Next Article in Journal
Characterization of the Complete Mitochondrial Genome of Harpalus sinicus and Its Implications for Phylogenetic Analyses
Next Article in Special Issue
The Role of Molecular Testing for the Indeterminate Thyroid FNA
Previous Article in Journal
The Expanded SWEET Gene Family Following Whole Genome Triplication in Brassica rapa
Previous Article in Special Issue
Molecular Alterations in Thyroid Cancer: From Bench to Clinical Practice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Thyroid Cancer in the Pediatric Population

by
Vera A. Paulson
1,*,
Erin R. Rudzinski
2 and
Douglas S. Hawkins
3
1
Department of Laboratory Medicine, University of Washington Medical Center, 1959 NE Pacific St, Box 357110, Seattle, WA 98105, USA
2
Department of Laboratories, Seattle Children’s Hospital, OC.8.720; 4800 Sandpoint Way NE, Seattle, WA 98105, USA
3
University of Washington Medical Center, Fred Hutchinson Cancer Research Center and Cancer and Blood Disorders Center, Seattle Children’s Hospital, MB.8.501, Seattle, WA 98105, USA
*
Author to whom correspondence should be addressed.
Submission received: 12 July 2019 / Revised: 11 September 2019 / Accepted: 12 September 2019 / Published: 18 September 2019
(This article belongs to the Special Issue Thyroid Cancer: Genetics and Targeted Therapies)

Abstract

:
Thyroid cancer is rare in the pediatric population, but thyroid carcinomas occurring in children carry a unique set of clinical, pathologic, and molecular characteristics. In comparison to adults, children more often present with aggressive, advanced stage disease. This is at least in part due to the underlying biologic and molecular differences between pediatric and adult thyroid cancer. Specifically, papillary thyroid carcinoma (which accounts for approximately 90% of pediatric thyroid cancer) has a high rate of gene fusions which influence the histologic subtypes encountered in pediatric thyroid tumors, are associated with more extensive extrathyroidal disease, and offer unique options for targeted medical therapies. Differences are also seen in pediatric follicular thyroid cancer, although there are few studies of non-papillary pediatric thyroid tumors published in the literature due to their rarity, and in medullary carcinoma, which is most frequently diagnosed in the pediatric population in the setting of prophylactic thyroidectomies for known multiple endocrine neoplasia syndromes. The overall shift in the spectrum of histotypes and underlying molecular alterations common in pediatric thyroid cancer is important to recognize as it may directly influence diagnostic test selection and therapeutic recommendations.

1. Introduction

Thyroid cancer is the leading cause of pediatric endocrine cancer, accounting for over 6% of all pediatric cancers from 2012 to 2016 [1]. This figure reflects the rising incidence of pediatric thyroid carcinoma over the last four decades [1]. Between 1975 and 1995, the annual percent change (APC) of pediatric thyroid carcinoma was +0.8%/year; with an accelerated +4.6%/year APC from 1996 to 2016 [1]. The majority of this increase is comprised of the most common subtype of thyroid cancer—papillary thyroid carcinoma (PTC). However, increases are also reported for follicular thyroid carcinoma (FTC) [2]. The contribution of medullary (MTC), anaplastic (ATC), and poorly differentiated (PDTC) thyroid carcinomas is minor given their rarity in the pediatric population [1,3,4,5]. Several studies suggest that the increasing incidence of papillary and follicular thyroid carcinoma is only partially accounted for by surveillance and enhanced detection, as there is also an increase in detection of large tumors and advanced-stage disease [3,6]. However, Chen et al. suggest that the increased rate of thyroid carcinoma may be the result of increased detection secondary to a “reservoir of clinically silent cancers”, increased use of medical imaging, and increased imaging sensitivity [7].
Historically, recommendations for the evaluation and management of thyroid cancer in the pediatric population have been extrapolated from adult guidelines. However, compared to adults, thyroid cancers in the pediatric population differ in clinical presentation, pathophysiology, and long-term outcomes [8]. The American Thyroid Association (ATA) has therefore published guidelines specifically addressing the management of children with thyroid nodules and differentiated thyroid cancer (DTC) in 2015 [8]. Herein we review the clinicopathologic presentation and treatment of pediatric thyroid carcinoma, highlighting key morphomolecular findings and their impact on clinical management.

2. Background and Epidemiology

Per the Surveillance, Epidemiology, and End Results program, 1.8% of the thyroid cancers diagnosed in the US are in children and adolescents [1]. Rarely neonatal thyroid carcinoma has been described [9], but most cases of pediatric thyroid carcinoma occur in the second decade [1]. Increased age also results in an increasingly disproportionate number of females affected compared to their male peers, with nearly a 6:1 ratio by 15–19 years of age [1]. The vast majority of these thyroid cancers are PTC (80–90%) followed by FTC (~10%), MTC (3–5%), and rarely ATC and PDTC [4,5,8,10,11]. Reported risk factors in the development of follicular-cell derived thyroid cancer in children include thyroid disease (e.g., autoimmune thyroid disorders) [8,12], nutritional (i.e., iodine) deficiency [13], and prior radiation exposure [8,14,15,16]. The frequency of malignancy in pediatric patients with Hashimoto’s thyroiditis has been reported to range from 0.67% to ~3%, which is above the background risk of ~0.02% in the general pediatric population (based on an incidence rate of 1.14/100,000 per year over the course of 19 years) [1,3,12,17,18,19]. Proposed mechanisms for the association between Hashimoto’s thyroiditis and PTC include over-production of thyroid stimulating hormone (TSH) or chronic inflammation resulting in proliferation, angiogenesis, and/or reduced apoptosis [12]. Data in pediatric Graves’ disease is more limited with respect to malignancy risk, with reports of follicular-cell derived thyroid cancer ranging from <1% to 22% of patients undergoing definitive surgical therapy; this number further decreases if non-surgically treated Graves patients are included in the denominator [20,21,22]. The link between iodine deficiency and FTC has also been more extensively studied in adults; however, of the few studies published in the pediatric population, several have suggested that iodine intake may be protective in the setting of radiation exposure and the development of PTC [13,23,24,25]. Radiation exposure, either environmental, as part of diagnostic tests, or following radiotherapy for a prior malignancy (most commonly Hodgkin lymphoma or central nervous system tumors) also increases the risk of DTC [8,14,15,16]. Indeed, thyroid cancer is one of the most common secondary malignancies among survivors of childhood cancer [8,14,15,16].
Rarely genetic predisposition syndromes including APC-associated polyposis, DICER1 syndrome, Carney Complex, PTEN hamartoma/Cowden syndrome, and Werner syndrome are associated with DTCs, accounting for approximately 5% of cases [6,8,26]. Cases of PTC and FTC have also been reported in association with Li–Fraumeni, Peutz–Jeghers, familial paragangliomas, McCune–Albright, and Beckwith–Wiedemann [8]. By comparison, the vast majority of MTC in the pediatric population are hereditary, the result of germline RET mutations resulting in multiple endocrine neoplasia (MEN) type 2A syndrome (90–95% of childhood MTC), MEN type 2B, or familial medullary thyroid carcinoma (FMTC) [6,27,28].

3. Clinical Presentation

A thyroid nodule in the pediatric patient typically manifests as an asymptomatic neck mass, with or without cervical lymphadenopathy, and may be accompanied by trouble breathing and/or hyperthyroidism. The identification of a nodule is clinically important due its increased malignant potential in the pediatric population. Nodules in the adult population are relatively common, occurring in 19–68% of the population, but are rarely malignant (5–10% of all thyroid nodules in adults, per most series) [8,29,30,31]. The opposite is true in children and adolescents. Approximately 1–3% of children harbor thyroid nodules, and more than a quarter (22–26%) of thyroid nodules occurring in pediatric patients are malignant [6,8,30,31].
A large hard nodule, especially one adherent to adjacent tissue, is concerning for cancer [6]. Equally concerning are nodules determined to have suspicious ultrasonographic features by ATA guidelines, including irregular margins, marked hypoechogenicity, and microcalcifications, or nodules accompanied by pathologic adenopathy [30,32]. Solid and predominantly solid nodules are also more likely to be malignant compared to cystic nodules [32]. It is worth noting that the diffuse sclerosing variant of PTC may present as diffusely infiltrating disease resulting not in a single nodule but in diffuse enlargement of a lobe or the entire gland [33,34]. Diffuse thyroid enlargement, especially if accompanied by cervical lymphadenopathy, must therefore prompt imaging for the microcalcifications that are invariably associated with this subtype [33,35]. Whether the ATA criteria for malignancy risk by ultrasound (US) features directly apply to pediatric thyroid lesions is a subject of continued investigation. Creo et al. demonstrated that revised criteria for risk of malignancy based on institutional report of local radiologists overall impression was more specific for a diagnosis of thyroid cancer in children as compared to ATA guidelines [31,36] but there was diminishing accuracy of these guidelines for predicting malignancy in younger patients [36].
Per recent pediatric management guidelines, the identification of a solitary or suspicious thyroid nodule on imaging (with features described above) or physical examination should prompt utilization of a diagnostic algorithm that includes evaluation of TSH (and nuclear thyroid scintigraphy in the setting of TSH suppression) to determine whether the nodule should undergo US-guided fine needle aspiration (FNA) (if hypofunctioning) or surgical resection (if hyperfunctioning) [8]. In the pediatric population, hyperfunctioning thyroid nodules by scintigraphy appear to carry a risk of malignancy of <10–15% [37,38,39], though some reports suggest a higher risk of malignancy than in adults (up to 30%) [40]. The Bethesda classification is used to predict the risk of malignancy based FNA findings, as discussed below [30,41].

4. Pathologic Evaluation

4.1. Fine Needle Aspiration

FNA has long been a mainstay in the diagnostic work-up of adult thyroid nodules, and recent literature supports its efficacy in the pediatric population, with 99% accuracy and sensitivity and specificity of 94% and 100% respectively, per one recent study [41]. The Bethesda system classifies thyroid FNAs into six categories, each category linked to a malignancy risk [30,41]. Although there is significant variability between studies, the risk of malignancy in non-diagnostic (Bethesda I), benign (Bethesda II), and suspicious/malignant (Bethesda V/VI) mirror adult risk assessment at 0% (range 0–10%), 5–8% (range 0–16%), and 100% respectively (Bethesda V range: 40–100%, Bethesda VI range: 100%). [42]. Several studies suggest that Bethesda III (atypia of uncertain significance/follicular lesion of uncertain significance; AUS/FLUS) and Bethesda IV (follicular neoplasm/suspicious for follicular neoplasm; FN/SFN) categories account for up to 40% (range: 13–43%) of all pediatric FNA diagnoses [42]. Further, these indeterminate aspirate diagnoses are associated with increased risk of malignancy in children versus adults (28% in children vs. 6–30% in adults for AUS/FLUS and 58% in children vs. 10–40% in adults for FN/SFN lesions) [42]. However, more recent studies indicate lower percentages of indeterminate FNA groups than previously reported (13–18% for indeterminate diagnoses combined) and a lower risk of malignancy after histologic resection within these groups (11–20% for AUS/FLUS; 25–28% for FN/SFN) [42].

4.2. Surgical Resection

Although diagnostic criteria for PTC are the same regardless of patient age, differences in histotypes and overall stage occur between adult and pediatric populations. Children tend to present with nodal disease and evidence of local or distant metastases [8]. They also have a higher risk of recurrence than adults with thyroid carcinoma. Small studies suggested that younger age, male sex, multifocality, and tumor size greater than 2 cm predicted extensive disease at diagnosis [43]. However, Balachandar et al. suggest that the primary predictor of extent of disease is the presence of extensive extrathyroidal extension, regardless of any other clinical or pathologic features [43]. High risk histologic subtypes of PTC are reported to occur in 15–37% of pediatric PTC, including 7–13% tall-cell variant, 7–16% diffuse sclerosing variant, 1–4% solid/trabecular variant and 2–6% poorly differentiated carcinoma. While some studies suggest the solid/trabecular variant may have an increased recurrence risk in children, most studies indicate that these “high-risk” histologic subtypes do not carry a worse event free survival in children [44,45]. This is in contrast to the increased risk of recurrence associated with these histologic subtypes reported in the adult population [43]. Low risk subtypes (classic and follicular variants of PTC) remain the most commonly encountered subtypes, while the newly described noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP) accounts for nearly 4.5% of pediatric PTC [46].
Follicular carcinoma in children is usually minimally invasive (90% of all FTC cases), rather than widely invasive [47,48]. Thus, as opposed to PTC, children with FTC have less aggressive disease than adults.
Medullary carcinoma is infrequently encountered in the pediatric population; however, prophylactic thyroidectomy is routinely performed for children with MEN2A, with surgical timing based on risk associated with specific mutations [28]. In this context, C-cell hyperplasia (CCH) is the most common finding [49]. Neoplastic CCH is considered distinct from physiologic CCH, and represents the precursor lesion of MTC. Cytologic atypia and integrity of the basement membrane are helpful in distinguishing the neoplastic pattern of CCH from either physiologic CCH or medullary microcarcinoma, respectively [50,51,52,53].

5. Molecular Diagnostic Testing

Molecular diagnostic testing has several emerging roles in the management of children with thyroid lesions. First, although data is scant in children, early studies suggest that indeterminate FNA diagnoses (AUS/FLUS and FN/SFN) may be further stratified by molecular findings [54]. Monaco et al. determined the presence of classic RAS and BRAF mutations as well as RET-PTC and PAX8-PPARG fusions in pediatric nodules that had undergone FNA [54]. They reported that all mutation positive FNAs were PTC on resection [54]. In general, however, given the difficulties of performing repeat FNA procedures in young children who may require sedation and the current ATA guidelines which recommend surgery (lobectomy plus isthmusectomy) for atypical thyroid nodules, molecular testing of FNA material is often deferred from routine practice [30]. More studies are needed to define the role of molecular testing in indeterminate thyroid nodules in the pediatric population.
The spectrum of genetic alterations found in pediatric thyroid carcinoma is shifted in comparison to what is found in the adult population. As most pediatric studies on the molecular landscape of thyroid carcinoma have been driven by findings reported in the adult population, the overall alterations described in pediatric and adult thyroid cancer populations are similar. Oncogenic gene fusions occur with increased frequency in pediatric thyroid carcinoma, with 50–60% of pediatric thyroid cancers harboring a fusion compared to 15% in the adult population (see Table 1). Correspondingly, point mutations occur with decreased frequency, with 30% of pediatric thyroid cancers demonstrating point mutations compared to nearly 70% of adult thyroid cancers [14,26]. These differences drive both potential diagnostic test selection (e.g., ensuring inclusion of fusion genes in molecular analysis of indeterminate aspirate samples) and therapeutic considerations.

5.1. PTC

Given its predominance in the pediatric population, the most extensively studied pediatric thyroid cancer, in relation to molecular drivers, is PTC. The molecular results of approximately 2000 “pediatric” PTCs have been reported in the literature (see Table 1); however, their characterization has been limited to small cohorts assessed for select alterations or in more recent years small multi-gene panels for the detection of point mutations or specific gene rearrangements (e.g., thyroseq) [26,51,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104]. The combination of these small cohorts with varying demographics (namely the age range included as “pediatric”), geographical and environmental factors, and method of assessment often results in a wide range of frequencies for the observed genetic alterations.
RET fusions are reported to be the most common observed alteration, and by our review appear to occur in approximately 25–30% of sporadic pediatric PTC (range 14–55%, Table 1), a figure that further increases to nearly 45% (range: 14–87%, Table 1) in patients exposed to radiation. While new RET fusion partners are continuously identified and reported, the most common are PTC1 and PTC3, though there is some variability in frequency due to detection methods and expression heterogeneity [26]. RET fusions are associated with classic, solid, and diffuse sclerosing PTC subtypes as well as increased extrathyroidal extension, spread to locoregional lymph nodes, and distant (pulmonary) metastases [26,103]. It should be noted that RET-PTC fusions have also been reported in benign thyroid samples, more commonly in those with than without prior radiation exposure [72,105,106].
Although less common than RET, other oncogenic fusions occur with increased frequency in sporadic pediatric PTC. Reported fusions include NTRK(1, 2 and 3), BRAF, and ALK. Although not comprehensively surveyed, these fusions have been identified in approximately 10% (range: 0–26%), 10% (range: 0–18%), and 5% (range: 0–21%) of pediatric PTC, respectively (see Table 1).
BRAF point mutations are reported to be the second most common single oncogenic driver in sporadic pediatric PTC, though our comprehensive review suggests BRAF mutations may occur as frequently as RET-PTC fusions (25–30% of lesions, range: 0–63%, Table 1). While more than 40 mutations have been identified in BRAF to date, >95% are V600E, though this may be skewed by methodology as many of these studies only assessed this single mutation. BRAFV600E mutations are associated with classic PTC histology and correlate with higher age at diagnosis; they do not appear to portend a more aggressive clinical course [96].
RAS mutations (specifically HRAS Q61R and NRAS Q61K) are present in <5% (range: 0–18%, Table 1) of sporadic pediatric PTC, and are more frequent in benign thyroid nodules (15–40%) [57,99]. RAS mutations were more commonly associated with the follicular variant of PTC, and undoubtedly some would be reclassified as NIFTP in light of current practices [26,87]. The clinical implications and impact of these RAS mutations are therefore difficult to discern given their identification in both benign and varied malignant lesions.
PAX8-PPARg fusions have also rarely been reported in pediatric PTC (<5%, range 0–9%, Table 1), predominantly the follicular variant though two solid variant PTCs were reported in Ricarte-Filho [26,80]. More comprehensive molecular panels have rarely identified mutations in DICER1 (up to 10% in one study), TP53, TERT, PIK3CA, and PTEN [57,80,86,100].

5.2. FTC and PDTC

Little is known about the drivers of pediatric FTC, with less than 100 cases scattered predominantly throughout reported cohorts of DTC (Table 1) [54,57,65,66,68,74,76,86,87,88,89,90,99,101]. A minority appear to harbor activating RAS point mutations (10–15%, range: 0–20%) and PAX8-PPARg fusions (<5%, range 0–20%), neither of which is unique to FTC (see Table 1). Cohort size (0–41) has limited meaningful analysis of the association of clinicopathologic and molecular findings. Even less is known about pediatric PDTC, with only three cases included in the analyzed cohorts [74,85,90].

5.3. MTC

Unlike follicular derived tumors, and MTCs in the adult population, over 95% of pediatric MTCs are hereditary, secondary to activating RET mutations [6,27]. Moreover, of the apparently sporadic cases of MTC another 6–10% are de novo germline RET mutations [28]. Gain of function RET mutations occur in two different ways. Either the mutation occurs in one of the six cysteine residues within the extracellular domains (Cys609, 611, 618, 620, 630, and 634) and promotes dimerization and ligand-independent activation, or the mutation occurs in the tyrosine kinase domain and confers ligand independent catalytic activity in monomeric form [28].

5.4. Molecular Summary

Compared to the landmark TCGA study in 2014, which reduced unknown oncogenic drivers in adult PTC from 25% to 3.5% through exome and whole genome DNA sequencing, RNA sequencing, miRNA sequencing, single nucleotide polymorphism (SNP) arrays, DNA methylation arrays, and reverse phase protein arrays of nearly 500 cases of PTCs (and of which only nine were pediatric), the pediatric PTC studies are quite limited [107]. Moreover, given the small pediatric cohorts, variety of methodologies used, and wide range of the molecular findings, it is difficult to know the true frequency of these alterations in the pediatric population and whether we have truly defined the molecular characteristics of this entity. If PTC is understudied, FTC and PDTC studies are virtually non-existent in pediatrics. Additional efforts with broader panels and larger cohorts are needed if we are to better define the genomic landscape of pediatric thyroid cancer and whether there are associations between histology and/or outcome. While molecular diagnostic testing is of uncertain utility in the setting of indeterminate FNAs and borderline histologic diagnoses; its role in treatment in certain settings is paramount.

6. Treatment

Total thyroidectomy is the treatment of choice for pediatric PTC and FTC due to the increased incidence of bilateral (30%) and multifocal (65%) disease [8], though a near-total thyroidectomy, in which a small amount of thyroid tissue near the recurrent laryngeal nerve or superior parathyroid glands may be spared to decrease the possibility of damage to those structures, is also an option. While data in the adult thyroid cancer population suggests there may be a role for less extensive surgery in certain clinical contexts, such an approach is less common in the pediatric population due to the increased risk of local recurrence in patients treated by lobectomy [108]. In the presence of central or lateral neck metastases, thyroidectomy should be accompanied by a central or lateral neck dissection. Neck dissection can also be considered prophylactically based on tumor size and focality though its use must be weighed against possible complications. The ATA task force further recommends adjuvant radioactive iodine (RAI) for unresectable iodine-avid persistent locoregional disease (due to invasion of vital structures) and/or distant metastases, which may be further guided by post-operative thyroglobulin levels [7,8]. The pediatric recommendations regarding indications for RAI differ from those published by the National Comprehensive Cancer Network for adults with PTC, which cite a variety of clinical features including primary tumor size >2–4 cm, gross extrathyroidal extension, and extensive or bulky regional nodal involvement as indications for adjuvant RAI [109]. In contrast, the guidelines for children with DTC recommend using post-thyroidectomy TSH-stimulated thyroglobulin levels (in addition to TSH-stimulated 123I imaging and the presence of distant metastases) to determine who should receive adjuvant RAI [8]. However, this algorithm assumes the absence of an anti-thyroglobulin antibody, with the recommendation to use only extensive local invasion or distant metastases as indications for RAI for patients with an anti-thyroglobulin antibody and potentially delay RAI to allow the anti-thyroglobulin antibody to clear prior to committing to RAI. There is no consensus on the preferred dose adjustment method to calculate a 131I for a child, with body weight and body surface area methods described. Whole body 131I dosimetry can also be used in patients with extensive metastases or who require repeat RAI to limit organ doses. For additional information regarding patient preparation for and administration of RAI, the authors recommend an excellent review of RAI by Parisi et al. [110].
Despite more advanced disease at presentation, 40–90% extensive regional node involvement and 7–30% distant lung metastasis, PTC has an excellent prognosis with greater than >98% survival, by most studies [7,46,111]. For patients with DTC who recur, RAI is usually effective. For patients with iodine non-avid or RAI-refractory disease or residual/metastatic MTC, molecularly targeted kinase inhibitors (TKI) may provide a medical therapeutic option.
Most of our knowledge of TKIs and their efficacy comes from the adult population extended to pediatric patients through ongoing clinical trials as well as case reports and small case series in the literature. To date, several kinase inhibitors (many of which are non-specific) have been approved for the treatment of thyroid cancer. Vandetanib and cabozantinib have received FDA approval for the treatment of MTC, while lenvatinib and sorafenib (regardless of mutational status), combination dabrafenib/trametinib (in the setting of anaplastic thyroid carcinoma and BRAFV600E), and larotrectinib (in the setting of NTRK fusions in any tumor type including thyroid) have been approved for DTC. Studies evaluating the efficacy of additional treatment options, including inhibitors of RET alterations and ALK fusions, are ongoing in both adult and pediatric patients with thyroid cancer. Table 2 provides a summary of the therapies (either with a specific approved indication or investigated in a recent clinical trial) according to their targeted molecular alterations and/or tumor type.
Several of the therapies mentioned above nonspecifically target RET, including vandetanib, cabozantinib, lenvatinib, and sorafenib. While their utility has been proven in RET mutation-positive thyroid cancers and thyroid cancer with unknown mutational status, their efficacy (along with that of ponatinib, sunitinib, LOXO-292, BLU-667) in RET fusion-positive thyroid cancer has not yet been fully established. Vandetanib demonstrated both a marked objective response (44% vs. 1%) and progression free survival (hazard ratio, 0.35) compared to placebo for the treatment of symptomatic or progressive unresectable, locally advanced, or metastatic medullary thyroid cancer in adult patients [112,113]. Children with locally advanced or metastatic MTC receiving vandetanib also showed a marked response with a confirmed objective partial response rate of 47%, as well as a median progression free survival of 6.7 years and a 5-year overall survival of 88.2% [114,115]. Advanced RAI-refractory DTC in adults has also shown promising results in a recent phase II trial of vandetanib (hazard ratio 0.63) though preliminary results of a phase 3 trial merely showed a trend favoring vandetanib and were not statistically significant [116]. Cabozantinib appears equally efficacious in adult patients with unresectable and/or locally advanced MTC with a 29% partial response and a statistically significant longer progression free survival (hazard ratio 0.28) [116,117]. To date only five pediatric patients with MTC have received cabozantinib as part of a larger study of dosage and pharmacodynamics; two demonstrated confirmed partial responses and two were observed to have stable disease [118]. Cabozantinib is also being investigated in the context of adult differentiated thyroid cancer, with promising phase I and II trial results, with a partial response of 45% and 40% respectively [116]. In the setting of adult late-stage metastatic and progressive radioactive iodine-refractory differentiated thyroid carcinoma, sorafenib and lenvatinib have both shown improved progression free survival compared to placebo, 10.8 months vs. 5.8 months (hazard ratio 0.59) and 18.3 months vs. 3.6 months (hazard ratio 0.21) respectively [116,119,120]. In the pediatric population, response to sorafenib has been reported both in progressive RAI refractory PTC, PTC with diffuse metastatic disease not amenable to upfront RAI, and as gap therapy in a patient who could not receive RAI in a timely fashion (one case each) [121,122,123], while stable disease has been reported in three pediatric patients with extensive bilateral metastatic pulmonary disease treated with lenvatinib (including one previously treated with sorafenib) [124]. Clinical trials evaluating the efficacy of lenvatinib in the treatment of children with refractory or relapsed solid malignancies (including thyroid cancer) are ongoing. More potent (and specific) RET inhibitors may be on the horizon according to interim clinical data reported at the 2018 American Thyroid Association (ATA) and American Association for Cancer Research (AACR) annual meetings. Both LOXO-292 [125] and BLU-667 [126] were reported to demonstrate robust efficacy in the treatment of RET-fusion (and mutation) positive thyroid cancers. The overall response rate in 31 thyroid cancers (29 MTC, two PTC) treated with BLU-667 was 40%, while LOXO-292 had a 45% objective response rate in RET-mutant MTC and a 100% objective response rate in RET-rearranged PTC [116].
Studies evaluating BRAF inhibitor therapy (vemurafenib/dabrafenib) with and without MEK inhibition (trametinib) are ongoing in a multitude of adult and pediatric malignancies. Adult patients with RAI-refractory metastatic or unresectable BRAFV600E-positive PTCs treated with vemurafenib alone had a response rate of 38.5%, while patients with metastatic or advanced BRAFV600E-positive anaplastic thyroid carcinoma receiving combination dabrafenib and trametinib had a response rate of 69% [127,128]. To the best of our knowledge, use of these inhibitors for the treatment of pediatric thyroid cancer has not been reported.
Most recently, multiple simultaneous studies evaluated the efficacy of TRK inhibitors in advanced solid tumors across three clinical trials comprised of a phase I trial in adults, a phase I/II trial in children (SCOUT), and a phase II basket trial in adults and adolescents (NAVIGATE) [129]. These trials contained a total of seven patients with NTRK-rearranged thyroid cancer. Of the patients with measurable disease (five adults), all demonstrated at least partial response (one responded completely) [129,130,131,132]. The two pediatric patients included in this cohort did not have measurable disease at the time of the trial; they remained stable on therapy at 16 months [130,131,132]. An additional case of pediatric thyroid cancer with TRK-inhibitor induced stabilization of disease was reported by Mahajan et al. [124]. Studies evaluating the efficacy of ALK inhibitors for tumors outside of ALK-rearranged non-small cell lung cancer (including thyroid cancer) are also underway [133], with a case report in the adult population demonstrating an excellent response to crizotinib (>90% reduction of pulmonary disease) [134].

7. Conclusions

In summary, pediatric thyroid carcinoma is a rare tumor but it is the most common carcinoma in children. The increased incidence of gene fusions in pediatric versus adult thyroid cancer is important to recognize as it has both potential implications for diagnostic molecular test selection (e.g., indeterminate FNA diagnoses) and offers novel therapeutic options. More data is needed to determine when alternative therapeutic strategies should be pursued, but targeted therapies have been used with great success in refractory pediatric thyroid cancer.

Author Contributions

V.P.: Writing—original draft preparation, Writing—reviewing and editing; Funding acquisition; E.R.: Writing—original draft preparation, Writing—reviewing and editing; D.H.: Writing—reviewing and editing.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Howlader, N.; Noone, A.M.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.R.; et al. (Eds.) SEER Cancer Statistics Review, 1975–2016; National Cancer Institute: Bethesda, MD, USA, 2019. [Google Scholar]
  2. Bernier, M.O.; Withrow, D.R.; Berrington de Gonzalez, A.; Lam, C.J.K.; Linet, M.S.; Kitahara, C.M.; Shiels, M.S. Trends in pediatric thyroid cancer incidence in the United States, 1998–2013. Cancer 2019. [Google Scholar] [CrossRef] [PubMed]
  3. Qian, Z.J.; Jin, M.C.; Meister, K.D.; Megwalu, U.C. Pediatric Thyroid Cancer Incidence and Mortality Trends in the United States, 1973–2013. JAMA Otolaryngol. Head Neck Surg. 2019. [Google Scholar] [CrossRef] [PubMed]
  4. Mon, R.; Newlon, J. Anaplastic carcinoma of the thyroid in a 12-year old girl. J. Pediatr. Surg. Case Rep. 2015, 3, 404–406. [Google Scholar] [CrossRef] [Green Version]
  5. Wu, Y.L.; Ting, W.H.; Wey, S.L.; Chen, C.K.; Huang, C.Y.; Cheng, S.P.; Lees, Y.J. Poorly differentiated thyroid carcinoma in a 9-year-old boy: Case report. J. Pediatr. Endocrinol. Metab. 2011, 24, 783–786. [Google Scholar] [CrossRef] [PubMed]
  6. Chan, C.M.; Young, J.; Prager, J.; Travers, S. Pediatric Thyroid Cancer. Adv. Pediatr. 2017, 64, 171–190. [Google Scholar] [CrossRef]
  7. Chen, A.Y.; Davies, L. Children and thyroid cancer: Interpreting troubling trends. Cancer 2019. [Google Scholar] [CrossRef]
  8. Francis, G.L.; Waguespack, S.G.; Bauer, A.J.; Angelos, P.; Benvenga, S.; Cerutti, J.M.; Dinauer, C.A.; Hamilton, J.; Hay, I.D.; Luster, M.; et al. Management Guidelines for Children with Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 2015, 25, 716–759. [Google Scholar] [CrossRef] [Green Version]
  9. Estevao-Costa, J.; Gil-Da-Costa, M.J.; Medina, A.M.; Sobrinho-Simoes, M. Thyroid carcinoma in a newborn: Clinical challenges in managing the first recorded case. Med. Pediatr. Oncol. 2000, 34, 290–292. [Google Scholar] [CrossRef]
  10. Dermody, S.; Walls, A.; Harley, E.H., Jr. Pediatric thyroid cancer: An update from the SEER database 2007–2012. Int. J. Pediatr. Otorhinolaryngol. 2016, 89, 121–126. [Google Scholar] [CrossRef]
  11. Hogan, A.R.; Zhuge, Y.; Perez, E.A.; Koniaris, L.G.; Lew, J.I.; Sola, J.E. Pediatric thyroid carcinoma: Incidence and outcomes in 1753 patients. J. Surg. Res. 2009, 156, 167–172. [Google Scholar] [CrossRef]
  12. Penta, L.; Cofini, M.; Lanciotti, L.; Leonardi, A.; Principi, N.; Esposito, S. Hashimoto’s Disease and Thyroid Cancer in Children: Are They Associated? Front. Endocrinol. 2018, 9, 565. [Google Scholar] [CrossRef] [PubMed]
  13. Zimmermann, M.B.; Galetti, V. Iodine intake as a risk factor for thyroid cancer: A comprehensive review of animal and human studies. Thyroid Res. 2015, 8, 8. [Google Scholar] [CrossRef] [PubMed]
  14. Pediatric Treatment Editorial Board. PDQ Childhood Thyroid Cancer Treatment. In PDQ Cancer Information Summaries; National Cancer Institute: Bethesda, MD, USA, 2002; (Updated 6/4/2019).
  15. Rose, J.; Wertheim, B.C.; Guerrero, M.A. Radiation treatment of patients with primary pediatric malignancies: Risk of developing thyroid cancer as a secondary malignancy. Am. J. Surg. 2012, 204, 881–886; discussion 886–887. [Google Scholar] [CrossRef] [PubMed]
  16. Turcotte, L.M.; Liu, Q.; Yasui, Y.; Arnold, M.A.; Hammond, S.; Howell, R.M.; Smith, S.A.; Weathers, R.E.; Henderson, T.O.; Gibson, T.M.; et al. Temporal Trends in Treatment and Subsequent Neoplasm Risk Among 5-Year Survivors of Childhood Cancer, 1970–2015. JAMA 2017, 317, 814–824. [Google Scholar] [CrossRef]
  17. Keskin, M.; Savas-Erdeve, S.; Aycan, Z. Co-Existence of Thyroid Nodule and Thyroid Cancer in Children and Adolescents with Hashimoto Thyroiditis: A Single-Center Study. Horm. Res. Paediatr. 2016, 85, 181–187. [Google Scholar] [CrossRef]
  18. Skarpa, V.; Kousta, E.; Tertipi, A.; Anyfandakis, K.; Vakaki, M.; Dolianiti, M.; Fotinou, A.; Papathanasiou, A. Epidemiological characteristics of children with autoimmune thyroid disease. Hormones 2011, 10, 207–214. [Google Scholar] [CrossRef] [Green Version]
  19. Corrias, A.; Cassio, A.; Weber, G.; Mussa, A.; Wasniewska, M.; Rapa, A.; Gastaldi, R.; Einaudi, S.; Baronio, F.; Vigone, M.C.; et al. Thyroid nodules and cancer in children and adolescents affected by autoimmune thyroiditis. Arch. Pediatr. Adolesc. Med. 2008, 162, 526–531. [Google Scholar] [CrossRef]
  20. Kovatch, K.J.; Bauer, A.J.; Isaacoff, E.J.; Prickett, K.K.; Adzick, N.S.; Kazahaya, K.; Sullivan, L.M.; Mostoufi-Moab, S. Pediatric Thyroid Carcinoma in Patients with Graves’ Disease: The Role of Ultrasound in Selecting Patients for Definitive Therapy. Horm. Res. Paediatr. 2015, 83. [Google Scholar] [CrossRef]
  21. Ohye, H.; Minagawa, A.; Noh, J.Y.; Mukasa, K.; Kunii, Y.; Watanabe, N.; Matsumoto, M.; Suzuki, M.; Yoshihara, A.; Ito, K.; et al. Antithyroid drug treatment for graves’ disease in children: A long-term retrospective study at a single institution. Thyroid 2014, 24, 200–207. [Google Scholar] [CrossRef]
  22. Breuer, C.K.; Solomon, D.; Donovan, P.; Rivkees, S.A.; Udelsman, R. Effect of patient Age on surgical outcomes for Graves’ disease: A case-control study of 100 consecutive patients at a high volume thyroid surgical center. Int. J. Pediatr. Endocrinol. 2013, 2013, 1. [Google Scholar] [CrossRef]
  23. Kumar, A.; Bal, C.S. Differentiated thyroid cancer. Indian J. Pediatr. 2003, 70, 707–713. [Google Scholar] [CrossRef] [PubMed]
  24. Woodruff, S.L.; Arowolo, O.A.; Akute, O.O.; Afolabi, A.O.; Nwariaku, F. Global variation in the pattern of differentiated thyroid cancer. Am. J. Surg. 2010, 200, 462–466. [Google Scholar] [CrossRef] [PubMed]
  25. Mishra, A.; Mishra, S.K.; Agarwal, A.; Das, B.K.; Agarwal, G.; Gambhir, S. Metastatic differentiated thyroid carcinoma: Clinicopathological profile and outcome in an iodine deficient area. World J. Surg. 2002, 26, 153–157. [Google Scholar] [CrossRef] [PubMed]
  26. Bauer, A.J. Molecular Genetics of Thyroid Cancer in Children and Adolescents. Endocrinol. Metab Clin. North. Am. 2017, 46, 389–403. [Google Scholar] [CrossRef] [PubMed]
  27. Waguespack, S.G.; Rich, T.A.; Perrier, N.D.; Jimenez, C.; Cote, G.J. Management of medullary thyroid carcinoma and MEN2 syndromes in childhood. Nat. Rev. Endocrinol. 2011, 7, 596–607. [Google Scholar] [CrossRef] [PubMed]
  28. Starenki, D.; Park, J.I. Pediatric Medullary Thyroid Carcinoma. J. Pediatr. Oncol. 2015, 3, 29–37. [Google Scholar] [CrossRef] [PubMed]
  29. Haugen, B.R.; Alexander, E.K.; Bible, K.C.; Doherty, G.M.; Mandel, S.J.; Nikiforov, Y.E.; Pacini, F.; Randolph, G.W.; Sawka, A.M.; Schlumberger, M.; et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 2016, 26, 1–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Karapanou, O.; Tzanela, M.; Vlassopoulou, B.; Kanaka-Gantenbein, C. Differentiated thyroid cancer in childhood: A literature update. Hormones 2017, 16, 381–387. [Google Scholar] [CrossRef] [PubMed]
  31. Creo, A.; Alahdab, F.; Al Nofal, A.; Thomas, K.; Kolbe, A.; Pittock, S.T. Ultrasonography and the American Thyroid Association Ultrasound-Based Risk Stratification Tool: Utility in Pediatric and Adolescent Thyroid Nodules. Horm. Res. Paediatr. 2018, 90, 93–101. [Google Scholar] [CrossRef] [PubMed]
  32. Lim-Dunham, J.E.; Erdem Toslak, I.; Alsabban, K.; Aziz, A.; Martin, B.; Okur, G.; Longo, K.C. Ultrasound risk stratification for malignancy using the 2015 American Thyroid Association Management Guidelines for Children with Thyroid Nodules and Differentiated Thyroid Cancer. Pediatr. Radiol. 2017, 47, 429–436. [Google Scholar] [CrossRef]
  33. Zhang, Y.; Xia, D.; Lin, P.; Gao, L.; Li, G.; Zhang, W. Sonographic findings of the diffuse sclerosing variant of papillary carcinoma of the thyroid. J. Ultrasound Med. 2010, 29, 1223–1226. [Google Scholar] [CrossRef] [PubMed]
  34. Jung, H.K.; Hong, S.W.; Kim, E.K.; Yoon, J.H.; Kwak, J.Y. Diffuse sclerosing variant of papillary thyroid carcinoma: Sonography and specimen radiography. J. Ultrasound Med. 2013, 32, 347–354. [Google Scholar] [CrossRef] [PubMed]
  35. Kwak, J.Y.; Kim, E.K.; Hong, S.W.; Oh, K.K.; Kim, M.J.; Park, C.S.; Cheong, W.Y. Diffuse sclerosing variant of papillary carcinoma of the thyroid: Ultrasound features with histopathological correlation. Clin. Radiol. 2007, 62, 382–386. [Google Scholar] [CrossRef] [PubMed]
  36. Creo, A.; Alahdab, F.; Al Nofal, A.; Thomas, K.; Kolbe, A.; Pittock, S. Diagnostic accuracy of the McGill thyroid nodule score in paediatric patients. Clin. Endocrinol. 2019, 90, 200–207. [Google Scholar] [CrossRef] [PubMed]
  37. Hodax, J.K.; Reinert, S.E.; Quintos, J.B. Autonomously Functioning Thyroid Nodules in Patients <21 Years of Age: The Rhode Island Hospital Experience from 2003–2013. Endocr. Pract. 2016, 22, 328–337. [Google Scholar] [CrossRef] [PubMed]
  38. Ly, S.; Frates, M.C.; Benson, C.B.; Peters, H.E.; Grant, F.D.; Drubach, L.A.; Voss, S.D.; Feldman, H.A.; Smith, J.R.; Barletta, J.; et al. Features and Outcome of Autonomous Thyroid Nodules in Children: 31 Consecutive Patients Seen at a Single Center. J. Clin. Endocrinol. Metab. 2016, 101, 3856–3862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Croom, R.D., 3rd; Thomas, C.G., Jr.; Reddick, R.L.; Tawil, M.T. Autonomously functioning thyroid nodules in childhood and adolescence. Surgery 1987, 102, 1101–1108. [Google Scholar] [PubMed]
  40. Niedziela, M.; Breborowicz, D.; Trejster, E.; Korman, E. Hot nodules in children and adolescents in western Poland from 1996 to 2000: Clinical analysis of 31 patients. J. Pediatr. Endocrinol. Metab. 2002, 15, 823–830. [Google Scholar] [CrossRef] [PubMed]
  41. Partyka, K.L.; Huang, E.C.; Cramer, H.M.; Chen, S.; Wu, H.H. Histologic and clinical follow-up of thyroid fine-needle aspirates in pediatric patients. Cancer Cytopathol. 2016, 124, 467–471. [Google Scholar] [CrossRef] [Green Version]
  42. Wang, H.; Mehrad, M.; Ely, K.A.; Liang, J.; Solorzano, C.C.; Neblett, W.W., 3rd; Coogan, A.C.; Weiss, V.L. Incidence and malignancy rates of indeterminate pediatric thyroid nodules. Cancer Cytopathol. 2019, 127, 231–239. [Google Scholar] [CrossRef] [PubMed]
  43. Balachandar, S.; La Quaglia, M.; Tuttle, R.M.; Heller, G.; Ghossein, R.A.; Sklar, C.A. Pediatric Differentiated Thyroid Carcinoma of Follicular Cell Origin: Prognostic Significance of Histologic Subtypes. Thyroid 2016, 26, 219–226. [Google Scholar] [CrossRef]
  44. Collini, P.; Mattavelli, F.; Pellegrinelli, A.; Barisella, M.; Ferrari, A.; Massimino, M. Papillary carcinoma of the thyroid gland of childhood and adolescence: Morphologic subtypes, biologic behavior and prognosis: A clinicopathologic study of 42 sporadic cases treated at a single institution during a 30-year period. Am. J. Surg. Pathol. 2006, 30, 1420–1426. [Google Scholar] [CrossRef] [PubMed]
  45. Onder, S.; Ozturk Sari, S.; Yegen, G.; Sormaz, I.C.; Yilmaz, I.; Poyrazoglu, S.; Sanli, Y.; Giles Senyurek, Y.; Kapran, Y.; Mete, O. Classic Architecture with Multicentricity and Local Recurrence, and Absence of TERT Promoter Mutations are Correlates of BRAF (V600E) Harboring Pediatric Papillary Thyroid Carcinomas. Endocr. Pathol. 2016, 27, 153–161. [Google Scholar] [CrossRef] [PubMed]
  46. Rossi, E.D.; Mehrotra, S.; Kilic, A.I.; Toslak, I.E.; Lim-Dunham, J.; Martini, M.; Fadda, G.; Lombardi, C.P.; Larocca, L.M.; Barkan, G.A. Noninvasive follicular thyroid neoplasm with papillary-like nuclear features in the pediatric age group. Cancer Cytopathol. 2018, 126, 27–35. [Google Scholar] [CrossRef] [PubMed]
  47. Spinelli, C.; Rallo, L.; Morganti, R.; Mazzotti, V.; Inserra, A.; Cecchetto, G.; Massimino, M.; Collini, P.; Strambi, S. Surgical management of follicular thyroid carcinoma in children and adolescents: A study of 30 cases. J. Pediatr. Surg. 2019, 54, 521–526. [Google Scholar] [CrossRef]
  48. Enomoto, K.; Enomoto, Y.; Uchino, S.; Yamashita, H.; Noguchi, S. Follicular thyroid cancer in children and adolescents: Clinicopathologic features, long-term survival, and risk factors for recurrence. Endocr. J. 2013, 60, 629–635. [Google Scholar] [CrossRef] [PubMed]
  49. Scognamiglio, T. C Cell and Follicular Epithelial Cell Precursor Lesions of the Thyroid. Arch. Pathol. Lab. Med. 2017, 141, 1646–1652. [Google Scholar] [CrossRef]
  50. Perry, A.; Molberg, K.; Albores-Saavedra, J. Physiologic versus neoplastic C-cell hyperplasia of the thyroid: Separation of distinct histologic and biologic entities. Cancer 1996, 77, 750–756. [Google Scholar] [CrossRef]
  51. Verga, U.; Ferrero, S.; Vicentini, L.; Brambilla, T.; Cirello, V.; Muzza, M.; Beck-Peccoz, P.; Fugazzola, L. Histopathological and molecular studies in patients with goiter and hypercalcitoninemia: Reactive or neoplastic C-cell hyperplasia? Endocr. Relat. Cancer 2007, 14, 393–403. [Google Scholar] [CrossRef]
  52. Krueger, J.E.; Maitra, A.; Albores-Saavedra, J. Inherited medullary microcarcinoma of the thyroid: A study of 11 cases. Am. J. Surg. Pathol. 2000, 24, 853–858. [Google Scholar] [CrossRef]
  53. McDermott, M.B.; Swanson, P.E.; Wick, M.R. Immunostains for collagen type IV discriminate between C-cell hyperplasia and microscopic medullary carcinoma in multiple endocrine neoplasia, type 2a. Hum. Pathol. 1995, 26, 1308–1312. [Google Scholar] [CrossRef]
  54. Monaco, S.E.; Pantanowitz, L.; Khalbuss, W.E.; Benkovich, V.A.; Ozolek, J.; Nikiforova, M.N.; Simons, J.P.; Nikiforov, Y.E. Cytomorphological and molecular genetic findings in pediatric thyroid fine-needle aspiration. Cancer Cytopathol. 2012, 120, 342–350. [Google Scholar] [CrossRef] [PubMed]
  55. Fugazzola, L.; Pilotti, S.; Pinchera, A.; Vorontsova, T.V.; Mondellini, P.; Bongarzone, I.; Greco, A.; Astakhova, L.; Butti, M.G.; Demidchik, E.P.; et al. Oncogenic rearrangements of the RET proto-oncogene in papillary thyroid carcinomas from children exposed to the Chernobyl nuclear accident. Cancer Res. 1995, 55, 5617–5620. [Google Scholar] [PubMed]
  56. Klugbauer, S.; Lengfelder, E.; Demidchik, E.P.; Rabes, H.M. High prevalence of RET rearrangement in thyroid tumors of children from Belarus after the Chernobyl reactor accident. Oncogene 1995, 11, 2459–2467. [Google Scholar] [PubMed]
  57. Nikiforov, Y.E.; Nikiforova, M.N.; Gnepp, D.R.; Fagin, J.A. Prevalence of mutations of ras and p53 in benign and malignant thyroid tumors from children exposed to radiation after the Chernobyl nuclear accident. Oncogene 1996, 13, 687–693. [Google Scholar] [PubMed]
  58. Bongarzone, I.; Fugazzola, L.; Vigneri, P.; Mariani, L.; Mondellini, P.; Pacini, F.; Basolo, F.; Pinchera, A.; Pilotti, S.; Pierotti, M.A. Age-related activation of the tyrosine kinase receptor protooncogenes RET and NTRK1 in papillary thyroid carcinoma. J. Clin. Endocrinol. Metab. 1996, 81, 2006–2009. [Google Scholar] [CrossRef] [PubMed]
  59. Williams, G.H.; Rooney, S.; Thomas, G.A.; Cummins, G.; Williams, E.D. RET activation in adult and childhood papillary thyroid carcinoma using a reverse transcriptase-n-polymerase chain reaction approach on archival-nested material. Br. J. Cancer 1996, 74, 585–589. [Google Scholar] [CrossRef] [Green Version]
  60. Nikiforov, Y.E.; Rowland, J.M.; Bove, K.E.; Monforte-Munoz, H.; Fagin, J.A. Distinct pattern of ret oncogene rearrangements in morphological variants of radiation-induced and sporadic thyroid papillary carcinomas in children. Cancer Res. 1997, 57, 1690–1694. [Google Scholar]
  61. Motomura, T.; Nikiforov, Y.E.; Namba, H.; Ashizawa, K.; Nagataki, S.; Yamashita, S.; Fagin, J.A. ret rearrangements in Japanese pediatric and adult papillary thyroid cancers. Thyroid 1998, 8, 485–489. [Google Scholar] [CrossRef]
  62. Suchy, B.; Waldmann, V.; Klugbauer, S.; Rabes, H.M. Absence of RAS and p53 mutations in thyroid carcinomas of children after Chernobyl in contrast to adult thyroid tumours. Br. J. Cancer 1998, 77, 952–955. [Google Scholar] [CrossRef]
  63. Pisarchik, A.V.; Ermak, G.; Fomicheva, V.; Kartel, N.A.; Figge, J. The ret/PTC1 rearrangement is a common feature of Chernobyl-associated papillary thyroid carcinomas from Belarus. Thyroid 1998, 8, 133–139. [Google Scholar] [CrossRef] [PubMed]
  64. Pisarchik, A.V.; Ermak, G.; Demidchik, E.P.; Mikhalevich, L.S.; Kartel, N.A.; Figge, J. Low prevalence of the ret/PTC3r1 rearrangement in a series of papillary thyroid carcinomas presenting in Belarus ten years post-Chernobyl. Thyroid 1998, 8, 1003–1008. [Google Scholar] [CrossRef] [PubMed]
  65. Fenton, C.; Anderson, J.; Lukes, Y.; Dinauer, C.A.; Tuttle, R.M.; Francis, G.L. Ras mutations are uncommon in sporadic thyroid cancer in children and young adults. J. Endocrinol. Invest. 1999, 22, 781–789. [Google Scholar] [CrossRef] [PubMed]
  66. Thomas, G.A.; Bunnell, H.; Cook, H.A.; Williams, E.D.; Nerovnya, A.; Cherstvoy, E.D.; Tronko, N.D.; Bogdanova, T.I.; Chiappetta, G.; Viglietto, G.; et al. 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, 4232–4238. [Google Scholar] [CrossRef] [PubMed]
  67. Beimfohr, C.; Klugbauer, S.; Demidchik, E.P.; Lengfelder, E.; Rabes, H.M. NTRK1 re-arrangement in papillary thyroid carcinomas of children after the Chernobyl reactor accident. Int J. Cancer 1999, 80, 842–847. [Google Scholar] [CrossRef]
  68. Fenton, C.L.; Lukes, Y.; Nicholson, D.; Dinauer, C.A.; Francis, G.L.; Tuttle, R.M. The ret/PTC mutations are common in sporadic papillary thyroid carcinoma of children and young adults. J. Clin. Endocrinol. Metab. 2000, 85, 1170–1175. [Google Scholar] [CrossRef] [PubMed]
  69. Santoro, M.; Thomas, G.A.; Vecchio, G.; Williams, G.H.; Fusco, A.; Chiappetta, G.; Pozcharskaya, V.; Bogdanova, T.I.; Demidchik, E.P.; Cherstvoy, E.D.; et al. Gene rearrangement and Chernobyl related thyroid cancers. Br. J. Cancer 2000, 82, 315–322. [Google Scholar] [CrossRef] [Green Version]
  70. Rabes, H.M.; Demidchik, E.P.; Sidorow, J.D.; 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, 1093–1103. [Google Scholar]
  71. Pauws, E.; Tummers, R.F.; Ris-Stalpers, C.; de Vijlder, J.J.; Voute, T. Absence of activating mutations in ras and gsp oncogenes in a cohort of nine patients with sporadic pediatric thyroid tumors. Med. Pediatr. Oncol. 2001, 36, 630–634. [Google Scholar] [CrossRef]
  72. Elisei, R.; Romei, C.; Vorontsova, T.; Cosci, B.; Veremeychik, V.; Kuchinskaya, E.; Basolo, F.; Demidchik, E.P.; Miccoli, P.; Pinchera, A.; et al. RET/PTC rearrangements in thyroid nodules: Studies in irradiated and not irradiated, malignant and benign thyroid lesions in children and adults. J. Clin. Endocrinol. Metab. 2001, 86, 3211–3216. [Google Scholar] [CrossRef]
  73. Nikiforova, M.N.; Ciampi, R.; Salvatore, G.; Santoro, M.; Gandhi, M.; Knauf, J.A.; Thomas, G.A.; Jeremiah, S.; Bogdanova, T.I.; Tronko, M.D.; et al. Low prevalence of BRAF mutations in radiation-induced thyroid tumors in contrast to sporadic papillary carcinomas. Cancer Lett. 2004, 209, 1–6. [Google Scholar] [CrossRef] [PubMed]
  74. Kumagai, A.; Namba, H.; Saenko, V.A.; Ashizawa, K.; Ohtsuru, A.; Ito, M.; Ishikawa, N.; Sugino, K.; Ito, K.; Jeremiah, S.; et al. Low frequency of BRAFT1796A mutations in childhood thyroid carcinomas. J. Clin. Endocrinol. Metab. 2004, 89, 4280–4284. [Google Scholar] [CrossRef] [PubMed]
  75. Lima, J.; Trovisco, V.; Soares, P.; Maximo, V.; Magalhaes, J.; Salvatore, G.; Santoro, M.; Bogdanova, T.; Tronko, M.; Abrosimov, A.; et al. BRAF mutations are not a major event in post-Chernobyl childhood thyroid carcinomas. J. Clin. Endocrinol. Metab. 2004, 89, 4267–4271. [Google Scholar] [CrossRef] [PubMed]
  76. Penko, K.; Livezey, J.; Fenton, C.; Patel, A.; Nicholson, D.; Flora, M.; Oakley, K.; Tuttle, R.M.; Francis, G. BRAF mutations are uncommon in papillary thyroid cancer of young patients. Thyroid 2005, 15, 320–325. [Google Scholar] [CrossRef] [PubMed]
  77. Rosenbaum, E.; Hosler, G.; Zahurak, M.; Cohen, Y.; Sidransky, D.; Westra, W.H. Mutational activation of BRAF is not a major event in sporadic childhood papillary thyroid carcinoma. Mod. Pathol. 2005, 18, 898–902. [Google Scholar] [CrossRef]
  78. Espadinha, C.; Santos, J.R.; Sobrinho, L.G.; Bugalho, M.J. Expression of iodine metabolism genes in human thyroid tissues: Evidence for age and BRAFV600E mutation dependency. Clin. Endocrinol 2009, 70, 629–635. [Google Scholar] [CrossRef] [PubMed]
  79. Sassolas, G.; Hafdi-Nejjari, Z.; Ferraro, A.; Decaussin-Petrucci, M.; Rousset, B.; Borson-Chazot, F.; Borbone, E.; Berger, N.; Fusco, A. Oncogenic alterations in papillary thyroid cancers of young patients. Thyroid 2012, 22, 17–26. [Google Scholar] [CrossRef] [PubMed]
  80. Ricarte-Filho, J.C.; Li, S.; Garcia-Rendueles, M.E.; Montero-Conde, C.; Voza, F.; Knauf, J.A.; Heguy, A.; Viale, A.; Bogdanova, T.; Thomas, G.A.; et al. Identification of kinase fusion oncogenes in post-Chernobyl radiation-induced thyroid cancers. J. Clin. Invest. 2013, 123, 4935–4944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Leeman-Neill, R.J.; Brenner, A.V.; Little, M.P.; Bogdanova, T.I.; Hatch, M.; Zurnadzy, L.Y.; Mabuchi, K.; Tronko, M.D.; Nikiforov, Y.E. RET/PTC and PAX8/PPARgamma chromosomal rearrangements in post-Chernobyl thyroid cancer and their association with iodine-131 radiation dose and other characteristics. Cancer 2013, 119, 1792–1799. [Google Scholar] [CrossRef] [PubMed]
  82. Leeman-Neill, R.J.; Kelly, L.M.; Liu, P.; Brenner, A.V.; Little, M.P.; Bogdanova, T.I.; Evdokimova, V.N.; Hatch, M.; Zurnadzy, L.Y.; Nikiforova, M.N.; et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer 2014, 120, 799–807. [Google Scholar] [CrossRef]
  83. Henke, L.E.; Perkins, S.M.; Pfeifer, J.D.; Ma, C.; Chen, Y.; DeWees, T.; Grigsby, P.W. BRAF V600E mutational status in pediatric thyroid cancer. Pediatr. Blood Cancer 2014, 61, 1168–1172. [Google Scholar] [CrossRef] [PubMed]
  84. Givens, D.J.; Buchmann, L.O.; Agarwal, A.M.; Grimmer, J.F.; Hunt, J.P. BRAF V600E does not predict aggressive features of pediatric papillary thyroid carcinoma. Laryngoscope 2014, 124, E389–E393. [Google Scholar] [CrossRef] [PubMed]
  85. Mitsutake, N.; Fukushima, T.; Matsuse, M.; Rogounovitch, T.; Saenko, V.; Uchino, S.; Ito, M.; Suzuki, K.; Suzuki, S.; Yamashita, S. BRAF(V600E) mutation is highly prevalent in thyroid carcinomas in the young population in Fukushima: A different oncogenic profile from Chernobyl. Sci. Rep. 2015, 5, 16976. [Google Scholar] [CrossRef] [PubMed]
  86. Ballester, L.Y.; Sarabia, S.F.; Sayeed, H.; Patel, N.; Baalwa, J.; Athanassaki, I.; Hernandez, J.A.; Fang, E.; Quintanilla, N.M.; Roy, A.; et al. Integrating Molecular Testing in the Diagnosis and Management of Children with Thyroid Lesions. Pediatr. Dev. Pathol. 2016, 19, 94–100. [Google Scholar] [CrossRef] [PubMed]
  87. Picarsic, J.L.; Buryk, M.A.; Ozolek, J.; Ranganathan, S.; Monaco, S.E.; Simons, J.P.; Witchel, S.F.; Gurtunca, N.; Joyce, J.; Zhong, S.; et al. Molecular Characterization of Sporadic Pediatric Thyroid Carcinoma with the DNA/RNA ThyroSeq v2 Next-Generation Sequencing Assay. Pediatr. Dev. Pathol. 2016, 19, 115–122. [Google Scholar] [CrossRef] [PubMed]
  88. Prasad, M.L.; Vyas, M.; Horne, M.J.; Virk, R.K.; Morotti, R.; Liu, Z.; Tallini, G.; Nikiforova, M.N.; Christison-Lagay, E.R.; Udelsman, R.; et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer 2016, 122, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
  89. Nikita, M.E.; Jiang, W.; Cheng, S.M.; Hantash, F.M.; McPhaul, M.J.; Newbury, R.O.; Phillips, S.A.; Reitz, R.E.; Waldman, F.M.; Newfield, R.S. Mutational Analysis in Pediatric Thyroid Cancer and Correlations with Age, Ethnicity, and Clinical Presentation. Thyroid 2016, 26, 227–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Alzahrani, A.S.; Qasem, E.; Murugan, A.K.; Al-Hindi, H.N.; AlKhafaji, D.; Almohanna, M.; Xing, M.; Alhomaidah, D.; AlSwailem, M. Uncommon TERT Promoter Mutations in Pediatric Thyroid Cancer. Thyroid 2016, 26, 235–241. [Google Scholar] [CrossRef] [PubMed]
  91. Gertz, R.J.; Nikiforov, Y.; Rehrauer, W.; McDaniel, L.; Lloyd, R.V. Mutation in BRAF and Other Members of the MAPK Pathway in Papillary Thyroid Carcinoma in the Pediatric Population. Arch. Pathol. Lab. Med. 2016, 140, 134–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Cordioli, M.I.; Moraes, L.; Carvalheira, G.; Sisdelli, L.; Alves, M.T.; Delcelo, R.; Monte, O.; Longui, C.A.; Cury, A.N.; Cerutti, J.M. AGK-BRAF gene fusion is a recurrent event in sporadic pediatric thyroid carcinoma. Cancer Med. 2016, 5, 1535–1541. [Google Scholar] [CrossRef]
  93. Cordioli, M.I.; Moraes, L.; Bastos, A.U.; Besson, P.; Alves, M.T.; Delcelo, R.; Monte, O.; Longui, C.; Cury, A.N.; Cerutti, J.M. Fusion Oncogenes Are the Main Genetic Events Found in Sporadic Papillary Thyroid Carcinomas from Children. Thyroid 2017, 27, 182–188. [Google Scholar] [CrossRef] [PubMed]
  94. Alzahrani, A.S.; Murugan, A.K.; Qasem, E.; Alswailem, M.; Al-Hindi, H.; Shi, Y. Single Point Mutations in Pediatric Differentiated Thyroid Cancer. Thyroid 2017, 27, 189–196. [Google Scholar] [CrossRef] [PubMed]
  95. Vanden Borre, P.; Schrock, A.B.; Anderson, P.M.; Morris, J.C., 3rd; Heilmann, A.M.; Holmes, O.; Wang, K.; Johnson, A.; Waguespack, S.G.; Ou, S.I.; et al. Pediatric, Adolescent, and Young Adult Thyroid Carcinoma Harbors Frequent and Diverse Targetable Genomic Alterations, Including Kinase Fusions. Oncologist 2017, 22, 255–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Hardee, S.; Prasad, M.L.; Hui, P.; Dinauer, C.A.; Morotti, R.A. Pathologic Characteristics, Natural History, and Prognostic Implications of BRAF(V600E) Mutation in Pediatric Papillary Thyroid Carcinoma. Pediatr. Dev. Pathol. 2017, 20, 206–212. [Google Scholar] [CrossRef] [PubMed]
  97. Oishi, N.; Kondo, T.; Nakazawa, T.; Mochizuki, K.; Inoue, T.; Kasai, K.; Tahara, I.; Yabuta, T.; Hirokawa, M.; Miyauchi, A.; et al. Frequent BRAF (V600E) and Absence of TERT Promoter Mutations Characterize Sporadic Pediatric Papillary Thyroid Carcinomas in Japan. Endocr. Pathol. 2017, 28, 103–111. [Google Scholar] [CrossRef] [PubMed]
  98. Vuong, H.G.; Kondo, T.; Oishi, N.; Nakazawa, T.; Mochizuki, K.; Miyauchi, A.; Hirokawa, M.; Katoh, R. Paediatric follicular thyroid carcinoma—Indolent cancer with low prevalence of RAS mutations and absence of PAX8-PPARG fusion in a Japanese population. Histopathology 2017, 71, 760–768. [Google Scholar] [CrossRef]
  99. Mostoufi-Moab, S.; Labourier, E.; Sullivan, L.; LiVolsi, V.; Li, Y.; Xiao, R.; Beaudenon-Huibregtse, S.; Kazahaya, K.; Adzick, N.S.; Baloch, Z.; et al. Molecular Testing for Oncogenic Gene Alterations in Pediatric Thyroid Lesions. Thyroid 2018, 28, 60–67. [Google Scholar] [CrossRef]
  100. Wasserman, J.D.; Sabbaghian, N.; Fahiminiya, S.; Chami, R.; Mete, O.; Acker, M.; Wu, M.K.; Shlien, A.; de Kock, L.; Foulkes, W.D. DICER1 Mutations Are Frequent in Adolescent-Onset Papillary Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2018, 103, 2009–2015. [Google Scholar] [CrossRef] [Green Version]
  101. Huang, M.; Yan, C.; Wei, H.; Lv, Y.; Ling, R. Clinicopathological characteristics and prognosis of thyroid cancer in northwest China: A population-based retrospective study of 2490 patients. Thorac. Cancer 2018, 9, 1453–1460. [Google Scholar] [CrossRef]
  102. Sisdelli, L.; Cordioli, M.; Vaisman, F.; Moraes, L.; Colozza-Gama, G.A.; Alves, P.A.G., Jr.; Araujo, M.L., Jr.; Alves, M.T.S.; Monte, O.; Longui, C.A.; et al. AGK-BRAF is associated with distant metastasis and younger age in pediatric papillary thyroid carcinoma. Pediatr. Blood Cancer 2019, 66, e27707. [Google Scholar] [CrossRef]
  103. Pekova, B.; Dvorakova, S.; Sykorova, V.; Vacinova, G.; Vaclavikova, E.; Moravcova, J.; Katra, R.; Vlcek, P.; Sykorova, P.; Kodetova, D.; et al. Somatic genetic alterations in a large cohort of pediatric thyroid nodules. Endocr. Connect. 2019. [Google Scholar] [CrossRef] [PubMed]
  104. Ronsley, R.; Rassekh, S.R.; Shen, Y.; Lee, A.F.; Jantzen, C.; Halparin, J.; Albert, C.; Hawkins, D.S.; Amed, S.; Rothstein, R.; et al. Application of genomics to identify therapeutic targets in recurrent pediatric papillary thyroid carcinoma. Cold Spring Harb. Mol. Case Stud. 2018, 4, a002568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Prescott, J.D.; Zeiger, M.A. The RET oncogene in papillary thyroid carcinoma. Cancer 2015, 121, 2137–2146. [Google Scholar] [CrossRef] [PubMed]
  106. Romei, C.; Elisei, R. RET/PTC Translocations and Clinico-Pathological Features in Human Papillary Thyroid Carcinoma. Front. Endocrinol. 2012, 3, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Cancer Genome Atlas Research, N. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef]
  108. Hay, I.D.; Gonzalez-Losada, T.; Reinalda, M.S.; Honetschlager, J.A.; Richards, M.L.; Thompson, G.B. Long-term outcome in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World J. Surg 2010, 34, 1192–1202. [Google Scholar] [CrossRef] [PubMed]
  109. Haddad, R.I.; Bischoff, L.; Busaidy, N.L.; Callender, G.; Dickson, P.; Duh, Q.Y.; Ehya, H.; Goldner, W.; Haymart, M.; Hoh, C.; et al. The NCCN Clinical Practice Guidelines in Oncology, Thyroid Carcinoma; (Version 1.2019); National Comprehensive Cancer Network: Fort Washington, PA, USA, 2019. [Google Scholar]
  110. Parisi, M.T.; Eslamy, H.; Mankoff, D. Management of Differentiated Thyroid Cancer in Children: Focus on the American Thyroid Association Pediatric Guidelines. Semin. Nucl. Med. 2016, 46, 147–164. [Google Scholar] [CrossRef] [PubMed]
  111. Cordioli, M.I.; Moraes, L.; Cury, A.N.; Cerutti, J.M. Are we really at the dawn of understanding sporadic pediatric thyroid carcinoma? Endocr. Relat. Cancer 2015, 22, R311–R324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Thornton, K.; Kim, G.; Maher, V.E.; Chattopadhyay, S.; Tang, S.; Moon, Y.J.; Song, P.; Marathe, A.; Balakrishnan, S.; Zhu, H.; et al. Vandetanib for the treatment of symptomatic or progressive medullary thyroid cancer in patients with unresectable locally advanced or metastatic disease: U.S. Food and Drug Administration drug approval summary. Clin. Cancer Res. 2012, 18, 3722–3730. [Google Scholar] [CrossRef]
  113. Wells, S.A., Jr.; Robinson, B.G.; Gagel, R.F.; Dralle, H.; Fagin, J.A.; Santoro, M.; Baudin, E.; Elisei, R.; Jarzab, B.; Vasselli, J.R.; et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: A randomized, double-blind phase III trial. J. Clin. Oncol. 2012, 30, 134–141. [Google Scholar] [CrossRef]
  114. Fox, E.; Widemann, B.C.; Chuk, M.K.; Marcus, L.; Aikin, A.; Whitcomb, P.O.; Merino, M.J.; Lodish, M.; Dombi, E.; Steinberg, S.M.; et al. Vandetanib in children and adolescents with multiple endocrine neoplasia type 2B associated medullary thyroid carcinoma. Clin. Cancer Res. 2013, 19, 4239–4248. [Google Scholar] [CrossRef] [PubMed]
  115. Kraft, I.L.; Akshintala, S.; Zhu, Y.; Lei, H.; Derse-Anthony, C.; Dombi, E.; Steinberg, S.M.; Lodish, M.; Waguespack, S.G.; Kapustina, O.; et al. Outcomes of Children and Adolescents with Advanced Hereditary Medullary Thyroid Carcinoma Treated with Vandetanib. Clin. Cancer Res. 2018, 24, 753–765. [Google Scholar] [CrossRef] [PubMed]
  116. Rao, S.N.; Cabanillas, M.E. Navigating Systemic Therapy in Advanced Thyroid Carcinoma: From Standard of Care to Personalized Therapy and Beyond. J. Endocr. Soc. 2018, 2, 1109–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kurzrock, R.; Sherman, S.I.; Ball, D.W.; Forastiere, A.A.; Cohen, R.B.; Mehra, R.; Pfister, D.G.; Cohen, E.E.; Janisch, L.; Nauling, F.; et al. Activity of XL184 (Cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J. Clin. Oncol. 2011, 29, 2660–2666. [Google Scholar] [CrossRef] [PubMed]
  118. Chuk, M.K.; Widemann, B.C.; Minard, C.G.; Liu, X.; Kim, A.; Bernhardt, M.B.; Kudgus, R.A.; Reid, J.M.; Voss, S.D.; Blaney, S.; et al. A phase 1 study of cabozantinib in children and adolescents with recurrent or refractory solid tumors, including CNS tumors: Trial ADVL1211, a report from the Children’s Oncology Group. Pediatr. Blood Cancer 2018, 65, e27077. [Google Scholar] [CrossRef] [PubMed]
  119. Brose, M.S.; Nutting, C.M.; Jarzab, B.; Elisei, R.; Siena, S.; Bastholt, L.; de la Fouchardiere, C.; Pacini, F.; Paschke, R.; Shong, Y.K.; et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: A randomised, double-blind, phase 3 trial. Lancet 2014, 384, 319–328. [Google Scholar] [CrossRef]
  120. Schlumberger, M.; Tahara, M.; Wirth, L.J.; Robinson, B.; Brose, M.S.; Elisei, R.; Habra, M.A.; Newbold, K.; Shah, M.H.; Hoff, A.O.; et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N. Engl. J. Med. 2015, 372, 621–630. [Google Scholar] [CrossRef] [PubMed]
  121. Waguespack, S.G.; Sherman, S.I.; Williams, M.D.; Clayman, G.L.; Herzog, C.E. The successful use of sorafenib to treat pediatric papillary thyroid carcinoma. Thyroid 2009, 19, 407–412. [Google Scholar] [CrossRef] [PubMed]
  122. Higuchi, Y.; Motoki, T.; Ishida, H.; Kanamitsu, K.; Washio, K.; Oyama, T.; Noda, T.; Tsurumaru, Y.; Okada, A.; Tsukahara, H.; et al. Sorafenib treatment for papillary thyroid carcinoma with diffuse lung metastases in a child with autism spectrum disorder: A case report. BMC Cancer 2017, 17, 775. [Google Scholar] [CrossRef] [PubMed]
  123. Iyer, P.; Mayer, J.L.; Ewig, J.M. Response to sorafenib in a pediatric patient with papillary thyroid carcinoma with diffuse nodular pulmonary disease requiring mechanical ventilation. Thyroid 2014, 24, 169–174. [Google Scholar] [CrossRef]
  124. Mahajan, P.; Dawrant, J.; Kheradpour, A.; Quintanilla, N.M.; Lopez, M.E.; Orth, R.C.; Athanassaki, I.; Venkatramani, R. Response to Lenvatinib in Children with Papillary Thyroid Carcinoma. Thyroid 2018, 28, 1450–1454. [Google Scholar] [CrossRef] [PubMed]
  125. Drilon, A.E.; Subbiah, V.; Oxnard, G.R.; Bauer, T.M.; Velcheti, V.; Lakhani, N.J.; Besse, B.; Park, K.; Patel, J.D.; Cabanillas, M.E.; et al. A phase 1 study of LOXO-292, a potent and highly selective RET inhibitor, in patients with RET-altered cancers. J. Clin. Oncol. 2018, 36, 102. [Google Scholar] [CrossRef]
  126. BLU-667 Targets RET-Altered Cancers. Cancer Discovery 2018, 8, OF8. [CrossRef] [PubMed]
  127. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.; Cabanillas, M.E.; Urbanowitz, G.; et al. Dabrafenib and Trametinib Treatment in Patients with Locally Advanced or Metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer. J. Clin. Oncol. 2018, 36, 7–13. [Google Scholar] [CrossRef] [PubMed]
  128. Brose, M.S.; Cabanillas, M.E.; Cohen, E.E.; Wirth, L.J.; Riehl, T.; Yue, H.; Sherman, S.I.; Sherman, E.J. Vemurafenib in patients with BRAF(V600E)-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: A non-randomised, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 1272–1282. [Google Scholar] [CrossRef]
  129. Brose, M.S.; Albert, C.M.; Waguespack, S.G.; Cabanillas, M.E.; Ma, P.C.; Sohal, D.; Cox, M.C.; Ku, N.C.; Wirth, L.J. Activity of Larotrectinib in Patients with Advanced TRK Fusion Thyroid Cancer. In Proceedings of the 88th Annual Meeting of the American Thyroid Association, Washington, DC, USA, 3–7 October 2018. [Google Scholar]
  130. Laetsch, T.W.; DuBois, S.G.; Mascarenhas, L.; Turpin, B.; Federman, N.; Albert, C.M.; Nagasubramanian, R.; Davis, J.L.; Rudzinski, E.; Feraco, A.M.; et al. Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: Phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol. 2018, 19, 705–714. [Google Scholar] [CrossRef]
  131. Chen, Y.; Chi, P. Basket trial of TRK inhibitors demonstrates efficacy in TRK fusion-positive cancers. J. Hematol. Oncol. 2018, 11, 78. [Google Scholar] [CrossRef]
  132. Drilon, A.; Laetsch, T.W.; Kummar, S.; DuBois, S.G.; Lassen, U.N.; Demetri, G.D.; Nathenson, M.; Doebele, R.C.; Farago, A.F.; Pappo, A.S.; et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N. Engl. J. Med. 2018, 378, 731–739. [Google Scholar] [CrossRef]
  133. Cao, Z.; Gao, Q.; Fu, M.; Ni, N.; Pei, Y.; Ou, W.B. Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives. Oncol. Lett. 2019, 17, 2020–2030. [Google Scholar] [CrossRef]
  134. Godbert, Y.; Henriques de Figueiredo, B.; Bonichon, F.; Chibon, F.; Hostein, I.; Perot, G.; Dupin, C.; Daubech, A.; Belleannee, G.; Gros, A.; et al. Remarkable Response to Crizotinib in Woman with Anaplastic Lymphoma Kinase-Rearranged Anaplastic Thyroid Carcinoma. J. Clin. Oncol. 2015, 33, e84–e87. [Google Scholar] [CrossRef]
Table 1. Molecular alterations in “pediatric” thyroid cancer by study.
Table 1. Molecular alterations in “pediatric” thyroid cancer by study.
AgeRadiation ExposureDiagnosisRET-FusionRET-MutationRAS-MutationBRAF-MutationBRAF-FusionNTRK-FusionALK-FusionPPARG-FusionAdditional TargetsNotes
Fugazzola L et al. [55]6–14 yearsPost-radiation6 PTC4/6 (67%)NENENENE0/6 (0%)NENE
Klugbauer et al. [56]<11 yearsPost-radiation12 PTC8/12 (67%)NENENENENENENE
Nikiforov et al. [57]5–19 yearsPost-radiation33 PTCNENE0/33 (0%)NENENENENETP53
1 FTCNENE1/1 (100%)NENENENENE
Bongarzone et al. [58]4–19 yearsSporadic *9 PTC6/9 (67%)NENENENE1/9 (11%)NENE * Fusion-+ cases
Williams et al. [59]7–14 yearsSporadic21 PTC10/21 (48%)NENENENENENENE
Nikiforov et al. [60]5–18 yearsPost-radiation38 PTC33/38 (87%)NENENENENENENE
Sporadic17 PTC12/17 (71%)NENENENENENENE
Motomura et al. [61]9–14 yearsSporadic10 PTC3/10 (30%)NENENENENENENE
Suchy et al. [62]“children”Post-radiation34 PTCNENE0/34 (0%)NENENENENETP53
Pisarchik et al. [63] Pisarchik et al. [64]<29 yearsPost-radiation32 PTC combined10/32 (31%)NENENENENENENE
Fenton et al. [65]6–21 yearsPost-radiation1 PTCNENE0/1 (0%)NENENENENE
Sporadic30 PTCNENE2/30 (7%)NENENENENE
2 MTCNENE0/2 (0%)NENENENENE
4 FTCNENE0/4 (0%)NENENENENE
Thomas et al. [66]6–18 yearsPost-radiation67 PTC37/67 (55%)NENENENENENENE
7 FTC0/7 (0%)NENENENENENENE
Beimfohr et al. [67]0–19 years *Post-radiation81 PTCRET NegativeNENENENE6/81 (7%)NENE * at exposure
Fenton et al. [68]6–21 yearsPost-radiation1 PTC1/1 (100%)NENENENENENENE
Sporadic33 PTC15/33 (45%)NENENENENENENE
1 MTC0/1 (0%)NENENENENENENE
8 FTC0/8 (0%)NENENENENENENE
Santoro et al. [69]<15 yearsPost-radiation106 PTC36/106 (34%)NE0/23–31 (0%)NENENENENETSHR, TP53
Rabes et al. [70]≤14 yearsPost-radiation99 PTC50/99 (50%)NENENENE4/99 (4%)NENE
>14 years92 PTC44/92 (48%)NENENENE2/92 (2%)NENE
Pauws et al. [71]9–16 yearsSporadic8 PTCNENE0/8 (0%)NENENENENE
Elisei et al. [72]<18 yearsPost-radiation25 PTC19/25 (76%)NENENENENENENE
Sporadic25 PTC10/25 (40%)NENENENENENENE
Nikifurova et al. [73]6–31yearsPost-radiation55 PTC32/55 (58%)NENE2/55 (4%)NENENENE
9–77 yearsSporadic82 PTC16/82 (20%)NENE30/82 (37%)NENENENE
Kumagai et al. [74]≤15 yearsPost-radiation48 PTC5/15 (33%)NE0/14 (0%)0/15 (0%)NENENENE
>15 years12/33 (36%)NE2/32–33 (6%)8/33 (24%)NENENENE
<15 yearsSporadic29 PTCNENE0/16–22 (0%)0/30 (0%)NENENENE
1 FTC
1 PDTC1/1 (100%)
Lima et al. [75]13–30 yearsPost-radiation34 PTC14/34 (41%)NENE4/34 (12%)NENENENE
<18 yearsSporadic17 PTCNENENE1/17 (6%)NENENENE
Penko et al. [76]10–21 yearsPost-radiation1 PTC1/1 (100%)NE0/1 (0%)0/1 (0%)NENENENE
Sporadic/Unknown13 PTC6/11 (55%)NE0/10 (0%)0/13 (0%)NENENENE
1 MTC0/1 (0%)NE0/1 (0%)0/1 (0%)NENENENE
4 FTC0/3 (0%)NE0/2 (0%)0/4 (0%)NENENENE
Rosenbaum et al. [77]10–17 yearsSporadic20 PTC *NENENE4/20 (20%)NENENENE * Select histology
Espadinha et al. [78]5–21 yearsUnknown15 PTCNENENE1/15 (7%)NENENENE
Monaco et al. [54]≤21 yearsUnknownX PTC *3/XNE4/X2/XNENENE2/X * 66 FNA samples
X FTC *0/XNE0/X0/XNENENE0/X
Sassolas et al. [79]8–19 yearsPost radiation5 PTC8/28 (29%) *NE1/28 (4%) *2/28 (7%)NE0/28 (0%)NENE * One case w/ two alterations
Sporadic23 PTC
Ricarte-Filho et al. [80]<23 yearsPost-radiation26 PTC15/26 (58%)0/26 (0%)0/26 (0%)3/26 (12%)2/26 (8%)3/26 (12%)NE2/26 (8%)TSHR, PIK3CA, AKT1one TSHR, two TERT
Sporadic27 PTC7/27 (26%)0/27 (0%)2/27 (7%)7/27 (26%)0/27 (0%)2/27 (7%)NE0/27 (0%)
Leeman et al. [81] Leeman et al. [82]14–35 yearsPost-radiation62 PTC22/62 (35%) *NE6/62 (10%) *9/62 (15%) *NE9/62 (15%) *NE2/62 (3%) *RNA-seq (subset)Four tumors w/ two alterations
Henke et al. [83]<22 yearsSporadic27 PTCNENENE17/27 (63%)NENENENE
Givens et al. [84]0–18 yearsUnknown19 PTCNENENE7/19 (37%)NENENENE
Mitusake et al. [85]9–22 yearsPost-radiation *67 PTC **7/67 (10%)NE0/67 (0%)43/67 (64%)0/67 (0%)4/67 (6%)NENETERT* includes cases dx shortly after exposure ** four w/ possible APC-FAP
1 PDTC0/1 (0%)NE0/1 (0%)0/1 (0%)0/1 (0%)0/1 (0%)NENE
Ballester et al. [86]10–18 yearsPost-radiation2 PTC1/2 (50%)0/1 (0%)0/1 (0%)0/2 (%)NE0/1 (0%)NE0/1 (0%)Ion Torrent AmpliSeq Cancer Hotspot Panel v.2
Sporadic23 PTC5/23 (22%)0/10 (0%)0/10 (0%)10/23 (43%)NE0/8 (0%)NE0/8 (0%)
1 MTC0/1 (0%)1/1 (100%)0/1 (0%)0/1 (0%)NENENENE
1 FTC0/1 (0%)0/1 (0%)0/1 (0%)0/1 (0%)NENENENEOne CTNNB1
Picarsic et al. [87]<18 yearsSporadic17 PTC3/17 (18%)0/17 (0%)3/17 (18%)3/17 (18%)0/17 (0%) *4/17 (24%) *0/17 (0%) *1/17 (6%)Thyroseq2 (subset)* combined denominator
1 FTC0/1 (0%)0/1 (0%)0/1 (0%)0/1 (0%)NENENE0/1 (0%)
Prasad et al. [88]6–18 yearsSporadic27 PTC6/27 (22%)0/27 (0%)0/27 (0%)13/27 (48%)0/27 (0%)7/27 (26%)0/27 (0%)0/27 (0%)Thyroseq2
Nikita et al. [89]7–18 yearsPost-radiation2 PTC0/2 (0%)NE0/2 (0%)0/2 (0%)NENENE0/2 (0%)TERT
Sporadic32 PTC6/32 (19%)NE1/32 (3%)9/32 (28%)NENENE3/32 (9%)
5 FTC0/5 (0%)NE1/5 (20%)0/5 (0%)NENENE1/5 (20%)
Alzahrani et al. [90]9–18 yearsUnknown52 PTCNENENE12/52 (23%)NENENENETERTOne TERT
2 FTCNENENENENENENENE
1 PDTCNENENE0/1 (0%)NENENENE
Gertz et al. [91]8–18 yearsSporadic14 PTC2/14 (14%) *NE0/14 (0%)5/13 (38%) *NENENENE One tumor w/ both
Cordioli et al. [92]4–18 yearsSporadic30 PTCNENENENE3/30 (10%)NENENE
Onder et al. [45]≤18 yearsPost-radiation3 PTCNENENE15/50 (30%)NENENENETERT
Sporadic47 PTC
Cordioli et al. [93]≤18 yearsPost-radiation3 PTC13/35 (37%) *NE0/35 (0%)3/35 (9%)4/35 (11%) *3/35 (9%) *NENE * Three cases w/ two alterations
Sporadic32 PTC
Alzahrani et al. [94]≤18 yearsUnknown72 cPTC
7 FVPTC
NENE2/79 (3%)19/79 (24%)NENENENEPTEN, PIK3CA, TERTone PTEN, two PIK3CA, one TERT
Vanden Borre et al. [95]≤21 yearsUnknown14 PTC5/14 (36%)0/14 (0%)0/14 (0%)2/14 (14%)0/14 (0%)0/14 (0%)3/14 (21%)NE236 cancer genes and 14/19 fusion genes
2 MTC0/2 (0%)2/2 (100%)0/2 (0%)0/2 (0%)0/2 (0%)0/2 (0%)0/2 (0%)NE
Hardee et al. [96]≤21 yearsUnknown50 PTCNENENE24/50 (48%)NENENENE
Oishi et al. [97]≤20 yearsSporadic81 PTCNENENE44/81 (54%)NENENENECTNNB1, PIK3CA, TERT
Vuong et al. [98]<21 yearsSporadic41 FTCNENE5/41 (12%)NENENENE0/39 (0%)
Mostoufi-Moab et al. [99]≤18 yearsPost-radiation3 PTC0/3 (0%)NE1/3 (33%)0/3 (0%)NENENE0/3 (0%)
Sporadic59 PTC12/59 (20%)NE3/59 (5%)12/59 (20%)NENENE2/59 (3%)
6 FTC0/6 (0%)NE2/6 (33%)0/6 (0%)NENENE0/6 (0%)
Wasserman et al. [100]<18 yearsPost-radiation2 PTC0/2 (0%)NE0/2 (0%)0/2 (0%)NENENE0/2 (0%)DICER1, TERTThree pts w/ DICER1 mutations
Sporadic28 PTC7/28 (25%)NE0/28 (0%)5/28 (18%)NENENE0/28 (0%)
Huang et al. [101]<21 years 59 PTCNENENE16/30 (53%)NENENENE
Unknown5 FTC
1 MTC
Sisdelli et al. [102]≤18 yearsPost-radiation3 PTCNENENE0/3 (0%)1/3 (33%)NENENE
Sporadic77 PTCNENENE12/77 (16%)14/77 (18%)NENENE
Pekova et al. [103]6–20 yearsPost-radiation2 PTC0/2 (0%)NE2/83 (2%)15/83 (18%)NENENENEIDH1, CHEK2, PPMID, TERT, EIF1AX, EZH1One Cowden’s patient
Sporadic81 PTC18/81 (22%)
Bolded results were used in the calculation of overall percentage of sporadic PTC harboring these alterations; *, ** asterisks correspond to information regarding the study within that row. cPTC = classic PTC, dx= diagnosed, FA = follicular adenoma, FTC = follicular thyroid carcinoma, FVPTC = follicular variant of PTC, IHC = immunohistochemistry, NE = not evaluated, PDTC = poorly differentiated thyroid carcinoma, PTC = papillary thyroid carcinoma, MTC = medullary thyroid carcinoma, w/ = with, X = unknown.
Table 2. Therapies available by molecular alteration and/or histology.
Table 2. Therapies available by molecular alteration and/or histology.
HistologyMolecular AlterationTargeted Therapy
(Clinicaltrials.gov ID)
MTCPresumed RET mutationvandetanib
cabozantinib
Thyroid Cancer (MTC, PTC)RET mutations and fusionsBLU-667 (NCT03037385)
LOXO-292 (NCT03157128; NCT03899792)
DTCNon-specificvandetanib (NCT01876784)
cabozantinib (NCT03690388)
sorafenib
lenvatinib
PTC/ATCBRAFV600Evemurafenib
dabrafenib/trametinib
Solid tumors (including thyroid)NTRK-fusionlarotrectinib
PTCALK-fusioncrizotinib (NCT02034981)
ceritinib (NCT02289144)
alectinib (NCT03194893)

Share and Cite

MDPI and ACS Style

Paulson, V.A.; Rudzinski, E.R.; Hawkins, D.S. Thyroid Cancer in the Pediatric Population. Genes 2019, 10, 723. https://0-doi-org.brum.beds.ac.uk/10.3390/genes10090723

AMA Style

Paulson VA, Rudzinski ER, Hawkins DS. Thyroid Cancer in the Pediatric Population. Genes. 2019; 10(9):723. https://0-doi-org.brum.beds.ac.uk/10.3390/genes10090723

Chicago/Turabian Style

Paulson, Vera A., Erin R. Rudzinski, and Douglas S. Hawkins. 2019. "Thyroid Cancer in the Pediatric Population" Genes 10, no. 9: 723. https://0-doi-org.brum.beds.ac.uk/10.3390/genes10090723

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop