Next Article in Journal
Genome-Wide Association Study of Phenolic Content and Antioxidant Properties in Eggplant Germplasm
Next Article in Special Issue
Soft Tissue and Bone Tumor Diagnostics: Harnessing the Power of Molecular Techniques
Previous Article in Journal
Ancient Mitochondrial Genomes Provide New Clues to the Origin of Domestic Cattle in China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Update on Molecular Diagnostics in Thyroid Pathology: A Review

Departments of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
*
Author to whom correspondence should be addressed.
Genes 2023, 14(7), 1314; https://doi.org/10.3390/genes14071314
Submission received: 31 May 2023 / Revised: 13 June 2023 / Accepted: 19 June 2023 / Published: 22 June 2023
(This article belongs to the Special Issue Molecular Pathology, Genetics and Genomics in Surgical Pathology)

Abstract

:
Thyroid nodules are quite common, and the determination of a nodule of concern is complex, involving serum testing, radiology and, in some cases, pathological evaluation. For those nodules that raise clinical concern of neoplasia, fine needle aspiration biopsy is the gold standard for evaluation; however, in up to 30% of cases, results are indeterminate for malignancy, and further testing is needed. Advances in molecular testing have shown it to be of benefit for both diagnostic and prognostic purposes, and its use has become an integral part of thyroid cancer management in the United States and in several global nations. After The Cancer Genome Atlas (TCGA) consortium published its molecular landscape of papillary thyroid carcinoma (PTC) and reduced the “black matter” in PTC from 25% to 3.5%, further work ensued to clarify the remaining fraction not neatly attributed to the BRAFV600E-like or RAS-like phenotypes of the TCGA. Over the past decade, commercial molecular platforms have been refined as data accrues, and they increasingly cover most genetic variants of thyroid carcinomas. Molecular reporting focuses on the nodule tested, including related clinical information for that nodule (size of nodule, Bethesda category, etc.). This results in a comprehensive report to physicians that may also include patient-directed, clear language that facilitates conversations about nodule management. In cases of advanced or recurrent disease, molecular testing may become essential for devising an individual therapeutic plan. In this review, we focus on the evolution of integrated molecular testing in thyroid nodules, and how our understanding of tumor genetics, combined with histopathology, is driving the next generation of rational patient management, particularly in the context of emerging small, targetable therapeutics.

1. Introduction

Increased knowledge about the molecular underpinnings of thyroid neoplasia has transformed the approach to diagnosis, prognosis, and therapeutics. Molecular profiles have become an integral part of thyroid cancer management. These sensitive and specific ancillary tools have been useful for subclassifying indeterminate (for malignancy) thyroid nodules by cytology (Bethesda III or IV) or ambiguous nodules by light microscopy (tumors of uncertain malignant potential), resulting in more optimal patient care [1]. The current (fifth edition) World Health Organization criteria categorizes thyroid tumors into follicular-derived neoplasms (benign, low-risk and malignant), C cell-derived carcinomas (medullary thyroid carcinoma; MTC), mixed medullary and follicular cell-derived carcinomas, and other rare non-follicular-, non-C cell-derived tumors (mucoepidermoid carcinoma (MEC), secretory carcinoma (SC), sclerosing mucoepidermoid carcinoma with eosinophilia (SMECE), cribriform morular thyroid carcinoma (CMTC), thyroblastoma (TB), etc.) [2]. Specific genetic variants have been described in some of these rare, primary tumors of the thyroid (CRTC1::MAML2 in MEC, ETV6::NTRK3 in SC, APC in CMTC and DICER1 in TB) [3,4,5,6], but description of these rare tumors is beyond the scope of this article. It is noted that some of these genes are aberrant in more common thyroid tumors and will be discussed further.
As the most frequent thyroid malignancy, papillary thyroid carcinoma (PTC) is most often classic PTC and is predominantly associated with BRAF p.V600E [7,8], although most prevalence studies were performed prior to a subset of PTC being relegated to other categories (non-invasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP), CMTC, well-differentiated tumor of uncertain malignant potential (WDT-UMP)) [8]. Following the publication of The Cancer Genome Atlas characterization of PTC in 2014 [9], which included a prominent subset of formerly classified follicular variant PTC, tumors were largely divided into those that were BRAFV600E-like and RAS-like [10]. A later study by Yoo et al. described the mutational landscape in follicular thyroid adenomas (FTA) and follicular carcinoma (FTC), with reference to PTC, including the previously described BRAF-like and RAS-like lesions along with a Non-Braf/Non-Ras (NBNR) group of follicular-derived tumors that behaved in an intermediate fashion. This NBNR group included neoplasias encompassing genetic fusion events involving tyrosine kinases (RET, NTRK, ALK, ROS, etc.) [11].
In the earlier part of the century, genetic testing was just emerging as a technology to complement standard histology and immunohistochemistry. For thyroid, the concept of routine preoperative molecular testing of thyroid nodules indeterminate for malignancy, following fine needle aspiration biopsy (FNAB), launched in 2010 [12]. Most classic PTC cases (BRAF-like) were diagnostic from routine FNAB, yet profiles picked up via indeterminate biopsies, including occasional differentiated thyroid carcinomas (DTC), which include PTC, FTC, and oncocytic thyroid carcinoma (OTC), largely included genetic RAS-like variants and those that were NBNR. Further, malignancies, such as MTC, which may be difficult to diagnose via FNAB unless suspected or accompanied by preoperative serum assays, along with rare thyroid malignancies of non-standard cytology, such as cribriform morular thyroid carcinoma, could be assessed [13]. Over the last decade, the preoperative molecular testing of FNAB material has played a significant role in optimizing patient management, triaging for minimal surgery and active surveillance as warranted [1]. However, for advanced thyroid cancers, including anaplastic thyroid carcinoma (ATC), some poorly differentiated thyroid carcinomas (PDTC), MTC and clinically aggressive DTC, naïve/untreated genetic analysis may be essential for determining the best treatment plan, including a subset of patients who may benefit from neoadjuvant therapy [14]. In this review, we focus on the molecular alterations and tests offered in the follicular-derived and C cell-derived thyroid tumors, and we will discuss the potential targeted therapies.

2. History of Molecular Tests in Thyroid Diagnostics

Lindsay et al. described PTC nuclear features in thyroid carcinomas in the 1960s, and the subsequent description of the follicular variant of PTC (FVPTC) in the late 1970s by Chen and Rosai were agnostic to molecular alterations. In the following several decades, there was a dramatic shift in the diagnosis of PTC based not only on the presence of architectural papillae in the presence of nuclear features, but also based on the nuclear features alone, even in the absence of architectural features [15]. The histories of molecular alterations and genetic testing in thyroid carcinomas are described in Table 1. In 1987, Fusco et al. first noted TRK and RET rearrangements in five PTC and their respective metastatic lymph nodes [16]. Inspired by their work, Santoro et al. conducted a study on 286 thyroid tumors (177 PTC, 37 FTC, 15 ATC, 18 MTC, 34 benign thyroid nodules and 5 cases of squamous cell carcinoma and sarcomatoid carcinoma of the thyroid), and found RET aberrations exclusively in 19% of PTC (33/177) [17]. In 1989, Lemoine et al. identified NRAS p.Q61R and HRAS p.Q61R in all stages of the thyroid neoplastic process (FTA, FTC and ATC) [18]. Namba et al. found further genetic variants within RAS subtypes (NRAS, HRAS and KRAS) in codons 12 and 13 in benign and neoplastic thyroid nodules [19]. In 2000, Kroll et al. reported PAX8::PPARG fusion in five of eight FTC but not in 20 FTA, 10 PTC or 10 multinodular hyperplasias [20]. Nikiforova et al. found the same fusion event in 8 of 15 (53%) FTC and 2 of 25 (8%) FTA, but not in 35 PTC (including 12 follicular variants), 12 OTC, 12 oncocytic thyroid adenoma (OTA), 2 ATC, 1 PDTC or 16 hyperplastic nodules [21].
At the turn of the twenty-first century, two distinct pathways were emerging in thyroid tumorigenesis, one being RAS-associated or PAX8::PPARG-related with a predominant follicular growth patten, and another not fully explained by RET or TRK aberrations. The inverse association between BRAF and RAS in alternative activation of MEK/MAPK signaling in many tumors [35], including colon [36] and ovarian tumors [37], inspired Cohen et al. to explore the role of BRAF in the predominantly RAS-negative PTC. Their work identified BRAF p.T1796A in 69% of PTC (24/35) [22]. Of brief note is that this original nomenclature referred to the nucleotide versus the codon, and the erroneous BRAF T1796A (then, BRAF p.V599E) eventually came to be recognized as BRAF T1799A (now, BRAF p.V600E) after the error was realized [38]. Since then, molecular studies have progressed to become a standard and integral part of our understanding of thyroid tumorigenesis and biological potential.

3. Fine Needle Aspiration and the Emergence of Molecular Testing Platforms

FNAB with cytological evaluation is the most reliable and commonly used tool for cancer diagnosis in thyroid nodules. The arrival of preoperative ancillary molecular diagnostics for nodules of indeterminate cytology was field-changing for just over the past decade [12]. Prior to molecular platforms for FNAB material, individual gene testing was more common in surgical resections, and testing was more expensive, assayed far fewer genes and worked with less reliable, longer genetic primers [39]. In 2009, Nikiforov et al. conducted a study on FNAB samples from thyroid nodules with Bethesda categories of benign, indeterminate and malignant. Their group analyzed BRAF variants (p.V600E, p.K601E), NRAS, KRAS and HRAS along with genetic fusion products RET::PTC1 (CCDC6::RET), RET::PTC3 (NCOA4::RET) and PAX8::PPARG—genetic changes commonly associated with thyroid neoplasia. Biopsies positive for genetic aberrations were nearly uniformly found to be malignant following excision [23], the results of which have been clarified over the years with broader panels, larger cohorts and refined diagnostic criteria for malignancy [28,31]. The evolution of the most common molecular tests in preoperative FNAB of thyroid nodules is illustrated in Figure 1. This discovery has greatly improved preoperative diagnosis for indeterminate thyroid nodules (ITN), confirming neoplasia and with increasing granularity around risk of malignancy.
In 2012, Alexander et al. conducted a prospective, multicenter validation study of 577 ITN, 413 of which had corresponding histopathological specimens. Using machine learning to interpret mRNA expression of 167 genes through microarray platforms, the Afirma Gene Expression Classifier (GEC) studied ITN and correctly identified 78 of the 85 excised malignant nodules as suspicious with a sensitivity of 92% and a specificity of 52%. The study aimed to eliminate unnecessary thyroid surgeries by ruling out thyroid neoplasms and recommended a conservative approach for most patients with ITN and benign nodules according to GEC results. Afirma GEC was the first rule-out test for thyroid neoplasms [12]. In 2018, the Afirma Genomic Sequencing Classifier (GSC) was developed to better describe the RNA transcriptome with additional sequencing of nuclear and mitochondrial genes and genomic copy number changes, including loss of heterozygosity. This resulted in a more robust genomic test. For ITN with non-oncocytic histology, the sensitivity was 92% and the specificity was 70% [29]. Although Afirma GEC was highly sensitive, certain thyroid tumors, like oncocytic neoplasms, were intentionally called suspicious, resulting in lower specificity for such neoplasms. With the improved GSC, a more comprehensive and updated GEC, Hao et al. used NGS of whole transcriptome RNA sequencing in FNA nodules with oncocytic features to identify mitochondrial and nuclear expression of genes related to oncocytic (formerly Hürthle cell) neoplasms. The specificity of this algorithmic classification increased from 12% to 59% [30].
In 2013, the first version of targeted NGS panel ThyroSeq was published [24]. ThyroSeq used NGS platforms for 12 genes to include 284 hot spot mutations to cover more than 95% of the reported variants in genes associated with thyroid neoplasms. In addition to some mutations detected by GEC (BRAF, RET, NRAS, KRAS and HRAS), ThyroSeq studied PIK3CA, TP53, TSHR, PTEN, GNAS, CTNNB1 and AKT1. PAX8::PPARG fusion and RET- and TRK-rearranged genes were not included in ThyroSeq v1 [24]. This panel was expanded as ThyroSeq v2.1 and was published in 2015 to include additional point mutations in EIF1AX and BRAFV601K and detection of over 40 other gene fusions including THADA, ALK, PAX8::PPARG, TRK1 and TRK3 genes. Finding BRAF mutations or PPARG, NTRK1, NTRK3 and ALK fusions were strong predictors of a higher risk of cancer (approaching 100%) while other mutations like RAS, PTEN and EIF1AX or THADA fusions were associated with a significant but lower risk of cancer [25]. The latest platform of ThyroSeq v3 is the current commercially available test [28].
Other commercially available molecular platforms for preoperative diagnosis of ITN include either a microRNA (miRNA) classifier such as RosettaGX Reveal and mirTHYtype or miRNA and somatic gene mutational platform such as Asuragen [26,27,41].

4. Molecular Tests and Clinical Practice

The workflow of thyroid nodules preoperatively is demonstrated in Figure 2. Thyroid FNAB predominantly occur in outpatient clinics and are performed by a range of physicians (endocrinologists, radiologists, surgeons and pathologists). When rapid on-site evaluation (ROSE) is available, adequacy and diagnosis can be made within a few minutes [42], and additional needle pass(es) for ITN can be obtained and sent out for molecular testing to Afirma, ThyroSeq or another desired platform. However, in settings where ROSE is unavailable, specimens are collected for subsequent cytologic and molecular evaluation. In the latter scenario, practices vary among institutions, but a “collect on all” protocol for possible molecular testing seems to be the most efficient for both patients and the lab [43] rather than a return visit and re-biopsy for an indeterminate biopsy. Both Afirma and ThyroSeq provide a send out service as a reflex to ITN FNA. Fresh FNA samples are preferred for optimal and successful testing, but ThyroSeq also offers molecular testing for tissue scrapings from FNA slides and from paraffin-embedded tissue [44].

5. Molecular Tests in ITN

Molecular tests were initially performed on ITN as a rule-out malignancy test in order to risk stratify for surgery (negative predictive value) [10]. The indications for these tests have evolved over the past decade. Three highly utilized, commercially available/validated platforms for thyroid nodules in the United States are Afirma Genomic Sequencing Classifier (Afirma GSC)/Afirma Xpression Atlas (XA), ThyGeNEXT/ThyraMIR (MPTX) and ThyroSeq v3.
Afirma GSC uses whole transcriptome RNA analysis using NGS combined with machine learning. Nodules evaluated by Afirma result in a benign or suspicious classification. Initially, samples sent to Afirma are tested for sufficient RNA. Samples are then tested against classifiers to detect parathyroid tissue, MTC, BRAF p.V600E, CCDC6::RET or NCOA4::RET. When all classifiers are negative, GSC analyzes > 10,000 genes and classifies samples as GSC-B (benign) or GSC-S (suspicious). Compared with Afirma GEC, Afirma GSC has enhanced diagnostic accuracy for oncocytic thyroid neoplasms [30]. Introduced in 2019 and updated in 2020, Afirma Xpression Atlas (XA) enumerates mutations in 593 genes, informing 905 variants and 235 fusions in suspicious Afirma GSC, and is extended to include Bethesda V and VI nodules. Comparing Afirma XA testing with whole-transcriptome RNA-, targeted RNA- and targeted DNA-sequencing, Angel et al. reported mutations and gene fusions associated with thyroid nodules with high reproducibility and accuracy. BRAF p.V600E was predominantly present in Bethesda V/VI FNA, and NRAS and HRAS variants were primarily seen in Bethesda III/IV FNA. In addition to point mutations in BRAF and RAS genes, Afirma XA reported mutations in TSHR p.M453T and SPOP p.P94R. The two mutations were mostly associated with benign nodules [45,46]. Afirma XA detected many fusions seen in thyroid neoplasms like PAX8::PPARG, ETV6::NTRK3, CCDC6::RET, NCOA4::RET, STRN::ALK, AGK::BRAF, SND1::BRAF and RBPMS::NTRK3. Fusion of PAX8::GLIS3 was also detected with Afirma XA, which is associated with hyalinizing trabecular tumor [47,48].
Afirma XA provides information about inherited syndromes with increased likelihood of thyroid malignancy, such as RET (multiple endocrine neoplasia 2; MEN2), PTEN (PTEN hamartoma tumor syndrome; Cowden syndrome), APC (APC-associated polyposis; familial adenomatous polyposis) and DICER1 (DICER1 syndrome). Other inherited mutations in oncogenes or tumor suppressor genes like BRCA1, BRCA2, RB1, WT1, NF2, COL3A1, TGFBR2, TP53 and MSH2 are also reported with Afirma XA. Since Afirma is not intended to test for germline alterations and mutations may be somatic, patients with clinical suspicion for inherited syndromes should be referred for genetic counseling [40].
MPTX includes the combination of a mutation panel (ThyGeNEXT) and an miRNA risk classifier (ThyraMIR). Results of MPTX are categorized as positive, moderate risk or negative for malignancy. The ThyGeNEXT portion uses targeted NGS for selected DNA gene mutations (ALK, BRAF, GNAS, HRAS, KRAS, NRAS, PIK3CA, PTEN, RET and TERT promoter genes) as well as mRNA fusion genes (ALK, BRAF, NTRK, PPARG, RET, PAX8, TBP, USP33 and THADA). When there is a strong driver mutation, the results of the test are flagged as positive. Further analysis with the miRNA classifier is carried out when a weak or absent driver mutation is detected. Several growth-suppressing miRNA (miR-29, -138, -139, -155, -204) or growth promoting miRNA (miR-31, -146, -222, -375, -551) are tested in the ThyraMIR portion of MPTX [32]. ThyGeNEXT has been updated to include coexisting driver mutations and increases detection of strong driver mutations (TERT promoters or BRAF p.V600E) or weak driver mutations (RAS), which are both correlated with positive miRNA results [33]. More recently, miR-21 has been added to the MPTX panel (MPTXv2) and shows a decrease in the moderate-risk cohort from 28% to 13% (p < 0.001) and an improved diagnostic accuracy of ITN risk stratification [34]. MTC diagnosis showed 100% accuracy with miRNA in the MPTX platform (4 MTC and 26 non-MTC samples) [49].
Thyroseqv3 is a DNA- and RNA-based targeted NGS platform that detects point mutations, gene fusions, copy number alterations and abnormal gene expression in 112 thyroid cancer-related genes. ThyroSeq v3 has a genomic classifier score that is the sum of all genetic alterations detected in the sample, to distinguish benign from malignant nodules and correctly classify most papillary, follicular and oncocytic thyroid neoplasias, MTC and parathyroid lesions with 94% sensitivity, 89% specificity and 92% accuracy [28]. Copy number alterations (CNA) have been reported in approximately 7% of PTC lacking other driver mutations and in other tumors such as oncocytic thyroid neoplasms, which show a nearly homozygous genome [50,51,52]. ThyroSeq v3 has a sensitivity of 93% and a specificity of 69% for oncocytic thyroid nodules. ThyroSeq v3 classifies nodules based on the likelihood of malignancy into six scores with a proposed cutoff of 1.5 for malignant lesions [28]. However, the ultimate arbiter is surgical pathology diagnosis and patient outcomes, in the context of molecular changes, combined with any incremental pathophysiologic data that may enhance this new baseline.
MPTX, Afirma GSC and ThyroSeq v3 were primarily designed as negative predictive value tests, although the use of molecular analysis in thyroid nodules has evolved. In one meta-analysis, ThyroSeq v3 and Afirma GSC showed optimal results to exclude malignancy. Both ThyroSeq v3 and Afirma GSC were not superior to ThyroSeq v2 in ruling out malignancy. MPTX reported similar results [53]. In two other meta-analyses by Lee et al. and Livhits et al., diagnostic performances of Afirma GSC and ThyroSeq v3 were not statistically different, and both showed similar specificity and reduced unnecessary diagnostic surgeries in 49% of ITN [54,55].
Regardless of the molecular platform used, the goal is to provide sufficient utility for optimal patient management. Lacking concomitant clinical concerns, a benign molecular finding in an ITN may be clinically observed. Although advances have been made in the molecular testing for oncocytic thyroid nodules and RAS-like genetic variant tumors (FTA, NIFTP or FTC), histology is still the gold standard in examining the entire periphery of the nodule to assess for capsular or angioinvasion, and such nodules are preferably treated with surgery, typically hemithyroidectomy/lobectomy barring concomitant clinical circumstances. For patients with higher-risk genetic or clinical profiles (radiologically high-risk +/− lateral neck disease), more comprehensive upfront surgery may be considered. Molecular testing is widespread in the United States; however, in most of the world, cytologic diagnosis alone, if available, is the primary determinant of risk of malignancy in thyroid nodules. There have been cost-effectiveness studies conducted that show great value in the cost of molecular testing versus unnecessary thyroid surgery for molecularly determined benign nodules [56]. In particular, clinical management of thyroid nodules in under-resourced populations is not well-studied internationally, with limited data for underserved areas of the United States [57]. As an alternative, immunocytochemistry on fine needle aspiration preparations or immunohistochemistry on core biopsies can provide some limited insight at a lower cost in some specific situations [58]. However, without molecular testing, the decision for surgery, including extent of surgery, is confined to the clinical scenario with possible input from FNAB interpretation, and this may result in a greater incidence of total thyroidectomy for a single ITN in up to 90% of cases depending upon geography [59].

6. Cancer Prognosis in the Era of Molecular Testing

Molecular alterations in thyroid neoplasms play an essential role in classification, prognosis and response to radioactive iodine (RAI). In 2014, The Cancer Genome Atlas (TCGA) consortium extensively investigated the molecular landscape of its selected PTC cohort and the distribution of data largely fell into a binary set of molecular alterations favoring a BRAFV600E-like or RAS-like phenotype. Although this cohort reduced the “black matter” in PTC from 25% to 3.5%, fusion genes like RET, NTRK1/3 and ALK showed neutral (non-BRAF-/non-RAS-like; NBNR) to weak BRAF-like signaling, thus questioning the correlation between such mutations and BRAF genes [9]. Focusing on FTA and minimally invasive FTC, Yoo et al. classified the molecular subtypes into three groups, BRAF-like, RAS-like and NBNR-mutated tumors. The BRAF-like mutated tumors were associated with a higher stage at presentation, larger tumors and lateral neck metastasis, compared with the RAS-like and NBNR groups. NBNR included tumors with DICER1, EIF11AX, PTEN and IDH1 mutations and PAX8::PPARG fusion [11].
In thyroid nodules preoperatively classified as malignant (Bethesda VI), BRAF p.V600E mutation was identified in 65–75% of tumors, followed by TERT promoter mutations that were seen in 11% [60]. The presence of both BRAF p.V600E and TERT promoter was significantly correlated with patient age, extrathyroidal extension, lymph node metastasis, distant metastasis, disease stage, tumor recurrence and mortality [61]. The presence of both mutations in PTC robustly predicts loss of radioiodine avidity in these tumors [62].
In one study, suspicious nodules (Bethesda V) were analyzed using ThyroSeq v3, and were categorized into low-, intermediate- and high-risk. Low-risk tumors were mostly associated with RAS-like mutations and focal chromosomal-type CNA, and intermediate risk comprised the BRAF-like mutations and genome haploidization-type CNA. The presence of TP53 and TERT promoter mutations was considered a high-risk mutation. High-risk mutations were associated with ATC, PDTC and tall cell PTC. Follow-up data on the high-risk cases showed more frequent recurrences than low-risk cases with limited risk of recurrence, affirming the predictive value of molecular profiling [63]. When PDTC was studied using ThyroSeq v3, high-, intermediate- and low-risk tumors were 60%, 23% and 17%, respectively, and high-risk tumors were associated with distant metastasis and worse overall survival, compared with low-risk, where no patient died of disease [64]. Genetic studies in higher grade tumors may be helpful in finding molecular targets as neoadjuvant therapy.

7. Molecular Identification of Targetable Alterations in Thyroid Cancer

The most common medications used for targetable mutations in thyroid carcinoma are summarized in Table 2. In the setting of advanced thyroid carcinomas, molecular testing is essential for identifying potential therapeutic options, including those tumors that were previously tested but have acquired a behavioral change. High-grade thyroid carcinomas, including PDTC, MTC and ATC, often present with advanced disease; however, 15% of DTC may also be locally aggressive, necessitating additional treatment, including RAI. In a study of metastatic non-anaplastic thyroid carcinomas (PTC, FTC, PDTC), Durante et al. found that only 30% of patients showed RAI uptake with remission, 39% demonstrated RAI uptake with no remission and in the remaining 31%, no RAI was detected. RAI treatment plays a crucial role in the treatment of advanced thyroid carcinomas. Overall survival rate at 10 years post-RAI was 92% in patients who achieved a negative study compared with 19% in patients who did not [65]. RAI-resistant patients can be treated with cytotoxic chemotherapy as a monotherapy or a combination of more than one agent; however, the response is partial, with troublesome side effects with a relatively narrow therapeutic index [66]. In this scenario, with the most advanced tumors resistant to traditional treatments like RAI or presenting with distant metastasis, expansion of treatment options to include therapies targeting specific genetic variants is promising.
Most thyroid neoplasia is related to activation of the mitogen-activated protein kinase (MAPK) and the PI3K pathways. When no targetable mutation is identified (NTRK, ALK, RET or BRAF) or the tumor is not sequenced, the antiangiogenic multi-targeted kinase inhibitors (aaMKIs) Sorafenib, Lenvatinib and Cabozantinib may be used. AaMKIs have been approved by the US Food and Drug Administration (FDA) for the treatment of aggressive RAI-resistant PTC or FTC. Both Sorafenib and Lenvatinib can be used as first-line treatment in RAI-resistant thyroid carcinomas. Lenvatinib is superior to Sorafenib in achieving longer progression-free median survival. Previously approved by the FDA for the treatment of MTC, Cabozantinib has been authorized as a second-line treatment when tumors fail to respond to Lenvatinib or Sorafenib, and it showed improved progression-free survival. Although aaMKIs slow the progression of disease, their toxicity profiles limit their clinical use due to nonspecific targeting [68,69,70,71,81].
A mutation-specific inhibitor should be considered when a specific driver mutation is identified (RET, NTRK, ALK or BRAF). Selpercatinib and Pralsetinib inhibit RET in MTC, but they can also block the RET fusion protein-mediated signaling found in PTC [80,82]. Although therapeutic resistance has been reported [84,85], RET inhibitors are better tolerated and more specific than aaMKIs.
In TRK fusion-positive thyroid cancers, Larotrectinib demonstrates rapid and durable disease control and a favorable safety profile in patients with advanced disease [83]. ROS1 inhibitors such as Entrectinib have also been used in one patient with ROS1 mutation thyroid carcinomas with liver metastasis and showed efficacy. Although this mutation is rare and no randomized clinical trials have been conducted on thyroid due to the rarity of this fusion, Entrectinib might be used in advanced cases with intractable disease [76].
Earlier reports of Dabrafenib and Vemurafenib show partial response to treatment in BRAF p.V600E-mutated PTC [72,73]. More recently, enhanced response to RAI and increased iodine uptake has been achieved with pretreatment of RAI-resistant PTC with Dabrafenib and Vemurafenib. This approach may represent a new potential treatment option for patients with RAI-resistant tumors [74,75]. Although ALK fusion represents <1% of thyroid carcinoma, it is more frequently seen in PDTC. There is no current FDA-approved treatment for ALK kinase fusion-related thyroid carcinomas; however, ALK inhibitors may be used for clinical trials or off-label treatment in advanced cases [67,86].
Although most thyroid carcinomas are microsatellite-stable and show low tumor mutational burden (TMB), a subset of follicular carcinomas (~2.5%) may show microsatellite instability or high TMB [87]. Programmed death (PD-1) inhibitors (including Pembrolizumab) can be also used, based on the agnostic approval for solid tumors [79].
ATC is a rare but lethal subtype of thyroid carcinoma with a historical 3–4 month median survival rate [88,89], a survival rate significantly improved with targeted therapeutics, now greater than one year [90]. In 58% of ATC, there is a history or a concurrent DTC, and in the remaining 42%, ATC is considered de novo. ATC tend to accumulate multiple mutations in genes readily present in DTC like BRAF or RAS, or kinase fusion-related thyroid carcinomas (NTRK, RET or ALK), in addition to higher risk genes like TERT promoter, TP53, PIK3CA and PTEN [91,92]. The treatment of ATC has been advanced in the past decade to include the FDA-approved usage of BRAF/MEK inhibitors (Dabrafenib/Trametinib) in BRAF p.V600E-mutated ATC [77,78]. The first report on vemurafenib to treat BRAF p.V600E-associated ATC was in 2013 [93]. The detection of BRAF p.V600E is crucial and time-sensitive in determining the course of treatment in ATC. Old and traditional tests like NGS (2–3 weeks) may delay treatment and rapid tests are needed to assess BRAFV p.V600E mutation and start treatment with BRAF/MEK inhibitors. BRAFV p.600E hot spot mutation can be detected either by mutation-specific immunohistochemistry on paraffin-embedded blocks of needle core biopsies or a cell block of FNA [94]. BRAF IHC is quite sensitive but may be accompanied by background staining. Results of BRAF IHC turn around is fast; it is <24 h for in-house testing [94]. BRAF p.V600E DNA can be detected using rapid PCR assay of an extract of paraffin blocks (2–3 days) or from the peripheral blood NGS (cell free DNA) [95,96]. Pyrosequencing is also an effective method for detecting the BRAF p.V600E mutation in FNA samples [97], and innovative methods for detection to obtain the BRAF p.V600E in less than 1 day have been developed [98].
Regardless of the method used to evaluate for BRAF p.V600E mutation, BRAF-directed therapy should be started as soon as possible to induce rapid and substantial disease regression. Advanced tumor stage ATC, which has been considered inoperable, can now profit from BRAF inhibitors alone or BRAF/MEK inhibitors. Surgery is a novel option in such patients with significant tumor reduction up to 100% locoregional control, 83% and 32% survival rate at one and two years, respectively [77,78,99]. Despite the actionable targeted therapies for BRAF p.V600E, MEK, NTRK, ALK, RET and ROS1, acquired resistance may develop following an initial response and biopsy of resistant lesions, and molecular testing may be appropriate in assisting with new therapeutic strategies [100].
MTC is a non-follicular origin thyroid tumor that may occur sporadically or be inherited, as part of the Multiple Endocrine Neoplasia 2 (MEN 2) syndrome and Familial Medullary Thyroid Carcinoma (FMTC) syndrome. RET mutations have been described in both germline and sporadic cases, while RAS mutations have been found only in sporadic cases [101]. At presentation, 58% of MTC presented with lymph node metastasis and 6% with distant metastasis [102]. Inherited MTC are associated with an autosomal dominant mutation in the RET protooncogene (40% of MTC). All patients diagnosed with MTC are candidates for genetic counselling with family testing for potential inherited RET mutations. In 47% of sporadic patients, RET mutations are sporadic, and no further treatment except follow-up is needed. RAS accounts for 26% of sporadic MTC (HRAS, KRAS and, less frequently, NRAS). In 19% of sporadic tumors, neither RET nor RAS mutations were identified [103]. One study showed that somatic RET-mutated MTC (exon 15 and 16) correlated with multiple lymph node metastasis, multiple tumors and presented with a higher clinical stage and increased metastatic rate compared with non-exon 15/16 and non-RET mutated MTC [104]. Cabozantinib and Vandetanib are RET-targeted therapies that have been approved by the FDA for progressive locally advanced and/or metastatic RET-mutated MTC and show slow progression of disease. Both medications are associated with significant toxicity due to a narrow therapeutic window and non-selective kinase inhibition [105,106]. Although non-MTC RET-mutated tumors have been associated with resistance to Vandetanib and Cabozantinib, RET-mutated MTC at codon V804 (gatekeeper mutation) showed indolent disease without classic MEN 2A features [107]. With more selective RET inhibitors like Selpercatinib, a considerable response was recorded in patients initially treated with Vandetanib and/or Cabozantinib, and it was recently approved by the FDA for treating advanced RET-mutated MTC. The most recent selective RET inhibitor approved by FDA is Pralsetinib. Advanced or metastatic MTC patients treated with Pralsetinib show improved response rates (65%), with RET-mutated tumors, including MKI-resistant and codon V804 mutations. Fewer side effects are recorded and mostly are reversable, which makes this medication a potential therapeutic for inoperable or advanced MTC [82].
In summary, molecular testing has become a standard tool in thyroid nodule management. With its reduced costs, there are increasing applications for its use, particularly in the space of emerging small, targetable therapeutics for advanced thyroid cancers, particularly those with variants in BRAF, RET or kinase fusions. Molecular testing frequently falls short, as does surgical pathology, in discerning follicular-patterned thyroid adenomas from carcinomas, and this remains an opportunity for refined risk stratification and therapeutic planning following molecular testing in these lesions.

Author Contributions

Conceptualization, B.A. and P.M.S.; methodology, B.A. and P.M.S.; data curation, B.A.; writing—original draft prepara-tion, B.A. and P.M.S.; writing—review and editing, B.A. and P.M.S.; visualization, B.A.; supervision, P.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

Sadow received salary support from the National Cancer Institute of the National Institutes of Health (5P01CA240239-04).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors have disclosed that they have no significant relationship with, or financial interest in, any commercial companies pertaining to this article.

References

  1. 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]
  2. Baloch, Z.W.; Asa, S.L.; Barletta, J.A.; Ghossein, R.A.; Juhlin, C.C.; Jung, C.K.; LiVolsi, V.A.; Papotti, M.G.; Sobrinho-Simoes, M.; Tallini, G.; et al. Overview of the 2022 WHO Classification of Thyroid Neoplasms. Endocr. Pathol. 2022, 33, 27–63. [Google Scholar] [CrossRef] [PubMed]
  3. Tirado, Y.; Williams, M.D.; Hanna, E.Y.; Kaye, F.J.; Batsakis, J.G.; El-Naggar, A.K. CRTC1/MAML2 fusion transcript in high grade mucoepidermoid carcinomas of salivary and thyroid glands and Warthin’s tumors: Implications for histogenesis and biologic behavior. Genes Chromosomes Cancer 2007, 46, 708–715. [Google Scholar] [CrossRef] [PubMed]
  4. Lam, A.K.; Fridman, M. Characteristics of cribriform morular variant of papillary thyroid carcinoma in post-Chernobyl affected region. Hum. Pathol. 2018, 74, 170–177. [Google Scholar] [CrossRef] [Green Version]
  5. Agaimy, A.; Witkowski, L.; Stoehr, R.; Cuenca, J.C.C.; Gonzalez-Muller, C.A.; Brutting, A.; Bahrle, M.; Mantsopoulos, K.; Amin, R.M.S.; Hartmann, A.; et al. Malignant teratoid tumor of the thyroid gland: An aggressive primitive multiphenotypic malignancy showing organotypical elements and frequent DICER1 alterations-is the term “thyroblastoma” more appropriate? Virchows Arch. 2020, 477, 787–798. [Google Scholar] [CrossRef]
  6. Saliba, M.; Mohanty, A.S.; Ho, A.L.; Drilon, A.; Dogan, S. Secretory Carcinoma of the Thyroid in a 49-Year-Old Man Treated with Larotrectinib: Protracted Clinical Course of Disease Despite the High-Grade Histologic Features. Head Neck Pathol. 2022, 16, 612–620. [Google Scholar] [CrossRef]
  7. Ding, Z.; Tao, X.; Deng, X.; Guo, B.; Kang, J.; Wu, B.; Yang, Z.; Chen, C.; Liu, P.; Zhang, Y.; et al. Genetic analysis and clinicopathologic features of locally advanced papillary thyroid cancers: A prospective observational study. J. Cancer Res. Clin. Oncol. 2023. [Google Scholar] [CrossRef]
  8. Elisei, R.; Grande, E.; Kreissl, M.C.; Leboulleux, S.; Puri, T.; Fasnacht, N.; Capdevila, J. Current perspectives on the management of patients with advanced RET-driven thyroid cancer in Europe. Front. Oncol. 2023, 13, 1141314. [Google Scholar] [CrossRef]
  9. The Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef] [Green Version]
  10. Asa, S.L.; Giordano, T.J.; LiVolsi, V.A. Implications of the TCGA genomic characterization of papillary thyroid carcinoma for thyroid pathology: Does follicular variant papillary thyroid carcinoma exist? Thyroid 2015, 25, 1–2. [Google Scholar] [CrossRef]
  11. Yoo, S.K.; Lee, S.; Kim, S.J.; Jee, H.G.; Kim, B.A.; Cho, H.; Song, Y.S.; Cho, S.W.; Won, J.K.; Shin, J.Y.; et al. Comprehensive Analysis of the Transcriptional and Mutational Landscape of Follicular and Papillary Thyroid Cancers. PLoS Genet. 2016, 12, e1006239. [Google Scholar] [CrossRef] [Green Version]
  12. Alexander, E.K.; Kennedy, G.C.; Baloch, Z.W.; Cibas, E.S.; Chudova, D.; Diggans, J.; Friedman, L.; Kloos, R.T.; LiVolsi, V.A.; Mandel, S.J.; et al. Preoperative diagnosis of benign thyroid nodules with indeterminate cytology. N. Engl. J. Med. 2012, 367, 705–715. [Google Scholar] [CrossRef] [Green Version]
  13. Randolph, G.W.; Sosa, J.A.; Hao, Y.; Angell, T.E.; Shonka, D.C., Jr.; LiVolsi, V.A.; Ladenson, P.W.; Blevins, T.C.; Duh, Q.Y.; Ghossein, R.; et al. Preoperative Identification of Medullary Thyroid Carcinoma (MTC): Clinical Validation of the Afirma MTC RNA-Sequencing Classifier. Thyroid 2022, 32, 1069–1076. [Google Scholar] [CrossRef]
  14. Bible, K.C.; Kebebew, E.; Brierley, J.; Brito, J.P.; Cabanillas, M.E.; Clark, T.J., Jr.; Di Cristofano, A.; Foote, R.; Giordano, T.; Kasperbauer, J.; et al. 2021 American Thyroid Association Guidelines for Management of Patients with Anaplastic Thyroid Cancer. Thyroid 2021, 31, 337–386. [Google Scholar] [CrossRef]
  15. Tallini, G.; Tuttle, R.M.; Ghossein, R.A. The History of the Follicular Variant of Papillary Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2017, 102, 15–22. [Google Scholar] [CrossRef] [Green Version]
  16. Fusco, A.; Grieco, M.; Santoro, M.; Berlingieri, M.T.; Pilotti, S.; Pierotti, M.A.; Della Porta, G.; Vecchio, G. A new oncogene in human thyroid papillary carcinomas and their lymph-nodal metastases. Nature 1987, 328, 170–172. [Google Scholar] [CrossRef]
  17. Santoro, M.; Carlomagno, F.; Hay, I.D.; Herrmann, M.A.; Grieco, M.; Melillo, R.; Pierotti, M.A.; Bongarzone, I.; Della Porta, G.; Berger, N.; et al. Ret oncogene activation in human thyroid neoplasms is restricted to the papillary cancer subtype. J. Clin. Investig. 1992, 89, 1517–1522. [Google Scholar] [CrossRef] [Green Version]
  18. Lemoine, N.R.; Mayall, E.S.; Wyllie, F.S.; Williams, E.D.; Goyns, M.; Stringer, B.; Wynford-Thomas, D. High frequency of ras oncogene activation in all stages of human thyroid tumorigenesis. Oncogene 1989, 4, 159–164. [Google Scholar]
  19. Namba, H.; Rubin, S.A.; Fagin, J.A. Point mutations of ras oncogenes are an early event in thyroid tumorigenesis. Mol. Endocrinol. 1990, 4, 1474–1479. [Google Scholar] [CrossRef] [Green Version]
  20. Kroll, T.G.; Sarraf, P.; Pecciarini, L.; Chen, C.J.; Mueller, E.; Spiegelman, B.M.; Fletcher, J.A. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma [corrected]. Science 2000, 289, 1357–1360. [Google Scholar] [CrossRef]
  21. Nikiforova, M.N.; Biddinger, P.W.; Caudill, C.M.; Kroll, T.G.; Nikiforov, Y.E. PAX8-PPARgamma rearrangement in thyroid tumors: RT-PCR and immunohistochemical analyses. Am. J. Surg. Pathol. 2002, 26, 1016–1023. [Google Scholar] [CrossRef] [PubMed]
  22. Cohen, Y.; Xing, M.; Mambo, E.; Guo, Z.; Wu, G.; Trink, B.; Beller, U.; Westra, W.H.; Ladenson, P.W.; Sidransky, D. BRAF mutation in papillary thyroid carcinoma. J. Natl. Cancer Inst. 2003, 95, 625–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Nikiforov, Y.E.; Steward, D.L.; Robinson-Smith, T.M.; Haugen, B.R.; Klopper, J.P.; Zhu, Z.; Fagin, J.A.; Falciglia, M.; Weber, K.; Nikiforova, M.N. Molecular testing for mutations in improving the fine-needle aspiration diagnosis of thyroid nodules. J. Clin. Endocrinol. Metab. 2009, 94, 2092–2098. [Google Scholar] [CrossRef] [PubMed]
  24. Nikiforova, M.N.; Wald, A.I.; Roy, S.; Durso, M.B.; Nikiforov, Y.E. Targeted next-generation sequencing panel (ThyroSeq) for detection of mutations in thyroid cancer. J. Clin. Endocrinol. Metab. 2013, 98, E1852–E1860. [Google Scholar] [CrossRef] [Green Version]
  25. Nikiforov, Y.E.; Carty, S.E.; Chiosea, S.I.; Coyne, C.; Duvvuri, U.; Ferris, R.L.; Gooding, W.E.; LeBeau, S.O.; Ohori, N.P.; Seethala, R.R.; et al. Impact of the Multi-Gene ThyroSeq Next-Generation Sequencing Assay on Cancer Diagnosis in Thyroid Nodules with Atypia of Undetermined Significance/Follicular Lesion of Undetermined Significance Cytology. Thyroid 2015, 25, 1217–1223. [Google Scholar] [CrossRef]
  26. Wylie, D.; Beaudenon-Huibregtse, S.; Haynes, B.C.; Giordano, T.J.; Labourier, E. Molecular classification of thyroid lesions by combined testing for miRNA gene expression and somatic gene alterations. J. Pathol. Clin. Res. 2016, 2, 93–103. [Google Scholar] [CrossRef]
  27. Benjamin, H.; Schnitzer-Perlman, T.; Shtabsky, A.; VandenBussche, C.J.; Ali, S.Z.; Kolar, Z.; Pagni, F.; Rosetta Genomics, G.; Bar, D.; Meiri, E. Analytical validity of a microRNA-based assay for diagnosing indeterminate thyroid FNA smears from routinely prepared cytology slides. Cancer Cytopathol. 2016, 124, 711–721. [Google Scholar] [CrossRef]
  28. Nikiforova, M.N.; Mercurio, S.; Wald, A.I.; Barbi de Moura, M.; Callenberg, K.; Santana-Santos, L.; Gooding, W.E.; Yip, L.; Ferris, R.L.; Nikiforov, Y.E. Analytical performance of the ThyroSeq v3 genomic classifier for cancer diagnosis in thyroid nodules. Cancer 2018, 124, 1682–1690. [Google Scholar] [CrossRef] [Green Version]
  29. Patel, K.N.; Angell, T.E.; Babiarz, J.; Barth, N.M.; Blevins, T.; Duh, Q.Y.; Ghossein, R.A.; Harrell, R.M.; Huang, J.; Kennedy, G.C.; et al. Performance of a Genomic Sequencing Classifier for the Preoperative Diagnosis of Cytologically Indeterminate Thyroid Nodules. JAMA Surg. 2018, 153, 817–824. [Google Scholar] [CrossRef]
  30. Hao, Y.; Duh, Q.Y.; Kloos, R.T.; Babiarz, J.; Harrell, R.M.; Traweek, S.T.; Kim, S.Y.; Fedorowicz, G.; Walsh, P.S.; Sadow, P.M.; et al. Identification of Hurthle cell cancers: Solving a clinical challenge with genomic sequencing and a trio of machine learning algorithms. BMC Syst. Biol. 2019, 13 (Suppl. S2), 27. [Google Scholar] [CrossRef] [Green Version]
  31. Hu, M.I.; Waguespack, S.G.; Dosiou, C.; Ladenson, P.W.; Livhits, M.J.; Wirth, L.J.; Sadow, P.M.; Krane, J.F.; Stack, B.C.; Zafereo, M.E.; et al. Afirma Genomic Sequencing Classifier and Xpression Atlas Molecular Findings in Consecutive Bethesda III-VI Thyroid Nodules. J. Clin. Endocrinol. Metab. 2021, 106, 2198–2207. [Google Scholar] [CrossRef]
  32. Sistrunk, J.W.; Shifrin, A.; Frager, M.; Bardales, R.H.; Thomas, J.; Fishman, N.; Goldberg, P.; Guttler, R.; Grant, E. Clinical performance of multiplatform mutation panel and microRNA risk classifier in indeterminate thyroid nodules. J. Am. Soc. Cytopathol. 2020, 9, 232–241. [Google Scholar] [CrossRef]
  33. Jackson, S.; Kumar, G.; Banizs, A.B.; Toney, N.; Silverman, J.F.; Narick, C.M.; Finkelstein, S.D. Incremental utility of expanded mutation panel when used in combination with microRNA classification in indeterminate thyroid nodules. Diagn. Cytopathol. 2020, 48, 43–52. [Google Scholar] [CrossRef] [Green Version]
  34. Finkelstein, S.D.; Sistrunk, J.W.; Malchoff, C.; Thompson, D.V.; Kumar, G.; Timmaraju, V.A.; Repko, B.; Mireskandari, A.; Evoy-Goodman, L.A.; Massoll, N.A.; et al. A Retrospective Evaluation of the Diagnostic Performance of an Interdependent Pairwise MicroRNA Expression Analysis with a Mutation Panel in Indeterminate Thyroid Nodules. Thyroid 2022, 32, 1362–1371. [Google Scholar] [CrossRef]
  35. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef] [Green Version]
  36. Rajagopalan, H.; Bardelli, A.; Lengauer, C.; Kinzler, K.W.; Vogelstein, B.; Velculescu, V.E. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature 2002, 418, 934. [Google Scholar] [CrossRef]
  37. Singer, G.; Oldt, R., 3rd; Cohen, Y.; Wang, B.G.; Sidransky, D.; Kurman, R.J.; Shih Ie, M. Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. J. Natl. Cancer Inst. 2003, 95, 484–486. [Google Scholar] [CrossRef] [Green Version]
  38. Rowe, L.R.; Bentz, B.G.; Bentz, J.S. Detection of BRAF V600E activating mutation in papillary thyroid carcinoma using PCR with allele-specific fluorescent probe melting curve analysis. J. Clin. Pathol. 2007, 60, 1211–1215. [Google Scholar] [CrossRef] [Green Version]
  39. Sadow, P.M.; Heinrich, M.C.; Corless, C.L.; Fletcher, J.A.; Nose, V. Absence of BRAF, NRAS, KRAS, HRAS mutations, and RET/PTC gene rearrangements distinguishes dominant nodules in Hashimoto thyroiditis from papillary thyroid carcinomas. Endocr. Pathol. 2010, 21, 73–79. [Google Scholar] [CrossRef]
  40. Krane, J.F.; Cibas, E.S.; Endo, M.; Marqusee, E.; Hu, M.I.; Nasr, C.E.; Waguespack, S.G.; Wirth, L.J.; Kloos, R.T. The Afirma Xpression Atlas for thyroid nodules and thyroid cancer metastases: Insights to inform clinical decision-making from a fine-needle aspiration sample. Cancer Cytopathol. 2020, 128, 452–459. [Google Scholar] [CrossRef]
  41. Santos, M.T.D.; Buzolin, A.L.; Gama, R.R.; Silva, E.; Dufloth, R.M.; Figueiredo, D.L.A.; Carvalho, A.L. Molecular Classification of Thyroid Nodules with Indeterminate Cytology: Development and Validation of a Highly Sensitive and Specific New miRNA-Based Classifier Test Using Fine-Needle Aspiration Smear Slides. Thyroid 2018, 28, 1618–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Michael, C.W.; Kameyama, K.; Kitagawa, W.; Azar, N. Rapid on-site evaluation (ROSE) for fine needle aspiration of thyroid: Benefits, challenges and innovative solutions. Gland. Surg. 2020, 9, 1708–1715. [Google Scholar] [CrossRef] [PubMed]
  43. Patel, J.; Klopper, J.; Cottrill, E.E. Molecular diagnostics in the evaluation of thyroid nodules: Current use and prospective opportunities. Front. Endocrinol. 2023, 14, 1101410. [Google Scholar] [CrossRef] [PubMed]
  44. Nikiforova, M.N.; Lepe, M.; Tolino, L.A.; Miller, M.E.; Ohori, N.P.; Wald, A.I.; Landau, M.S.; Kaya, C.; Malapelle, U.; Bellevicine, C.; et al. Thyroid cytology smear slides: An untapped resource for ThyroSeq testing. Cancer Cytopathol. 2021, 129, 33–42. [Google Scholar] [CrossRef] [PubMed]
  45. Ye, L.; Zhou, X.; Huang, F.; Wang, W.; Qi, Y.; Xu, H.; Yang, S.; Shen, L.; Fei, X.; Xie, J.; et al. The genetic landscape of benign thyroid nodules revealed by whole exome and transcriptome sequencing. Nat. Commun. 2017, 8, 15533. [Google Scholar] [CrossRef] [Green Version]
  46. Guan, H.; Matonis, D.; Toraldo, G.; Lee, S.L. Clinical Significance of Thyroid-Stimulating Hormone Receptor Gene Mutations and/or Sodium-Iodine Symporter Gene Overexpression in Indeterminate Thyroid Fine Needle Biopsies. Front. Endocrinol. 2018, 9, 566. [Google Scholar] [CrossRef]
  47. Nikiforova, M.N.; Nikitski, A.V.; Panebianco, F.; Kaya, C.; Yip, L.; Williams, M.; Chiosea, S.I.; Seethala, R.R.; Roy, S.; Condello, V.; et al. GLIS Rearrangement is a Genomic Hallmark of Hyalinizing Trabecular Tumor of the Thyroid Gland. Thyroid 2019, 29, 161–173. [Google Scholar] [CrossRef]
  48. Angell, T.E.; Wirth, L.J.; Cabanillas, M.E.; Shindo, M.L.; Cibas, E.S.; Babiarz, J.E.; Hao, Y.; Kim, S.Y.; Walsh, P.S.; Huang, J.; et al. Analytical and Clinical Validation of Expressed Variants and Fusions From the Whole Transcriptome of Thyroid FNA Samples. Front. Endocrinol. 2019, 10, 612. [Google Scholar] [CrossRef]
  49. Ciarletto, A.M.; Narick, C.; Malchoff, C.D.; Massoll, N.A.; Labourier, E.; Haugh, K.; Mireskandari, A.; Finkelstein, S.D.; Kumar, G. Analytical and clinical validation of pairwise microRNA expression analysis to identify medullary thyroid cancer in thyroid fine-needle aspiration samples. Cancer Cytopathol. 2021, 129, 239–249. [Google Scholar] [CrossRef]
  50. Corver, W.E.; Ruano, D.; Weijers, K.; den Hartog, W.C.; van Nieuwenhuizen, M.P.; de Miranda, N.; van Eijk, R.; Middeldorp, A.; Jordanova, E.S.; Oosting, J.; et al. Genome haploidisation with chromosome 7 retention in oncocytic follicular thyroid carcinoma. PLoS ONE 2012, 7, e38287. [Google Scholar] [CrossRef] [Green Version]
  51. Gopal, R.K.; Kubler, K.; Calvo, S.E.; Polak, P.; Livitz, D.; Rosebrock, D.; Sadow, P.M.; Campbell, B.; Donovan, S.E.; Amin, S.; et al. Widespread Chromosomal Losses and Mitochondrial DNA Alterations as Genetic Drivers in Hurthle Cell Carcinoma. Cancer Cell 2018, 34, 242–255.e5. [Google Scholar] [CrossRef] [Green Version]
  52. Ganly, I.; Makarov, V.; Deraje, S.; Dong, Y.; Reznik, E.; Seshan, V.; Nanjangud, G.; Eng, S.; Bose, P.; Kuo, F.; et al. Integrated Genomic Analysis of Hurthle Cell Cancer Reveals Oncogenic Drivers, Recurrent Mitochondrial Mutations, and Unique Chromosomal Landscapes. Cancer Cell 2018, 34, 256–270.e5. [Google Scholar] [CrossRef] [Green Version]
  53. Silaghi, C.A.; Lozovanu, V.; Georgescu, C.E.; Georgescu, R.D.; Susman, S.; Nasui, B.A.; Dobrean, A.; Silaghi, H. Thyroseq v3, Afirma GSC, and microRNA Panels Versus Previous Molecular Tests in the Preoperative Diagnosis of Indeterminate Thyroid Nodules: A Systematic Review and Meta-Analysis. Front. Endocrinol. 2021, 12, 649522. [Google Scholar] [CrossRef]
  54. Lee, E.; Terhaar, S.; McDaniel, L.; Gorelik, D.; Gerhard, E.; Chen, C.; Ma, Y.; Joshi, A.S.; Goodman, J.F.; Thakkar, P.G. Diagnostic performance of the second-generation molecular tests in the assessment of indeterminate thyroid nodules: A systematic review and meta-analysis. Am. J. Otolaryngol. 2022, 43, 103394. [Google Scholar] [CrossRef]
  55. Livhits, M.J.; Zhu, C.Y.; Kuo, E.J.; Nguyen, D.T.; Kim, J.; Tseng, C.H.; Leung, A.M.; Rao, J.; Levin, M.; Douek, M.L.; et al. Effectiveness of Molecular Testing Techniques for Diagnosis of Indeterminate Thyroid Nodules: A Randomized Clinical Trial. JAMA Oncol. 2021, 7, 70–77. [Google Scholar] [CrossRef]
  56. Hu, Q.L.; Schumm, M.A.; Zanocco, K.A.; Yeh, M.W.; Livhits, M.J.; Wu, J.X. Cost analysis of reflexive versus selective molecular testing for indeterminate thyroid nodules. Surgery 2022, 171, 147–154. [Google Scholar] [CrossRef]
  57. Chen, D.W.; Haymart, M.R. Disparities Research in Thyroid Cancer: Challenges and Strategies for Improvement. Thyroid 2020, 30, 1231–1235. [Google Scholar] [CrossRef]
  58. Xiong, Y.; Li, X.; Liang, L.; Li, D.; Yan, L.; Li, X.; Di, J.; Li, T. Application of biomarkers in the diagnosis of uncertain samples of core needle biopsy of thyroid nodules. Virchows Arch. 2021, 479, 961–974. [Google Scholar] [CrossRef]
  59. Alzumaili, B.A.; Krumeich, L.N.; Collins, R.; Kravchenko, T.; Ababneh, E.I.; Fisch, A.S.; Faquin, W.C.; Nose, V.; Martinez-Lage, M.; Randolph, G.W.; et al. A Comprehensive Study on the Diagnosis and Management of Noninvasive Follicular Thyroid Neoplasm with Papillary-Like Nuclear Features. Thyroid 2023, 33, 566–577. [Google Scholar] [CrossRef]
  60. Labourier, E.; Fahey, T.J., 3rd. Preoperative molecular testing in thyroid nodules with Bethesda VI cytology: Clinical experience and review of the literature. Diagn. Cytopathol. 2021, 49, E175–E180. [Google Scholar] [CrossRef]
  61. Niciporuka, R.; Nazarovs, J.; Ozolins, A.; Narbuts, Z.; Miklasevics, E.; Gardovskis, J. Can We Predict Differentiated Thyroid Cancer Behavior? Role of Genetic and Molecular Markers. Medicina 2021, 57, 1131. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, J.; Liu, R.; Shen, X.; Zhu, G.; Li, B.; Xing, M. The Genetic Duet of BRAF V600E and TERT Promoter Mutations Robustly Predicts Loss of Radioiodine Avidity in Recurrent Papillary Thyroid Cancer. J. Nucl. Med. 2020, 61, 177–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Skaugen, J.M.; Taneja, C.; Liu, J.B.; Wald, A.I.; Nikitski, A.V.; Chiosea, S.I.; Seethala, R.R.; Ohori, N.P.; Karslioglu-French, E.; Carty, S.E.; et al. Performance of a Multigene Genomic Classifier in Thyroid Nodules with Suspicious for Malignancy Cytology. Thyroid 2022, 32, 1500–1508. [Google Scholar] [CrossRef] [PubMed]
  64. Hescot, S.; Al Ghuzlan, A.; Henry, T.; Sheikh-Alard, H.; Lamartina, L.; Borget, I.; Hadoux, J.; Baudin, E.; Dupuy, C.; Nikitski, A.V.; et al. Prognostic of recurrence and survival in poorly differentiated thyroid cancer. Endocr. Relat. Cancer 2022, 29, 625–634. [Google Scholar] [CrossRef]
  65. Durante, C.; Haddy, N.; Baudin, E.; Leboulleux, S.; Hartl, D.; Travagli, J.P.; Caillou, B.; Ricard, M.; Lumbroso, J.D.; De Vathaire, F.; et al. Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: Benefits and limits of radioiodine therapy. J. Clin. Endocrinol. Metab. 2006, 91, 2892–2899. [Google Scholar] [CrossRef]
  66. Sherman, S.I. Cytotoxic chemotherapy for differentiated thyroid carcinoma. Clin. Oncol. (R. Coll. Radiol.) 2010, 22, 464–468. [Google Scholar] [CrossRef]
  67. Kelly, L.M.; Barila, G.; Liu, P.; Evdokimova, V.N.; Trivedi, S.; Panebianco, F.; Gandhi, M.; Carty, S.E.; Hodak, S.P.; Luo, J.; et al. Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 4233–4238. [Google Scholar] [CrossRef] [Green Version]
  68. 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] [Green Version]
  69. Ferrari, S.M.; Politti, U.; Spisni, R.; Materazzi, G.; Baldini, E.; Ulisse, S.; Miccoli, P.; Antonelli, A.; Fallahi, P. Sorafenib in the treatment of thyroid cancer. Expert. Rev. Anticancer Ther. 2015, 15, 863–874. [Google Scholar] [CrossRef]
  70. Ferrari, S.M.; Elia, G.; Ragusa, F.; Paparo, S.R.; Mazzi, V.; Miccoli, M.; Galdiero, M.R.; Varricchi, G.; Foddis, R.; Guglielmi, G.; et al. Lenvatinib: An investigational agent for the treatment of differentiated thyroid cancer. Expert Opin. Investig. Drugs 2021, 30, 913–921. [Google Scholar] [CrossRef]
  71. 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] [Green Version]
  72. Falchook, G.S.; Millward, M.; Hong, D.; Naing, A.; Piha-Paul, S.; Waguespack, S.G.; Cabanillas, M.E.; Sherman, S.I.; Ma, B.; Curtis, M.; et al. BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer. Thyroid 2015, 25, 71–77. [Google Scholar] [CrossRef] [Green Version]
  73. 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] [Green Version]
  74. Rothenberg, S.M.; McFadden, D.G.; Palmer, E.L.; Daniels, G.H.; Wirth, L.J. Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clin. Cancer Res. 2015, 21, 1028–1035. [Google Scholar] [CrossRef] [Green Version]
  75. Dunn, L.A.; Sherman, E.J.; Baxi, S.S.; Tchekmedyian, V.; Grewal, R.K.; Larson, S.M.; Pentlow, K.S.; Haque, S.; Tuttle, R.M.; Sabra, M.M.; et al. Vemurafenib Redifferentiation of BRAF Mutant, RAI-Refractory Thyroid Cancers. J. Clin. Endocrinol. Metab. 2019, 104, 1417–1428. [Google Scholar] [CrossRef]
  76. Liu, S.V.; Macke, L.A.; Colton, B.S.; Imran, S.S.; Christiansen, J.; Chow-Maneval, E.; Hornby, Z.; Multani, P.S. Response to Entrectinib in Differentiated Thyroid Cancer With a ROS1 Fusion. JCO Precis. Oncol. 2017, 1, 1–5. [Google Scholar] [CrossRef] [Green Version]
  77. 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] [Green Version]
  78. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.C.; Cabanillas, M.E.; Boran, A.; et al. Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: Updated analysis from the phase II ROAR basket study. Ann. Oncol. 2022, 33, 406–415. [Google Scholar] [CrossRef]
  79. Marcus, L.; Lemery, S.J.; Keegan, P.; Pazdur, R. FDA Approval Summary: Pembrolizumab for the Treatment of Microsatellite Instability-High Solid Tumors. Clin. Cancer Res. 2019, 25, 3753–3758. [Google Scholar] [CrossRef] [Green Version]
  80. Wirth, L.J.; Sherman, E.; Robinson, B.; Solomon, B.; Kang, H.; Lorch, J.; Worden, F.; Brose, M.; Patel, J.; Leboulleux, S.; et al. Efficacy of Selpercatinib in RET-Altered Thyroid Cancers. N. Engl. J. Med. 2020, 383, 825–835. [Google Scholar] [CrossRef]
  81. Brose, M.S.; Robinson, B.; Sherman, S.I.; Krajewska, J.; Lin, C.C.; Vaisman, F.; Hoff, A.O.; Hitre, E.; Bowles, D.W.; Hernando, J.; et al. Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2021, 22, 1126–1138. [Google Scholar] [CrossRef] [PubMed]
  82. Subbiah, V.; Hu, M.I.; Wirth, L.J.; Schuler, M.; Mansfield, A.S.; Curigliano, G.; Brose, M.S.; Zhu, V.W.; Leboulleux, S.; Bowles, D.W.; et al. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): A multi-cohort, open-label, registrational, phase 1/2 study. Lancet Diabetes Endocrinol. 2021, 9, 491–501. [Google Scholar] [CrossRef] [PubMed]
  83. Waguespack, S.G.; Drilon, A.; Lin, J.J.; Brose, M.S.; McDermott, R.; Almubarak, M.; Bauman, J.; Casanova, M.; Krishnamurthy, A.; Kummar, S.; et al. Efficacy and safety of larotrectinib in patients with TRK fusion-positive thyroid carcinoma. Eur. J. Endocrinol. 2022, 186, 631–643. [Google Scholar] [CrossRef]
  84. Solomon, B.J.; Tan, L.; Lin, J.J.; Wong, S.Q.; Hollizeck, S.; Ebata, K.; Tuch, B.B.; Yoda, S.; Gainor, J.F.; Sequist, L.V.; et al. RET Solvent Front Mutations Mediate Acquired Resistance to Selective RET Inhibition in RET-Driven Malignancies. J. Thorac. Oncol. 2020, 15, 541–549. [Google Scholar] [CrossRef] [Green Version]
  85. Zhu, V.W.; Madison, R.; Schrock, A.B.; Ou, S.I. Emergence of High Level of MET Amplification as Off-Target Resistance to Selpercatinib Treatment in KIF5B-RET NSCLC. J. Thorac. Oncol. 2020, 15, e124–e127. [Google Scholar] [CrossRef] [PubMed]
  86. Su, C.; Jiang, Y.; Jiang, W.; Wang, H.; Liu, S.; Shao, Y.; Zhao, W.; Ning, R.; Yu, Q. STRN-ALK Fusion in Lung Adenocarcinoma with Excellent Response Upon Alectinib Treatment: A Case Report and Literature Review. OncoTargets Ther. 2020, 13, 12515–12519. [Google Scholar] [CrossRef]
  87. Genutis, L.K.; Tomsic, J.; Bundschuh, R.A.; Brock, P.L.; Williams, M.D.; Roychowdhury, S.; Reeser, J.W.; Frankel, W.L.; Alsomali, M.; Routbort, M.J.; et al. Microsatellite Instability Occurs in a Subset of Follicular Thyroid Cancers. Thyroid 2019, 29, 523–529. [Google Scholar] [CrossRef]
  88. Janz, T.A.; Neskey, D.M.; Nguyen, S.A.; Lentsch, E.J. Is the incidence of anaplastic thyroid cancer increasing: A population based epidemiology study. World J. Otorhinolaryngol. Head Neck Surg. 2019, 5, 34–40. [Google Scholar] [CrossRef]
  89. Lin, B.; Ma, H.; Ma, M.; Zhang, Z.; Sun, Z.; Hsieh, I.Y.; Okenwa, O.; Guan, H.; Li, J.; Lv, W. The incidence and survival analysis for anaplastic thyroid cancer: A SEER database analysis. Am. J. Transl. Res. 2019, 11, 5888–5896. [Google Scholar]
  90. Lang, M.; Longerich, T.; Anamaterou, C. Targeted therapy with vemurafenib in BRAF(V600E)-mutated anaplastic thyroid cancer. Thyroid Res. 2023, 16, 5. [Google Scholar] [CrossRef]
  91. Xu, B.; Fuchs, T.; Dogan, S.; Landa, I.; Katabi, N.; Fagin, J.A.; Tuttle, R.M.; Sherman, E.; Gill, A.J.; Ghossein, R. Dissecting Anaplastic Thyroid Carcinoma: A Comprehensive Clinical, Histologic, Immunophenotypic, and Molecular Study of 360 Cases. Thyroid 2020, 30, 1505–1517. [Google Scholar] [CrossRef]
  92. Maniakas, A.; Zafereo, M.; Cabanillas, M.E. Anaplastic Thyroid Cancer: New Horizons and Challenges. Endocrinol. Metab. Clin. North. Am. 2022, 51, 391–401. [Google Scholar] [CrossRef]
  93. Rosove, M.H.; Peddi, P.F.; Glaspy, J.A. BRAF V600E inhibition in anaplastic thyroid cancer. N. Engl. J. Med. 2013, 368, 684–685. [Google Scholar] [CrossRef]
  94. Smith, A.L.; Williams, M.D.; Stewart, J.; Wang, W.L.; Krishnamurthy, S.; Cabanillas, M.E.; Roy-Chowdhuri, S. Utility of the BRAF p.V600E immunoperoxidase stain in FNA direct smears and cell block preparations from patients with thyroid carcinoma. Cancer Cytopathol. 2018, 126, 406–413. [Google Scholar] [CrossRef] [Green Version]
  95. Sandulache, V.C.; Williams, M.D.; Lai, S.Y.; Lu, C.; William, W.N.; Busaidy, N.L.; Cote, G.J.; Singh, R.R.; Luthra, R.; Cabanillas, M.E. Real-Time Genomic Characterization Utilizing Circulating Cell-Free DNA in Patients with Anaplastic Thyroid Carcinoma. Thyroid 2017, 27, 81–87. [Google Scholar] [CrossRef] [Green Version]
  96. Khatami, F.; Tavangar, S.M. Liquid Biopsy in Thyroid Cancer: New Insight. Int. J. Hematol. Oncol. Stem Cell Res. 2018, 12, 235–248. [Google Scholar]
  97. Yeo, M.K.; Liang, Z.L.; Oh, T.; Moon, Y.; An, S.; Kim, M.K.; Kim, K.S.; Shong, M.; Kim, J.M.; Jo, Y.S. Pyrosequencing cut-off value identifying BRAFV600E mutation in fine needle aspiration samples of thyroid nodules. Clin. Endocrinol. 2011, 75, 555–560. [Google Scholar] [CrossRef]
  98. Huber, F.; Lang, H.P.; Glatz, K.; Rimoldi, D.; Meyer, E.; Gerber, C. Fast Diagnostics of BRAF Mutations in Biopsies from Malignant Melanoma. Nano Lett. 2016, 16, 5373–5377. [Google Scholar] [CrossRef] [Green Version]
  99. Wang, J.R.; Zafereo, M.E.; Dadu, R.; Ferrarotto, R.; Busaidy, N.L.; Lu, C.; Ahmed, S.; Gule-Monroe, M.K.; Williams, M.D.; Sturgis, E.M.; et al. Complete Surgical Resection Following Neoadjuvant Dabrafenib Plus Trametinib in BRAF(V600E)-Mutated Anaplastic Thyroid Carcinoma. Thyroid 2019, 29, 1036–1043. [Google Scholar] [CrossRef] [Green Version]
  100. Gild, M.L.; Bullock, M.; Tsang, V.; Clifton-Bligh, R.J.; Robinson, B.G.; Wirth, L.J. Challenges and Strategies to Combat Resistance Mechanisms in Thyroid Cancer Therapeutics. Thyroid 2023, 33, 682–690. [Google Scholar] [CrossRef]
  101. Ceolin, L.; Duval, M.; Benini, A.F.; Ferreira, C.V.; Maia, A.L. Medullary thyroid carcinoma beyond surgery: Advances, challenges, and perspectives. Endocr. Relat. Cancer 2019, 26, R499–R518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Alzumaili, B.; Xu, B.; Spanheimer, P.M.; Tuttle, R.M.; Sherman, E.; Katabi, N.; Dogan, S.; Ganly, I.; Untch, B.R.; Ghossein, R.A. Grading of medullary thyroid carcinoma on the basis of tumor necrosis and high mitotic rate is an independent predictor of poor outcome. Mod. Pathol. 2020, 33, 1690–1701. [Google Scholar] [CrossRef] [PubMed]
  103. Boichard, A.; Croux, L.; Al Ghuzlan, A.; Broutin, S.; Dupuy, C.; Leboulleux, S.; Schlumberger, M.; Bidart, J.M.; Lacroix, L. Somatic RAS mutations occur in a large proportion of sporadic RET-negative medullary thyroid carcinomas and extend to a previously unidentified exon. J. Clin. Endocrinol. Metab. 2012, 97, E2031–E2035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Moura, M.M.; Cavaco, B.M.; Pinto, A.E.; Domingues, R.; Santos, J.R.; Cid, M.O.; Bugalho, M.J.; Leite, V. Correlation of RET somatic mutations with clinicopathological features in sporadic medullary thyroid carcinomas. Br. J. Cancer 2009, 100, 1777–1783. [Google Scholar] [CrossRef] [Green Version]
  105. Degrauwe, N.; Sosa, J.A.; Roman, S.; Deshpande, H.A. Vandetanib for the treatment of metastatic medullary thyroid cancer. Clin. Med. Insights Oncol. 2012, 6, 243–252. [Google Scholar] [CrossRef]
  106. Nagilla, M.; Brown, R.L.; Cohen, E.E. Cabozantinib for the treatment of advanced medullary thyroid cancer. Adv. Ther. 2012, 29, 925–934. [Google Scholar] [CrossRef]
  107. Frisco, N.A.; Gunn, A.H.; Thomas, S.M.; Stang, M.T.; Scheri, R.P.; Kazaure, H.S. Medullary thyroid cancer with RET V804M mutation: More indolent than expected? Surgery 2023, 173, 260–267. [Google Scholar] [CrossRef]
Figure 1. Timeline of the two most common molecular tests for preoperative evaluation of thyroid nodules for fine needle aspiration biopsy; Afirma and ThyroSeq. Nikiforov et al. published the earliest molecular testing on FNA in 2009 [23] but the test became available in 2013 as ThyroSeq V1 [24]. ThyroSeq V2.1 followed in 2015 [25], and the current commercially available test is ThyroSeq V3 [20]. Afirma GEC started in 2012 [12] and updated as Afirma GSC in 2018 [29]. The latest version that is currently available is Afirma XA [40]. GEC: Gene Expression Classifier, GSC: Genetic Sequencing Classifier and XA: Xpression Atlas.
Figure 1. Timeline of the two most common molecular tests for preoperative evaluation of thyroid nodules for fine needle aspiration biopsy; Afirma and ThyroSeq. Nikiforov et al. published the earliest molecular testing on FNA in 2009 [23] but the test became available in 2013 as ThyroSeq V1 [24]. ThyroSeq V2.1 followed in 2015 [25], and the current commercially available test is ThyroSeq V3 [20]. Afirma GEC started in 2012 [12] and updated as Afirma GSC in 2018 [29]. The latest version that is currently available is Afirma XA [40]. GEC: Gene Expression Classifier, GSC: Genetic Sequencing Classifier and XA: Xpression Atlas.
Genes 14 01314 g001
Figure 2. Algorithm for preoperative evaluation of thyroid nodules following fine needle aspiration biopsy (FNAB). Nodules of Bethesda III and IV (indeterminate thyroid nodules) are sent for molecular testing for further classification. Nodules of Bethesda V and VI (suspicious and malignant) can be sent for Afirma XA or ThyroSeq v3 to study the tumor landscape and potential high-risk mutations (TERT promotor, TP53, etc.). Most FNAB are sent out and most labs have a reflex for molecular testing for Bethesda III or IV. When rapid on-site evaluation is available, samples can be sent directly for molecular testing. NGS: Next Generation Sequencing, XA: Xpression Atlas, RNA: ribonucleic acid, DNA: Deoxyribonucleic acid and miRNA: micro ribonucleic acid.
Figure 2. Algorithm for preoperative evaluation of thyroid nodules following fine needle aspiration biopsy (FNAB). Nodules of Bethesda III and IV (indeterminate thyroid nodules) are sent for molecular testing for further classification. Nodules of Bethesda V and VI (suspicious and malignant) can be sent for Afirma XA or ThyroSeq v3 to study the tumor landscape and potential high-risk mutations (TERT promotor, TP53, etc.). Most FNAB are sent out and most labs have a reflex for molecular testing for Bethesda III or IV. When rapid on-site evaluation is available, samples can be sent directly for molecular testing. NGS: Next Generation Sequencing, XA: Xpression Atlas, RNA: ribonucleic acid, DNA: Deoxyribonucleic acid and miRNA: micro ribonucleic acid.
Genes 14 01314 g002
Table 1. History of Molecular Diagnostics.
Table 1. History of Molecular Diagnostics.
Year
Authors
Molecular Alterations or Genetic TestingDiagnosis
1987
Fusco et al. [16]
TRK and RET rearrangementsPTC
1989
Lemoine et al. [18]
NRAS p.Q61R and HRAS p.Q61RThyroid neoplastic process (FTA, FTC and ATC)
1990
Namba et al. [19]
NRAS, HRAS and KRAS at codons 12 and 13Benign and neoplastic thyroid nodules
1992
Santoro et al. [17]
RET aberrations PTC
2000
Kroll et al. [20]
PAX8::PPARG fusionFTC
2002
Nikiforova et al. [21]
PAX8::PPARG fusionFTC and FTA
2003
Cohen et al. [22]
BRAF p.T1796A point mutationPTC
2009
Nikiforov et al. [23]
BRAF (p.V600E, p.K601E), NRAS, KRAS and HRAS and RET::PTC1, RET::PTC3 and PAX8::PPARG gene fusionBethesda III and IV
2012
Alexander et al. [12]
Afirma GEC: (using machine learning to interpret the expression of mRNA of 167 genes through microarray platforms)Bethesda III and IV
2013
Nikiforova et al. [24]
ThyroSeq V1: NGS (BRAF, RET, NRAS, KRAS, HRAS, PIK3CA, TP53, TSHR, PTEN, GNAS, CTNNB1 and AKT1)Bethesda III and IV
2014
The Cancer Genome Atlas (TCGA) [9]
BRAFV600E-like or RAS-like phenotypePTC including follicular variant
2015
Yoo et al. [11]
BRAF-like, RAS-like and non-BRAF-/non-RAS-like (NBNR)FTA and minimally invasive FTC
2015
Nikiforov et al. [25]
ThyroSeq V2.1: ThyroSeq V1 and EIF1AX and BRAFV601K and 40 gene fusions including THADA, ALK, PAX8::PPARG, TRK1 and TRK3Bethesda III and IV
2016
Wylie et al. [26]
RosettaGX Reveal (miRNA classifier)Bethesda III and IV
2016
Benjamin et al. [27]
Asuragen (miRNA and somatic gene mutational platform)Bethesda III and IV
2018
Nikiforova et al. [28]
ThyroSeq V3: NGS of 112 thyroid cancer-related genesBethesda III and IV
2018
Patel et al. [29]
Afirma GSC (describing RNA transcriptome with additional sequencing of nuclear and mitochondrial genes, changes in genomic copy number including loss of heterozygosity)Bethesda III and IV
2019
Hao et al. [30]
Afirma GSC (NGS of whole transcriptome RNA sequencing)Oncocytic cell neoplasms
2019/2020
Hu et al. [31]
Afirma Xpression Atlas (enumerates mutations in 593 genes informing 905 variants and 235 fusions in suspicious Afirma GSC)Bethesda III, IV, V and VI
2020
Sistrunk et al. [32]
Jackson et al. [33]
Finkelstein et al. [34]
MPTX (ThyGeNEXT/ThyraMIR): NGS (ALK, BRAF, GNAS, HRAS, KRAS, NRAS, PIK3CA, PTEN, RET and TERT promoter genes) as well as mRNA fusion genes (ALK, BRAF, NTRK, PPARG, RET, PAX8, TBP, USP33 and THADA) and miRNA- (21, 29, 31, 138, 139, 155, 146, 204, 222, 375, 551)Bethesda III and IV
PTC: papillary thyroid carcinoma, FTA: follicular thyroid adenoma, FTC: follicular thyroid carcinoma, ATC: anaplastic thyroid carcinoma, mRNA: messenger ribonucleic acid, miRNA: micro ribonucleic acid, NGS: Next Generation Sequencing, GEC: Gene Expression Classifier and GSC: Genetic Sequencing Classifier.
Table 2. FDA-approved medications for different subtypes of thyroid carcinoma.
Table 2. FDA-approved medications for different subtypes of thyroid carcinoma.
MedicationMolecular TargetMechanism of ActionYearReference
VandetanibInhibits RET in MTCSelective RET TKI2012Degrauwe et al. [10]
Alectinib *ALK1 fusion-positive thyroid carcinomaALK-TKI2014Kelly et al. [67]
SorafenibAggressive RAI-resistant PTC or FTCAnti-angiogenic multi-targeted kinase inhibitor (aaMKI)2014
2015
Brose and Nutting et al. [68]
Ferrari, Politti et al. [69]
Lenvatinib Aggressive RAI-resistant PTC or FTCAnti-angiogenic multi-targeted kinase inhibitor (aaMKI)2015
2021
Ferrari and Elia et al. [70]
Schlumberger et al. [71]
DabrafenibBRAF p.V600E-mutated PTCBRAF inhibitor2015
2016
Falchook et al. [72]
Brose and Cabanillas et al. [73]
Dabrafenib and VemurafenibBRAF p.V600E-mutated PTCBRAF inhibitor2015
2019
Rothenberg et al. [74]
Dunn et al. [75]
EntrectinibROS1 fusion-positive thyroid carcinomaMultikinase inhibitor (NTRK1/2/3, ROS1 and ALK)2017Liu et al. [76]
Dabrafenib/
Trametinib
BRAF p.V600E-mutated ATCBRAF/
MEK inhibitors
2018
2022
Subbiah and Kreitman et al. [77]
Subbiah and Kreitman et al. [78]
PembrolizumabSolid tumors including thyroid tumorsProgrammed death (PD-1) inhibitors2019Marcus et al. [79]
Selpercatinib-Inhibits RET in MTC
-RET fusion-positive thyroid carcinoma
Selective RET TKI2020Wirth et al. [80]
CabozantinibSecond line for treatment of aggressive RAI-resistant PTC or FTCAntiangiogenic multi-targeted kinase inhibitor (aaMKIs)2021Brose and Robinson et al. [81]
Pralsetinib-Inhibits RET in MTC
-RET fusion-positive thyroid carcinoma
TKI2021Subbiah and Hu et al. [82]
LarotrectinibTRK fusion-positive thyroid carcinomaSelective TRK inhibitor2022Waguespack et al. [83]
* Approved for non-small cell lung carcinoma. FDA: the Food and Drug Administration, RAI: Radioactive iodine, PTC: Papillary thyroid carcinoma, FTC: Follicular thyroid carcinoma, ATC: Anaplastic thyroid carcinoma and MTC: Medullary thyroid carcinoma. TKI: tyrosine kinase inhibitor.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alzumaili, B.; Sadow, P.M. Update on Molecular Diagnostics in Thyroid Pathology: A Review. Genes 2023, 14, 1314. https://doi.org/10.3390/genes14071314

AMA Style

Alzumaili B, Sadow PM. Update on Molecular Diagnostics in Thyroid Pathology: A Review. Genes. 2023; 14(7):1314. https://doi.org/10.3390/genes14071314

Chicago/Turabian Style

Alzumaili, Bayan, and Peter M. Sadow. 2023. "Update on Molecular Diagnostics in Thyroid Pathology: A Review" Genes 14, no. 7: 1314. https://doi.org/10.3390/genes14071314

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