Next Article in Journal
The Beneficial Effects of Essential Oils in Anti-Obesity Treatment
Next Article in Special Issue
Extracellular Vesicles as Signal Carriers in Malignant Thyroid Tumors?
Previous Article in Journal
Differential p53-Mediated Cellular Responses to DNA-Damaging Therapeutic Agents
Previous Article in Special Issue
Modifier Role of Common RET Variants in Sporadic Medullary Thyroid Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Inhibitor-Based Therapies for Thyroid Cancer—An Update

by
Maciej Ratajczak
1,
Damian Gaweł
2,3,* and
Marlena Godlewska
3,*
1
Centre of Postgraduate Medical Education, Department of Endocrinology, Marymoncka 99/103, 01-813 Warsaw, Poland
2
Centre of Postgraduate Medical Education, Department of Immunohematology, Marymoncka 99/103, 01-813 Warsaw, Poland
3
Centre of Postgraduate Medical Education, Department of Biochemistry and Molecular Biology, Marymoncka 99/103, 01-813 Warsaw, Poland
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(21), 11829; https://doi.org/10.3390/ijms222111829
Submission received: 27 September 2021 / Revised: 27 October 2021 / Accepted: 28 October 2021 / Published: 31 October 2021
(This article belongs to the Special Issue Thyroid Disease and Thyroid Cancer)

Abstract

:
Thyroid cancers (TCs) are the most common tumors of the endocrine system and a constant rise in the number of TC cases has been observed for the past few decades. TCs are one of the most frequent tumors in younger adults, especially in women, therefore early diagnosis and effective therapy are especially important. Ultrasonography examination followed by fine needle biopsy have become the gold standard for diagnosis of TCs, as these strategies allow for early-stage detection and aid accurate qualification for further procedures, including surgical treatment. Despite all the advancements in detection and treatment of TCs, constant mortality levels are still observed. Therefore, a novel generation line of targeted treatment strategies is being developed, including personalized therapies with kinase inhibitors. Recent molecular studies on TCs demonstrate that kinase inhibitor-based therapies might be considered as the most promising. In the past decade, new kinase inhibitors with different mechanisms of action have been reported and approved for clinical trials. This review presents an up-to-date picture of new approaches and challenges of inhibitor-based therapies in treatment of TCs, focusing on the latest findings reported over the past two years.

1. Introduction

1.1. Thyroid Cancer: Origins and Classification

The thyroid gland consists of two types of cells: thyrocytes (epithelial, follicular cells) and C cells (parafollicular cells). Thyrocytes form thyroid follicles, which are the structural and functional units of the mature thyroid gland. Thyroid hormones (THs), which are involved in regulation of metabolism, are synthetized and stored in the follicular lumen. Production and secretion of THs is controlled by the thyroid-stimulating hormone (TSH) [1]. C cells are located adjacently to thyroid follicles and are responsible for production of various biologically-active substances, such as calcitonin, carcinoembryonic antigen, prostaglandins and serotonin [2].
Classification of thyroid cancer is based on the histological type of thyroid cells. Differentiated thyroid cancer (DTC) and de-differentiated thyroid cancer (DeTC) arise from follicular cells. DTC is subclassified into papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC) and Hürtle cell carcinoma (HCC) [3]. HCC, which was previously recognized as FTC, is now classified as a subtype of DTC [3,4]. The rarest, but most aggressive forms of TC, anaplastic thyroid carcinoma (ATC) and poorly differentiated thyroid carcinoma (PDTC) are classified as DeTCs. PDTCs are a subset of thyroid tumors intermediate between DTCs and ATCs. Medullary thyroid cancer (MTC) is classified as the forth main type of TC and in contrast to DTC and DeTC, is a neuroendocrine tumor originating from parafollicular C cells. The Cancer Genome Atlas (TCGA) Research Network [5] data and other subsequent studies [6,7,8] underlined the need for reclassification of thyroid cancers into molecular- and genotype-based subtypes that would predict their prognosis and be helpful in administration of treatment strategies. Therefore, molecular and genetic aberrations were additionally considered and implemented to the World Health Organization (WHO) Classification of Tumours of Endocrine Organs that was published in 2017 [3].

1.2. Thyroid Cancer Epidemiology

Thyroid cancers (TCs) account for ~3% of all malignancies worldwide. Among cancers of the endocrine system, TCs are the most common (approximately 95% of all cases). For the past several decades, a substantial increase in cancer morbidity has been observed and the same tendency has been found for TCs. According to GLOBOCAN 2020, the WHO cancer statistics for 185 countries worldwide, 586,202 new TC cases were reported with mortality of all cancer types reaching 0.4% [9]. This ranks TCs in the 11th place for all cancer incidences. Until 2020, the morbidity of TCs was the most rapidly increasing among all cancers in the United States. In 2021, in the USA alone, the number of TCs was estimated to be about 44,280 cases and ~2200 patients were expected to die [10].
DTCs account for more than 90% of all TC cases, where ~84% of incidences are PTCs, while diagnoses of FTCs and HCCs stand for approximately 2% each. ATC also represents ~2% of thyroid malignancies, and as the most aggressive thyroid tumor, is responsible for 50% of annual thyroid cancer deaths. ATC generally affects 60–70 year-old patients. PDTC is a relatively rare disease, accounting for slightly more than 2% of all new cases of TCs each year. MTC accounts for about 4–5% of thyroid cancers. Most MTCs (70–80%) are observed in 50 to 60 year-old patients and result from sporadic mutations. Over 50 different point mutations have been linked with development of MTC [11]. A hereditary pattern of MTC is present in 20–30% of cases, transmitted as an autosomal dominant trait, while a mutation in the RET gene at codon 634 is responsible for development of familial MTC in 85% of patients. MTC is associated with multiple endocrine neoplasia (MEN) type IIA and IIB syndrome. Approximately 35% of patients with MTC have lymph node metastases at the time of diagnosis, while about 20% have distant metastases [3,12,13].

1.3. Thyroid Cancer: Risk Factors

Incidences of TC are most common among younger adults [14,15]. Women are three times more likely to develop TC than men [10,16]. This sex-dependent disparity is mostly observed for PTCs and FTCs [17]. The possible relationship between female hormonal and reproductive processes (menstrual cycle, pregnancy, menopause and hormone replacement therapy) has been studied, but no direct correlation with higher risk of developing thyroid cancer was established [18,19]. According to the American Thyroid Association (ATA), a high-risk factors for TC are: (i) history of TC in one or more first degree relatives, (ii) exposure to external beam radiation in the childhood, (iii) exposure to ionizing radiation in the childhood or adolescence, (iv) prior hemithyroidectomy with discovery of TC, (v) 18F-fluoro-2-deoxy-D-glucose (18FDG) avidity on positron emission tomography (PET) scanning in thyroid nodule, (vi) calcitonin blood level more than 100 pg/mL [20]. Moreover, there are several genetic syndromes predisposing to TC development, like multiple endocrine neoplasia type 2 (MEN2), familial adenomatous polyposis (FAP), Cowden disease, Carney complex and Werner syndrome/progeria [20,21]. Thyroid dysfunctions, especially hypothyroidism, are postulated as factors associated with increased incidence of TC [22,23]. The association between elevated body mass index (BMI) and aggressive clinicopathologic features of PTC has also been reported [24].

1.4. Role of Genetic Factors in Pathogenesis of TC

Progression of TC relies on both phenotypic diversity and genetic alterations. DNA sequencing studies of TC have revealed that most tumors harbor genetic aberrations affecting the mitogen-activated protein kinase (MAPK) or phosphatidylinositol 3-kinase (PI3K)/AKT signal transduction pathways [5]. These genetic alterations are predominantly point mutations in critical trigger genes of these pathways, such as BRAF, RAS or phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3CA). RET/PTC rearrangements are the other class of aberrations frequently observed in PTCs.
Pathogenesis of TC can also be associated with mutations of receptor tyrosine kinases (RTKs) and other less commonly reported pathways, including Wnt/β-catenin, FOXO3 and NF-ĸB [25,26].
Genetic dysfunctions of components of these pathways, accompanied by epigenetic modifications, lead to progression of normal thyroid follicular cells to PTC or FTC [27]. The so-called “early” molecular driver alterations, BRAF V600E or RAS, are the most common [28]. Co-occurrence of other mutations and/or rearrangements with increased signaling of the MAPK and/or PI3K/AKT pathways likely promotes progression to PDTC [27,29,30]. Further accumulation of epigenetic and genetic alterations, as well as infiltration of the thyroid gland by immune cells, seem to be the major triggers of ATC (Figure 1) [31].

1.4.1. MAPK Pathway

The MAPK pathway connects extracellular signals to intracellular transduction cascades that play a central role in regulation of cell growth. The major processes governed by the MAPK signaling chain include: proliferation, migration, apoptosis, cytoskeletal integrity, survival and cellular differentiation. In TCs, particularly in PTC, MAPK stimuli play a crucial role, as MAPK-activating mutations in genes encoding for both B-Raf proto-oncogene (BRAF) kinase and RAS kinase, or RET/PTC rearrangements, are frequently observed [32,33].
Substitution of valine with glutamic acid in codon 600 (V600E) of the BRAF gene encoding the BRAF serine/threonine kinase results from a thymine-to-adenine point mutation at position 1799 (T1799A). Oncogenic BRAF is the most frequently mutated gene in TCs and presence of the V600E alteration is reported in up to 80% of PTC cases, as well as 20–30% of ATCs [34,35,36]. The BRAF V600E-mutated kinase acts as the constitutive trigger of signal transmission in the MAPK pathway, independent of its upstream target, RAS, ultimately leading to increased activation of ERK kinases (Figure 2). Hyperactivation of signaling pathways, mediated by the BRAF V600E allele, causes impaired expression of sodium iodide symporter (NIS) and plays a key role in the radioiodine refractory (RAIR) phenomenon [37,38,39]. Some studies associate BRAF V600E with development of more aggressive and resistant forms of TCs [30,40]. The BRAF V600E allele is a useful prognostic marker determining tumor malignancy, especially in countries with a high prevalence of this genetic aberration [41,42].

1.4.2. PI3K/AKT Pathway

PI3K/AKT is another signaling pathway that plays major and diverse roles in regulation of cell growth, proliferation, apoptosis, metabolism, motility, angiogenesis and resistance to treatment. Genetic alterations affecting signal transduction in this pathway involve: (i) mutations in RAS isoform-encoding genes, (ii) mutations or amplifications in the alpha catalytic subunit of PIK3CA, (iii) mutations in the AKT gene and (iv) mutations in the phosphatase and tensin homolog phosphatase PTEN [28,43]. Genetic changes in RAS family genes are considered as “early” alterations and are characteristic for DTCs, especially for FTC. However, mutations of downstream effectors of the pathway are also commonly identified in less-differentiated TCs [30,44].
The RAS family of cell membrane-anchored GTPases (HRAS, NRAS and KRAS) activates downstream proteins (e.g., PI3K and BRAF kinases) when bound to guanosine triphosphate (GTP). Constitutively activated (mutated) GTP-bound RAS isoforms stimulate downstream effector pathways, even in the absence of extracellular stimuli (Figure 2) [45,46]. The utility of RAS mutations as a diagnostic tool is rather low [44]. RAS mutations occur, on average, in 30–40% of FTCs, 30–45% of the follicular variant of PTCs (FVPTCs), in approximately 25% of ATCs and PDTCs, in 15% of HCCs and in 10–45% of MTCs. Rare cases of mutations in the classical variant of PTC have also been reported [47,48,49,50].
PTEN is a tumor suppressor that decreases the cellular levels of phosphatidylinositol 3-phosphate (PIP3) to down-regulate the activity of the PI3K/AKT pathway. PTEN loss through gene deletion, mutations and epigenetic modifications results in dysregulation of the PI3K/AKT pathway, leading to uncontrolled cell proliferation, motility and protein synthesis. PTEN alterations have been described in FTC (14%), PDTC (4%), ATC (15%), PTC (2%) and HCC (5%) [4,5,50,51,52].
Activating (missense) mutations in PIK3CA or copy number gains lead to increased PIK3CA expression. Nevertheless, its role in carcinogenesis is still not well-defined. PIK3CA genetic alterations are common in de-differentiated TCs, like ATCs (18%), and less frequent in FTCs (1%) and PDTCs (2%) [53,54].
The AKT kinase family is comprised of three highly homologous isoforms: AKT1, AKT2 and AKT3. Activating mutations in genes encoding these protein kinases have been detected in FTCs, PDTCs and ATCs [53,55,56]. Murine TC model-based studies have shown that AKT1 is the primary promoter of TC development and local invasion, while vascular invasion and metastatic progression are dependent predominantly on AKT1 and AKT3, and to a lesser extent, on AKT2 [57,58].

1.4.3. Molecular Alterations in Receptor Tyrosine Kinases

RTKs are key regulators of cellular processes that are controlled by a variety of positive and negative feedback loops [59]. Various alterations may lead to their constant activation and triggering of signaling cascades, including MAPK and PI3K/AKT pathways, that might contribute to carcinogenesis [29,50]. The RTK family encompasses single-pass transmembrane proteins, such as RET, TRK, VEGFR and ALK. The most frequent and well-described genetic aberrations in RTK genes include rearrangements, duplications and point mutations in RET, ALK and NTRK [60].
RET is a transforming proto-oncogene. RET fusions or point mutations are potent oncogenic drivers in TCs. Up to a quarter of PTCs harbor RET fusions, where RET/PTC1 and RET/PTC3 rearrangements are most common. It has been reported that their prevalence is increased in patients previously exposed to high-dose radiation. RET fusions are also found in PDTCs and rarely in ATCs. In about 20–30% of all MTCs, germline mutations of the RET gene lead to hereditary MTC in the course of MEN2, whereas somatic RET mutations are mainly associated with sporadic MTC (approximately 50% of cases). The RET alteration determines distinct phenotypes of MTC that may differ in terms of age of disease onset and aggressiveness. For example, malignancies harboring the RET M918T somatic mutation that is the most frequent genetic change in sporadic MTCs, are related to a more aggressive MTC course and worse survival [61,62].
The anaplastic lymphoma kinase (ALK) is a transmembrane tyrosine kinase of the insulin receptor family that, upon ligand binding to its extracellular domain, promotes activation of multiple downstream signaling pathways, such as MAPK, PI3K/AKT and JAK/STAT. ALK gene activation, driven by mutations/rearrangements and gene fusions, is predominantly reported in PDTCs, ATCs and less frequently in PTCs and is linked with disease progression and aggressiveness [54,63,64,65].
Tropomyosin receptor kinases A, B and C (TRKA, TRKB and TRKC) are a group of receptor tyrosine kinases that play a crucial role in control and promotion of cell proliferation, survival and differentiation through the MAPK, PI3K/AKT and phospholipase C (PLC-γ) pathways. Activating DNA rearrangements that affect genes encoding TRKs (NTRK1, NTRK2 and NTRK3) as oncogenic drivers were reported in PTCs, PDTCs and ATCs [66,67,68,69]. Recently, determination of the NTRK activation status has gained great clinical utility since the emergence of targeted inhibitor therapy, as discussed below.

1.4.4. Other Molecular Alterations

PAX8-PPARγ, a nuclear transcription factor enhancing apoptosis, is the product of a gene fusion between paired box 8 (PAX8) and peroxisome proliferator activated receptor γ (PPARγ). It is suggested that the PAX8-PPARγ protein exerts a dominant negative effect on endogenous PPARγ and/or leads to the activation of subsets of PPARγ and PAX8 inducible genes. The fusion gene is detected in approximately 30% of FTCs and approximately 5% of FVPTCs, and rarely in PDTCs, HCCs and benign follicular adenoma [5,70,71,72].
TP53 is a suppressor gene controlling the cell cycle and apoptosis, and its mutations promote tumor development and progression. TP53 mutations are found in more than half of ATCs and in a small fraction of well-differentiated cancers [50,73,74].
The telomerase reverse transcriptase (TERT) gene encodes the catalytic subunit of telomerase, the enzyme responsible for adding tandem arrays of simple-sequence repeats to the ends of chromosomes and thereby preventing replicative senescence. Activating mutations in the promoter of TERT are believed to represent a “late” driver molecular event in the development of well-differentiated TCs. These genetic alterations are associated with tumor aggressiveness and poor outcomes in TC patients. The frequency of TERT mutations is 40–70% in ATCs, 40% in PDTCs, 32% in aggressive forms of HCCs, 20% in FTCs and 10% in PTCs [51,75,76]. It has been observed that enhanced activity of mutant TERT, which acts as an oncoprotein, accelerates further progression of cancer cells and tumor development, especially when BRAF V600E is already present [77]. Moreover, co-occurrence of TERT and BRAF mutations leads to failure of MAPK-targeted therapies [78].
The EIF1AX gene encodes for an eukaryotic translation initiation factor 1A (eIF1A), which is responsible for assembly of the ribosomal pre-initiation complex, mRNA binding, scanning and ribosomal subunit joining. Mutations in EIF1AX have been detected in 11% of PDCTs, 9% of ATCs and 1–2% of PTCs [5,79]. It seems that different EIF1AX mutations correspond to distinct phenotypes in thyroid cancer and, when they co-occur with RAS in advanced TCs, are related to higher tumor aggressiveness [80,81].
Aberrant activation of the Wnt/β-catenin signaling pathway, commonly observed during PTC initiation and progression, contributes to therapeutic resistance in cancer cells via increased expression of resistance-related genes. Among these targets of the Wnt/β-catenin signaling pathway, proteins functioning as extrusion pumps are found, such as P-glycoprotein encoded by the ABCB1 gene [82,83]. Upregulation of the pathway confers resistance to vemurafenib, a MEK inhibitor [83]. Mutations of components of the Wnt/β-catenin signaling cascade, like β-catenin (CTNNB1), adenomatous polyposis coli (APC) or axis inhibition protein 1 (AXIN1), are common in less-differentiated TCs, i.e., in 60–65% of ATCs and 25% of PDTCs [82,84].
Functionally, isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) play a key role within the Krebs cycle and produce α-ketoglutarate by catalyzing the oxidative decarboxylation of isocitrate. Mutations in IDH1-encoding genes are present in 16% of TCs, in particular in ATCs (11%) and FTCs (5%), whereas IDH2 mutations have been identified in only 3% of ATCs [85,86,87]. Unlike in other tumors, IDH1 and BRAF (or RAS) mutations in TCs are not mutually exclusive. Until now, no association between the IDH mutational status and clinical characteristics has been reported [86].

2. Current Diagnostic and Treatment Strategies

Diagnosis of TC is required when a patient presents symptoms, like palpable thyroid lesions, enlargement of the thyroid gland (especially in a short period of time), exposure to ionizing factors in childhood or a history of thyroid disease (adenoma, goiter or thyroiditis) [12,88].
In most TC incidents it is advised to perform surgical thyroidectomy and dissection of local metastases, if present, in the shortest possible time from diagnosis. After surgery, the extent of spread of cancer is assessed using the TNM Classification of Malignant Tumors. Recommendations of surgical intervention depend on the TC type and are discussed in the next subsection. Additionally, based on the TC type, various adjuvant therapies can be applied. For ATC and MTC, complementary chemotherapy and/or radiotherapy can be performed, as well as new targeted therapies, which strictly rely on the type of molecular alterations. In patients with DTC and PDTC, the recommended adjuvant therapy is radioactive iodine/radioiodine (RAI) treatment [12,13,88].

2.1. Diagnosis of TC: Ultrasonography (USG) and Fine-Needle Aspiration Biopsy (FNAB)

Palpation is the first line examination of the thyroid gland, while the best tool for the diagnosis of thyroid disorders is ultrasonography examination. The frequency of diagnosed nodules in palpation is 2–6%, while USG can detect thyroid nodules even in 19–68% of randomly selected individuals [89,90]. Thyroid nodules are found in 8–65% of autopsy specimens [91]. Among the nodules detected by palpation or USG, approximately 5–15% are malignant [92,93].
USG scan is recommended as the first-line thyroid imagining technique, preceding computer tomography (CT), magnetic resonance imaging (MRI) and others. USG enables identification of features, such as shape (taller than wide), echogenity, echostructure, margin and microcalcifications, that aid differentiation between benign and malignant thyroid nodules [94]. Nevertheless, there is a need to create a pattern of thyroid cancer USG images that will enable better TC risk stratification and decrease the rate of fine-needle aspiration biopsy.
Since 2009, when Horvath published the Thyroid Imaging Reporting and Data System (TIRADS) [95], other classifications of thyroid nodules have been published, including the ATA guideline in 2015 [20], the Korean Thyroid Association (K-TIRADS) guideline in 2016 [96], the European Thyroid Association (EU-TIRADS) guideline in 2017 [97], the American College of Radiology (ACR-TIRADS) guideline in 2017 and the Chinese (C-TIRADS) guideline in 2020 [98].
Indication to FNAB is made based on the clinical and USG examination. The material obtained during FNAB is analyzed and the risk of malignancy is estimated using The Bethesda System for Reporting Thyroid Cytopathology (TBSRTC) [99]. Also, depending on the result of FNAB, the decision of surgical thyroid excision is made.

2.2. Surgical Treatment of TC

Although DTC patients have a more favorable prognosis than most other cancers, with an overall survival (OS) rate of 98.3% at 5 years for the majority of cases, local recurrence occurs in about 20% of cases and distant metastases are found in approximately 10% of patients [100]. PDTC has a higher risk of persistence and mortality [101,102]. According to recent guidelines [20], surgical treatment is the gold standard in TC therapy. Before cervical surgery, a detailed USG of the neck with evaluation of lymph nodes should be performed. If any suspicious lymph nodes are recognized, they should be removed simultaneously with the gland. The range of surgery depends on the type of TC and on the stratification of recurrence risk. DTC patients with low risk of recurrence can be treated with total thyroidectomy or lobectomy with isthmusectomy. DTC and PDTC patients with intermediate or high risk of recurrent disease undergo total thyroidectomy and therapeutic neck dissection with or without prophylactic central neck dissection. Patients with intermediate or high risk of recurrence or with distant metastases should be treated with radioiodine [13].
ATC is an undifferentiated subtype of TCs that is resistant to therapy with the iodine-131 radioisotope. Due to an inauspicious prognosis, TNM classification for ATC only provides stage IV, which is subdivided into three classes: IVA, IVB and IVC. Surgery, followed by chemotherapy and radiotherapy, is recommended as the first choice of intervention for management of ATC, provided that a full resection of the tumor can be obtained and no distant metastases are identified (stage IVA). Patients with unresectable ATCs or with local invasion and small-volume metastases confirmed in the neck (IVB) should be treated with radiation therapy and adjuvant chemotherapy, without surgical intervention. Furthermore, systemic therapies with or without palliative radiotherapy should be considered for patients with large-volume or distant metastases. Recent ATA Guidelines for Management of Patients with ATC, which were released in March 2021, incorporate targeted therapies with kinase inhibitors in their recommendations [103], as discussed below.
Patients with MTC have poor prognosis, with a 10-year survival rate at approximately 50% and distant metastases observed in 7–23% of cases at the moment of recognition. The first-choice treatment is surgical thyroidectomy and central compartment neck dissection, but it is curative only for localized MTCs. In case of widespread regional or metastatic disease, surgical interventions are not associated with a higher cure rate, therefore, less aggressive local treatment procedures should be preferred. Systemic therapy is employed in multi-metastatic and rapidly progressing malignancies [61,104,105].

2.3. RAI Treatment

The thyroid gland has the unique ability to uptake and concentrate iodide, which is a critical step in biosynthesis of TH. Iodide is actively transported from the blood through the basolateral plasma membrane by the NIS protein, whereas the anion transporter pendrin and other proteins mediate iodide efflux to the follicular lumen. Thyroid peroxidase (TPO) attaches oxidized iodide to tyrosine residues of thyroglobulin (Tg) producing monoiodotyrosine (MIT) and diiodotyrosine (DIT). Subsequently, TPO couples a MIT and a DIT residue to form triiodothyronine (T3) or two DIT residues to form thyroxine (T4) [1,106].
The ability to uptake the iodine-131 radioisotope is preserved in most cases of DTC, therefore RAI has become a basic treatment modality in DTC. The accumulated radioiodine predominantly emits β-particles, which penetrate a short distance of only a few millimeters causing death of cancer cells. RAI is recommended as remnant ablation for patients with intermediate-risk DTC and as an adjuvant therapy for patients with high-risk DTC. For patients with low-risk DTC, who have undergone less than total thyroidectomy, RAI is not recommended [13,20,107].
De-differentiated TCs, like ATC and some PDTCs, lose their functional ability to transport and accumulate iodine due to several alterations. The same phenomenon occurs in 5–15% of DTC incidents [108], while approximately 50% of metastatic DTCs are refractory to RAI treatment [109,110].
DTCs that are metastatic or locally advanced and have one or more metastatic sites without any radioiodine uptake, or for which significant progression of the disease is observed during the year after radioiodine treatment, are considered as RAIR. RAIR-DTC significantly reduces the mean life expectancy by 3–5 years [111].
The other molecular mechanism harming iodine uptake is an underpinning disfunction of NIS, which occurs predominantly in BRAF-mutated tissue. However, other aberrant activations of MAPK and PI3K/AKT pathways are also associated with this phenomenon. Much effort has been made to identify small molecule inhibitors that would be able to restore and enhance NIS protein expression and its physiological functions [112,113]. It seems that BRAF/MEK inhibitors are the most promising agents for selected DTCs, nevertheless, prospective multicenter trials are required to evaluate the efficacy and safety of iodine uptake restoration on a larger cohort of patients [37,39].

3. Currently Used and Investigated Targeted Therapies in TCs

Although most TC cases can be cured by combined surgical and RAI treatment, all ATCs and MTCs, as well as some DTC incidents, require further therapy. Standard chemotherapy, if recommended, is used in ATC treatment, whereas in DTC, only doxorubicin has been approved for RAIR-DTC patients. Further treatment of MTC relies on removal of metastases (e.g., external beam radiation therapy, EBRT) and no systemic chemotherapy is recommended. Due to numerous adverse events and lack of survival rate enhancement, standard chemotherapy is insufficient in treatment of TCs. Over the last decade, knowledge about the molecular basis of TC carcinogenesis has led to the development of kinase inhibition-based therapies, which allow for targeting of tumor cells and are thus considered as a novel tool for personalized medicine [43].
Kinase inhibitors (KIs) are small molecular compounds. Most of the Food and Drug Administration (FDA)-approved inhibitors for DTC, ATC and MTC competitively block the ATP-binding pocket of protein kinase(s), thereby inhibiting cell signaling and proliferation. First generation KIs, such as lenvatinib, sorafenib, vandetanib and cabozantinib, present a broad-spectrum of activity and are known as multi-kinase inhibitors (MKIs). Due to their low selectivity, MKIs have more off-target side effects leading to certain adverse events (AEs). The higher selectivity of the second generation of specific kinase inhibitors (SKIs) that target individual protein kinases, e.g., BRAF-specific inhibitors, makes those drugs more tolerable and less AEs-triggering [114,115,116].
Ten years have passed since the first targeted kinase inhibitor, vandetanib, was approved and registered for treatment of symptomatic or progressive MTCs. Since then, other MKIs, such as sorafenib, lenvatinib, vandetanib and cabozantinib, were approved for TC treatment by both the FDA and the European Medical Agency (EMA). Lenvatinib and sorafenib are used for treatment of advanced RAIR-DTC, while vandetanib and cabozantinib are administered in MTC therapy. Apart from the approved indications, MKIs are used in other subtypes of TCs with variable effect [28,114,115].
Larotrectinib, entrectinib and selpercatinib represent selective FDA- and EMA-approved inhibitors. Pralsetinib (BLU-667), a selective inhibitor targeting mutated and fused forms of RET, has been approved by the FDA and is under evaluation by the EMA [115]. Combinatory therapy using dabrafenib and trametinib, which are selective inhibitors of MEK and BRAF, respectively, has been approved by the FDA for ATC treatment in patients with locally advanced or metastasized disease [103]. Kinase inhibitors that have been approved for TC therapy are summarized in Table 1. The clinical and preclinical approaches from the last two years regarding the use of kinase inhibitors in the treatment of TCs are discussed below.

3.1. Targeted Therapies for DTC Treatment

Guidelines for DTC treatment that were developed in 2015 [20], include recommendations for KI usage in metastatic, rapidly progressive and symptomatic RAIR-DTC cases. A variety of novel agents and combinatory treatments are being tested as candidate clinical approaches in DTC and progressive metastatic RAIR-DTC (summarized in Table 2). Special attention is paid to inhibitors directed against BRAF, RET, MEK and TRK kinases, which will “re-sensitize” DTC and RAI-refractory tumors.

3.1.1. MKI-Based Therapies of DTC

Recent studies on the use of MKIs in RAIR-DTCs showed that treatment with lenvatinib or sorafenib results in an improvement of progression-free survival (PFS) [117], objective tumor response rate and OS. However, during treatment, dose modifications are required to reduce AEs [118]. A long-term evaluation of apatinib-based therapy in a phase II trial for progressive RAIR-DTC displayed sustainable efficacy and a tolerable safety profile, therefore, it can be considered as a promising treatment option [119]. The benefit from apatinib therapy may result from the effective inhibition of proliferation and migration of PTC cells, as well as from induction of apoptosis, cell cycle arrest and autophagy, as was recently demonstrated in an in vitro study [120]. Donafenib tested in a randomized, open-label, multicenter phase II trial of progressive locally advanced or metastatic RAIR-DTC, demonstrated improvement in PFS, partial response (PR) and the overall response rate (ORR). That effect was dose-dependent and effective for both used regimens, but a higher rate of AEs was reported in the higher dose group [121]. In a randomized, double-blind, placebo-controlled phase III trial, treatment with cabozantinib (an agent already approved for MTC therapy) prolonged PFS in RAIR-DTC patients, but at the same time accounted for a relatively high rate of AEs [122].
The results of Real-World Studies (RWS) showed that treatment with lenvatinib, sorafenib and pazopanib, followed or accompanied by another therapy, may improve the clinical outcome (i.e., higher PFS) of patients with RAIR-DTC [123,124,125]. However, the observed AEs, most frequently fatigue, asthenia and hypertension (HT), forced to reduce the MKI dose. Despite that, a dose reduction likely did not abrogate the apparent efficacy of therapy [126]. A recently published report, comparing sorafenib and lenvatinib for RAIR-DTC treatment, showed that in the group treated with lenvatinib, PR to treatment was significantly increased, and was accompanied by a drop in serum Tg levels [127]. Even though MKIs are considered as a promising treatment for RAIR-DTC, the use of them should be cautious due to a high frequency of AEs [128].
There is increasing interest in MKI-based therapies in combination with immunotherapy (e.g., NCT04560127, NCT03732495), MAPK- or PI3K/AKT-SKIs (e.g., NCT02143726, NCT01947023) or radioiodine (NCT04952493). In the recently reported preliminary studies on a small, randomized, multi-institutional phase II study (NCT02143726), where everolimus (mTOR inhibitor) and sorafenib co-therapy was applied for patients with RAI refractory HCC, improved PFS was observed [129]. Similarly, in a preclinical study, DTC cells treated with a combined therapy, including lenvatinib and radiation, synergistically induced apoptosis and G2/M phase arrest, as well as inhibited colony formation in vitro (cell lines) and tumor growth in nude mice [130].

3.1.2. Single Kinase-Targeted Therapies of DTC

Since MAPK activation, mediated by a BRAF single-nucleotide mutation, is one of the most critical events in DTC development, there are ongoing efforts to investigate BRAF-specific SKIs either in monotherapy or in combination with other agents. For example, a recent in vitro study performed on BRAF V600E-positive TC cell lines indicated antiproliferative and re-differentiative effects of dabrafenib and vemurafenib [131]. Moreover, vemurafenib-treated BRAF V600E-positive PTC cells exhibited an increased apoptosis level [132], whereas ascorbic acid (vitamin C) was found to sensitize BRAF V600E-positive thyroid cancer cells to this agent [133]. Recent reports suggest, that co-therapy based on dabrafenib and trametinib may lead to durable disease control and prolonged benefit in patients with a BRAF-mutated PTC [134]. This combination of drugs is currently under investigation in a panel of clinical trials, such as NCT03244956, NCT04619316 and NCT04554680 (Table 2). Simultaneous treatment with BRAF and MEK inhibitors upregulates NIS expression, suggesting it may improve RAI responsiveness [135]. According to preclinical data, this effect may be amplified when either dabrafenib or vemurafenib is used in combination with follistatin or vactosertib, SMAD pathway inhibitors [136]. A prospective, multicentric, open-label phase II trial (MERAIODE, NCT03244956) evaluating the efficacy and tolerance of trametinib and dabrafenib treatment followed by administration of RAI in patients with metastatic DTC provided promising results. The therapy restored radioiodine accumulation in BRAF-mutated patients and led to tumor control in 90% of cases [137].
In a phase I, open label study of lifirafenib (BGB-283), a RAF family kinase inhibitor targeting BRAF V600E, EGFR and RAS proteins, four patients with BRAF V600E-positive TCs were enrolled, including one in the dose-escalation phase and three in the dose-expansion phase. Lifirafenib demonstrated antitumor activity with PR and an acceptable risk benefit profile in BRAF V600E-positive solid tumors [138]. Recently, enrollment for a phase I/II trial for assessment of the safety and efficacy of lifirafenib in combination with a MEK inhibitor (mirdametinib) in patients with BRAF- and RAS-mutated tumors was commenced (NCT03905148).
The utility of therapies based on inhibitors specific for tropomyosin receptor kinases in NTRK gene fusion-positive solid tumors was also investigated. Entrectinib (RXDX-101), a potent inhibitor of TRKA, TRKB and TRKC, was reported to induce durable and clinically meaningful responses in patients with NTRK fusion-positive solid tumors [139]. A phase I study recruitment of patients with PTC (STARTRK-1, NCT02097810) was completed in June 2021 and a phase II trial (STARTRK-2, NCT02568267) is still recruiting. Another TRK inhibitor, larotrectinib, was found to restore radioiodine avidity in PTC-pediatric patients, leading to inhibition of tumor growth [140].
The common activating RET alterations in PTC cases encouraged researchers to use RET kinase inhibitors for PTC therapy. The results of selpercatinib treatment in two independent case reports of patients with RET-altered PTC (NCOA4-RET and CCDC6-RET fusions) showed restored radioiodine avidity and a re-differentiation effect suppressing Tg after six months of treatment [140,141].
A report on ALK-targeted therapy in RAIR-PTC patients harboring an EML4-ALK gene fusion variant 3 has just been released. In the experimental approach, the authors established a cell line derived from a PTC patient to select the most potent ALK-specific inhibitor in vitro. Tumor cell line data showed that lorlatinib (3rd generation ALK TKI) was a more potent drug than the previously administered crizotinib (1st generation ALK TKI). After lorlatinib therapy, the patient exhibited a significant decrease in Tg levels, a partial drop in PR, with a decrease in the sum of the target lesions [142].

3.2. Targeted Therapies for ATC Treatment

Recent ATA guidelines for management of ATC patients with grade IVB and IVC recommends mutation-guided individualized targeted therapeutic strategies based on KIs. Therefore, extended molecular profiling of ATC cases is strongly recommended, as it may reveal promising possibilities for target-specific therapies [143]. If it is possible, patient participation in clinical trials should be taken under consideration [103]. The list of on-going clinical trials for treatment of ATC is provided in Table 3.

3.2.1. MKI-Based Therapies of ATC

Over the last two years, a few studies have reported the results of MKI-based approaches in ATC treatment. Lenvatinib, an oral multitarget inhibitor, has been used in adjuvant therapy after thyroidectomy with neck dissection and postoperative chemoradiotherapy. Therapy extended the time to progression, but some AEs, like HT fatigue, anorexia and severe drug-induced hepatitis, forced investigators to taper the drug dose [144,145]. Results of treatment of ATC patients with lenvatinib (phase II trial) confirmed tumor size reduction (in more than half of evaluated patients), PR to treatment (n = 1) and >30% reduction in the total size of the target lesion. Nevertheless, lack of efficacy per prespecified criteria in interim analysis led to end the study at the enrolment stage. The authors suggested that lenvatinib, when used alone, may not be an effective treatment for ATC [146]. Another preclinical study demonstrated that combination of lenvatinib with the MEK inhibitor selumetinib (AZD6244) enhanced the antitumor effects of monotherapy in vitro and in an ATC mouse model. These effects may occur through the PI3K/AKT and MAPK signaling pathways [147]. Sorafenib, an oral MKI, exerts activity against ATC cells, especially in combination with other drugs. Simultaneous administration of sorafenib and an agent restoring molecular function of the p53 protein (CP-31398) was reported to decrease viability of ATC-derived cells (SW579) [148]. Another study showed that combination of N-hydroxy-7-(2-naphthylthio) heptanamide (HNHA), sorafenib and radiation was effective in inducing apoptosis and cell cycle arrest, leading to significant suppression of tumor growth in a mouse xenograft model and may be considered as a potential approach to ATC treatment [149].

3.2.2. Single Kinase-Targeted Therapies of ATC

Since AEs are a significant limitation of the currently available MKI-based therapies, identification of novel molecule inhibitors for ATC management is needed. CTOM-DHP is a promising SKI specifically inhibiting both MAPK and PI3K/AKT signaling pathways. Administration of this compound to the ATC-originated cell line and tumor xenograft mice model led to increased NIS promotor expression, RAI avidity (both I124 and I131 uptake) and cytotoxicity. Therefore, CTOM-DHP may be considered as a potent compound for restoring RAI avidity in ATC [112]. Furthermore, it is postulated that combinatory approaches, rather than single inhibitor-based therapies, may provide more profits for patients. As already mentioned above, co-administration of selective BRAF and MEK inhibitors (dabrafenib and trametinib, respectively) has received FDA approval for treatment of locally advanced and metastatic BRAF V600E-positive ATCs [103]. A retrospective study on such combinatory treatment showed prolonged survival of BRAF-mutant ATC patients [150]. Additionally, a recently released case report demonstrated a good rapid response in a patient treated with dabrafenib and trametinib who had already received other BRAF-targeted inhibitors [151]. Moreover, co-administration of BRAF-directed agents with other drugs, like check point inhibitors (pembrolizumab), may prolong survival of patients [152]. In in vitro studies, exposure of BRAF V600E-positive ATC cell lines to dabrafenib and melatonin showed synergistic inhibition of hTERT, and in consequence, cell arrest in the G1 phase and decreased cell viability [153]. Similarly, usage of another SKI, vemurafenib with a STAT3 pathway inhibitor on ATC-derived cell lines (sphere and monolayer) and mouse xenografts resulted in decreased viability and increased rate of cell apoptosis. These results indicate that inhibition of other pathways, like STAT3, could enhance the sensitivity of kinase inhibitors in ATC cells [154].

3.3. Targeted Therapies for MTC Treatment

Due to their origins, MTCs are unable to accumulate radioiodine, thus only surgical- and systemic-based treatments may be applicable in clinical practice. Most MTCs occur sporadically, but in approximately one-fifth of cases, they are familial and caused by germline mutations of the RET proto-oncogene. Somatic RET mutations are found in approximately half of patients with sporadic MTCs. RAS gene mutations, and less often ALK fusions, are alternative genetic events spotted in sporadic MTCs. Apart from these alterations of the proto-oncogene, overexpression of vascular endothelial growth factor (VEGF) receptors is often detected in the disease. The advent of targeted small-molecule KIs has revolutionized medical treatment of MTCs. As already mentioned, current guidelines on management of MTC patients recommend cabozantinib and vandetanib as the first-line single-agent systemic therapy in patients with advanced progressive MTC, based on their documented ability to improve PFS (Table 1). Both drugs inhibit RET kinase activity to some extent, however, their major anticancer effect is due to their strong inhibition of angiogenesis [13,61,105,155]. The list of on-going clinical trials for treatment of MTC is provided in Table 4.

3.3.1. MKI-Based Therapies of MTC

In 2021, the results of a comprehensive retrospective study, which aimed to identify whether approval of MKIs for MTC is associated with changes in systemic therapy administration or changes in overall survival, was published [156]. The clinicopathologic comparisons were conducted between pre-multikinase (2005–2010) and post-multikinase inhibitor (2011–2016) approval groups. A total of 2891 patients were enrolled, including 1265 cases in the pre-MKI and 1626 cases in the post-MKI approval group. The results showed that after approval of MKIs, the rate of systemic treatment administration significantly increased (8.3% and 11.3% in pre-MKI and post-MKI, respectively). Nevertheless, no improvements in OS were detected between those two groups of patients. In 2020, a post hoc analysis phase III trial (ZETA trial) involving patients with advanced MTCs treated with vandetanib demonstrated increased PFS and improved ORR compared to a placebo; however, OS did not significantly differ between vandetanib- and placebo-treated patients. In contrast, time to worsening of pain, which was predefined as an endpoint to assess symptomatic benefits of the trial, was found to be extended in the vandetanib group [157]. According to recent reports [158,159], one in four patients treated with vandetanib or cabozantinib developed AEs, which required withdrawal of the drug, or exhibited drug resistance. Therefore, extensive efforts are made to broaden the panel of MKI-based therapies for MTC patients. For example, Matrone et al. (2021) evaluated the impact of the off-label use of lenvatinib in a group of ten patients affected by locally advanced, non-resectable, metastatic MTCs with previous failure of other TKIs [160]. Lenvatinib induced substantial stabilization of metastatic lesions and disease control. The observed AEs were consistent with other studies and could be managed by personalization of the initial dose and one or more prompt dose reductions. It seems that this agent could be effective as salvage therapy in cases, where no other treatment strategies are available. Moreover, due to the absence of cross-resistance between vandetanib and lenvatinib, second- and third-line kinase inhibitor treatment should always be considered [160]. A multicenter, randomized, double-blind, placebo-controlled phase IIB study investigated the usefulness of a novel MKI, anlotinib, in the therapy of locally advanced or metastatic MTCs. Drug treatment, compared to placebo, significantly improved the median PFS and ORR, with no significant difference in OS between the tested groups [161].

3.3.2. Single Kinase-Targeted Therapies of MTC

Since the release of the MTC management guidelines [105] in 2020, the FDA has approved two new, highly potent RET-selective tyrosine kinase inhibitors, pralsetinib and selpercatinib. Both agents are destined for individuals 12 years or older with advanced, metastatic RET-mutant MTCs or other RET fusion-positive TCs, which require systemic therapy. Selpercatinib has also been approved by the EMA for patients aged ≥ 12 years, who were previously treated with cabozantinib and/or vandetanib, while pralsetinib is still awaiting a positive opinion. Pralsetinib and selpercatinib recommendations were based on multicenter, open label, multi-cohort clinical trials, ARROW, NCT03037385 [162] and LIBRETTO-001, NCT03157128, respectively, that investigated their efficacy in MTC patients with RET gene alterations. These potent inhibitors not only improved ORR, but also higher PFS and OS, with a lower rate of AEs was observed [162,163]. Unfortunately, in a number of cases, there were no improvements after treatment and resistance-associated mutations were identified [62,163,164]. Several recent studies have shown that both pralsetinib and selpercatinib are inefficient in MTC patients with RET non-gate mutations at the front and at the hinge of the receptor. However, strong pralsetinib-resistant L730V/I mutations, located at the roof of the solvent front of the RET ATP-binding site, remained sensitive to selpercatinib in the preclinical study [165]. These findings highlight the need to develop a next-generation of drugs covering both gatekeeper and non-gatekeeper mutations for on-target resistance, in addition to deciphering patterns of off-target resistance by alternative mechanisms for combinatory therapies [62,164].

4. Treatment-Related Toxicities

MKI-related toxicities (listed in Table 5) are very common and likely reflect target-binding affinities specific to each drug [62,166,167,168,169]. Among the AEs observed during treatment, HT appears to be the most frequent and is managed using standard antihypertensive drugs. Early diagnosis of HT may help avoid serious complications and prevent premature termination of MKI-based therapies in patients [38,168]. Administration of MKIs is also frequently associated with an increased risk of palmar-plantar erythrodysesthesia syndrome (PPES), characterized by tingling and tenderness, with more serious symptoms including symmetrical redness, swelling and pain on the palms and soles. The etiology of PPES is still unclear and its management is predominantly symptomatic with dose reduction or interruption for severe cases. Other reported muco-cutaneous AEs, such as rash, alopecia and oral stomatitis, are specifically related to inhibition of the VEGFR/EGFR pathway [162,168]. Proteinuria is a common renal side effect of antiangiogenic MKIs [166]. All MKI treatments evaluated in TCs are related to increased risk of gastrointestinal toxicities, including nausea, vomiting, diarrhoea, mucositis, weight loss and hepatic impairment [122,166]. Fatigue, also defined as asthenia, can arise as a common AE during treatment with MKIs [168]. Recently, Monti et al. (2021) suggested that adrenal insufficiency may be responsible for lenvatinib-associated fatigue (especially in patients experiencing extreme fatigue) [170]. Preliminary findings suggest that early diagnosis of primary adrenal insufficiency is important, since cortisone acetate replacement therapy can improve fatigue in patients without the need of dose reduction of MKIs [170]. Thyroid function and thyroid hormone metabolism are the most commonly reported endocrine toxicities caused by MKIs, especially those targeting VEGFR [171]. The majority of severe side effects occur early in the course of treatment, and if managed, patients may experience persistent long-term disease control [172]. Side effects occur more frequently in older patients and may differ according to the patient’s ethnicity [169].
The most frequent AEs in BRAF- and MEK-targeted therapies are fatigue, fever, diarrhoea, HT and hyperproliferative cutaneous events [38,173]. Most are grade 1 and 2 and are consistent with those reported for BRAF or MEK inhibitors alone [173]. The incidence of AEs in combinatory therapy with dabrafenib and trametinib is highest during the initial six months of treatment and declines thereafter [173]. These findings emphasize the need for proper management of AEs during early treatment, similarly to MKI-based therapies, to avoid poor adherence and early discontinuation from the treatment [166,167,173]. RET-targeted therapies, based on selpercatinib and pralsetinib, seem to exhibit lower side effects than other FDA-approved treatments (Table 5). The majority of patients tolerate these drugs well, while only a few percent of patients discontinue the therapy due to treatment-related AEs [62,162,163]. The most commonly observed AEs of selpercatinib are HT, diarrhoea, fatigue and dry mouth [163], whereas pralsetinib administration is associated with increased risk of neutropenia and liver impairment as well as decreased white blood cells (WBCs) count [162]. Overall, comprehensive education of patients to increase their awareness of signs and symptoms of possible side effects and continued monitoring is critical.

5. Conclusions and Future Perspectives

Novel tyrosine kinase inhibitor-based approaches have been studied, alone or in combination, to improve the inauspicious prognosis of TC patients. The results of recent studies highlight new treatment opportunities for RAIR-DTC, ATC and MTC patients. Prolonged progression-free and overall survival are possible with targeted therapeutic agents. Prospectively, information on molecular aberrations will be more important than the histological type of TC and will be crucial for clinical decision-making.
Implementation of kinase inhibitors promotes approaches, which are related to the individual needs of patients. Such personalized, low-risk therapies, in which the administrated selective KIs target certain specific aberrations, are possible due to the increasing availability of advanced molecular tests that allow for rapid mutational analysis of TCs. Moreover, therapy of TCs will be based upon selection of chemotherapeutics previously used in other cancer types, while the aim of treatment will focus on the point of action, i.e., place and type of mutation.
Despite remarkable progress, there continue to be two broad categories of barriers in clinical usage of targeted therapeutics in TCs, i.e., limiting adverse effects and development of resistance. Targeted agents usually have less severe and long-term AEs than conventional chemotherapy. However, they should be considered as chronic approaches, and appropriately managed to prolong treatment duration in patients exhibiting a clinical benefit [38]. The aquired drug-resistance phenomenon, which results from systemic therapies, remains a concern. For example, up to half of patients treated with BRAF-targeted SKIs eventually develop resistance. It is typically mediated through reactivation of the MAPK pathway and can occur through several mechanisms, including upstream activating mutations (e.g., mutations in RAS-encoding genes), downstream MAPK pathway alterations (e.g., MEK and ERK mutations), activation of parallel signaling pathways (e.g., PI3K/AKT), increase in the expression level of RTKs (e.g., EGFR) and BRAF gene amplification and alternative splicing. The failure of targeted therapies could also result from overexpression of ATP binding cassette (ABC) drug efflux pumps [173,174]. Recent studies demonstrated that overexpression of ABCB1 is sufficient to confer resistance of cancer cells to kinase inhibitors [175,176]. The phenomenon of multidrug resistance requires use of combinatory therapies that are composed of drugs with differing molecular targets, mechanisms of action and various signaling pathways. Thus, both non-selective and selective kinase inhibitors, as well as immunomodulatory drugs, check-point inhibitors and agents that influence other signaling pathways will be implemented in management and therapy of TCs.

Author Contributions

Conceptualization, M.R., D.G. and M.G.; writing—original draft preparation, M.R.; writing—review and editing, D.G. and M.G.; visualization, M.R.; supervision, D.G. and M.G.; funding acquisition, M.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by the National Science Centre, grant numbers 2018/29/B/NZ3/02642 and 2017/01/X/NZ3/00045.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AEsadverse events
ALKanaplastic lymphoma kinase
ASTaspartate aminotransferase
ATAAmerican Thyroid Association
ATCanaplastic thyroid cancer
BMIbody mass index
BRAFB-Raf proto-oncogene
CTcomputer tomography
CTLA-4cytotoxic T-lymphocyte antigen-4
DeTCdedifferentiated thyroid cancer
DITdiiodotyrosine
DTCdifferentiated thyroid cancer
EBRTexternal beam radiation therapy
eIF1Aeukaryotic translation initiation factor 1A
EMAEuropean Medical Agency
FAPfamilial adenomatous polyposis
FDAFood and Drug Administration
18FDG18F-fluoro-2-deoxy-D-glucose
FNABfine-needle aspiration biopsy
FTCfollicular thyroid carcinoma
FVPTCfollicular variant of PTC
GTPguanosine triphosphate
HCCHürtle cell carcinoma
HNHAN-hydroxy-7-(2-naphthylthio) heptanamide
HThypertension
IDH1isocitrate dehydrogenase 1
IDH2isocitrate dehydrogenase 2
IMRTintensity-modulated radiation therapy
KIskinase inhibitors
MAPKmitogen-activated protein kinase
MENmultiple endocrine neoplasia
MEN2multiple endocrine neoplasia type 2
MITmonoiodotyrosine
MKIsmulti-kinase inhibitors
MRImagnetic resonance imaging
MTCmedullary thyroid cancer
NDnot defined
NISsodium iodide symporter
ORRoverall response rate
OSoverall survival
PAX8paired box 8
PDprogressive disease
PDCTpoorly differentiated thyroid carcinoma
PETpositron emission tomography
PFSprogression-free survival
PI3Kphosphatidylinositol 3-kinase
PIK3CAphosphatidylinositol-4,5-bisphosphate 3-kinase
PLC-γphospholipase C
PPARγperoxisome proliferator activated receptor γ
PPESpalmar-plantar erythrodysesthesia syndrome
PRpartial response
PTCpapillary thyroid carcinoma
RAIradioactive iodine; radioiodine
RAIRradioactive iodine refractory; radioiodine refractory
RANKLreceptor activator of nuclear factor κ-Β ligand
RETfRET fusion-positive
RETmRET mutation-positive
RTKreceptor tyrosine kinase
RWSReal-Word Studies
SDstable disease
SKIsspecific kinase inhibitors
T3triiodothyronine
T4thyroxine
TBSRTCThe Bethesda System for Reporting Thyroid Cytopathology
TCthyroid cancer
TCGAThe Cancer Genome Atlas
TERTtelomerase reverse transcriptase
Tgthyroglobulin
THsthyroid hormones
TPOthyroid peroxidase
TRKtropomyosin receptor kinase
TSHthyroid-stimulating hormone
USGultrasonography
WBCwhite blood cell
WHOWorld Health Organization
wtwild type

References

  1. Godlewska, M.; Banga, P.J. Thyroid peroxidase as a dual active site enzyme: Focus on biosynthesis, hormonogenesis and thyroid disorders of autoimmunity and cancer. Biochimie 2019, 160, 34–45. [Google Scholar] [CrossRef]
  2. Ceolin, L.; Duval, M.A.D.S.; 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] [Green Version]
  3. WHO. WHO Classification of Tumours of Endocrine Organs, 4th ed.; Lloyd, R., Osamura, R., Klèoppel, G., Rosai, J., Eds.; International Agency for Research on Cancer: Lyon, France, 2017; Volume 10. [Google Scholar]
  4. Kure, S.; Ohashi, R. Thyroid Hürthle Cell Carcinoma: Clinical, Pathological, and Molecular Features. Cancers 2020, 13, 26. [Google Scholar] [CrossRef] [PubMed]
  5. Agrawal, N.; Akbani, R.; Aksoy, B.A.; Ally, A.; Arachchi, H.; Asa, S.; Auman, J.T.; Balasundaram, M.; Balu, S.; Baylin, S.B.; et al. Integrated Genomic Characterization of Papillary Thyroid Carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef] [Green Version]
  6. 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] [PubMed]
  7. 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] [PubMed]
  8. Pietrowska, M.; Diehl, H.C.; Mrukwa, G.; Kalinowska-Herok, M.; Gawin, M.; Chekan, M.; Elm, J.; Drazek, G.; Krawczyk, A.; Lange, D.; et al. Molecular profiles of thyroid cancer subtypes: Classification based on features of tissue revealed by mass spectrometry imaging. Biochim. Biophys. Acta (BBA)-Proteins Proteomic 2017, 1865, 837–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  10. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics. CA A Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  11. Clayman, G.L. Medullary Thyroid Cancer Genetics. Available online: https://www.thyroidcancer.com/thyroid-cancer/medullary/genetics (accessed on 27 September 2021).
  12. Cabanillas, M.E.; McFadden, D.G.; Durante, C. Thyroid cancer. Lancet 2016, 388, 2783–2795. [Google Scholar] [CrossRef]
  13. Filetti, S.; Durante, C.; Hartl, D.; Leboulleux, S.; Locati, L.; Newbold, K.; Papotti, M.; Berruti, A. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 1856–1883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Moses, W.; Weng, J.; Khanafshar, E.; Duh, Q.-Y.; Clark, O.H.; Kebebew, E. Multiple Genetic Alterations in Papillary Thyroid Cancer are Associated with Younger Age at Presentation. J. Surg. Res. 2010, 160, 179–183. [Google Scholar] [CrossRef] [PubMed]
  15. Lee, J.-H.; Youn, S.; Jung, S.; Kim, K.; Chai, Y.J.; Chung, Y.S.; Park, W.S.; Lee, K.E.; Yi, K.H. A national database analysis for factors associated with thyroid cancer occurrence. Sci. Rep. 2020, 10, 1–7. [Google Scholar] [CrossRef]
  16. Miller, K.D.; Ortiz, A.P.; Pinheiro, P.S.; Bandi, P.; Minihan, A.; Fuchs, H.E.; Tyson, D.M.; Tortolero-Luna, G.; Fedewa, S.A.; Jemal, A.M.; et al. Cancer statistics for the US Hispanic/Latino population. CA Cancer J. Clin. 2021, 1–22. [Google Scholar] [CrossRef]
  17. LeClair, K.; Bell, K.J.L.; Furuya-Kanamori, L.; Doi, S.A.; Francis, D.O.; Davies, L. Evaluation of Gender Inequity in Thyroid Cancer Diagnosis. JAMA Intern. Med. 2021, 181, 1351. [Google Scholar] [CrossRef]
  18. Rahbari, R.; Zhang, L.; Kebebew, E. Thyroid cancer gender disparity. Futur. Oncol. 2010, 6, 1771–1779. [Google Scholar] [CrossRef] [Green Version]
  19. Peterson, E.; De, P.; Nuttall, R. BMI, Diet and Female Reproductive Factors as Risks for Thyroid Cancer: A Systematic Review. PLoS ONE 2012, 7, e29177. [Google Scholar] [CrossRef]
  20. 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] [Green Version]
  21. Liu, Y.; Su, L.; Xiao, H. Review of Factors Related to the Thyroid Cancer Epidemic. Int. J. Endocrinol. 2017, 2017, 1–9. [Google Scholar] [CrossRef] [Green Version]
  22. Tran, T.-V.-T.; Kitahara, C.M.; De Vathaire, F.; Boutron-Ruault, M.-C.; Journy, N. Thyroid dysfunction and cancer incidence: A systematic review and meta-analysis. Endocr. Relat. Cancer 2020, 27, 245–259. [Google Scholar] [CrossRef]
  23. Nagayama, Y. Thyroid Autoimmunity and Thyroid Cancer—The Pathogenic Connection: A 2018 Update. Horm. Metab. Res. 2018, 50, 922–931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Economides, A.; Giannakou, K.; Mamais, I.; Economides, P.A.; Papageorgis, P. Association Between Aggressive Clinicopathologic Features of Papillary Thyroid Carcinoma and Body Mass Index: A Systematic Review and Meta-Analysis. Front. Endocrinol. 2021, 12, 692879. [Google Scholar] [CrossRef]
  25. Jin, S.; Borkhuu, O.; Bao, W.; Yang, Y.-T. Signaling Pathways in Thyroid Cancer and Their Therapeutic Implications. J. Clin. Med. Res. 2016, 8, 284–296. [Google Scholar] [CrossRef] [Green Version]
  26. Zhao, C.; Mo, L.; Li, C.; Han, S.; Zhao, W.; Liu, L. FOXN3 suppresses the growth and invasion of papillary thyroid cancer through the inactivation of Wnt/β-catenin pathway. Mol. Cell. Endocrinol. 2020, 515, 110925. [Google Scholar] [CrossRef]
  27. Nikiforov, Y.E.; Nikiforova, M.N. Molecular genetics and diagnosis of thyroid cancer. Nat. Rev. Endocrinol. 2011, 7, 569–580. [Google Scholar] [CrossRef]
  28. Dralle, H.; Machens, A.; Basa, J.; Fatourechi, V.; Franceschi, S.; Hay, I.D.; Nikiforov, Y.E.; Pacini, F.; Pasieka, J.L.; Sherman, S.I. Follicular cell-derived thyroid cancer. Nat. Rev. Dis. Prim. 2015, 1, 15077. [Google Scholar] [CrossRef] [PubMed]
  29. Lorusso, L.; Cappagli, V.; Valerio, L.; Giani, C.; Viola, D.; Puleo, L.; Gambale, C.; Minaldi, E.; Campopiano, M.; Matrone, A.; et al. Thyroid Cancers: From Surgery to Current and Future Systemic Therapies through Their Molecular Identities. Int. J. Mol. Sci. 2021, 22, 3117. [Google Scholar] [CrossRef] [PubMed]
  30. Chmielik, E.; Rusinek, D.; Oczko-Wojciechowska, M.; Jarząb, M.; Krajewska, J.; Czarniecka, A.; Jarząb, B. Heterogeneity of Thyroid Cancer. Pathobiology 2018, 85, 117–129. [Google Scholar] [CrossRef]
  31. Riesco-Eizaguirre, G.; Santisteban, P. ENDOCRINE TUMOURS: Advances in the molecular pathogenesis of thyroid cancer: Lessons from the cancer genome. Eur. J. Endocrinol. 2016, 175, R203–R217. [Google Scholar] [CrossRef]
  32. Zaman, A.; Wu, W.; Bivona, T.G. Targeting Oncogenic BRAF: Past, Present, and Future. Cancers 2019, 11, 1197. [Google Scholar] [CrossRef] [Green Version]
  33. Nylén, C.; Mechera, R.; Maréchal-Ross, I.; Tsang, V.; Chou, A.; Gill, A.J.; Clifton-Bligh, R.J.; Robinson, B.G.; Sywak, M.S.; Sidhu, S.B.; et al. Molecular Markers Guiding Thyroid Cancer Management. Cancers 2020, 12, 2164. [Google Scholar] [CrossRef]
  34. Kim, S.W.; Lee, J.I.; Kim, J.-W.; Ki, C.-S.; Oh, Y.L.; Choi, Y.-L.; Shin, J.H.; Kim, H.K.; Jang, H.W.; Chung, J.H. BRAFV600E Mutation Analysis in Fine-Needle Aspiration Cytology Specimens for Evaluation of Thyroid Nodule: A Large Series in aBRAFV600E-Prevalent Population. J. Clin. Endocrinol. Metab. 2010, 95, 3693–3700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Rusinek, D.; Pfeifer, A.; Krajewska, J.; Oczko-Wojciechowska, M.; Handkiewicz-Junak, D.; Pawlaczek, A.; Zebracka-Gala, J.; Kowalska, M.; Cyplinska, R.; Zembala-Nozynska, E.; et al. Coexistence of TERT Promoter Mutations and the BRAF V600E Alteration and Its Impact on Histopathological Features of Papillary Thyroid Carcinoma in a Selected Series of Polish Patients. Int. J. Mol. Sci. 2018, 19, 2647. [Google Scholar] [CrossRef] [Green Version]
  36. Chen, T.; Lorch, J.H.; Wong, K.S.; Barletta, J.A. Histological features of BRAF V600E-mutant anaplastic thyroid carcinoma. Histopathology 2020, 77, 314–320. [Google Scholar] [CrossRef]
  37. Jhiang, S.M.; Konda, B.; Sipos, J.A.; Nabhan, F.A. Prospects for Redifferentiating Agents in the Use of Radioactive Iodine Therapy for Thyroid Cancer. Thyroid 2020, 30, 471–473. [Google Scholar] [CrossRef]
  38. Fullmer, T.; Cabanillas, M.E.; Zafereo, M. Novel Therapeutics in Radioactive Iodine-Resistant Thyroid Cancer. Front. Endocrinol. 2021, 12, 720723. [Google Scholar] [CrossRef]
  39. Lamartina, L.; Anizan, N.; Dupuy, C.; Leboulleux, S.; Schlumberger, M. Redifferentiation-facilitated radioiodine therapy in thyroid cancer. Endocr. Relat. Cancer 2021, 28, T179–T191. [Google Scholar] [CrossRef]
  40. Czarniecka, A.; Oczko-Wojciechowska, M.; Barczyński, M. BRAF V600E mutation in prognostication of papillary thyroid cancer (PTC) recurrence. Gland. Surg. 2016, 5, 495–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Oczko-Wojciechowska, M.; Kotecka-Blicharz, A.; Krajewska, J.; Rusinek, D.; Barczyński, M.; Jarząb, B.; Czarniecka, A. European perspective on the use of molecular tests in the diagnosis and therapy of thyroid neoplasms. Gland. Surg. 2020, 9, S69–S76. [Google Scholar] [CrossRef]
  42. Skubisz, K.; Januszkiewicz-Caulier, J.; Cybula, P.; Bakuła-Zalewska, E.; Goryca, K.; Paziewska, A.; Ambrożkiewicz, F.; Woliński, K.; Mikula, M.; Ostrowski, J.; et al. Higher EU-TIRADS-Score Correlated with BRAF V600E Positivity in the Early Stage of Papillary Thyroid Carcinoma. J. Clin. Med. 2021, 10, 2304. [Google Scholar] [CrossRef] [PubMed]
  43. Laha, D.; Nilubol, N.; Boufraqech, M. New Therapies for Advanced Thyroid Cancer. Front. Endocrinol. 2020, 11, 82. [Google Scholar] [CrossRef] [PubMed]
  44. Marotta, V.; Bifulco, M.; Vitale, M. Significance of RAS Mutations in Thyroid Benign Nodules and Non-Medullary Thyroid Cancer. Cancers 2021, 13, 3785. [Google Scholar] [CrossRef] [PubMed]
  45. Nussinov, R.; Zhang, M.; Maloney, R.; Jang, H. Ras isoform-specific expression, chromatin accessibility, and signaling. Biophys. Rev. 2021, 13, 489–505. [Google Scholar] [CrossRef]
  46. Tang, D.; Kroemer, G.; Kang, R. Oncogenic KRAS blockade therapy: Renewed enthusiasm and persistent challenges. Mol. Cancer 2021, 20, 1–24. [Google Scholar] [CrossRef]
  47. Ciampi, R.; Mian, C.; Fugazzola, L.; Cosci, B.; Romei, C.; Barollo, S.; Cirello, V.; Bottici, V.; Marconcini, G.; Rosa, P.M.; et al. Evidence of a Low Prevalence ofRASMutations in a Large Medullary Thyroid Cancer Series. Thyroid 2013, 23, 50–57. [Google Scholar] [CrossRef]
  48. Censi, S.; Cavedon, E.; Bertazza, L.; Galuppini, F.; Watutantrige-Fernando, S.; De Lazzari, P.; Nacamulli, D.; Pennelli, G.; Fassina, A.; Iacobone, M.; et al. Frequency and Significance of Ras, Tert Promoter, and Braf Mutations in Cytologically Indeterminate Thyroid Nodules: A Monocentric Case Series at a Tertiary-Level Endocrinology Unit. Front. Endocrinol. 2017, 8, 273. [Google Scholar] [CrossRef]
  49. Ciampi, R.; Romei, C.; Ramone, T.; Prete, A.; Tacito, A.; Cappagli, V.; Bottici, V.; Viola, D.; Torregrossa, L.; Ugolini, C.; et al. Genetic Landscape of Somatic Mutations in a Large Cohort of Sporadic Medullary Thyroid Carcinomas Studied by Next-Generation Targeted Sequencing. iScience 2019, 20, 324–336. [Google Scholar] [CrossRef] [Green Version]
  50. Tirrò, E.; Martorana, F.; Romano, C.; Vitale, S.R.; Motta, G.; Di Gregorio, S.; Massimino, M.; Pennisi, M.S.; Stella, S.; Puma, A.; et al. Molecular Alterations in Thyroid Cancer: From Bench to Clinical Practice. Genes 2019, 10, 709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. 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 Hürthle Cell Cancer Reveals Oncogenic Drivers, Recurrent Mitochondrial Mutations, and Unique Chromosomal Landscapes. Cancer Cell 2018, 34, 256–270.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Pozdeyev, N.; Gay, L.M.; Sokol, E.S.; Hartmaier, R.; Deaver, K.E.; Davis, S.; French, J.D.; Borre, P.V.; LaBarbera, D.V.; Tan, A.C.; et al. Genetic Analysis of 779 Advanced Differentiated and Anaplastic Thyroid Cancers. Clin. Cancer Res. 2018, 24, 3059–3068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Liu, Z.; Hou, P.; Ji, M.; Guan, H.; Studeman, K.; Jensen, K.; Vasko, V.; El-Naggar, A.K.; Xing, M. Highly Prevalent Genetic Alterations in Receptor Tyrosine Kinases and Phosphatidylinositol 3-Kinase/Akt and Mitogen-Activated Protein Kinase Pathways in Anaplastic and Follicular Thyroid Cancers. J. Clin. Endocrinol. Metab. 2008, 93, 3106–3116. [Google Scholar] [CrossRef] [Green Version]
  54. Murugan, A.K.; Xing, M. Anaplastic Thyroid Cancers Harbor Novel Oncogenic Mutations of the ALK Gene. Cancer Res. 2011, 71, 4403–4411. [Google Scholar] [CrossRef] [Green Version]
  55. Ricarte-Filho, J.C.; Ryder, M.; Chitale, D.; Rivera, M.; Heguy, A.; Ladanyi, M.; Janakiraman, M.; Solit, D.; Knauf, J.; Tuttle, R.M.; et al. Mutational Profile of Advanced Primary and Metastatic Radioactive Iodine-Refractory Thyroid Cancers Reveals Distinct Pathogenetic Roles for BRAF, PIK3CA, and AKT. Cancer Res. 2009, 69, 4885–4893. [Google Scholar] [CrossRef] [Green Version]
  56. Paulsson, J.O.; Rafati, N.; DiLorenzo, S.; Chen, Y.; Haglund, F.; Zedenius, J.; Juhlin, C.C. Whole-genome Sequencing of Follicular Thyroid Carcinomas Reveal Recurrent Mutations in MicroRNA Processing Subunit DGCR. J. Clin. Endocrinol. Metab. 2021, 106, 3265–3282. [Google Scholar] [CrossRef]
  57. Saji, M.; Narahara, K.; McCarty, S.K.; Vasko, V.V.; La Perle, K.M.; Porter, K.; Jarjoura, D.; Lu, C.; Cheng, S.-Y.; Ringel, M.D. Akt1 deficiency delays tumor progression, vascular invasion, and distant metastasis in a murine model of thyroid cancer. Oncogene 2011, 30, 4307–4315. [Google Scholar] [CrossRef] [PubMed]
  58. Saji, M.; Kim, C.S.; Wang, C.; Zhang, X.; Khanal, T.; Coombes, K.; La Perle, K.; Cheng, S.-Y.; Tsichlis, P.N.; Ringel, M.D. Akt isoform-specific effects on thyroid cancer development and progression in a murine thyroid cancer model. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
  59. Sabbah, M.; Najem, A.; Krayem, M.; Awada, A.; Journe, F.; Ghanem, G. RTK Inhibitors in Melanoma: From Bench to Bedside. Cancers 2021, 13, 1685. [Google Scholar] [CrossRef]
  60. Esteban-Villarrubia, J.; Soto-Castillo, J.J.; Pozas, J.; Román-Gil, M.S.; Orejana-Martín, I.; Torres-Jiménez, J.; Carrato, A.; Alonso-Gordoa, T.; Molina-Cerrillo, J. Tyrosine Kinase Receptors in Oncology. Int. J. Mol. Sci. 2020, 21, 8529. [Google Scholar] [CrossRef] [PubMed]
  61. Oczko-Wojciechowska, M.; Czarniecka, A.; Gawlik, T.; Jarzab, B.; Krajewska, J. Current status of the prognostic molecular markers in medullary thyroid carcinoma. Endocr. Connect. 2020, 9, R251–R263. [Google Scholar] [CrossRef]
  62. Thein, K.Z.; Velcheti, V.; Mooers, B.H.; Wu, J.; Subbiah, V. Precision therapy for RET-altered cancers with RET inhibitors. Trends Cancer 2021. [Google Scholar] [CrossRef]
  63. Pérot, G.; Soubeyran, I.; Ribeiro, A.; Bonhomme, B.; Savagner, F.; Boutet-Bouzamondo, N.; Hostein, I.; Bonichon, F.; Godbert, Y.; Chibon, F. Identification of a Recurrent STRN/ALK Fusion in Thyroid Carcinomas. PLoS ONE 2014, 9, e87170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Landa, I.; Ibrahimpasic, T.; Boucai, L.; Sinha, R.; Knauf, J.A.; Shah, R.; Dogan, S.; Ricarte-Filho, J.C.; Krishnamoorthy, G.P.; Xu, B.; et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J. Clin. Investig. 2016, 126, 1052–1066. [Google Scholar] [CrossRef] [Green Version]
  65. Yakushina, V.D.; Lerner, L.V.; Lavrov, A.V. Gene Fusions in Thyroid Cancer. Thyroid 2018, 28, 158–167. [Google Scholar] [CrossRef]
  66. Leeman-Neill, R.J.; Bs, L.M.K.; Liu, P.; Brenner, A.V.; Leeman-Neill, R.J.; 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] [PubMed] [Green Version]
  67. 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. NTRKfusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer 2016, 122, 1097–1107. [Google Scholar] [CrossRef] [Green Version]
  68. Marino, F.Z.; Pagliuca, F.; Ronchi, A.; Cozzolino, I.; Montella, M.; Berretta, M.; Errico, M.E.; Donofrio, V.; Bianco, R.; Franco, R. NTRK Fusions, from the Diagnostic Algorithm to Innovative Treatment in the Era of Precision Medicine. Int. J. Mol. Sci. 2020, 21, 3718. [Google Scholar] [CrossRef]
  69. Chu, Y.-H.; Dias-Santagata, D.; Farahani, A.A.; Boyraz, B.; Faquin, W.C.; Nosé, V.; Sadow, P.M. Clinicopathologic and molecular characterization of NTRK-rearranged thyroid carcinoma (NRTC). Mod. Pathol. 2020, 33, 2186–2197. [Google Scholar] [CrossRef]
  70. Armstrong, M.J.; Yang, H.; Yip, L.; Ohori, N.P.; McCoy, K.L.; Stang, M.T.; Hodak, S.P.; Nikiforova, M.N.; Carty, S.E.; Nikiforov, Y.E. PAX8/PPARγ Rearrangement in Thyroid Nodules Predicts Follicular-Pattern Carcinomas, in Particular the Encapsulated Follicular Variant of Papillary Carcinoma. Thyroid 2014, 24, 1369–1374. [Google Scholar] [CrossRef] [Green Version]
  71. Raman, P.; Koenig, R.J. Pax-8–PPAR-γ fusion protein in thyroid carcinoma. Nat. Rev. Endocrinol. 2014, 10, 616–623. [Google Scholar] [CrossRef]
  72. Giordano, T.J.; Haugen, B.R.; Sherman, S.I.; Shah, M.H.; Caoili, E.M.; Koenig, R.J. Pioglitazone Therapy of PAX8-PPARγ Fusion Protein Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2018, 103, 1277–1281. [Google Scholar] [CrossRef] [PubMed]
  73. Manzella, L.; Stella, S.; Pennisi, M.S.; Tirrò, E.; Massimino, M.; Romano, C.; Puma, A.; Tavarelli, M.; Vigneri, P. New Insights in Thyroid Cancer and p53 Family Proteins. Int. J. Mol. Sci. 2017, 18, 1325. [Google Scholar] [CrossRef] [PubMed]
  74. Nikitski, A.V.; Nikiforova, M.N.; Yip, L.; Karslioglu-French, E.; Carty, S.E.; Nikiforov, Y.E. Can TP53-mutant follicular adenoma be a precursor of anaplastic thyroid carcinoma? Endocr. Relat. Cancer 2021, 28, 621–630. [Google Scholar] [CrossRef]
  75. Liu, R.; Xing, M. TERT promoter mutations in thyroid cancer. Endocr. Relat. Cancer 2016, 23, R143–R155. [Google Scholar] [CrossRef] [Green Version]
  76. Rusinek, D.; Pfeifer, A.; Cieslicka, M.; Kowalska, M.; Pawlaczek, A.; Krajewska, J.; Szpak-Ulczok, S.; Tyszkiewicz, T.; Halczok, M.; Czarniecka, A.; et al. TERT Promoter Mutations and Their Impact on Gene Expression Profile in Papillary Thyroid Carcinoma. Cancers 2020, 12, 1597. [Google Scholar] [CrossRef]
  77. Liu, R.; Zhang, T.; Zhu, G.; Xing, M. Regulation of mutant TERT by BRAF V600E/MAP kinase pathway through FOS/GABP in human cancer. Nat. Commun. 2018, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  78. Tan, J.; Liu, R.; Zhu, G.; Umbricht, C.B.; Xing, M. TERTpromoter mutation determines apoptotic and therapeutic responses ofBRAF-mutant cancers to BRAF and MEK inhibitors: Achilles Heel. Proc. Natl. Acad. Sci. USA 2020, 117, 15846–15851. [Google Scholar] [CrossRef]
  79. Karunamurthy, A.; Panebianco, F.; Hsiao, S.J.; Vorhauer, J.; Nikiforova, M.N.; Chiosea, S.; Nikiforov, Y.E. Prevalence and phenotypic correlations of EIF1AX mutations in thyroid nodules. Endocr. Relat. Cancer 2016, 23, 295–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Simões-Pereira, J.; Moura, M.M.; Marques, I.J.; Rito, M.; Cabrera, R.A.; Leite, V.; Cavaco, B.M. The role of EIF1AX in thyroid cancer tumourigenesis and progression. J. Endocrinol. Investig. 2019, 42, 313–318. [Google Scholar] [CrossRef] [PubMed]
  81. Krishnamoorthy, G.P.; Davidson, N.R.; Leach, S.D.; Zhao, Z.; Lowe, S.W.; Lee, G.; Landa, I.; Nagarajah, J.; Saqcena, M.; Singh, K.; et al. EIF1AX and RAS Mutations Cooperate to Drive Thyroid Tumorigenesis through ATF4 and c-MYC. Cancer Discov. 2019, 9, 264–281. [Google Scholar] [CrossRef] [Green Version]
  82. Martin-Orozco, E.; Sanchez-Fernandez, A.; Ortiz-Parra, I.; Nicolas, M.A.-S. WNT Signaling in Tumors: The Way to Evade Drugs and Immunity. Front. Immunol. 2019, 10, 2854. [Google Scholar] [CrossRef]
  83. Wang, N.; Wen, J.; Ren, W.; Wu, Y.; Deng, C. Upregulation of TRIB2 by Wnt/β-catenin activation in BRAFV600E papillary thyroid carcinoma cells confers resistance to BRAF inhibitor vemurafenib. Cancer Chemother. Pharmacol. 2021, 88, 155–164. [Google Scholar] [CrossRef]
  84. Xing, M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat. Rev. Cancer 2013, 13, 184–199. [Google Scholar] [CrossRef]
  85. Murugan, A.K.; Bojdani, E.; Xing, M. Identification and functional characterization of isocitrate dehydrogenase 1 (IDH1) mutations in thyroid cancer. Biochem. Biophys. Res. Commun. 2010, 393, 555–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Hemerly, J.P.; Bastos, A.U.; Cerutti, J.M. Identification of several novel non-p.R132 IDH1 variants in thyroid carcinomas. Eur. J. Endocrinol. 2010, 163, 747–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Murugan, A.K.; Qasem, E.; Al-Hindi, H.; Alzahrani, A.S. Analysis of ALK, IDH1, IDH2 and MMP8 somatic mutations in differentiated thyroid cancers. Mol. Clin. Oncol. 2021, 15, 1–8. [Google Scholar] [CrossRef] [PubMed]
  88. Fagin, J.A.; Wells, S.A. Biologic and Clinical Perspectives on Thyroid Cancer. N. Engl. J. Med. 2016, 375, 1054–1067. [Google Scholar] [CrossRef] [Green Version]
  89. Guth, S.; Theune, U.; Aberle, J.; Galach, A.; Bamberger, C.M. Very high prevalence of thyroid nodules detected by high frequency (13 MHz) ultrasound examination. Eur. J. Clin. Investig. 2009, 39, 699–706. [Google Scholar] [CrossRef]
  90. Dean, D.S.; Gharib, H. Epidemiology of thyroid nodules. Best Pr. Res. Clin. Endocrinol. Metab. 2008, 22, 901–911. [Google Scholar] [CrossRef]
  91. Fresilli, D.; David, E.; Pacini, P.; Del Gaudio, G.; Dolcetti, V.; Lucarelli, G.; Di Leo, N.; Bellini, M.; D’Andrea, V.; Sorrenti, S.; et al. Thyroid Nodule Characterization: How to Assess the Malignancy Risk. Update of the Literature. Diagnostics 2021, 11, 1374. [Google Scholar] [CrossRef]
  92. Papini, E.; Guglielmi, R.; Bianchini, A.; Crescenzi, A.; Taccogna, S.; Nardi, F.; Panunzi, C.; Rinaldi, R.; Toscano, V.; Pacella, C.M. Risk of Malignancy in Nonpalpable Thyroid Nodules: Predictive Value of Ultrasound and Color-Doppler Features. J. Clin. Endocrinol. Metab. 2002, 87, 1941–1946. [Google Scholar] [CrossRef]
  93. Nam-Goong, I.S.; Kim, H.Y.; Gong, G.; Lee, H.K.; Shong, Y.K.; Kim, W.B. Ultrasonography-guided fine-needle aspiration of thyroid incidentaloma: Correlation with pathological findings. Clin. Endocrinol. 2004, 60, 21–28. [Google Scholar] [CrossRef]
  94. Moon, W.-J.; Jung, S.L.; Lee, J.H.; Na, D.G.; Baek, J.-H.; Lee, Y.H.; Kim, J.; Kim, H.S.; Byun, J.S.; Lee, D.H. Benign and Malignant Thyroid Nodules: US Differentiation—Multicenter Retrospective Study. Radiology 2008, 247, 762–770. [Google Scholar] [CrossRef]
  95. Horvath, E.; Majlis, S.; Rossi, R.; Franco, C.; Niedmann, J.P.; Castro, A.; Dominguez, M. An Ultrasonogram Reporting System for Thyroid Nodules Stratifying Cancer Risk for Clinical Management. J. Clin. Endocrinol. Metab. 2009, 94, 1748–1751. [Google Scholar] [CrossRef] [PubMed]
  96. Shin, J.H.; Baek, J.H.; Chung, J.; Ha, E.J.; Kim, J.-H.; Lee, Y.H.; Lim, H.K.; Moon, W.-J.; Na, D.G.; Park, J.S.; et al. Ultrasonography Diagnosis and Imaging-Based Management of Thyroid Nodules: Revised Korean Society of Thyroid Radiology Consensus Statement and Recommendations. Korean J. Radiol. 2016, 17, 370–395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Russ, G.; Bonnema, S.J.; Erdogan, M.F.; Durante, C.; Ngu, R.; Leenhardt, L. European Thyroid Association Guidelines for Ultrasound Malignancy Risk Stratification of Thyroid Nodules in Adults: The EU-TIRADS. Eur. Thyroid. J. 2017, 6, 225–237. [Google Scholar] [CrossRef] [Green Version]
  98. Zhou, J.; Yin, L.; Wei, X.; Zhang, S.; Song, Y.; Luo, B.; Li, J.; Qian, L.; Cui, L.; Chen, W.; et al. 2020 Chinese guidelines for ultrasound malignancy risk stratification of thyroid nodules: The C-TIRADS. Endocrine 2020, 70, 256–279. [Google Scholar] [CrossRef]
  99. Cibas, E.S.; Ali, S.Z. The 2017 Bethesda System for Reporting Thyroid Cytopathology. Thyroid 2017, 27, 1341–1346. [Google Scholar] [CrossRef] [PubMed]
  100. Eustatia-Rutten, C.F.A.; Corssmit, E.P.M.; Biermasz, N.R.; Pereira, A.M.; Romijn, J.A.; Smit, J.W. Survival and Death Causes in Differentiated Thyroid Carcinoma. J. Clin. Endocrinol. Metab. 2006, 91, 313–319. [Google Scholar] [CrossRef]
  101. Ibrahimpasic, T.; Ghossein, R.; Carlson, D.L.; Chernichenko, N.; Nixon, I.; Palmer, F.L.; Lee, N.Y.; Shaha, A.R.; Patel, S.G.; Tuttle, R.M.; et al. Poorly Differentiated Thyroid Carcinoma Presenting with Gross Extrathyroidal Extension: 1986–2009 Memorial Sloan-Kettering Cancer Center Experience. Thyroid 2013, 23, 997–1002. [Google Scholar] [CrossRef]
  102. Ho, A.S.; Luu, M.; Ba, L.B.; Balzer, B.L.; Bose, S.; Fan, X.; Walgama, E.; Clair, J.M.-S.; Alam Bs, U.; Shafqat, I.; et al. Prognostic Impact of Histologic Grade for Papillary Thyroid Carcinoma. Ann. Surg. Oncol. 2021, 28, 1731–1739. [Google Scholar] [CrossRef]
  103. 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]
  104. Schlumberger, M.; Bastholt, L.; Dralle, H.; Jarząb, B.; Pacini, F.; Smit, J.; European Thyroid Association Task Force. 2012 European Thyroid Association Guidelines for Metastatic Medullary Thyroid Cancer. Eur. Thyroid. J. 2012, 1, 5–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wells, S.A.; Asa, S.; Dralle, H.; Elisei, R.; Evans, D.B.; Gagel, R.F.; Lee, N.Y.; Machens, A.; Moley, J.F.; Pacini, F.; et al. Revised American Thyroid Association Guidelines for the Management of Medullary Thyroid Carcinoma. Thyroid 2015, 25, 567–610. [Google Scholar] [CrossRef] [PubMed]
  106. Godlewska, M.; Gawel, D.; Buckle, A.M.; Banga, J.P. Thyroid Peroxidase Revisited—What’s New? Horm. Metab. Res. 2019, 51, 765–769. [Google Scholar] [CrossRef] [Green Version]
  107. Mayson, S.E.; Chan, C.M.; Haugen, B.R. Tailoring the approach to radioactive iodine treatment in thyroid cancer. Endocr. Relat. Cancer 2021, 28, T125–T140. [Google Scholar] [CrossRef] [PubMed]
  108. Zarnegar, R.; Brunaud, L.; Kanauchi, H.; Wong, M.; Fung, M.; Ginzinger, D.; Duh, Q.-Y.; Clark, O.H. Increasing the effectiveness of radioactive iodine therapy in the treatment of thyroid cancer using Trichostatin A, a histone deacetylase inhibitor. Surgery 2002, 132, 984–990. [Google Scholar] [CrossRef] [PubMed]
  109. Worden, F. Treatment strategies for radioactive iodine-refractory differentiated thyroid cancer. Ther. Adv. Med Oncol. 2014, 6, 267–279. [Google Scholar] [CrossRef] [Green Version]
  110. Xing, M.; Haugen, B.R.; Schlumberger, M. Progress in molecular-based management of differentiated thyroid cancer. Lancet 2013, 381, 1058–1069. [Google Scholar] [CrossRef] [Green Version]
  111. Jin, Y.; Van Nostrand, D.; Cheng, L.; Liu, M.; Chen, L. Radioiodine refractory differentiated thyroid cancer. Crit. Rev. Oncol. 2018, 125, 111–120. [Google Scholar] [CrossRef] [PubMed]
  112. Oh, M.J.M.; Baek, S.H.; Gangadaran, P.; Hong, C.M.; Rajendran, R.L.; Lee, H.W.; Zhu, L.; Gopal, A.; Kalimuthu, S.; Jeong, S.Y.; et al. A Novel Tyrosine Kinase Inhibitor Can Augment Radioactive Iodine Uptake Through Endogenous Sodium/Iodide Symporter Expression in Anaplastic Thyroid Cancer. Thyroid 2020, 30, 501–518. [Google Scholar] [CrossRef]
  113. Riesco-Eizaguirre, G.; Santisteban, P.; De la Vieja, A. The complex regulation of NIS expression and activity in thyroid and extrathyroidal tissues. Endocr. Relat. Cancer 2021, 28, T141–T165. [Google Scholar] [CrossRef]
  114. Gild, M.L.; Tsang, V.H.M.; Clifton-Bligh, R.J.; Robinson, B.G. Multikinase inhibitors in thyroid cancer: Timing of targeted therapy. Nat. Rev. Endocrinol. 2021, 17, 225–234. [Google Scholar] [CrossRef]
  115. Ringel, M.D. New Horizons: Emerging Therapies and Targets in Thyroid Cancer. J. Clin. Endocrinol. Metab. 2021, 106, e382–e388. [Google Scholar] [CrossRef] [PubMed]
  116. Lubitz, C.C.; Sadow, P.M.; Daniels, G.H.; Wirth, L.J. Progress in Treating Advanced Thyroid Cancers in the Era of Targeted Therapy. Thyroid 2021, 31, 1451–1462. [Google Scholar] [CrossRef]
  117. Kiyota, N.; Schlumberger, M.; Muro, K.; Ando, Y.; Takahashi, S.; Kawai, Y.; Wirth, L.J.; Robinson, B.G.; Sherman, S.; Suzuki, T.; et al. Subgroup analysis of Japanese patients in a phase 3 study of lenvatinib in radioiodine-refractory differentiated thyroid cancer. Cancer Sci. 2015, 106, 1714–1721. [Google Scholar] [CrossRef] [Green Version]
  118. Fleeman, N.; Houten, R.; Bagust, A.; Richardson, M.; Beale, S.; Boland, A.; Dundar, Y.; Greenhalgh, J.; Hounsome, J.; Duarte, R.; et al. Lenvatinib and sorafenib for differentiated thyroid cancer after radioactive iodine: A systematic review and economic evaluation. Health Technol. Assess. 2020, 24, 1–180. [Google Scholar] [CrossRef]
  119. Lin, Y.-S.; Zhang, X.; Wang, C.; Liu, Y.-Q.; Guan, W.-M.; Liang, J. Long-Term Results of a Phase II Trial of Apatinib for Progressive Radioiodine Refractory Differentiated Thyroid Cancer. J. Clin. Endocrinol. Metab. 2021, 106, 3027. [Google Scholar] [CrossRef] [PubMed]
  120. Meng, X.; Wang, H.; Zhao, J.; Hu, L.; Zhi, J.; Wei, S.; Ruan, X.; Hou, X.; Li, D.; Zhang, J.; et al. Apatinib Inhibits Cell Proliferation and Induces Autophagy in Human Papillary Thyroid Carcinoma via the PI3K/Akt/mTOR Signaling Pathway. Front. Oncol. 2020, 10, 217. [Google Scholar] [CrossRef] [Green Version]
  121. Lin, Y.-S.; Yang, H.; Ding, Y.; Cheng, Y.-Z.; Shi, F.; Tan, J.; Deng, Z.-Y.; Chen, Z.-D.; Wang, R.-F.; Ji, Q.-H.; et al. Donafenib in Progressive Locally Advanced or Metastatic Radioactive Iodine-Refractory Differentiated Thyroid Cancer: Results of a Randomized, Multicenter Phase II Trial. Thyroid 2021, 31, 607–615. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. Kish, J.K.; Chatterjee, D.; Wan, Y.; Yu, H.-T.; Liassou, D.; Feinberg, B.A. Lenvatinib and Subsequent Therapy for Radioactive Iodine-Refractory Differentiated Thyroid Cancer: A Real-World Study of Clinical Effectiveness in the United States. Adv. Ther. 2020, 37, 2841–2852. [Google Scholar] [CrossRef] [PubMed]
  124. Takahashi, S.; Tahara, M.; Ito, K.; Tori, M.; Kiyota, N.; Yoshida, K.; Sakata, Y.; Yoshida, A. Safety and Effectiveness of Lenvatinib in 594 Patients with Unresectable Thyroid Cancer in an All-Case Post-Marketing Observational Study in Japan. Adv. Ther. 2020, 37, 3850–3862. [Google Scholar] [CrossRef]
  125. Koehler, V.F.; Berg, E.; Adam, P.; Weber, G.-L.; Pfestroff, A.; Luster, M.; Kutsch, J.M.; Lapa, C.; Sandner, B.; Rayes, N.; et al. Real world efficacy and safety of multi-tyrosine kinase inhibitors in radioiodine refractory thyroid cancer. Thyroid 2021, 31, 1531–1541. [Google Scholar] [CrossRef] [PubMed]
  126. Song, E.; Kim, M.; Kim, E.Y.; Kim, B.H.; Shin, D.Y.; Kang, H.-C.; Ahn, B.-C.; Kim, W.B.; Shong, Y.K.; Jeon, M.J.; et al. Lenvatinib for Radioactive Iodine-Refractory Differentiated Thyroid Carcinoma and Candidate Biomarkers Associated with Survival: A Multicenter Study in Korea. Thyroid 2020, 30, 732–738. [Google Scholar] [CrossRef]
  127. Ito, Y.; Onoda, N.; Kudo, T.; Masuoka, H.; Higashiyama, T.; Kihara, M.; Miya, A.; Miyauchi, A. Sorafenib and Lenvatinib Treatment for Metastasis/Recurrence of Radioactive Iodine-refractory Differentiated Thyroid Carcinoma. In Vivo 2021, 35, 1057–1064. [Google Scholar] [CrossRef] [PubMed]
  128. Feng, G.; Luo, Y.; Zhang, Q.; Zeng, F.; Xu, J.; Zhu, J. Sorafenib and radioiodine-refractory differentiated thyroid cancer (RR-DTC): A systematic review and meta-analysis. Endocrine 2020, 68, 56–63. [Google Scholar] [CrossRef]
  129. Sherman, E.J.; Foster, N.R.; Su, Y.B.; Shergill, A.; Ho, A.L.; Konda, B.; Ghossein, R.A.; Ganly, I.; Schwartz, G.K. Randomized phase II study of sorafenib with or without everolimus in patients with radioactive iodine refractory Hürthle cell thyroid cancer (HCC) (Alliance A091302/ ITOG 1706). J. Clin. Oncol. 2021, 39, 6076. [Google Scholar] [CrossRef]
  130. Suzuki, K.; Iwai, H.; Utsunomiya, K.; Kono, Y.; Kobayashi, Y.; Van Bui, D.; Sawada, S.; Yun, Y.; Mitani, A.; Kondo, N.; et al. Combination therapy with lenvatinib and radiation significantly inhibits thyroid cancer growth by uptake of tyrosine kinase inhibitor. Exp. Cell Res. 2021, 398, 112390. [Google Scholar] [CrossRef]
  131. Bonaldi, E.; Gargiuli, C.; De Cecco, L.; Micali, A.; Rizzetti, M.; Greco, A.; Borrello, M.; Minna, E. BRAF Inhibitors Induce Feedback Activation of RAS Pathway in Thyroid Cancer Cells. Int. J. Mol. Sci. 2021, 22, 5744. [Google Scholar] [CrossRef] [PubMed]
  132. Park, K.S.; Saindane, M.; Yang, E.Y.; Jin, T.; Rallabandi, H.R.; Heil, A.; Nam, S.E.; Yoo, Y.B.; Yang, J.-H.; Bin Kim, J.; et al. Selective inhibition of V600E-mutant BRAF gene induces apoptosis in thyroid carcinoma cell lines. Ann. Surg. Treat. Res. 2021, 100, 127–136. [Google Scholar] [CrossRef]
  133. Su, X.; Li, P.; Han, B.; Jia, H.; Liang, Q.; Wang, H.; Gu, M.; Cai, J.; Li, S.; Zhou, Y.; et al. Vitamin C sensitizes BRAFV600E thyroid cancer to PLX4032 via inhibiting the feedback activation of MAPK/ERK signal by PLX. J. Exp. Clin. Cancer Res. 2021, 40, 1–12. [Google Scholar] [CrossRef]
  134. Salama, A.K.S.; Li, S.; Macrae, E.R.; Park, J.-I.; Mitchell, E.P.; Zwiebel, J.A.; Chen, H.X.; Gray, R.J.; McShane, L.M.; Rubinstein, L.V.; et al. Dabrafenib and Trametinib in Patients With Tumors With BRAFV600E Mutations: Results of the NCI-MATCH Trial Subprotocol H. J. Clin. Oncol. 2020, 38, 3895–3904. [Google Scholar] [CrossRef]
  135. Ullmann, T.M.; Liang, H.; Moore, M.D.; Al-Jamed, I.; Gray, K.D.; Limberg, J.; Stefanova, D.; Buicko, J.L.; Finnerty, B.; Beninato, T.; et al. Dual inhibition of BRAF and MEK increases expression of sodium iodide symporter in patient-derived papillary thyroid cancer cells in vitro. Surgery 2020, 167, 56–63. [Google Scholar] [CrossRef] [PubMed]
  136. Luckett, K.A.; Cracchiolo, J.R.; Krishnamoorthy, G.P.; Leandro-Garcia, L.J.; Nagarajah, J.; Saqcena, M.; Lester, R.; Im, S.Y.; Zhao, Z.; Lowe, S.W.; et al. Co-inhibition of SMAD and MAPK signaling enhances 124I uptake in BRAF-mutant thyroid cancers. Endocr. Relat. Cancer 2021, 28, 391–402. [Google Scholar] [CrossRef]
  137. Leboulleux, S.; Cao, C.D.; Zerdoud, S.; Attard, M.; Bournaud, C.; Benisvy, D.; Taieb, D.; Bardet, S.; Terroir-Cassou-Mounat, M.; Betrian, S.; et al. MERAIODE: A Redifferentiation Phase II Trial With Trametinib and Dabrafenib Followed by Radioactive Iodine Administration for Metastatic Radioactive Iodine Refractory Differentiated Thyroid Cancer Patients with a BRAFV600E Mutation (NCT 03244956). J. Endocr. Soc. 2021, 5, A876. [Google Scholar] [CrossRef]
  138. Desai, J.; Gan, H.; Barrow, C.; Jameson, M.; Atkinson, V.; Haydon, A.; Millward, M.; Begbie, S.; Brown, M.; Markman, B.; et al. Phase I, Open-Label, Dose-Escalation/Dose-Expansion Study of Lifirafenib (BGB-283), an RAF Family Kinase Inhibitor, in Patients with Solid Tumors. J. Clin. Oncol. 2020, 38, 2140–2150. [Google Scholar] [CrossRef]
  139. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  140. Lee, Y.A.; Lee, H.; Im, S.-W.; Song, Y.S.; Oh, D.-Y.; Kang, H.J.; Won, J.-K.; Jung, K.C.; Kwon, D.; Chung, E.-J.; et al. NTRK and RET fusion–directed therapy in pediatric thyroid cancer yields a tumor response and radioiodine uptake. J. Clin. Investig. 2021, 131, 131. [Google Scholar] [CrossRef]
  141. Groussin, L.; Bessiene, L.; Arrondeau, J.; Garinet, S.; Cochand-Priollet, B.; Lupo, A.; Zerbit, J.; Clerc, J.; Huillard, O. Selpercatinib-Enhanced Radioiodine Uptake in RET-Rearranged Thyroid Cancer. Thyroid 2021, 31, 1603–1604. [Google Scholar] [CrossRef]
  142. Aydemirli, M.D.; van Eendenburg, J.D.; van Wezel, T.; Oosting, J.; Corver, W.; Kapiteijn, E.; Morreau, H. Targeting EML4-ALK gene fusion variant 3 in thyroid cancer. Endocr. Relat. Cancer 2021, 28, 377–389. [Google Scholar] [CrossRef]
  143. Moore, A.; Bar, Y.; Maurice-Dror, C.; Finkel, I.; Goldvaser, H.; Dudnik, E.; Goldstein, D.A.; Gordon, N.; Billan, S.; Gutfeld, O.; et al. Next-generation sequencing in thyroid cancers: Do targetable alterations lead to a therapeutic advantage? Medicine 2021, 100, e26388. [Google Scholar] [CrossRef] [PubMed]
  144. Hamamoto, T.; Kono, T.; Taruya, T.; Ishino, T.; Ueda, T.; Takeno, S. A long survival patient of anaplastic thyroid carcinoma treated with lenvatinib. Auris Nasus Larynx 2020. [Google Scholar] [CrossRef]
  145. Tanaka, A.; Uemura, H.; Masui, T.; Kanazawa, S.; Yoshii, Y.; Kanno, M.; Morita, G.; Obayashi, C.; Yamanaka, T.; Kitahara, T. Anaplastic thyroid cancer with long-term survival with lenvatinib therapy and preservation of laryngeal function after one-stage reconstruction: A case report. Mol. Clin. Oncol. 2021, 15, 1–5. [Google Scholar] [CrossRef]
  146. Wirth, L.J.; Brose, M.S.; Sherman, E.J.; Licitra, L.; Schlumberger, M.; Sherman, S.I.; Bible, K.C.; Robinson, B.; Rodien, P.; Godbert, Y.; et al. Open-Label, Single-Arm, Multicenter, Phase II Trial of Lenvatinib for the Treatment of Patients With Anaplastic Thyroid Cancer. J. Clin. Oncol. 2021, 39, JCO2003093. [Google Scholar] [CrossRef] [PubMed]
  147. Enomoto, K.; Hirayama, S.; Kumashiro, N.; Jing, X.; Kimura, T.; Tamagawa, S.; Matsuzaki, I.; Murata, S.-I.; Hotomi, M. Synergistic Effects of Lenvatinib (E7080) and MEK Inhibitors against Anaplastic Thyroid Cancer in Preclinical Models. Cancers 2021, 13, 862. [Google Scholar] [CrossRef]
  148. Wu, J.; Lin, C.; Huang, C.; Cheng, Y.; Chien, C.; Sung, Y. Potential synergistic effects of sorafenib and CP-31398 for treating anaplastic thyroid cancer with p53 mutations. Oncol. Lett. 2020, 19, 3021–3026. [Google Scholar] [CrossRef]
  149. Yun, H.J.; Kim, H.J.; Kim, J.; Kim, S.Y.; Chang, H.-S.; Park, C.S.; Chang, H.-J.; Park, K.C. Synergistic Anticancer Activity of N-Hydroxy-7-(2-Naphthylthio) Heptanomide, Sorafenib, and Radiation Therapy in Patient-Derived Anaplastic Thyroid Cancer Models. Int. J. Mol. Sci. 2021, 22, 536. [Google Scholar] [CrossRef]
  150. Park, J.; Jung, H.A.; Shim, J.H.; Park, W.-Y.; Kim, T.H.; Lee, S.-H.; Kim, S.W.; Ahn, M.-J.; Park, K.; Chung, J.H. Multimodal treatments and outcomes for anaplastic thyroid cancer before and after tyrosine kinase inhibitor therapy: A real-world experience. Eur. J. Endocrinol. 2021, 184, 837–845. [Google Scholar] [CrossRef]
  151. Arıkan, R.; Telli, T.A.; Demircan, N.C.; Başoğlu, T.; Ercelep, Ö.; Atasoy, B.M.; Özgüven, S.; Dane, F.; Yumuk, P.F. Rechallenge with dabrafenib plus trametinib in anaplastic thyroid cancer: A case report and review of literature. Curr. Probl. Cancer 2021, 45, 100668. [Google Scholar] [CrossRef]
  152. Lungulescu, C. Durable Response in a Case of Metastatic Anaplastic Thyroid Cancer Using a Combination of Tyrosine Kinase Inhibitors and a Check Point Inhibitor. Acta Endocrinol. 2020, 16, 236–241. [Google Scholar] [CrossRef] [PubMed]
  153. Liao, Y.; Gao, Y.; Chang, A.; Li, Z.; Wang, H.; Cao, J.; Gu, W.; Tang, R. Melatonin synergizes BRAF-targeting agent dabrafenib for the treatment of anaplastic thyroid cancer by inhibiting AKT/hTERT signalling. J. Cell. Mol. Med. 2020, 24, 12119–12130. [Google Scholar] [CrossRef]
  154. Wang, Y.; Hu, Z.; Ma, W.; Niu, Y.; Su, J.; Zhang, L.; Zhao, P. Signal transducer and activator of transcription 3 inhibition alleviates resistance to BRAF inhibition in anaplastic thyroid cancer. Investig. New Drugs 2021, 39, 764–774. [Google Scholar] [CrossRef]
  155. Subbiah, V.; Yang, D.; Velcheti, V.; Drilon, A.; Meric-Bernstam, F. State-of-the-Art Strategies for Targeting RET-Dependent Cancers. J. Clin. Oncol. 2020, 38, 1209–1221. [Google Scholar] [CrossRef]
  156. Thiesmeyer, J.W.; Limberg, J.; Ullmann, T.M.; Stefanova, D.; Bains, S.; Beninato, T.; Zarnegar, R.; Fahey, T.J.; Finnerty, B.M. Impact of multikinase inhibitor approval on survival and physician practice patterns in advanced or metastatic medullary thyroid carcinoma. Surgery 2021, 169, 50–57. [Google Scholar] [CrossRef] [PubMed]
  157. Kreissl, M.C.; Bastholt, L.; Elisei, R.; Haddad, R.; Hauch, O.; Jarząb, B.; Robinson, B.; Colzani, R.; Foster, M.; Weiss, R.; et al. Efficacy and Safety of Vandetanib in Progressive and Symptomatic Medullary Thyroid Cancer: Post Hoc Analysis from the ZETA Trial. J. Clin. Oncol. 2020, 38, 2773–2781. [Google Scholar] [CrossRef]
  158. Valerio, L.; Bottici, V.; Matrone, A.; Piaggi, P.; Viola, D.; Cappagli, V.; Agate, L.; Molinaro, E.; Ciampi, R.; Tacito, A.; et al. Medullary thyroid cancer treated with vandetanib: Predictors of a longer and durable response. Endocr. Relat. Cancer 2020, 27, 97–110. [Google Scholar] [CrossRef]
  159. Koehler, V.F.; Adam, P.; Frank-Raue, K.; Raue, F.; Berg, E.; Hoster, E.; Allelein, S.; Schott, M.; Kroiss, M.; Spitzweg, C.; et al. Real-World Efficacy and Safety of Cabozantinib and Vandetanib in Advanced Medullary Thyroid Cancer. Thyroid 2021, 31, 459–469. [Google Scholar] [CrossRef]
  160. Matrone, A.; Prete, A.; Nervo, A.; Ragni, A.; Agate, L.; Molinaro, E.; Giani, C.; Valerio, L.; Minaldi, E.; Piovesan, A.; et al. Lenvatinib as a salvage therapy for advanced metastatic medullary thyroid cancer. J. Endocrinol. Investig. 2021, 44, 2139–2151. [Google Scholar] [CrossRef] [PubMed]
  161. Li, D.; Chi, Y.; Chen, X.; Ge, M.-H.; Zhang, Y.; Guo, Z.; Wang, J.; Chen, J.; Zhang, J.; Cheng, Y.; et al. Anlotinib in Locally Advanced or Metastatic Medullary Thyroid Carcinoma: A Randomized, Double-Blind Phase IIB Trial. Clin. Cancer Res. 2021, 27, 3567–3575. [Google Scholar] [CrossRef]
  162. 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]
  163. 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]
  164. Subbiah, V.; Shen, T.; Terzyan, S.; Liu, X.; Hu, X.; Patel, K.; Hu, M.; Cabanillas, M.; Behrang, A.; Meric-Bernstam, F.; et al. Structural basis of acquired resistance to selpercatinib and pralsetinib mediated by non-gatekeeper RET mutations. Ann. Oncol. 2021, 32, 261–268. [Google Scholar] [CrossRef]
  165. Shen, T.; Hu, X.; Liu, X.; Subbiah, V.; Mooers, B.H.M.; Wu, J. The L730V/I RET roof mutations display different activities toward pralsetinib and selpercatinib. NPJ Precis. Oncol. 2021, 5, 1–4. [Google Scholar] [CrossRef]
  166. Resteghini, C.; Cavalieri, S.; Galbiati, D.; Granata, R.; Alfieri, S.; Bergamini, C.; Bossi, P.; Licitra, L.; Locati, L. Management of tyrosine kinase inhibitors (TKI) side effects in differentiated and medullary thyroid cancer patients. Best Pract. Res. Clin. Endocrinol. Metab. 2017, 31, 349–361. [Google Scholar] [CrossRef] [PubMed]
  167. Brose, M.S.; Bible, K.C.; Chow, L.Q.; Gilbert, J.; Grande, C.; Worden, F.; Haddad, R. Management of treatment-related toxicities in advanced medullary thyroid cancer. Cancer Treat. Rev. 2018, 66, 64–73. [Google Scholar] [CrossRef]
  168. Cabanillas, M.E.; Takahashi, S. Managing the adverse events associated with lenvatinib therapy in radioiodine-refractory differentiated thyroid cancer. Semin. Oncol. 2019, 46, 57–64. [Google Scholar] [CrossRef]
  169. Verburg, F.A.; Amthauer, H.; Binse, I.; Brink, I.; Buck, A.; Darr, A.; Dierks, C.; Koch, C.; König, U.; Kreissl, M.C.; et al. Questions and Controversies in the Clinical Application of Tyrosine Kinase Inhibitors to Treat Patients with Radioiodine-Refractory Differentiated Thyroid Carcinoma: Expert Perspectives. Horm. Metab. Res. 2021, 53, 149–160. [Google Scholar] [CrossRef] [PubMed]
  170. Monti, S.; Presciuttini, F.; Deiana, M.G.; Motta, C.; Mori, F.; Renzelli, V.; Stigliano, A.; Toscano, V.; Pugliese, G.; Poggi, M. Cortisol deficiency in Lenvatinib treatment of thyroid cancer: An underestimated, common adverse event. Thyroid 2021. [Google Scholar] [CrossRef] [PubMed]
  171. Basolo, A.; Matrone, A.; Elisei, R.; Santini, F. Effects of tyrosine kinase inhibitors on thyroid function and thyroid hormone metabolism. Semin. Cancer Biol. 2021. [Google Scholar] [CrossRef] [PubMed]
  172. Porcelli, T.; Luongo, C.; Sessa, F.; Klain, M.; Masone, S.; Troncone, G.; Bellevicine, C.; Schlumberger, M.; Salvatore, D. Long-term management of lenvatinib-treated thyroid cancer patients: A real-life experience at a single institution. Endocrine 2021, 73, 358–366. [Google Scholar] [CrossRef]
  173. Subbiah, V.; Baik, C.; Kirkwood, J.M. Clinical Development of BRAF plus MEK Inhibitor Combinations. Trends Cancer 2020, 6, 797–810. [Google Scholar] [CrossRef]
  174. Robey, R.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef] [PubMed]
  175. Wu, Z.-X.; Yang, Y.; Wang, J.-Q.; Zhou, W.-M.; Chen, J.; Fu, Y.-G.; Patel, K.; Chen, Z.-S.; Zhang, J.-Y. Elevated ABCB1 Expression Confers Acquired Resistance to Aurora Kinase Inhibitor GSK-1070916 in Cancer Cells. Front. Pharmacol. 2021, 11, 615824. [Google Scholar] [CrossRef] [PubMed]
  176. Wang, J.; Yang, D.-H.; Yang, Y.; Wang, J.-Q.; Cai, C.-Y.; Lei, Z.-N.; Teng, Q.-X.; Wu, Z.-X.; Zhao, L.; Chen, Z.-S. Overexpression of ABCB1 Transporter Confers Resistance to mTOR Inhibitor WYE-354 in Cancer Cells. Int. J. Mol. Sci. 2020, 21, 1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of TC progression showing critical genes and factors involved in de-differentiation. Modified from [29]. ATC, anaplastic thyroid carcinoma; FTC, follicular thyroid carcinoma; PDCT, poorly differentiated thyroid carcinoma; PTC, papillary thyroid carcinoma.
Figure 1. Schematic representation of TC progression showing critical genes and factors involved in de-differentiation. Modified from [29]. ATC, anaplastic thyroid carcinoma; FTC, follicular thyroid carcinoma; PDCT, poorly differentiated thyroid carcinoma; PTC, papillary thyroid carcinoma.
Ijms 22 11829 g001
Figure 2. Schematic representation of major molecular targets in kinase inhibition-based therapies for thyroid cancer. RTK, receptor tyrosine kinase.
Figure 2. Schematic representation of major molecular targets in kinase inhibition-based therapies for thyroid cancer. RTK, receptor tyrosine kinase.
Ijms 22 11829 g002
Table 1. Kinase inhibitors currently approved for thyroid cancer (TC) treatment.
Table 1. Kinase inhibitors currently approved for thyroid cancer (TC) treatment.
Cancer TypeAgent
(Brand Name(s))
Time of ApprovalPoint of ActionIndication
DTC
Entrectinib
(Rozlytrek)
FDA 2019
EMA 2020
NTRKSolid tumors with a NTRK gene fusion without a drug-resistance mutation in certain TRK proteins
Larotrectinib
(Vitrakvi)
FDA 2018
EMA 2019
NTRKSolid tumors with a NTRK gene fusion without a drug-resistance mutation in certain TRK proteins
Lenvatinib
(Lenvima)
FDA 2015
EMA 2014
MKIProgressive, recurrent or metastatic RAIR-DTC
Pralsetinib
(Gavreto)
FDA 2020RETMetastatic or advanced TCs with a RET fusion gene
Selpercatinib
(Retevmo,
Retsevmo)
FDA 2020
EMA 2021
RETFDA: metastatic or advanced RAIR-DTC patients aged ≥ 12 years with a RET fusion gene; EMA: advanced DTC patients with changes in RET gene previously treated with sorafenib and/or lenvatinib
Cabozantinib
(Cabometyx,
Cometriq)
FDA 2021MKIAdvanced or metastatic RAIR-DTC that has progressed following prior VEGFR-targeted therapy
Sorafenib
(Nexavar)
FDA 2005
EMA 2006
MKIProgressive, recurrent or metastatic RAIR-TC
ATC
Dabrafenib
(Tafinlar)
FDA 2018BRAFLocally advanced or metastasized ATC that cannot receive local treatment; Therapy in combination with trametinib
Entrectinib
(Rozlytrek)
FDA 2019
EMA 2020
NTRKSolid tumors with a NTRK gene fusion without a drug-resistance mutation in certain TRK proteins
Larotrectinib
(Vitrakvi)
FDA 2018
EMA 2019
NTRKSolid tumors with a NTRK gene fusion without a drug-resistance mutation in certain TRK proteins
Pralsetinib
(Gavreto)
FDA 2020RETMetastatic or advanced TCs with a RET fusion gene
Selpercatinib
(Retevmo,
Retsevmo)
FDA 2020
EMA 2021
RETFDA: metastatic or advanced ATC patients aged ≥ 12 years with a RET fusion gene; EMA: advanced ATC patients with changes in RET gene previously treated with sorafenib and/or lenvatinib
Trametinib
(Mekinist)
FDA 2018MEKLocally advanced or metastasized ATC that cannot receive local treatment; Therapy in combination with dabrafenib
MTC
Cabozantinib
(Cabometyx, Cometriq)
FDA 2012
EMA 2014
MKIProgressive or metastasized MTC
Pralsetinib
(Gavreto)
FDA 2020RETAdvanced or metastatic MTC with a mutation in the RET gene
Selpercatinib
(Retevmo,
Retsevmo)
FDA 2020
EMA 2021
RETFDA: advanced or metastatic MTC aged ≥ 12 years with a mutation in the RET gene; EMA: advanced MTC patients aged ≥ 12 years with changes in RET gene previously treated with cabozantinib and/or vandetanib
Vandetanib
(Caprelsa)
FDA 2011
EMA 2013
MKILocally advanced or metastasized MTC
ATC, anaplastic thyroid carcinoma; DTC, differentiated thyroid carcinoma; EMA, European Medical Agency; FDA, Food and Drug Administration; MKI, multikinase inhibitor; MTC, medullary thyroid carcinoma; RAIR, radioiodine refractory.
Table 2. Kinase inhibitors currently investigated in clinical trials for DTC treatment (registered under ClinicalTrials.gov and clinicaltrialsregister.eu).
Table 2. Kinase inhibitors currently investigated in clinical trials for DTC treatment (registered under ClinicalTrials.gov and clinicaltrialsregister.eu).
Agent
(Drug Name)
Alternative/Control TreatmentStudy PhaseSpecific Inclusion CriteriaStudy Start/ Estimated Completion DateStatusTrial Number
MKIs
AnlotinibNDIIND01.12.2020/
07.12.2022
RecruitingNCT05007093
ApatinibNDIIVEGFR210.04.2019/
30.05.2020
RecruitingNCT04180007
(APT-01)
ApatinibPlaceboIIIVEGFR212.2016/
06.2022
Active,
not recruiting
NCT03048877
CabozantinibNDIIND01.2014/
05.2022
Active,
not recruiting
NCT02041260
CabozantinibPlaceboIIIND05.10.2018/
12.2022
Active,
not recruiting
NCT03690388
DonafenibPlaceboIIIND29.04.2018/
12.2021
RecruitingNCT03602495
LenvatinibNDI/IIFTC, PTC29.12.2014/
31.03.2022
Active,
not recruiting
NCT02432274
(in Cohort 2A)
LenvatinibNDIIND01.05.2021/
01.01.2024
Not yet recruitingNCT04858867
LenvatinibNDIIIND11.01.2017/
30.10.2021
Active,
not recruiting
NCT02966093
EntrectinibNDIINTRK, ROS1 or ALK
PTC
01.12.2022/
02.12.2024
RecruitingNCT02568267
(STARTRK-2)
ImatinibNDIPTC18.09.2018/
30.12.2021
RecruitingNCT03469011
VandetanibPlaceboIIFTC, PTC29.09.2007/
12.2021
Active,
not recruiting
NCT00537095
VandetanibPlaceboIIIND17.09.2013/
31.12.2021
Active,
not recruiting
NCT01876784
TRK inhibitors
LarotrectinibNDIINTRK30.09.2015/
30.09.2025
RecruitingNCT02576431
(NAVIGATE)
ALK inhibitors
AlectinibNDIIALK-mutant PTC24.05.2021/
01.12.2025
RecruitingNCT04644315
RepotrectinibNDIALK, ROS1 or NTRK
rearrangements
27.02.2017/
12.2022
RecruitingNCT03093116
(TRIDENT-1)
MEK and BRAF inhibitors (combination therapies)
Cobimetinib/
ABM-1310
NDIBRAF V600E16.06.2020/
12.2021
RecruitingNCT04190628
Encorafenib/
Binimetinib
Encorafenib/
Binimetinib/
Nivolumab 1
IIBRAF V600E/M30.10.2020/
01.08.2024
RecruitingNCT04061980
Trametinib/I131NDIIRAS-mutant or RAS/BRAF wt14.08.2014/
31.12.2021
Active,
not recruiting
NCT02152995
Dabrafenib/
Trametinib
NDIIBRAF V600E
or RAS
27.12.2017/
12.2022
RecruitingNCT03244956
(MERAIODE)
TrametinibTrametinib/
Dabrafenib
IIBRAF wt (Trametinib)
and BRAF V600E
(Trametinib/
Dabrafenib)
05.02.2018/
30.06.2022
RecruitingNCT04619316
(ERRITI)
Dabrafenib/
Trametinib
NDIIBRAF V600E
or RAS
30.12.2020/
04.2022
RecruitingNCT04554680
TrametinibTrametinib/
Dabrafenib
I
(pilot
study)
BRAF wt (Trametinib) and BRAF V600E (Trametinib/
Dabrafenib) RAIR-PTC
NDOngoing2016-002941-4
(ERRITI)
Dabrafenib/
Trametinib
PlaceboIIIBRAF V600E01.10.2021/
30.10.2026
Not yet recruitingNCT04940052
VemurafenibNDIIPTC07.11.2012/
30.11.2020
Active,
not recruiting
NCT01709292
Other combination therapies
Anlotinib/I131NDIIND15.08.2021/
20.07.2024
Not yet recruitingNCT04952493
Apatinib/
Camrelizumab 1
NDIIND23.09.2020/
30.06.2023
RecruitingNCT04560127
Cabozantinib/
Nivolumab 1/
Ipilimumab 2
NDIIND15.07.2019/
15.01.2022
RecruitingNCT03914300
Cabozantinib/
Atezolizumab 1
NDIND05.09.2017/
12.2022
RecruitingNCT03170960
Lenvatinib/
Pembrolizumab 1
NDIIND08.10.2021/
30.09.2022
RecruitingNCT02973997
Lapatinib/
Dabrafenib
NDIBRAF29.08.2013/
01.07.2022
Active,
not recruiting
NCT01947023
Lenvatinib/
Denosumab 3
NDIIND26.07.2019/
15.06.2022
RecruitingNCT03732495
(LENVOS)
Sorafenib/
Everolimus 4
NDIITC06.2010/
06.2022
Active,
not recruiting
NCT01141309
Sorafenib/
Everolimus 4
ND IIDTC progressed on
monotherapy with
Sorafenib
10.2010/
03.2021
Active,
not recruiting
NCT01263951
Regorafenib/
Avelumab 1
NDI/IIND04.05.2018/
05.2022
RecruitingNCT03475953 (REGOMUNE)
Sorafenib/
Everolimus 4
Sorafenib aloneIIRAIR-HCC01.10.2014/
28.01.2021
Active,
not recruiting
NCT02143726
SelumetinibSelumetinib/I131IIND04.05.2015/
20.02.2022
Active,
not recruiting
NCT02393690
Surufatinib/
Toripalimab 1
NDIIND01.10.2020/
30.09.2022
Not yet recruitingNCT04524884
DabrafenibDabrafenib/
Lenvatinib
IIBRAF07.11.2012/
31.12.2021
Active,
not recruiting
NCT01723202
Trametinib/I131Trametinib/
Dabrafenib/
I131
NDRAS (Trametinib/I131) or
BRAF (Trametinib/
Dabrafenib/I131)
NDOngoingNCT03244956 (MERAIODE)
PDR001 1/
Trametinib
(Cohort A) or
Dabrafenib
(Cohort B)
NDIICohort A: BRAF wt
Cohort B: BRAF-mutant, resistant to previous
BRAF inhibitors
02.09.2020/
02.09.2022
RecruitingNCT04544111
Vemurafenib/
Copanlisib 5
NDIBRAF V600E26.06.2020/
06.2022
RecruitingNCT04462471
1 Anti-PD-1 monoclonal antibody; 2 anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4) monoclonal antibody; 3 anti-receptor activator of nuclear factor κ-Β ligand (RANKL) monoclonal antibody; 4 mTOR inhibitor; 5 phosphatidylinositol 3-kinase (PI3K) inhibitor; wt, wild type; ND, not defined.
Table 3. Kinase inhibitors currently investigated in clinical trials for ATC treatment (registered under ClinicalTrials.gov and clinicaltrialsregister.eu).
Table 3. Kinase inhibitors currently investigated in clinical trials for ATC treatment (registered under ClinicalTrials.gov and clinicaltrialsregister.eu).
Agent
(Drug Name)
Alternative/Control TreatmentStudy
Phase
Specific Inclusion CriteriaStudy Start/
Estimated
Completion Date
StatusTrial Number
MEK and BRAF inhibitors (combination therapies)
Dabrafenib/
Trametinib
NDIIMutated BRAF22.01.2021/
01.2026
RecruitingNCT04739566
(ANAPLAST-NEO)
Dabrafenib/
Trametinib
NDIBRAF V600E04.05.2020/
30.04.2025
RecruitingNCT03975231
Cobimetinib/
ABM-1310
ABM-1310IBRAF V600E16.06.2020/
12.2021
RecruitingNCT04190628
RET inhibitors
SelpercatinibNDIIMutated RET26.02.2021/
10.09.2024
RecruitingNCT04759911
TRK inhibitors
LarotrectinibNDIINTRK30.09.2015/
30.09.2025
RecruitingNCT02576431
(NAVIGATE)
MKIs in combination with other agents
MKI
(ND)/
Anti-PD-1
antibody
(ND)
NDIIArm C: ATC30.12.2019/
30.06.2023
RecruitingNCT04521348
Cabozantinib/
Atezolizumab 1
NDIIND07.10.2020/
03.2024
RecruitingNCT04400474
(CABETEN)
Lenvatinib/
Pembrolizumab 1
NDIIND02.03.2021/
31.08.2022
Not yet recruitingNCT04171622
Pazopanib/
Paclitaxel 2/IMRT
Placebo/
Paclitaxel 2/IMRT
IIND28.10.2010/
lack of data
Active, not recruitingNCT01236547
SKIs in combination with other agents
RepotrectinibNDIALK, ROS1, NTRK rearrangements27.02.2017/
12.2022
RecruitingNCT03093116 (TRIDENT-1)
Cobimetinib/
Atezolizumab 1
(/Vemurafenib if
BRAF V600E) (Cohort A)
Cobimetinib/
Atezolizumab 1
(Cohort B)
IIMutated BRAF (Cohort A) vs. RAS, NF1, NF2
and MAPK pathway
proteins at or above
MEK mutations
(Cohort B)
27.07.2017/
27.07.2023
RecruitingNCT03181100
Dabrafenib/
Trametinib/
Cemiplimab 1
NDIIBRAF V600E20.01.2020/
20.06.2022
RecruitingNCT04238624
Trametinib/
Dabrafenib/
Pembrolizumab 1
NDIIMutated BRAF24.06.2021/
30.06.2024
RecruitingNCT04675710
Trametinib/
Paclitaxel 2
NDI (pilot
study)
ND03.2017/
09.2023
RecruitingNCT03085056
1 Anti-PD-1 monoclonal antibody; 2 taxan; IMRT, intensity-modulated radiation therapy.
Table 4. Kinase inhibitors currently investigated in clinical trials for MTC treatment (registered under ClinicalTrials.gov and clinicaltrialsregister.eu).
Table 4. Kinase inhibitors currently investigated in clinical trials for MTC treatment (registered under ClinicalTrials.gov and clinicaltrialsregister.eu).
Agent
(Drug Name)
Alternative/Control TreatmentStudy
Phase
Specific Inclusion CriteriaStudy Start/
Estimated
Completion Date
StatusTrial Number
MKIs
AnlotinibNDIIND01.01.2019/
01.06.2022
Active,
not recruiting
NCT04309136
CabozantinibNDIIPediatric MTC08.05.2017/
01.07.2022
Active,
not recruiting
NCT02867592
Cabozantinib
60 mg
Cabozantinib
140 mg
IVND12.2014/
12.2022
Active,
not recruiting
NCT01896479
(EXAMINER)
PonatinibNDIIND26.07.2019/
06.2022
RecruitingNCT03838692
RegorafenibNDIIND01.2016/
10.2022
RecruitingNCT02657551
SorafenibNDIIHereditary
vs. sporadic MTC
05.10.2006/
30.01.2017
Active,
not recruiting
NCT00390325
SunitinibNDIIND08.08.2006/
31.12.2016
Active,
not recruiting
NCT00381641
VandetanibPlaceboIIIND30.11.2016/
30.12.2021
Active,
not recruiting
NCT00410761
Vandetanib
150 mg
Vandetanib
300 mg
IVND06.2014/
30.12.2020
Active,
not recruiting
NCT01496313
SKIs
BOS172738NDIRET; MTC12.12.2018/
06.2022
Active, not recruitingNCT03780517
PralsetinibNDI/IIRET; MTC and PTC17.03.2017/
29.02.2024
RecruitingNCT03037385
(ARROW)
TPX-0046NDI/IIRET; MTC and other TCs16.12.2019/
03.2025
RecruitingNCT04161391
SelpercatinibNDIIRET; MTC and other TCs16.03.2020/
20.10.2025
Active, not recruitingNCT04280081
(LIBRETTO-321)
SelpercatinibNDI/IIRET; MTC and other TCs02.05.2017/
21.11.2023
RecruitingNCT03157128
(LIBRETTO-001)
SelpercatinibNDIIRET; MTC26.02.2021/
10.09.2024
Recruiting2017-000800-59
Kinase inhibitors in combination with other agents
MKIs
MKI
(not precised)/
Anti-PD-1 antibody
(not precised)
NDIIArm B: MTC30.12.2019/
30.06.2023
RecruitingNCT04521348
Apatinib/
Camrelizumab 1
NDIIND01.12.2020/
31.12.2023
Not yet recruitingNCT04612894
SKIs vs. MKIs
PralsetinibCabozantinib or
Vandetanib
IIIMutated RET01.09.2021/
15.04.2028
Not yet recruitingNCT04760288
(AcceleRET-MTC)
SelpercatinibCabozantinib or
Vandetanib
IIIMutated RET30.12.2019/
30.06.2023
RecruitingNCT04211337
(LIBRETTO-531)
2019-001978-28
1 Anti-PD-1 monoclonal antibody.
Table 5. Most common adverse events (AEs) reported for kinase inhibitors in TC treatment over the past two years.
Table 5. Most common adverse events (AEs) reported for kinase inhibitors in TC treatment over the past two years.
ResearchStudy PhaseAgent
(Dose or Group)
No of Patients:
Total/
Dropouts due to AEs
Dose
Reduction
(No of Patients)
ORRDCRCRPRSDPDOS
(Months)
PFS
(Months)
All-Grade Most Common
AEs (%)
MKIs
Li et al.
[161]
IIBAnlotinib vs. placebo vs.
open label
62 vs.
29 vs.
12
10 vs.
1 vs.
1
48% vs.
0% vs.
33%
89% vs.
86% vs.
83%
NDNDNDND50 vs.
19 vs.
ND
21 vs.
11 vs.
15
PPES (63% vs. 10% vs. ND)
Proteinuria (61% vs. 10% vs. ND)
Hypertriglyceridemia (48% vs. 24% vs. ND)
Lin et al.
[119]
IIApatinib
(500 mg vs.
750 mg)
10 vs.
10
5 vs.
9
70% vs.
90%
90% vs.
100%
0% vs. 0%70% vs.
90%
20% vs.
10%
ND34 vs.
52
14 vs.
35
PPES (95%)
Proteinuria (90%)
HT (80%)
Brose et al.
[122]
IIICabozantinib vs. placebo125/6 vs.
62/0
70 vs.
3
35% vs.
2%
43% vs.
16%
0% vs. 0%9% vs.
0%
61% vs.
34%
6% vs.
50%
ND6 vs.
2
Diarrhoea (51% vs. 3%)
PPES (45% vs. 0%)
HT (28% vs. 5%)
Lin et al.
[121]
IIDonafenib
(200 mg vs.
300 mg)
17/1 vs.
18/3
8 vs.
13
13% vs.
13%
100% vs.
100%
ND13% vs.
13%
88% vs.
87%
NDND9 vs.
15
PPES (88% vs. 78%)
Alopecia (65% vs. 78%)
HT (47% vs. 44%)
Wirth et al.
[146]
IILenvatinib34/6143%53%0%3%50%27%33HT (56%)
Decreased appetite (29%)
Fatigue (29%)
Stomatitis (29%)
Takahashi et al. [124] NDLenvatinib
(DTC or ATC
or MTC)
442 or
124 or
28
105 or
37 or
6
59% or
44% or
45%
92% or
76% or
100%
3% or
3% or
5%
57% or
41% or
40%
33% vs.
32% vs.
55%
4% vs.
24% vs.
0%
ND or
ND or
4
NDHT (79% or 70% or 64%)
Proteinuria (43% or 30% or 39%)
PPES (39% or 26% or 50%)
Song et al.
[126]
NDLenvatinib43/630ND98%ND65%56%2%ND22Fatigue or asthenia (72%)
Diarrhoea (67%)
HT (63%)
Proteinuria (58%)
Porcelli et al.
[172]
NDLenvatinib23/323NDNDND26%61%4%4625HT (78%)
Fatigue (74%)
Weight loss (65%)
Matrone et al.
[160]
NDLenvatinib9736%80%ND11%89%NDNDNDAsthenia (50%)
Nausea (40%)
Diarrhoea (30%)
Weight loss (30%)
Ito et al.
[127]
NDSorafenib vs.
Lenvatinib
21 vs.
18
NDNDNDND22% vs.
65%
72% vs.
24%
6% vs.
2%
NDNDPPES (81% vs. 39%)
HT (19% vs. 94%)
Thrombocytopenia (ND vs. 50%)
Hypocalcemia (38% vs. ND)
Proteinuria (33% vs 33%)
Koehler et al.
[125]
NDSorafenib vs.
Lenvatinib vs.
Pazopanib
33/14 vs.
53/23 vs.
15/4
16 vs.
31 vs.
6
18% vs.
70% vs.
60%
ND0% vs.
2% vs.
7%
18% vs.
68% vs.
53%
21% vs.
9% vs.
13%
ND37 vs.
47 vs.
34
9 vs.
12 vs.
12
Loss of appetite/weight (46% vs. 49% vs. 25%)
Diarrhoea (43% vs. 44% vs. 42%)
Fatigue (ND vs. 44% vs. 29%)
Valerio et al.
[158]
NDVandetanib
(short-term vs.
long-term treatment)
25 vs.
54
NDNDNDNDND vs.
26%
ND vs.
72%
ND vs.
2%
ND47 vs.
87
Hypothyroidism (92% vs. 100%)
Rash (28% vs. 48%)
Diarrhoea (12% vs. 39%)
SKIs
Salama et al.
[134]
IIDabrafenib and
Trametinib
29ND38%76ND38%38%7%2911Fatigue (74%)
Nausea (57%)
Chills (54%)
Doebele et al.
[139]
I /IIEntrectinib68/32757%ND7%50%17%7%2111Dysgeusia (47%)
Fatigue (35%)
Constipation (28%)
Desai et al.
[138]
ILifirafenib
(dose escalation vs.
dose expansion)
35/5 vs.
96/19
NDNDNDNDNDNDNDNDNDFatigue (69% vs. 49%)
Acneiform dermatitis (43% vs. 18%)
Decreased appetite (40% vs. 36%)
Constipation (40% vs. 27%)
Subbiah et al.
[162]
I/IIPralsetinib
(RETm MTC previously treated or
RETm MTC with no previous treatment or RETf TC)
55 or
21 or
9
5 (and 8 deaths)60% or
71% or
89%
93% or
100% or
100%
2% or
5% or
0%
58% or
67% or
89%
33% vs.
29% vs.
11%
4% vs.
0% vs.
0%
17 or
19 or
16
15 or
15 or
13
Neutropenia (34%)
Decreased WBC count (34%)
Increased AST level (34%)
Wirth et al.
[163]
I/IISelpercatinib
(RETm MTC previously treated or
RETm MTC with no previous treatment or RETf TC previously treated)
55 or
88 or
19
ND62% or
71% or
58%
69% or
67% or
58%
5% or
3% or
0%
56% or
67% or
58%
29% vs.
27% vs.
37%
5% vs.
0% vs.
0%
ND27 or
24 or
ND
Dry mouth (46%)
HT (43%)
Diarrhoea (38%)
Fatigue (38%)
AST, aspartate aminotransferase; CR, complete response; DCR, disease control rate; HT, hypertension; ORR, overall response rate; OS, overall survival; PD, progressive disease; PFS, progression-free survival; PPES, palmar-plantar erythrodysesthesia syndrome; PR, partial response; RETf, RET fusion-positive; RETm, RET mutation-positive; SD, stable disease; WBC, white blood cell.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ratajczak, M.; Gaweł, D.; Godlewska, M. Novel Inhibitor-Based Therapies for Thyroid Cancer—An Update. Int. J. Mol. Sci. 2021, 22, 11829. https://doi.org/10.3390/ijms222111829

AMA Style

Ratajczak M, Gaweł D, Godlewska M. Novel Inhibitor-Based Therapies for Thyroid Cancer—An Update. International Journal of Molecular Sciences. 2021; 22(21):11829. https://doi.org/10.3390/ijms222111829

Chicago/Turabian Style

Ratajczak, Maciej, Damian Gaweł, and Marlena Godlewska. 2021. "Novel Inhibitor-Based Therapies for Thyroid Cancer—An Update" International Journal of Molecular Sciences 22, no. 21: 11829. https://doi.org/10.3390/ijms222111829

APA Style

Ratajczak, M., Gaweł, D., & Godlewska, M. (2021). Novel Inhibitor-Based Therapies for Thyroid Cancer—An Update. International Journal of Molecular Sciences, 22(21), 11829. https://doi.org/10.3390/ijms222111829

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