and GTP in response to TSH-receptor binding. This results sequentially in stimulation of adenylyl cyclase activity, increased cyclic AMP production, and activation of protein kinases that stimulate thyroid cell division and hormone production. Adenylyl cyclase activity decreases when the GTP is hydrolyzed by the intrinsic GTPase activity of Gs-alpha. (See "Thyroid hormone synthesis and physiology".)

Activating mutations of the TSH receptor gene — Activating mutations of the TSH receptor produce constitutive activation of adenylyl cyclase in the absence of TSH [1,2]. The thyroid follicular cell with this TSH-receptor mutation divides and produces thyroid hormone without TSH stimulation, eventually becoming clinically recognized as a hot nodule. Although TSH (via the TSH receptor) also stimulates the inositol triphosphate pathway, most activating mutations of the TSH receptor do not stimulate signaling through this pathway [2-4].

Among patients with an autonomously functioning thyroid adenoma, the frequency of TSH-receptor mutations in the adenoma varies from approximately 5 to 80 percent [2-7]. The large differences in frequency may be related to the sensitivity of the methods used to screen for the mutations or interpatient differences. Since the mutations are scattered throughout the receptor, studies of the entire receptor are most likely to identify a mutation. (In rare families, germline mutations in the TSH receptor cause hereditary hyperthyroidism, initially with a diffuse goiter but ultimately with a nodular goiter with multiple hot nodules [8]).

Gs-alpha gene mutations that activate adenylyl cyclase — Gs-alpha mutations that lead to adenylyl cyclase activation have the same effects on cell division and thyroid hormone production as those in the TSH receptor (figure 1) [9,10]. These mutations code for changes in either an arginine residue at position 201 or a glutamine residue at position 227 of the molecule. The mutant Gs-alpha molecules have decreased GTPase activity; as a result, the Gs-alpha-GTP complex is more stable, leading to constitutive activation of adenylyl cyclase.

Among autonomously functioning thyroid adenomas, these mutations are less common than TSH-receptor mutations, occurring in 0 to 25 percent [7,9,11]. Gs-alpha mutations are also found in other endocrine tumors such as somatotroph adenomas [11]. The occurrence of these mutations during embryogenesis causes some of the manifestations of the McCune-Albright syndrome, including hyperthyroidism (with no goiter or a diffuse or multinodular goiter) [12-15] (see "Definition, etiology, and evaluation of precocious puberty", section on 'McCune-Albright syndrome'). Germline Gs-alpha mutations that activate adenylyl cyclase have not been reported and are probably lethal.

No autonomously functioning thyroid adenomas have been identified with mutations in both proteins, and some have no mutations in either. There are no clinical characteristics that distinguish patients with TSH-receptor mutations, Gs-alpha mutations, and no detectable mutations.

OTHER BENIGN THYROID NODULES — The most common benign thyroid tumors are the nodules of multinodular goiters (colloid nodules) and follicular adenomas. The oncogene changes accounting for these benign thyroid nodules are not well delineated.

Multinodular goiters — If a tumor arises from a single cell, it is clonal. If it arises from all or most cells in a tissue, it is polyclonal. The nodules of multinodular goiters seemed polyclonal by morphologic studies [16], but many are clonal by molecular studies [17,18]. The two types of studies, taken together, indicate that regions of phenotypic or functional heterogeneity can develop within a clonal thyroid nodule.

Multinodular goiters are occasionally familial, which means that the patient has at least one germline mutation. One familial form of nontoxic multinodular goiter has been linked to DNA markers on chromosome 14q [19], but the etiologic gene is not known.

Follicular adenomas — As anticipated from pathologic studies, follicular adenomas are clonal [20]. Point mutations in the HRAS, KRAS, and NRAS proto-oncogenes have been identified in both follicular adenomas and follicular cancers [21-25].

Approximately one-quarter of sporadic follicular adenomas have a hemizygous deletion of a chromosome region containing PTEN, the tumor suppressor gene in which germline defects cause Cowden's disease (multiple hamartomas and breast, thyroid, and other tumors). Because defects in both alleles of a tumor suppressor gene are needed for tumor formation, it remains unclear whether inactivation of PTEN itself or a different tumor suppressor gene on chromosome 10q is a cause of sporadic follicular thyroid adenomas. (See "PTEN hamartoma tumor syndromes, including Cowden syndrome".)

Chromosomal translocations involving 19q13 and 2p21 are found in follicular adenomas and do not co-occur in the same neoplasm. Based upon analysis of a small number of follicular adenomas, the 19q13 rearranged gene is ZNF331 (HUGO nomenclature) [26], and the 2q21 rearranged gene is referred to as thyroid adenoma-associated gene (THADA) [27]. THADA may be a death receptor-interacting protein. Both genes may be rearranged with different partners.

PAPILLARY THYROID CANCER — The Cancer Genome Atlas summarizes the most common genetic alterations in papillary thyroid cancer [28]. The genetic alterations of papillary thyroid cancer activate the mitogen-activated protein kinase (MAPK) pathway that promotes cell division. Rearrangements of the genes coding for RET and NTRK1 tyrosine kinases, activating mutations of BRAF, and activating mutations of RAS are sequential components leading to activation of MAPK (figure 2). Additional drivers include anaplastic lymphoma kinase (ALK) rearrangements, EIF1AX mutations, and mutations of the promoter region of the telomerase reverse transcriptase gene (TERT) [28]. In general, any given papillary thyroid cancer carries only a single one of these genetic changes [29]. However, approximately 9 percent of papillary thyroid cancers harbor both a TERT promoter mutation plus either a BRAF or RAS activating mutation. These tumors are more aggressive than those papillary thyroid cancers carrying only a single driver [30].

The RAS mutations are referred to as weak drivers since they are not unique to thyroid cancer and are frequently found in benign thyroid neoplasms. In contrast, the rearrangements of RET, NTRK, and ALK, as well as mutations of BRAF, EIF1AX, and the TERT promoter are referred to as strong drivers. When strong drivers are identified in thyroid nodules, the nodule is almost always malignant. These concepts apply to the DNA analysis of thyroid nodules that are currently offered by a number of different commercial laboratories.

RET and NTRK — Papillary thyroid cancers are associated with rearrangements of three different transmembrane tyrosine kinase genes: RET, NTRK1, and NTRK3. These rearrangements result in the production of chimeric proteins with constitutive tyrosine kinase activity that contributes to the development of the malignant phenotype (figure 3). The chimeric genes resulting from RET rearrangements are referred to as RET/PTC and those resulting from NTRK1 rearrangements as TRK (table 2).

RET codes for a glial cell line-derived neurotrophic factor receptor with tyrosine kinase activity. Germline RET mutations cause the multiple endocrine neoplasia type 2 (MEN2) syndromes. (See "Classification and genetics of multiple endocrine neoplasia type 2".)

NTRK1 and NTRK3 code for a nerve growth factor receptor that also has tyrosine kinase activity [31].

Neither of these genes is expressed in normal thyroid epithelial cells. As a result, patients with MEN2 syndromes do not have an increased incidence of papillary thyroid cancer.

In somatic rearrangements of the RET or NTRK genes, the attachment of new genetic material to the 5' end of the tyrosine kinase domain of the gene leads to a constitutive increase in tyrosine kinase activity that activates growth pathways and may be sufficient to cause the papillary cancer phenotype (figure 3). This etiologic role has been confirmed in transgenic mice, in which an activated RET gene in thyroid epithelial cells causes papillary cancer [32,33].

The frequency of gene rearrangements involving RET and NTRK1 in papillary thyroid cancers has varied in different studies. In adults, approximately 40 percent of sporadic papillary cancers have these rearrangements, with those involving RET being approximately three times more common [34]. The incidence of RET rearrangements in papillary cancers is higher in children (approximately 60 percent) [35,36] and is approximately 80 percent in the papillary cancers of children exposed to external x-irradiation and those exposed to radiation after the Chernobyl nuclear accident in 1986 [35,37].

Over 20 chimeric RET and TRK genes have been identified in papillary thyroid cancer. Six of the more common rearrangements are summarized in the table (table 2). Their clinical importance seems to be similar [34,35,37-43]. For those few patients with these rearrangements and aggressive disease, directed tyrosine kinase inhibitors are now available [44-46]. (See "Differentiated thyroid cancer refractory to standard treatment: Systemic therapy", section on 'Mutation-specific kinase inhibitor' and "TRK fusion-positive cancers and TRK inhibitor therapy", section on 'Treatment with TRK inhibitors'.)

BRAF mutations — The BRAF isoform of RAF has been implicated in the pathogenesis of papillary thyroid cancer, but not of benign or follicular neoplasms [29]. The RAF proteins are serine-threonine kinases that activate the RAF/MEK/MAPK signaling pathway. The T1799A mutation of the BRAF gene, which was originally found in over 50 percent of malignant melanomas and a smaller percentage of colon cancers, occurs in 29 to 69 percent of papillary thyroid cancers [29,47-49]. The predicted protein product BRAF V600E has increased basal kinase activity and transforms NIH3T3 cells with a higher efficiency than does the wild-type BRAF. Transgenic mice expressing this mutation develop papillary thyroid cancer [50].

In one report, BRAF mutations were found in 219 of 500 patients with papillary thyroid cancer (44 percent) [51]. BRAF V600E, the most prevalent mutation, was associated with invasive tumor growth and the follicular variant of papillary cancer.

BRAF mutations may confer a worse clinical prognosis than for papillary thyroid cancer without the BRAF mutation [52-54]. Recurrence occurs more frequently when BRAF mutations are present [55]. In addition, BRAF mutations are associated with extrathyroidal invasion, lymph node metastases, and advanced tumor stage at initial surgery, although for these findings, conventional histologic evaluation was as effective in predicting outcome as was the presence of the BRAF mutation [53,56]. Interestingly, some lymph node metastases may acquire additional BRAF mutations [57,58].

BRAF point mutations appear to be much less common in childhood thyroid cancers, including those that developed after the Chernobyl accident [59,60]. However, an oncogene (AKAP9-BRAF) that derives from a paracentric inversion of the long arm of chromosome 7 has been identified in radiation-associated papillary thyroid cancers developing after a short latency [61].

Because BRAF mutations are not found in follicular carcinomas and benign thyroid nodules, identification of a BRAF mutation may aid in the diagnosis and management of papillary thyroid cancer. In a preliminary report, BRAF mutations were detected in blood samples from patients with papillary thyroid cancer [62]. BRAF positivity correlated with the presence of residual or metastatic disease. In addition, the detection of BRAF gene mutations in fine-needle aspiration biopsy specimens may prove useful in the assessment of follicular lesions. This topic is discussed in detail elsewhere.

In thyroid cancer cell lines harboring activated BRAF, the addition of selective inhibitors of BRAF inhibited proliferation of BRAF mutant cell lines, suggesting a possible role of such inhibitors in the treatment of patients with papillary thyroid cancer and BRAF mutations [63,64].

RAS mutations — Although activating RAS mutations are found in follicular adenomas, they have also been reported in follicular variant of papillary thyroid cancers [65]. NRAS mutations are more common than HRAS mutations [66].

TERT promoter mutations — Two promoter region mutations of the TERT have been reported in many malignancies. The chr5:1,295,228C>T (C228T) mutation was found in approximately 10 percent of sporadic papillary thyroid cancers, and the chr5:1,295250C>T (C250T) was found in approximately 2 percent of sporadic papillary thyroid cancers [28]. When TERT promoter mutations coexist with either the V600E BRAF mutation or with a RAS mutation, the thyroid cancers are more aggressive [67].

Other genetic abnormalities — MicroRNAs (miRNAs) modify gene expression by binding to specific targets in the 3' untranslated region and are being implicated in tumorigenesis of a variety of tissues. Several miRNAs (miR-221, miR-222, miR-146, and others) are upregulated in papillary thyroid cancer and may contribute to tumorigenesis [68]. The expression profiles in the TCGA database confirmed the upregulation of these miRNAs as well as miR-181a/b/d, miR-34a, and miR-424 [28]. It also identified downregulation of miR-363, miR-138, mir-363, miR-20b, miR-195, and miR-152 [69]. In addition, mutations were identified in PPM1D and CHEK2, both DNA repair genes [28].

Common germline sequence variants — Large population studies have identified multiple different single nucleotide polymorphisms (SNPs) that are associated with a clinically modest, but statistically significant, increased risk of well-differentiated thyroid carcinoma, including the following:

A G/C heterozygosity within the precursor of miRNA-146a predisposes to papillary thyroid cancer (odds ratio [OR] 1.62) [70]. This polymorphism alters the miRNA sequence and induces miRNA overexpression with new miRNA forms that alter target gene expression [71].

SNPs at 9q22.33 that are close to FOXE1 (TTF2) and at 14q13.3 that are close to NKX2-1 (TTF1) [72]. Both FOXE1 and NKX2-1 are known to regulate gene transcription within the thyroid cells.

SNPs found in CHEK2, NBS1, XRCC3, and the promoter of RAD51 have all been associated with an increased risk of differentiated thyroid cancer, which are mostly papillary thyroid cancers [73-76].

Methylation often decreases gene expression and may contribute to tumorigenesis. Increased methylation of several tumor suppressor genes (TIMP3, SLC5A8, DAPK, and RARbeta2) is associated with features of papillary thyroid cancer aggressiveness [77,78].

There may be an increased incidence of papillary cancer in patients with familial adenomatous polyposis [79]. This disorder is caused by germline mutations of the APC gene, a tumor suppressor gene of unknown function (see "Epidemiology and risk factors for colorectal cancer"). In comparison, somatic mutations of this gene are uncommon in sporadic papillary cancers [80]. The papillary thyroid cancers of patients with familial adenomatous polyposis may have rearrangements of RET [81].

Papillary thyroid cancers are usually sporadic, but approximately 5 percent have a familial predisposition [82-85]. Surprisingly, powerful genetic linkage methodologies have been relatively ineffective in identifying the predisposing gene abnormalities. A single kindred has a mutation in a long-term enhancer element in 4Q32 that predisposes to thyroid cancer [86]. An increased incidence of papillary thyroid cancer is found in patients with congenital hypothyroidism with huge goiters caused by thyroglobulin gene mutations [87]. Other potential regions of linkage include chromosome regions 1q21 [88], 8q24 [89], and 19p13.2.

FOLLICULAR THYROID CANCER — Follicular thyroid cancers tend to be caused by mutations that signal through the AKT pathway (figure 4) [90]. Since activating RAS mutations occur in both follicular and papillary thyroid cancer and participate in both the MAPK and AKT pathways, this is an oversimplification.

RAS mutations — Approximately 30 percent of follicular thyroid cancers have RAS mutations with the NRAS mutations being more common than HRAS and KRAS [66]. Overexpression of normal c-myc and c-fos genes, as well as mutations of HRAS, NRAS, and KRAS proto-oncogenes, are found in follicular adenomas, follicular cancers, and occasionally papillary cancers [21-23,25,91]. These abnormalities may confer a growth-promoting effect that is not specific for tumor type.

PAX8 fusion — A chromosomal translocation has been identified in some follicular cancers that distinguishes them from papillary cancers and follicular adenomas [92]. The translocation (t(2;3)[q13;p25]) results in fusion of part of the DNA-binding segment of the PAX8 gene and the peroxisome proliferator-activated receptor gamma 1 (PPAR-gamma-1) gene; PAX8 is a thyroid transcription factor, and PPAR-gamma-1 is a transcription factor that stimulates cell differentiation and inhibits cell growth.

The product of the fusion gene blocks the action of the PPAR-gamma-1, an effect that might inhibit cell differentiation and stimulate cell growth. In one study, the translocation was found in 5 of 8 follicular cancers and 0 of 20 follicular adenomas [92], and in another study, it was found in 5 of 9 cancers and 2 of 16 adenomas, suggesting that it may be useful for distinguishing between follicular cancers and follicular adenomas [93].

TERT promoter mutations — The two telomerase reverse transcriptase (TERT) promoter mutations described above in the discussion of papillary thyroid cancer have also been described in follicular thyroid cancer [28,67]. The chr5:1,295,228C>T (C228T) mutation was found in approximately 30 percent of follicular thyroid cancers, and the chr5:1,295250C>T (C250T) was found in approximately 5 percent of follicular thyroid cancers. Thyroid cancers harboring both a TERT promoter mutation and a RAS mutation are more aggressive [30].

PTEN — Germline mutations in PTEN, a tumor suppressor gene, have been described in a variety of rare syndromes that are collectively known as the PTEN hamartoma tumor syndromes (PHTS). Cowden syndrome is the best described syndrome within PHTS. In addition to benign follicular adenomas, individuals with Cowden syndrome have an approximately 70-fold increased incidence of thyroid cancer relative to the general population [94]. Both follicular and papillary neoplasms may occur. (See "PTEN hamartoma tumor syndromes, including Cowden syndrome", section on 'Thyroid'.)

RASSF1A — Hypermethylation of RASSF1A, a known tumor suppressor gene, has been described in 75 percent of follicular thyroid cancers (9 of 12) as well as in a smaller percentage of benign adenomas (44 percent), and papillary thyroid cancers (20 percent) [95]. This may be an early step in thyroid epithelial tumorigenesis.

HÜRTHLE CELL TUMORS — Some benign and malignant thyroid tumors are found to have an increased number of mitochondria and are referred to as oxyphil or Hürthle cell tumors. Hürthle cell carcinomas were once thought to be a variant of follicular thyroid carcinomas, but more recent analyses demonstrate that these are much more complex tumors with both nuclear and mitochondrial DNA alterations. Multiple driver mutations have been identified and include mutations of EIF1AX, MADCAM1, OR4L1, and ATXN1 in addition to multiple others. Whole chromosome duplication of chromosomes 5 and 7 occurs as well as widespread loss of heterozygosity. In the mitochondrial genome, there are a large number of disruptive mutations of the protein-coding and tRNAs encoding regions [96].

NONINVASIVE FOLLICULAR THYROID NEOPLASM WITH PAPILLARY-LIKE NUCLEAR FEATURES (NIFTP) — An indolent variant of a follicular cell-derived neoplasm has been identified, termed noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP) [97]. Preliminary studies suggest that the most frequent gene mutations are activating RAS mutations [98,99]. (See "Papillary thyroid cancer: Clinical features and prognosis", section on 'Variant forms'.)

ANAPLASTIC AND POORLY DIFFERENTIATED THYROID CANCER — Anaplastic and poorly differentiated thyroid cancers are the most aggressive thyroid cancers. Combined pathology/molecular studies have identified regions within the anaplastic thyroid cancer that are well differentiated and contain the same BRAF or RAS mutation as the regions of anaplastic thyroid cancer [100]. More recent genetic analyses suggest that these tumors arise from well-differentiated thyroid cancers with BRAF V600E or RAS mutations that have accumulated a greater mutational burden [101]. The differentiated thyroid cancers [100-102] with RET, NTRK [101,103], or PAX8-PPAR-gamma-1 rearrangements do not appear to progress to these aggressive malignancies [101,103].

Subsequent mutational burden leading to aggressive behavior and found in both poorly differentiated thyroid cancer and anaplastic thyroid cancer occur in the TERT promoter, P53, EIF1AX, PI3K/AKT/MTOR pathway effectors, the SWI/SNF subunits, histone methyltransferases, exon 3 of the beta-catenin gene, and the anaplastic lymphoma kinase gene (ALK) [101,104-106]. Mutations of the p53 tumor suppressor gene that lead to production of an inactive p53 protein occur in many anaplastic thyroid cancers, but not well-differentiated thyroid cancers [101,107]. The protein product of p53 is a transcription factor that regulates both apoptosis and the cell cycle.

Therapies targeted at specific gene mutations have emerged, so that molecular profiling of anaplastic thyroid cancer is recommended by the American Thyroid Association at the time of diagnosis [108]. (See "Anaplastic thyroid cancer", section on 'Molecular testing' and "Anaplastic thyroid cancer", section on 'BRAF V600E mutation identified'.)

SUMMARY

General principles – Our knowledge of the molecular pathogenesis of benign and malignant thyroid neoplasia is evolving. The molecular profiling of thyroid neoplasms is commercially available to help distinguish benign from malignant thyroid nodules using DNA, mRNA, and microRNA from fine-needle aspiration (FNA) biopsy samples. The accuracy of this molecular testing continues to improve. Molecular profiling is now clinically indicated to direct the initial therapy of biopsy-proven anaplastic thyroid cancers. Furthermore, identification of RET and TRK rearrangements can now direct therapy in aggressive differentiated thyroid cancers. (See 'Introduction' above.)

Autonomously functioning adenomas – Autonomously functioning thyroid nodules are benign tumors that produce thyroid hormone. Many of these tumors are caused by somatic mutations in genes that code for the thyroid-stimulating hormone (TSH) receptor or the alpha subunit of the guanyl nucleotide stimulatory protein (Gs). (See 'Autonomously functioning thyroid adenomas' above.)

Other benign nodules – The most common benign thyroid tumors are the nodules of multinodular goiters (colloid nodules) and follicular adenomas. The oncogene changes accounting for these benign thyroid nodules are not well delineated. (See 'Other benign thyroid nodules' above.)

Papillary thyroid cancer – Papillary thyroid cancers frequently carry gene mutations and rearrangements that lead to activation of the mitogen-activated protein kinase (MAPK) that promotes cell division. Rearrangements of RET and NTRK1, activating mutations of BRAF, and activating mutations of RAS are sequential components leading to activation of MAPK (figure 2). Any given papillary thyroid cancer carries only a single one of these genetic changes. Approximately 9 percent of papillary thyroid cancers harbor both a TERT promoter mutation and one of the driver mutations and are more aggressive. Molecular profiling of aggressive papillary thyroid cancer not responsive to conventional therapies is indicated, since directed therapies are available for those with rearrangements of RET or NTRK1. (See 'Papillary thyroid cancer' above and "Differentiated thyroid cancer refractory to standard treatment: Systemic therapy", section on 'Mutation-specific kinase inhibitor'.)

Follicular thyroid cancer – Follicular thyroid cancers frequently carry gene mutations that lead to activation of the AKT pathway (figure 4). PAX8/PPAR-gamma-1 rearrangements and mutations of HRAS, NRAS, and KRAS proto-oncogenes are found in follicular thyroid cancers and occasionally papillary cancers. (See 'Follicular thyroid cancer' above.)

Hürthle cell thyroid cancers – Once thought to be a variant of follicular thyroid cancer, Hürthle cell thyroid cancers are now recognized to be a distinct tumor type with complex alterations in both the nuclear and mitochondrial DNA. (See 'Hürthle cell tumors' above.)

NIFTPRAS mutations are common in noninvasive follicular thyroid neoplasms with papillary-like nuclear features (NIFTP), an indolent thyroid neoplasm. (See 'Noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP)' above and "Papillary thyroid cancer: Clinical features and prognosis", section on 'Variant forms'.)

Anaplastic thyroid cancer – Anaplastic thyroid cancers and poorly differentiated thyroid cancers are the most aggressive of the thyroid cancers. Many of these arise from well-differentiated thyroid carcinomas that originally contained BRAF or RAS driver mutations and subsequently acquired mutations conferring increased aggressiveness. Additional mutations include those in the TERT promoter region, p53, and PIK3CA. Molecular profiling is clinically indicated at the time of diagnosis to assist in directing therapy. (See 'Anaplastic and poorly differentiated thyroid cancer' above.)

  1. Porcellini A, Ruggiano G, Pannain S, et al. Mutations of thyrotropin receptor isolated from thyroid autonomous functioning adenomas confer TSH-independent growth to thyroid cells. Oncogene 1997; 15:781.
  2. Parma J, Duprez L, Van Sande J, et al. Somatic mutations in the thyrotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature 1993; 365:649.
  3. Paschke R, Tonacchera M, Van Sande J, et al. Identification and functional characterization of two new somatic mutations causing constitutive activation of the thyrotropin receptor in hyperfunctioning autonomous adenomas of the thyroid. J Clin Endocrinol Metab 1994; 79:1785.
  4. Porcellini A, Ciullo I, Laviola L, et al. Novel mutations of thyrotropin receptor gene in thyroid hyperfunctioning adenomas. Rapid identification by fine needle aspiration biopsy. J Clin Endocrinol Metab 1994; 79:657.
  5. Russo D, Arturi F, Wicker R, et al. Genetic alterations in thyroid hyperfunctioning adenomas. J Clin Endocrinol Metab 1995; 80:1347.
  6. Russo D, Arturi F, Suarez HG, et al. Thyrotropin receptor gene alterations in thyroid hyperfunctioning adenomas. J Clin Endocrinol Metab 1996; 81:1548.
  7. Führer D, Holzapfel HP, Wonerow P, et al. 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; 82:3885.
  8. Kopp P, van Sande J, Parma J, et al. Brief report: congenital hyperthyroidism caused by a mutation in the thyrotropin-receptor gene. N Engl J Med 1995; 332:150.
  9. O'Sullivan C, Barton CM, Staddon SL, et al. Activating point mutations of the gsp oncogene in human thyroid adenomas. Mol Carcinog 1991; 4:345.
  10. Suarez HG, du Villard JA, Caillou B, et al. gsp mutations in human thyroid tumours. Oncogene 1991; 6:677.
  11. Lyons J, Landis CA, Harsh G, et al. Two G protein oncogenes in human endocrine tumors. Science 1990; 249:655.
  12. Schwindinger WF, Francomano CA, Levine MA. Identification of a mutation in the gene encoding the alpha subunit of the stimulatory G protein of adenylyl cyclase in McCune-Albright syndrome. Proc Natl Acad Sci U S A 1992; 89:5152.
  13. Weinstein LS, Shenker A, Gejman PV, et al. Activating mutations of the stimulatory G protein in the McCune-Albright syndrome. N Engl J Med 1991; 325:1688.
  14. Malchoff CD, Reardon G, MacGillivray DC, et al. An unusual presentation of McCune-Albright syndrome confirmed by an activating mutation of the Gs alpha-subunit from a bone lesion. J Clin Endocrinol Metab 1994; 78:803.
  15. Mastorakos G, Mitsiades NS, Doufas AG, Koutras DA. Hyperthyroidism in McCune-Albright syndrome with a review of thyroid abnormalities sixty years after the first report. Thyroid 1997; 7:433.
  16. Ramelli F, Studer H, Bruggisser D. Pathogenesis of thyroid nodules in multinodular goiter. Am J Pathol 1982; 109:215.
  17. Apel RL, Ezzat S, Bapat BV, et al. Clonality of thyroid nodules in sporadic goiter. Diagn Mol Pathol 1995; 4:113.
  18. Aeschimann S, Kopp PA, Kimura ET, et al. Morphological and functional polymorphism within clonal thyroid nodules. J Clin Endocrinol Metab 1993; 77:846.
  19. Bignell GR, Canzian F, Shayeghi M, et al. Familial nontoxic multinodular thyroid goiter locus maps to chromosome 14q but does not account for familial nonmedullary thyroid cancer. Am J Hum Genet 1997; 61:1123.
  20. Namba H, Matsuo K, Fagin JA. Clonal composition of benign and malignant human thyroid tumors. J Clin Invest 1990; 86:120.
  21. Lemoine NR, Mayall ES, Wyllie FS, et al. High frequency of ras oncogene activation in all stages of human thyroid tumorigenesis. Oncogene 1989; 4:159.
  22. Namba H, Rubin SA, Fagin JA. Point mutations of ras oncogenes are an early event in thyroid tumorigenesis. Mol Endocrinol 1990; 4:1474.
  23. Suarez HG, du Villard JA, Severino M, et al. Presence of mutations in all three ras genes in human thyroid tumors. Oncogene 1990; 5:565.
  24. Bos JL. ras oncogenes in human cancer: a review. Cancer Res 1989; 49:4682.
  25. Karga H, Lee JK, Vickery AL Jr, et al. Ras oncogene mutations in benign and malignant thyroid neoplasms. J Clin Endocrinol Metab 1991; 73:832.
  26. Belge G, Rippe V, Meiboom M, et al. Delineation of a 150-kb breakpoint cluster in benign thyroid tumors with 19q13.4 aberrations. Cytogenet Cell Genet 2001; 93:48.
  27. Rippe V, Drieschner N, Meiboom M, et al. Identification of a gene rearranged by 2p21 aberrations in thyroid adenomas. Oncogene 2003; 22:6111.
  28. Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014; 159:676.
  29. Kimura ET, Nikiforova MN, Zhu Z, et al. 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.
  30. Shen X, Liu R, Xing M. A six-genotype genetic prognostic model for papillary thyroid cancer. Endocr Relat Cancer 2017; 24:41.
  31. Indo Y, Mardy S, Tsuruta M, et al. Structure and organization of the human TRKA gene encoding a high affinity receptor for nerve growth factor. Jpn J Hum Genet 1997; 42:343.
  32. Jhiang SM, Sagartz JE, Tong Q, et al. Targeted expression of the ret/PTC1 oncogene induces papillary thyroid carcinomas. Endocrinology 1996; 137:375.
  33. Santoro M, Chiappetta G, Cerrato A, et al. Development of thyroid papillary carcinomas secondary to tissue-specific expression of the RET/PTC1 oncogene in transgenic mice. Oncogene 1996; 12:1821.
  34. Bongarzone I, Vigneri P, Mariani L, et al. RET/NTRK1 rearrangements in thyroid gland tumors of the papillary carcinoma family: correlation with clinicopathological features. Clin Cancer Res 1998; 4:223.
  35. Nikiforov YE, Rowland JM, Bove KE, et al. Distinct pattern of ret oncogene rearrangements in morphological variants of radiation-induced and sporadic thyroid papillary carcinomas in children. Cancer Res 1997; 57:1690.
  36. Bongarzone I, Fugazzola L, Vigneri P, et al. Age-related activation of the tyrosine kinase receptor protooncogenes RET and NTRK1 in papillary thyroid carcinoma. J Clin Endocrinol Metab 1996; 81:2006.
  37. Bounacer A, Wicker R, Caillou B, et al. High prevalence of activating ret proto-oncogene rearrangements, in thyroid tumors from patients who had received external radiation. Oncogene 1997; 15:1263.
  38. Bongarzone I, Butti MG, Fugazzola L, et al. Comparison of the breakpoint regions of ELE1 and RET genes involved in the generation of RET/PTC3 oncogene in sporadic and in radiation-associated papillary thyroid carcinomas. Genomics 1997; 42:252.
  39. Greco A, Miranda C, Pagliardini S, et al. Chromosome 1 rearrangements involving the genes TPR and NTRK1 produce structurally different thyroid-specific TRK oncogenes. Genes Chromosomes Cancer 1997; 19:112.
  40. Grieco M, Santoro M, Berlingieri MT, et al. PTC is a novel rearranged form of the ret proto-oncogene and is frequently detected in vivo in human thyroid papillary carcinomas. Cell 1990; 60:557.
  41. Klugbauer S, Lengfelder E, Demidchik EP, Rabes HM. A new form of RET rearrangement in thyroid carcinomas of children after the Chernobyl reactor accident. Oncogene 1996; 13:1099.
  42. Fugazzola L, Pierotti MA, Vigano E, et al. Molecular and biochemical analysis of RET/PTC4, a novel oncogenic rearrangement between RET and ELE1 genes, in a post-Chernobyl papillary thyroid cancer. Oncogene 1996; 13:1093.
  43. Wynford-Thomas D. Origin and progression of thyroid epithelial tumours: cellular and molecular mechanisms. Horm Res 1997; 47:145.
  44. Wirth LJ, Sherman E, Robinson B, et al. Efficacy of Selpercatinib in RET-Altered Thyroid Cancers. N Engl J Med 2020; 383:825.
  45. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med 2018; 378:731.
  46. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020; 21:271.
  47. Cohen Y, Xing M, Mambo E, et al. BRAF mutation in papillary thyroid carcinoma. J Natl Cancer Inst 2003; 95:625.
  48. Cohen Y, Rosenbaum E, Clark DP, et al. Mutational analysis of BRAF in fine needle aspiration biopsies of the thyroid: a potential application for the preoperative assessment of thyroid nodules. Clin Cancer Res 2004; 10:2761.
  49. Fugazzola L, Mannavola D, Cirello V, et al. BRAF mutations in an Italian cohort of thyroid cancers. Clin Endocrinol (Oxf) 2004; 61:239.
  50. Knauf JA, Ma X, Smith EP, et al. Targeted expression of BRAFV600E in thyroid cells of transgenic mice results in papillary thyroid cancers that undergo dedifferentiation. Cancer Res 2005; 65:4238.
  51. Lupi C, Giannini R, Ugolini C, et al. Association of BRAF V600E mutation with poor clinicopathological outcomes in 500 consecutive cases of papillary thyroid carcinoma. J Clin Endocrinol Metab 2007; 92:4085.
  52. Caronia LM, Phay JE, Shah MH. Role of BRAF in thyroid oncogenesis. Clin Cancer Res 2011; 17:7511.
  53. Xing M, Alzahrani AS, Carson KA, et al. Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA 2013; 309:1493.
  54. Kim TH, Park YJ, Lim JA, et al. The association of the BRAF(V600E) mutation with prognostic factors and poor clinical outcome in papillary thyroid cancer: a meta-analysis. Cancer 2012; 118:1764.
  55. Xing M, Westra WH, Tufano RP, et al. BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J Clin Endocrinol Metab 2005; 90:6373.
  56. Cappola AR, Mandel SJ. Molecular testing in thyroid cancer: BRAF mutation status and mortality. JAMA 2013; 309:1529.
  57. Vasko V, Hu S, Wu G, et al. High prevalence and possible de novo formation of BRAF mutation in metastasized papillary thyroid cancer in lymph nodes. J Clin Endocrinol Metab 2005; 90:5265.
  58. Oler G, Ebina KN, Michaluart P Jr, et al. Investigation of BRAF mutation in a series of papillary thyroid carcinoma and matched-lymph node metastasis reveals a new mutation in metastasis. Clin Endocrinol (Oxf) 2005; 62:509.
  59. Lima J, Trovisco V, Soares P, et al. BRAF mutations are not a major event in post-Chernobyl childhood thyroid carcinomas. J Clin Endocrinol Metab 2004; 89:4267.
  60. Kumagai A, Namba H, Saenko VA, et al. Low frequency of BRAFT1796A mutations in childhood thyroid carcinomas. J Clin Endocrinol Metab 2004; 89:4280.
  61. Ciampi R, Knauf JA, Kerler R, et al. Oncogenic AKAP9-BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J Clin Invest 2005; 115:94.
  62. Cradic KW, Milosevic D, Rosenberg AM, et al. Mutant BRAF(T1799A) can be detected in the blood of papillary thyroid carcinoma patients and correlates with disease status. J Clin Endocrinol Metab 2009; 94:5001.
  63. Salerno P, De Falco V, Tamburrino A, et al. Cytostatic activity of adenosine triphosphate-competitive kinase inhibitors in BRAF mutant thyroid carcinoma cells. J Clin Endocrinol Metab 2010; 95:450.
  64. Ball DW. Selectively targeting mutant BRAF in thyroid cancer. J Clin Endocrinol Metab 2010; 95:60.
  65. Zhu Z, Gandhi M, Nikiforova MN, et al. 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.
  66. Fagin JA, Mitsiades N. Molecular pathology of thyroid cancer: diagnostic and clinical implications. Best Pract Res Clin Endocrinol Metab 2008; 22:955.
  67. Liu X, Qu S, Liu R, et al. TERT promoter mutations and their association with BRAF V600E mutation and aggressive clinicopathological characteristics of thyroid cancer. J Clin Endocrinol Metab 2014; 99:E1130.
  68. He H, Jazdzewski K, Li W, et al. The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci U S A 2005; 102:19075.
  69. Cong D, He M, Chen S, et al. Expression profiles of pivotal microRNAs and targets in thyroid papillary carcinoma: an analysis of The Cancer Genome Atlas. Onco Targets Ther 2015; 8:2271.
  70. Jazdzewski K, Murray EL, Franssila K, et al. Common SNP in pre-miR-146a decreases mature miR expression and predisposes to papillary thyroid carcinoma. Proc Natl Acad Sci U S A 2008; 105:7269.
  71. Jazdzewski K, Liyanarachchi S, Swierniak M, et al. Polymorphic mature microRNAs from passenger strand of pre-miR-146a contribute to thyroid cancer. Proc Natl Acad Sci U S A 2009; 106:1502.
  72. Gudmundsson J, Sulem P, Gudbjartsson DF, et al. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nat Genet 2009; 41:460.
  73. Sturgis EM, Zhao C, Zheng R, Wei Q. Radiation response genotype and risk of differentiated thyroid cancer: a case-control analysis. Laryngoscope 2005; 115:938.
  74. Cybulski C, Górski B, Huzarski T, et al. CHEK2 is a multiorgan cancer susceptibility gene. Am J Hum Genet 2004; 75:1131.
  75. Adjadj E, Schlumberger M, de Vathaire F. Germ-line DNA polymorphisms and susceptibility to differentiated thyroid cancer. Lancet Oncol 2009; 10:181.
  76. Bastos HN, Antão MR, Silva SN, et al. Association of polymorphisms in genes of the homologous recombination DNA repair pathway and thyroid cancer risk. Thyroid 2009; 19:1067.
  77. Hu S, Liu D, Tufano RP, et al. Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. Int J Cancer 2006; 119:2322.
  78. Xing M. Gene methylation in thyroid tumorigenesis. Endocrinology 2007; 148:948.
  79. Giardiello FM, Offerhaus GJ, Lee DH, et al. Increased risk of thyroid and pancreatic carcinoma in familial adenomatous polyposis. Gut 1993; 34:1394.
  80. Zeki K, Spambalg D, Sharifi N, et al. Mutations of the adenomatous polyposis coli gene in sporadic thyroid neoplasms. J Clin Endocrinol Metab 1994; 79:1317.
  81. Cetta F, Chiappetta G, Melillo RM, et al. The ret/ptc1 oncogene is activated in familial adenomatous polyposis-associated thyroid papillary carcinomas. J Clin Endocrinol Metab 1998; 83:1003.
  82. Houlston RS, Stratton MR. Genetics of non-medullary thyroid cancer. QJM 1995; 88:685.
  83. Loh KC. Familial nonmedullary thyroid carcinoma: a meta-review of case series. Thyroid 1997; 7:107.
  84. Malchoff CD, Malchoff DM. Familial nonmedullary thyroid carcinoma. Semin Surg Oncol 1999; 16:16.
  85. Malchoff CD, Malchoff DM. The genetics of hereditary nonmedullary thyroid carcinoma. J Clin Endocrinol Metab 2002; 87:2455.
  86. He H, Li W, Wu D, et al. Ultra-rare mutation in long-range enhancer predisposes to thyroid carcinoma with high penetrance. PLoS One 2013; 8:e61920.
  87. Hishinuma A, Fukata S, Kakudo K, et al. High incidence of thyroid cancer in long-standing goiters with thyroglobulin mutations. Thyroid 2005; 15:1079.
  88. Malchoff CD, Sarfarazi M, Tendler B, et al. Papillary thyroid carcinoma associated with papillary renal neoplasia: genetic linkage analysis of a distinct heritable tumor syndrome. J Clin Endocrinol Metab 2000; 85:1758.
  89. McKay JD, Lesueur F, Jonard L, et al. Localization of a susceptibility gene for familial nonmedullary thyroid carcinoma to chromosome 2q21. Am J Hum Genet 2001; 69:440.
  90. Xing M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer 2013; 13:184.
  91. Terrier P, Sheng ZM, Schlumberger M, et al. Structure and expression of c-myc and c-fos proto-oncogenes in thyroid carcinomas. Br J Cancer 1988; 57:43.
  92. Kroll TG, Sarraf P, Pecciarini L, et al. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma [corrected]. Science 2000; 289:1357.
  93. Marques AR, Espadinha C, Catarino AL, et al. Expression of PAX8-PPAR gamma 1 rearrangements in both follicular thyroid carcinomas and adenomas. J Clin Endocrinol Metab 2002; 87:3947.
  94. Ngeow J, Mester J, Rybicki LA, et al. Incidence and clinical characteristics of thyroid cancer in prospective series of individuals with Cowden and Cowden-like syndrome characterized by germline PTEN, SDH, or KLLN alterations. J Clin Endocrinol Metab 2011; 96:E2063.
  95. Xing M, Cohen Y, Mambo E, et al. Early occurrence of RASSF1A hypermethylation and its mutual exclusion with BRAF mutation in thyroid tumorigenesis. Cancer Res 2004; 64:1664.
  96. Ganly I, Makarov V, Deraje S, et al. Integrated Genomic Analysis of Hürthle Cell Cancer Reveals Oncogenic Drivers, Recurrent Mitochondrial Mutations, and Unique Chromosomal Landscapes. Cancer Cell 2018; 34:256.
  97. Nikiforov YE, Seethala RR, Tallini G, et al. Nomenclature Revision for Encapsulated Follicular Variant of Papillary Thyroid Carcinoma: A Paradigm Shift to Reduce Overtreatment of Indolent Tumors. JAMA Oncol 2016; 2:1023.
  98. Lee SE, Hwang TS, Choi YL, et al. Molecular Profiling of Papillary Thyroid Carcinoma in Korea with a High Prevalence of BRAFV600E Mutation. Thyroid 2017; 27:802.
  99. Jiang XS, Harrison GP, Datto MB. Young Investigator Challenge: Molecular testing in noninvasive follicular thyroid neoplasm with papillary-like nuclear features. Cancer Cytopathol 2016; 124:893.
  100. Shi X, Liu R, Qu S, et al. Association of TERT promoter mutation 1,295,228 C>T with BRAF V600E mutation, older patient age, and distant metastasis in anaplastic thyroid cancer. J Clin Endocrinol Metab 2015; 100:E632.
  101. Landa I, Ibrahimpasic T, Boucai L, et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J Clin Invest 2016; 126:1052.
  102. Quiros RM, Ding HG, Gattuso P, et al. Evidence that one subset of anaplastic thyroid carcinomas are derived from papillary carcinomas due to BRAF and p53 mutations. Cancer 2005; 103:2261.
  103. Tallini G, Santoro M, Helie M, et al. 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; 4:287.
  104. Garcia-Rostan G, Camp RL, Herrero A, et al. Beta-catenin dysregulation in thyroid neoplasms: down-regulation, aberrant nuclear expression, and CTNNB1 exon 3 mutations are markers for aggressive tumor phenotypes and poor prognosis. Am J Pathol 2001; 158:987.
  105. García-Rostán G, Costa AM, Pereira-Castro I, et al. Mutation of the PIK3CA gene in anaplastic thyroid cancer. Cancer Res 2005; 65:10199.
  106. Murugan AK, Xing M. Anaplastic thyroid cancers harbor novel oncogenic mutations of the ALK gene. Cancer Res 2011; 71:4403.
  107. Fagin JA, Matsuo K, Karmakar A, et al. High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas. J Clin Invest 1993; 91:179.
  108. Bible KC, Kebebew E, Brierley J, et al. 2021 American Thyroid Association Guidelines for Management of Patients with Anaplastic Thyroid Cancer. Thyroid 2021; 31:337.
Topic 7819 Version 18.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟