Next Article in Journal
HLA-DQB1 and HLA-DRB1 Variants Confer Susceptibility to Latent Autoimmune Diabetes in Adults: Relative Predispositional Effects among Allele Groups
Next Article in Special Issue
Thyroid Cancer in the Pediatric Population
Previous Article in Journal
The First Highly Contiguous Genome Assembly of Pikeperch (Sander lucioperca), an Emerging Aquaculture Species in Europe
Previous Article in Special Issue
Twenty-Five Years Experience on RET Genetic Screening on Hereditary MTC: An Update on The Prevalence of Germline RET Mutations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Alterations in Thyroid Cancer: From Bench to Clinical Practice

1
Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
2
Center of Experimental Oncology and Hematology, A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
3
Department of Medical Oncology A.O.U. Policlinico-Vittorio Emanuele, 95123 Catania, Italy
4
Department of Clinical and Experimental Medicine, Section of Endocrinology, University of Catania, Garibaldi-Nesima Medical Center, 95122 Catania, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2019, 10(9), 709; https://doi.org/10.3390/genes10090709
Submission received: 12 July 2019 / Revised: 26 August 2019 / Accepted: 10 September 2019 / Published: 13 September 2019
(This article belongs to the Special Issue Thyroid Cancer: Genetics and Targeted Therapies)

Abstract

:
Thyroid cancer comprises different clinical and histological entities. Whereas differentiated (DTCs) malignancies are sensitive to radioiodine therapy, anaplastic (ATCs) and medullary (MTCs) tumors do not uptake radioactive iodine and display aggressive features associated with a poor prognosis. Moreover, in a majority of DTCs, disease evolution leads to the progressive loss of iodine sensitivity. Hence, iodine-refractory DTCs, along with ATCs and MTCs, require alternative treatments reflective of their different tumor biology. In the last decade, the molecular mechanisms promoting thyroid cancer development and progression have been extensively studied. This has led to a better understanding of the genomic landscape, displayed by thyroid malignancies, and to the identification of novel therapeutic targets. Indeed, several pharmacological compounds have been developed for iodine-refractory tumors, with four multi-target tyrosine kinase inhibitors already available for DTCs (sorafenib and lenvatinib) and MTCs (cabozantib and vandetanib), and a plethora of drugs currently being evaluated in clinical trials. In this review, we will describe the genomic alterations and biological processes intertwined with thyroid cancer development, also providing a thorough overview of targeted drugs already tested or under investigation for these tumors. Furthermore, given the existing preclinical evidence, we will briefly discuss the potential role of immunotherapy as an additional therapeutic strategy for the treatment of thyroid cancer.

1. Introduction

Thyroid cancer represents 2.5% of all cancers and about 90% of endocrine tumors [1]. The incidence of this neoplasia, three-fold higher in women and in individuals aged 25 to 65 years [2], has increased by 4.5% per year over the last decade [3], possibly because of diagnostic techniques improvement, environmental (e.g., radiation, pollution) and lifestyle changes [4,5]. Histologically, thyroid tissue consists of two different epithelial populations, the follicular and para-follicular [also known as C (clear)] cells, with different embryological origins and function. Follicular cells, organized in functional units called follicles, synthetize and secrete thyroid hormones (thyroxine and triiodothyronine), whereas C-cells, nestled between the follicles, secrete the hormone calcitonin [6].
Thyroid cancers are classified in follicular-derived and C-cell-derived, according to their cell of origin. Follicular-derived thyroid malignancies are subdivided in differentiated (DTCs) and anaplastic (ATCs) thyroid carcinomas. DTCs include the papillary (PTCs) and follicular (FTCs) histotypes, Hurtle cell thyroid cancers and the more aggressive poorly differentiated carcinomas (PDTCs) [7,8]. ATC accounts for about 2% of all thyroid cancers and is constituted by undifferentiated cells with very low similarity to normal thyroid tissue [9].
Lastly, medullary thyroid cancer (MTC) stemming from para-follicular C-cells represents 2–5% of all thyroid carcinomas [10]. It is a neuroendocrine tumor that can be either sporadic (75%) or familial (25%) the latter defined as part of Multiple Endocrine Neoplasia type 2 (MEN2) syndrome [11]. MEN2 comprises MEN2A, which features familial MTC (FMTC) [12] or MTC plus pheochromocytoma and hyper-parathyroidism/parathyroid adenoma, and MEN2B also characterized by MTC and pheochromocytoma, but with mucosal ganglioneuromas and marfanoid habitus.
The vast majority of DTCs usually display an indolent course, and standard upfront treatment involves surgery followed by adjuvant hormone replacement and radioactive iodine (131I-based RAI) therapy for high-risk diseases [13]. Despite their overall good prognosis, 10–20% of DTC patients present distant metastases at diagnosis or will subsequently develop them [14]. The majority of these patients are eligible for RAI, with a 40% chance of achieving a complete and durable response [15]. However, the remaining 60% display primary or acquired resistance to RAI (RAIR), thus needing further treatment options [16].
Unlike DTC, ATC grows rapidly and does not maintain the features of follicular cells, including iodine uptake. Indeed, ATC shows suppression of the sodium iodide symporter (NIS) expression/function and RAI refractoriness. Hence, radiotherapy and chemotherapy are the only treatment options for this disease, even though reported outcomes are dismal [17,18].
Since para-follicular cells intrinsically lack 131I avidity, thyroidectomy with subsequent hormone replacement is the treatment of choice for localized MTC. However, targeted therapies or, less frequently, chemotherapy may be an option for locally advanced or metastatic patients [19].
Thyroid cancers are characterized by molecular alterations, such as activating/inactivating mutations, rearrangements and copy number variations in genes responsible for cell proliferation, differentiation and apoptosis [20]. In recent years, the discovery of disease-specific molecular targets has led to the approval of new drugs (e.g., sorafenib, lenvatinib, vandetanib and cabozantinib), which are currently available for metastatic RAIR DTCs and MTCs [21,22]. Aim of this review is to discuss the molecular alterations associated with DTCs, ATCs and MTCs and provide an update on recently published studies and ongoing trials testing targeted therapies and immunotherapy in advanced thyroid carcinomas.

2. Molecular Alterations in Thyroid Cancers

As thyroid cancer progresses, the accumulation of molecular alterations disrupting multiple normal cell functions results in RAIR development, due to impaired NIS expression [23,24,25]. Indeed, dysregulation of different receptor-tyrosine kinase (RTK)-dependent signaling and proliferation pathways—such as the mitogen-activated protein kinase (MAPK), the phosphoinositide 3 kinase (PI3K), the Wingless/Integrated (WNT), the p53 and p73 pathways—are involved in the multistep tumorigenic process of thyroid cancer [25,26,27] (Figure 1). Alterations of these cascades can be linked to different mechanisms, including genetic and epigenetic modifications in pathway receptors and effectors [28,29]. Moreover, distinct, mutually exclusive molecular alterations may be associated with specific disease stages or histotypes [30].
In order to better classify the molecular alterations detected in thyroid cancer, we will initially discuss RTK-related upstream signaling pathways involved in tumorigenesis and subsequently focus on the effectors of these pathways. Finally, we will describe alterations contributing to thyroid carcinogenesis that involve pivotal cellular functions.

2.1. Alterations in RTKs

Rearrangements, copy number gains and point mutations are the genetic alterations more frequently observed in RTKs. The main consequence of these alterations is increased protein expression and downstream activation of different signaling pathways involved in thyroid cancer progression [31,32,33].
ALK: The anaplastic lymphoma kinase (ALK) may undergo both activating mutations in exon 23 (L1198F and G1201E) [34] and gene rearrangements (especially in PTCs, 1–3%) [35]. While the most common rearrangement involves the striatin (STRN) gene (STRN-ALK), ALK may also rearrange with the echinoderm microtubule-associated protein-like 4 (EML4) gene (EML4-ALK) [36,37]. Furthermore, two novel ALK fusions have been recently identified (GTF2IRD1-ALK and MALAT1-ALK) [30]. Both ALK mutants and cytoplasmic ALK fusion proteins promote the activation of MAPK, PI3K and JAK/STAT downstream pathways (Figure 2). ALK mutations and rearrangements are mainly found in PDTCs and ATCs, as they contribute to disease progression and aggressiveness [34,38,39,40].
NTRK: The Neurotrophic tyrosine kinase receptor (NTRK) gene encodes for the tropomyosine-related kinase (Trk)-family of proteins known as TrkA (encoded by NTRK1), TrkB (encoded by NTRK2) or TrkC (encoded by NTRK3). In PTCs, chromosomal rearrangements, due to environmental factors (e.g., ionizing radiations) cause NTRK fusions with different partners [41]. Cytoplasmic Trk fusion proteins activate downstream signaling via PI3K, MAPK and phospholipase C-gamma (PLCγ) that control cell-cycle progression, proliferation, apoptosis and survival (Figure 2). The major NTRK fusions occur in PTCs between NTRK3 and ETS Variant 6 (ETV6-NTRK3) [41], but have also been identified in 25% of pediatric PTCs [42]. However, in PTCs, NTRK1 may also rearrange with tropomyosin 3 (TPM3), translocated promoter region (TPR) and trafficking from ER to Golgi regulator (TFG) [43].
RET: The Rearranged during Transfection (RET) proto-oncogene is frequently altered in thyroid cancer. Specifically, gene translocations identified as RET/PTC rearrangements are prevalent in PTCs (5–25%), while RET mutations are the primary molecular mechanism underlying MTC tumorigenesis [44]. These events share a common downstream effect as they lead to RET constitutive activation and improper stimulation of both the MAPK and PI3K pathways (Figure 3). To date, at least 19 different rearrangements between the 3’ portion of RET (containing the tyrosine kinase domain) and the 5’ portion of partner genes have been described, [30]. The most frequent fusions are RET-PTC1 (60% of RET-rearranged PTCs), involving the coiled-coil domain-containing gene 6 (CCDC6), RET-PTC3 (30%), generated by the fusion with the nuclear receptor co-activator 4 (NCOA4) and, less frequently (5%) RET-PTC2, involving the protein kinase cAMP-dependent type I regulatory subunit alpha (PRKAR1A). These rearrangements determine loss of the RET transmembrane domain leading to the cytosolic localization of the protein [45]. While RET recombinations are more frequent in young PTC patients (45–60%) as well in radiation-related tumors (up to 80%) [46], they have also been identified in PDTCs, ATCs and MTCs [35].
Several gain-of-function germline or somatic RET mutations arise in hereditary or sporadic MTC patients, respectively [47,48]. In most cases, mutations causing MEN2A involve cysteines within the cysteine-rich extracellular domain (exons 10 and 11) at codon 634 (C634R; 80% frequency) or codons 609, 611, 618, 620 and 630 [49]. These single nucleotide variations cause constitutive dimerization and activation of the receptor, in a ligand-independent manner. The most frequent substitution found in MEN2B patients (95%) is the M918T mutation in exon 16 that induces constitutive kinase activation in the absence of dimerization [50]. Other rare mutations involve codons 634, 691, 838, 883 and 904 [48]. In 95% of FMTC patients, mutations occur at codon 620, although rare substitutions have been reported in other codons, including 611 and 618 [49]. Finally, about 40% of sporadic MTC patients present a somatic RET mutation that in 80% of cases is M918T [51].
Others RTKs: Copy number gains in several other RTKs [epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor A/B (PDGFRA/B), vascular endothelial growth factor receptor 1,2 (VEGFR1,2), Mast/Stem Cell Growth Factor Receptor Kit (c-KIT) and MET Proto-Oncogene, Receptor Tyrosine Kinase (MET)] have been identified in different subtypes of thyroid cancer [25,31,52]. These alterations are associated with increased phosphorylation of AKT, leading to activation of the PI3K pathway [31] (Figure 3). Interestingly, no mutations have been reported in these genes, with the exception of a single case of PTC displaying the G735S EGFR point mutation, which causes a conformational change of the kinase domain leading to its constitutive activation [53]. Fibroblast growth factor receptor 2 (FGFR2) and FMS-like tyrosine kinase 3 (FLT3) missense mutations have been identified in 11% and 17% of PDTCs, respectively [54]. Lastly, FGFR2 fusions may occur in PTCs with very low frequency (<1%) [30,35], while FGFR4 may be overexpressed in PTCs, FTCs and MTCs [52].

2.2. Alterations in the PI3K Pathway

Enhanced PI3K signaling is a common feature of thyroid cancer, in particular in the FTC subtype [25] (Figure 3). Alterations in this pathway involve the GTPase RAS, the alpha catalytic subunit of phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3CA), the serine-threonine protein kinase AKT and the phosphatase and tensin homolog phosphatase (PTEN). While RAS mutations are considered an early event in thyroid cell tumorigenesis, alterations in other downstream effectors of the pathway characterize the less differentiated thyroid cancer histotypes [55].
AKT: Activating mutations in AKT (e.g., the single hotspot E17K mutation promoting constitutive localization to the plasma membrane) inhibit apoptosis in thyroid cells [39]. AKT copy number gains have also been reported [31]. As for PIK3CA, AKT mutations represent a late event in thyroid tumorigenesis; hence, they are more frequent in PDTCs (19%) [56].
PIK3CA: PIK3CA may exhibit activating mutations or undergo copy number gains. Missense mutations take place in exons 9 and 20 (E542K, E545K and H1047R) and are less frequent than amplifications occurring at chromosome site 3q26.3 [57]. These events increase PIK3CA protein expression, yet their tumorigenic role is not well defined. PIK3CA mutations and copy number gains are mutually exclusive in WDTCs, but can co-occur in less differentiated tumors, where they drive disease progression [58,59]. PIK3CA alterations are common in ATCs (18%) and less frequent in FTCs (1%) and PDTCs (2%) [31,39].
PTEN: Alterations involving the tumor suppressor PTEN lead to constitutive activation of the PI3K pathway, causing an increase in cell proliferation, motility and protein synthesis. PTEN inactivating mechanisms include mutations, loss of heterozygosis, deletions and epigenetic modifications, resulting in the loss of PTEN expression [60]. PTEN alterations are described in FTCs [31], and their frequency increases with thyroid tumor progression (4% in PDTCs and 15% in ATCs) [39].
RAS: The RAS oncoprotein is a common effector of both the PI3K and MAPK pathways, although RAS mutations in thyroid cancer prevalently alter the PI3K cascade [31]. Alterations can occur in codons G12, G13 and Q61 and may involve one of the three RAS genes (KRAS, HRAS and NRAS), albeit the latter is predominantly mutated in thyroid tumors [61]. The effect of these mutations is to lock RAS in its active GTP-bound form. RAS point mutations mainly characterize FTCs (30–50%), but are also frequent in RET wild-type sporadic MTCs (10–45%), since point mutations in these two proteins are mutually exclusive [62]. Furthermore, RAS mutations have also been found in PTCs (5%), PDTCs and ATCs (both around 25%) [39,61].

2.3. Alterations in the MAPK Pathway

The MAPK pathway is frequently altered in thyroid cancer, particularly in PTCs [63]. In most cases, mutations involve RAS (previously described) and the B-Raf proto-oncogene (BRAF), in addition to the upstream receptors described above, which activate different signaling cascades (Figure 3).
BRAF: Alterations in BRAF are an early tumorigenic event in PTCs (40–80%) although they have also been reported in PDTCs (5–35%) and ATCs (10–50%) [64]. Point mutations in this serine/threonine kinase activate the MAPK pathway resulting in loss of differentiation, tumor progression and inhibition of apoptosis [65,66]. The most common BRAF mutation is the V600E substitution, found in 45% of PTCs, which causes constitutive activation of the proto-oncogene [67]. Rare mutations may occur around codon 600, the most frequent the K601E substitution, and display an inferior oncogenic potential [30,67]. BRAF fusions have also been found in radiation-associated PTCs and, at a lower frequency, in PDTCs and ATCs [30,35]. The first to be identified was the AKAP9-BRAF rearrangement, found in 10% of radiation-induced PTCs, resulting in a fusion protein lacking the auto-inhibitory N-terminal portion of BRAF that exhibits elevated kinase activity [68].

2.4. Alterations in the WNT Pathway

Mutations in genes encoding members of the WNT signaling pathway—i.e., Catenin Beta 1 (CTNNB1), AXIN1 and Adenomatous Polyposis Coli (APC)—are hallmarks of less differentiated thyroid carcinomas, in particular, ATCs [69]. Mutations in the transcription factor CTNNB1 are frequent events (>60%) [70,71], that modify its phosphorylation leading to protein stabilization because of reduced degradation [69]. These alterations become more frequent with loss of thyroid cancer differentiation (25% in PDTCs and 60–65% in ATCs) [25] (Figure 3).

2.5. Alterations in the TP53 Pathway

The tumor suppressor TP53 is a transcription factor involved in the control of the cell cycle and apoptosis (Figure 3). More than 75% of TP53 mutations are small nucleotide changes that inactivate the protein’s function. These changes are mostly located in the DNA-binding domain (residues 92–292) [72]. Considering the high prevalence of TP53 mutations in PDTCs (10–35%) and ATCs (40–80%), TP53 inactivation is considered a final step in tumor progression. Indeed, p53 deficiency, in association with activating mutations of oncogenes, such as RAS and BRAF, accounts for the high proliferation rate and increased aggressiveness of the more aggressive forms of thyroid cancer [73].

2.6. Other Molecular Alterations in Thyroid Cancer

EIF1AX: Mutations in the eukaryotic translation initiation factor 1A (EIF1AX) cause defects in the formation of the 43S pre-initiation complex for protein translation (Figure 3). Alterations of this gene are clustered in exons 2, 5 and 6, and the most common is the A113 splice mutation at the intron 5/exon 6 splice site, followed by a cluster of mutations in exon 2 [74]. EIF1AX mutations were detected in 1–2% of PTCs, largely occurring in a mutually exclusive manner with BRAF and RAS mutations [30], and more frequently in PDTCs (11%) and ATCs (9%) in which, on the contrary, they are strongly associated with RAS mutations [39].
IDH1: Mutations in the isocitrate dehydrogenase 1 (IDH1) gene are highly prevalent in thyroid carcinoma (16%) in particular in FTCs (5%) and ATCs (11%) [75,76] (Figure 3). Even if the most recurrent IDH1 mutation involves arginine at codon 132, no R132 amino acid change has been reported in thyroid tumors. All IDH1 substitutions found in thyroid samples concern five hotspot mutations in exon 4: G70D, G123R, I130M, H133Q and A134D. For some of these mutants, a reduced enzymatic activity has been demonstrated, suggesting a potential tumorigenic role of the IDH1 system in thyroid cancer [76].
PPARγ:Peroxisome proliferator activated receptor gamma (PPAR-γ) is a nuclear transcription factor that enhances apoptosis by activating caspases, up-regulating Bax and down-regulating bcl-2, survivin and c-myc [77,78]. Besides RAS mutations, rearrangements of the PPARγ gene are the most frequent alterations found in FTCs (20–50%) and may represent an initiating event in the transformation of follicular-derived cells [79]. PPARγ fusions may involve PAX8 and CREB3L2, but the PAX8-PPARγ rearrangement is the most frequent [35,80]. The PAX8-PPAR fusion protein (PPFP) acts as a dominant negative inhibitor of wild type PPARγ, thereby constitutively activating the transcription of a subset of PPARγ and PAX8 responsive genes [80] (Figure 3).
TERT: Activating mutations in the promoter of the telomerase reverse transcriptase (TERT) are mostly a late event in thyroid tumorigenesis [39,81]. They are more common in PDTCs (40%) and ATCs (70%), even if they can also be found in PTCs (10%) and FTCs (20%) [81], where they are associated with a poor prognosis [82]. Two mutually exclusive TERT promoter mutations are recurrent in thyroid cancer, one at position -124 (c228t) and one at position -146 (c250t) upstream of the TERT translation start site. Both mutations generate a consensus-binding site in the TERT promoter for E-twenty-six (ETS) transcription factors, which increase TERT transcriptional activities [81] (Figure 3). Finally, TERT mutations may co-occur with BRAF and RAS mutations in PDTCs and ATCs [82,83].
Additional alterations occurring in advanced follicular-derived thyroid cancer concern: (i) Members of the DNA Mismatch Repair pathway (MSH2, MSH6, and MLH1), mutated in 2% of PDTCs and 12% of ATCs that induce a “hypermutator phenotype”; (ii) histone methyl-transferases (HMTs), altered in 7% of PDTCs and 24% of ATCs; (iii) genes encoding for members of the SWI-SNF chromatin remodeling complex, mutated in 6% of PDTCs and 36% of ATCs [39]. Finally, specific histone deacetylase (HDAC) subtypes are associated with different thyroid cancer characteristics and behaviors [84] (Figure 3).

3. Targeted Therapies in Thyroid Cancer

Due to an improved understanding of thyroid cancer biology, in recent years, a plethora of targeted molecules have been tested while other compounds are currently under investigation. Herein, we thoroughly review the current landscape of targeted therapies for thyroid cancer. Ongoing trials awaiting preliminary results are also reported.

3.1. Tyrosine Kinase Inhibitors

3.1.1. Multi-Target Agents

Several molecules inhibiting tyrosine kinases involved in cell proliferation, survival and angiogenesis have shown clinical efficacy in both advanced RAIR DTCs and MTCs, while promising results are also beginning to emerge in ATCs (Figure 4 and Figure 5). To date, four drugs have received FDA approval, two for advanced RAIR DTCs (sorafenib and lenvatinib) and two for metastatic MTCs (vandetanib and cabozantinib). However, numerous trials have investigated additional tyrosine kinase inhibitors (TKIs), whereas many others are currently ongoing (Table 1).
Anlotinib: A multi-target TKI, displaying an affinity for VEGFR 2-3, FGFR 1-4, PDGFR α/β, c-KIT and RET [85]. After a phase I trial defined an MTD of 12 mg daily in a 2/1 schedule, a phase IIA study tested the drug on 58 chinese patients with advanced MTC. Partial responses (PR) occurred in 57% of patients, whereas median PFS was not reached at the time of data cut-off [85,86]. Results from a phase IIB randomized trial on 91 metastatic MTC patients were recently reported: mPFS was 20.7 months in the experimental arm and 11 months in the placebo arm (HR 0.53; p < 0.03), with a considerable benefit in terms of ORR (48% with anlotinib vs. 3.5% with placebo, p < 0.0001). Overall survival data are still immature. Adverse events were consistent throughout the trials, with hand-foot syndrome, hypertension and hyper-triglyceridemia representing the more frequent toxicities [87].
Axitinib: A VEGFR1-2-3 inhibitor [88] that also targets PDGFR-β and c-KIT. Its activity in thyroid tumors of any histology has been explored in two phase-II trials, comprehensively, including 112 patients. Results were consistent in the two studies, with an ORR of 30% in the first one and of 35% in the second, and median PFS of 18.1 and 16 months, respectively [89,90].
Cabozantinib: A selective inhibitor of MET, VEGFR-2 and RET [91]. In a phase I trial cabozantinib showed encouraging results in a cohort of 37 heavily pre-treated MTC patients [92]. These promising findings were further confirmed in the phase III EXAM study that randomized 330 patients with hereditary or sporadic advanced MTC to receive cabozantinib 140 mg daily or placebo, demonstrating a significant PFS benefit for subjects in the experimental arm (11.2 months vs. 4 months, HR 0.28, CI 0.19–0.40, p < 0.001). However, OS did not differ significantly between the two arms. The drug seems more effective in patients carrying a RET mutation [93,94]. In 2012 FDA approved cabozantinib for the treatment of advanced MTC [95]. More recently, the potential role of cabozantinib as salvage treatment in RAIR DTCs progressing on a VEGFR inhibitor has been explored in a phase II trial. Twenty-five enrolled patients presented a 40% ORR and a 92% disease control rate (DCR), with 12.7 months of median PFS and 34.47 months of median OS [96].
Imatinib: A multiple kinases inhibitor that targets ABL, c-KIT and PDGFR. This drug was tested both in advanced or relapsed ATCs and MTCs. A pilot trial with 11 ATC patients with proven PDGFR overexpression by IHC showed that 800 mg Imatinib/daily determined 2 PR and four assessments of stable disease [97]. However, in two additional studies, a total of 24 advanced MTCs received imatinib 600 mg daily with dismal results (i.e., no OR and sporadic disease stabilization) [98,99].
Lenvatinib: The drug inhibits several targets, including VEGFR 1-2-3, FGFR 1-2-3-4, PDGFR-α, RET and c-KIT [100]. Its activity in advanced RAIR DTCs has been proven in a phase II trial enrolling 58 patients, treatment naïve or pre-treated, and then confirmed in phase III SELECT trial, which randomized 261 subjects to receive 24 mg lenvatinib daily or placebo. Median PFS was 18.3 months with lenvatinib versus 3.6 months with placebo (HR 0.21; 95% CI 0.14 to 0.31; p < 0.0001), with a 64% OR rate, including four complete responses (CR). Median OS was not reached at the time of data cutoff. Most frequent adverse events were hypertension, diarrhea, fatigue, appetite and weight loss and nausea, with a 14% discontinuation rate [101,102]. Because of its favorable efficacy and safety profile, in 2015 lenvatinib was granted approval for the treatment of advanced RAIR DTCs and is currently the preferred therapeutic choice for this disease in the first line setting [103]. The drug also showed activity in advanced MTCs, since a phase II trial on 59 patients reported 36% OT rates (CI 24–49%) with 80% DCR (CI 67–89%) and nine months of median PFS (CI seven months-NE) [104].
Motesanib: A VEGFR 1-2-3, PDGFR, RET and c-KIT inhibitor [105] that demonstrated activity in two phase II trials enrolling RAIR DTCs and MTCs, respectively. Ninety-three patients with advanced RAIR DTCs received motesanib (125 mg daily)—13 (14%) experienced a PR, whereas 33 additional patients (35%) had disease stabilization >24 weeks [106]. The second trial recruited 91 patients with advanced symptomatic or progressive MTCs: only two patients (3%) achieved a PR, while 44 (48%) had an SD >24 weeks [107].
Pazopanib: A multi-kinase inhibitor targeting VEGFR 1-2-3, PDGFR-α and β, c-KIT and FGFR 1-3-4 [108]. Thirty-seven patients with RAIR DTCs, either pre-treated or TKI naïve, received the drug—at the daily dose of 800 mg—in a phase II trial. Results were encouraging, with almost 50% ORR, durable responses and a median PFS of 11.7 months [109]. Conversely, pazopanib showed negligible activity in 15 ATCs [110]. Finally, in 35 patients with progressive MTC, the drug demonstrated moderate efficacy, inducing PR in five patients (14%) and a 9.4 months median PFS [111].
Sorafenib: A multi-kinase inhibitor used against VEGFR 1-2-3, RET, RAF, PDGFR-β, c-KIT and FLT3 [112]. In a single arm phase II trial, Schneider and colleagues reported 18 months of median PFS [mPFS (CI 7–29 months)] and 34.5 months median OS (CI 19–50 months) in 31 patients with advanced RAIR DTC [113]. The subsequent phase III, multicenter, randomized, double blinded, placebo-controlled DECISION trial enrolled 417 advanced RAIR DTC patients in the first line setting. It showed significantly longer PFS in the experimental arm (10.8 months) compared with the placebo arm (5.8 months) [HR 0.59; 95% CI 0.45 to 0.76; p < 0.0001] and a 12% ORR, but failed to demonstrate a survival benefit in the group of patients treated with sorafenib 400 mg bis in die, which reported considerable toxicities leading to dose reduction or treatment discontinuation in more than 60% of cases [114]. Nevertheless, sorafenib is currently approved both by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) for the treatment of advanced RAIR DTC. Additionally, in a cohort of 20 pre-treated ATC patients, sorafenib induced durable PR in two patients and stable disease in five subjects, whereas it led to 1 PR and 14 disease stabilizations in 16 sporadic advanced MTCs [115,116]. However, the use of this drug in anaplastic and medullary thyroid carcinomas remains off label.
Sunitinib: A compound that displays a broad spectrum of activity against VEGFR 1-2, c-KIT, RET, PDGFR-β and FLT3 [117]. Preliminary results from a phase II trial on 33 evaluable patients with advanced RAIR DTC (n = 26) or MTC (n = 7) receiving sunitinib 37.5 mg on a continuous schedule indicated that 11 patients (31%) experienced a PR and 16 patients (46%) had SD, with a median time to progression of 12 months [118]. Another phase II study of continuous sunitinib in 23 RAIR DTC patients confirmed a good activity profile in this subset of patients. In fact, ORR was 26% and clinical benefit rate (CBR) 83% [119]. Additionally, the phase II THYSU trial investigated sunitinib activity at 50 mg/daily four weeks on followed by two weeks off, in 71 previously untreated advanced thyroid carcinomas of any histology. Nine out of 39 evaluable DTC patients had a disease response (1 CR and 8 PR; ORR 22%), whereas 10 of the 24 MTC patients reported a PR (ORR 38.5%). Of the four enrolled patients with ATC, only two were evaluable and experienced disease stabilization. PFS was 13.1 months and 16.5 months in DTCs and MTCs, respectively [120].
Vandetanib: A RET, VEGFR 2-3, c-KIT and EGFR inhibitor primarily tested in MTC [121,122]. Preliminary efficacy data came from a phase II trial on 30 locally advanced or metastatic hereditary MTC patients, 22 of which yielded PR or disease stabilization >24 weeks (73% DCR) with vandetanib 300 mg/daily [123]. The following phase III randomized, placebo-controlled study (ZETA trial) included 331 patients with hereditary or sporadic advanced MTC and confirmed the drug’s efficacy. Median PFS was 30.5 months in the vandetanib arm compared with 19.3 months in the placebo arm [HR 0.46, CI 0.31–0-69, p = 0.001], while OS results were still immature. Overall, a trend of enhanced efficacy in RET mutated disease emerged. Diarrhea, skin rash, nausea, hypertension and headache were the most frequent adverse reactions. Additionally, QT prolongation, potentially evolving in torsade de pointes and sudden death, represented an infrequent, but critical adverse event [124]. In 2011 vandetanib was the first FDA-approved drug for the treatment of symptomatic or progressive MTC in patients with unresectable, locally advanced, or metastatic disease. A special “black box” warning about QTc prolongation risk exists, hence the drug can only be prescribed by certified physicians [125]. Even though vandetanib is not indicated in advanced RAIR DTCs, it has been tested in a phase II randomized trial in this population, demonstrating a good activity profile (median PFS 11.1 months vs 5.9 months in patients receiving placebo, HR 0.63, 95% CI 0.54–0.74, p = 0.008) [126]. A phase III trial (VERIFY, NCT01876784) randomizing 238 subjects with RAIR DTC to receive vandetanib or placebo completed accrual, but no results are yet available.

3.1.2. Single-Target Agents

Although multi-kinase inhibitors represent the most studied agents for the treatment of thyroid cancer, several drugs selectively blocking a single altered protein may also be effective in oncogene-addicted disease (Figure 6). Different trials have already investigated the role of single target agents, whereas many others are still ongoing (Table 2). However, in the absence of a reliable predictive biomarker, the use of single-target molecules generally leads to unsatisfactory results.
Apatinib: Two small chinese trials evaluated apatinib, a selective VEGFR-2 inhibitor, in 30 patients with advanced RAIR DTC. The drug showed promising activity, both in terms of thyroglobulin levels reduction and tumor shrinkage [127,128]. Hence, further studies (phase II and III) assessing the role of apatinib for RAIR DTC treatment are currently ongoing (Table 2).
Dabrafeniband Trametinib: Combining a BRAF inhibitor (dabrafenib) with a MEK inhibitor (trametinib) may be an effective strategy for BRAFV600E mutated ATC, as reported in a phase II trial recruiting 16 pre-treated ATC patients. The reported 69% ORR, including a CR, along with the estimated 90% rate of ongoing responses at 12 months, represent unprecedented results for this aggressive disease [129,130]. Clinical trials employing dabrafenib in combination with trametinib or lapatinib are ongoing (Table 2).
Selumetinib: The drug targets MAP kinases MEK-1 and 2 and reversed iodine refractoriness in 8 of 20 patients with metastatic thyroid cancer, as assessed by 124I-PET. Selumetinib-treated patients subsequently received RAI achieving PR (n = 5) or SD (n = 3) after the radio-metabolic treatment. Additionally, the NRAS mutation seems to be a predictive biomarker of selumetinib efficacy [131]. Further studies are ongoing (Table 2).
Vemurafenib: A selective BRAF inhibitor, which showed promising results in a phase II trial conducted on 51 patients with advanced RAIR PTC displaying BRAFV600E. The study population comprised two cohorts according to previous treatment with an anti-VEGFR. Ten out of 26 (38.5%) TKI naïve patients achieved a PR, whereas nine patients (35%) maintained an SD for six months or longer, determining a 73% CBR, with a median PFS of 18.2 months. In the second TKI-exposed cohort, six of the 22 evaluable patients (27%) experienced a PR, while other six had SD for at least six months (CBR 54.6%) and a median PFS of 8.9 months [132]. A further study is ongoing (Table 2).

3.1.3. ALK Inhibitors

Due to its oncogenic activity, ALK represents a potential therapeutic target in many solid and hematologic cancers [133,134]. Currently, the therapeutic options for ALK-rearranged tumors include first- (crizotinib) or second-generation (ceritinib, alectinib and brigatinib) inhibitors or immunotherapeutic drugs directed against activated ALK (Figure 5). Ongoing trials employing ALK inhibitors are described in Table 2.
Ceritinib: A second-generation inhibitor that overcomes secondary resistance, due to acquired ALK mutations, amplification or activation of alternative—ALK-independent—survival pathways (e.g., EGF, IGF, RAS/SRC and AKT/mTOR signaling pathways) [134,135,136]. A study by Guan at al demonstrated limited efficacy of ceritinib in ATC patients with the ALKL1198F mutation in full-length ALK or the EML4-ALK fusion protein [137]. An additional trial is currently evaluating this drug in patients harboring ALK mutations or fusions (NCT02289144) (Table 2).
Crizotinib: A second generation TKI targeting ALK, MET and ROS1. This drug has been extensively investigated in ALK-fusion-positive tumors [138]. One ALK-translocated ATC patient treated with crizotinib achieved a PR [139].
Jun Ho Ji and colleagues reported a dramatic response with crizotinib in an advanced MTC patient harboring an ALK fusion (NCT01121588) [140]. Additionally, in a phase Ib study (PROFILE 1013; NCT01121588), which enrolled 44 ALK-positive metastatic patients, one individual diagnosed with MTC experienced a PR lasting 16.1 weeks [138].

3.1.4. NTRK Inhibitors

Selective inhibition of TRK signaling may be useful for patients with thyroid tumors that harbor an oncogenic NTRK translocation. Currently, four first and second generations TRK-inhibitors (entrectinib, larotrectinib, LOXO-195 and TPX-0005) have been developed and tested in clinical trials [141,142] (Table 2) (Figure 5).
Entrectinib: A pan-TRK inhibitor with additional activity against ROS1 and ALK [143]. A phase I/Ib study in children or young adults (aged 2–22 years) with or without NTRK, ROS1 or ALK fusions (STARTRK-NG, NCT02650401) and a phase II “basket trial” in adults with the same rearrangements (STARTRK-2, NCT02568267) enrolled patients with thyroid cancer. Results are still pending.
Larotrectinib: An ATP-competitive pan-TRK inhibitor, which received agnostic approval by the FDA for patients with advanced solid tumors harboring an NTRK gene fusion [144]. Fifty-five patients received larotrectinib in three different trials: A phase I in adults (NCT02122913), a phase I/II in pediatric patients (SCOUT, NCT02637687) and a phase II in adolescents and adults (NAVIGATE, NCT02576431), including five patients (9%) with thyroid cancer [145]. Although data specifically concerning the thyroid population are unavailable, promising results emerged in the intention-to-treat population, whit a 75% ORR (95% CI, 61% to 85%) and 71% of patients free from progression at 12 months. Median DOR and PFS were not reached after a median follow-up of 8.3 months and 9.9 months, respectively.
LOXO-195: A selective TRK inhibitor specifically developed to overcome acquired resistance that may occur in subjects receiving larotrectinib or other TRK inhibitors [145]. LOXO-195 is currently under evaluation in a phase I/II trial (NCT03215511) enrolling patients with NTRK-rearranged tumors—including thyroid cancer—previously treated with a TRK inhibitor.

3.2. PI3K/AKT/mTOR Pathway Inhibitors

Activation of the PI3K/AKT/mTOR pathway is a common feature in thyroid cancer [31,146,147]. Preclinical and clinical data suggest that targeting this pathway can be an effective strategy for the treatment of patients with advanced RAIR DTCs and MTCs [148]. To date, several trials have employed mTOR inhibitors in thyroid cancer (Table 3), whereas the phase II MATCH studies are testing PI3K (e.g., taselisib, copanlisib) and AKT-inhibitors (e.g., capivasertib) (Figure 6).
Buparlisib: A pan-class I PI3K inhibitor that failed to show a significant PFS benefit in 43 advanced, RAIR DTCs. Despite a reduction in tumor growth, the drug did not induce any objective response in the overall population, and 48.8% of patients had progressed after six months. The decrease in tumor growth may suggest an incomplete inhibition of the PI3K oncogenic pathway [149].
Everolimus: A phase II trial tested this mTOR inhibitor in 38 patients with advanced RAIR DTC. The authors reported an 81% DCR and a median PFS of 47 weeks [150]. Two additional phase II studies analyzed everolimus safety and efficacy on seven subjects with MTC, 28 patients with metastatic or locally advanced DTC and seven individuals with ATC. Five patients (71.4%) showed SD, and 4 (57.1%) had an SD lasting >24 weeks [151]. In the second study, 17 patients (65%) showed SD; in 15 of these 17 patients (58%), the response lasted >24 weeks. Median PFS and OS were 9 and 18 months, respectively [152]. A further phase II trial evaluated everolimus efficacy in patients with RAIR thyroid cancer and correlated tumor mutational profiling with response. Median PFS were 12.9, 13.1 and 2.2 for DTC, MTC and ATC cohorts, respectively, and patients with mutations in the PI3K pathway appeared to benefit most from drug treatment [153].
Since activation of the somatostatin receptor (SSTR1-5) also inhibits PI3K/AKT signaling, the somatostatin analog pasireotide has been tested in combination with everolimus [154]. Pasireotide activates SSTRs, in particular, subtype 2, which is the most expressed somatostatin receptor in thyroid cancers [155]. In a phase II trial, 19 patients with advanced MTC began pasireotide, achieving a median PFS of 36 months (95% CI 19.5–52.5). Seven patients with tumor progression received everolimus in combination with pasireotide, experiencing a median PFS of nine months (95% CI 0–21.83) [156]. Another phase II trial combining pasireotide with everolimus completed accrual and results are still awaited (Table 3).
Sirolimus: A retrospective study reported that this drug combined with cyclophosphamide generated PFS rates comparable to the standard of care for RAIR DTC. One-year PFS probability was 0.45 in the sirolimus plus cyclophosphamide cohort and 0.30 in the control population [157].
Temsirolimus: In a phase II trial temsirolimus was combined with sorafenib for the treatment of 36 RAIR thyroid cancer patients. Radiographic response rate was the primary endpoint. A PR was observed in 22% of cases, stable disease in 58% and progressive disease in 3% of patients. Individuals with any prior systemic treatment had a response rate of 10% compared to 38% for subjects with no prior systemic treatment [158].

3.3. PPAR-γ Agonist—Efatutazone

Different studies suggest that PPAR-γ agonists may inhibit tumor growth through the induction of terminal cell differentiation, cell cycle arrest, apoptosis, and angiogenesis inhibition [78,159]. Efatutazone, is a PPAR-γ agonist evaluated in combination with paclitaxel in a phase I study accruing patients diagnosed with ATC. Safety, potential effectiveness, and maximally tolerated dose were the end point of this trial. Results demonstrated that the combination was safe, with no dose-limiting toxicities (DLTs) and preliminary evidence of efficacy. Thus, these findings supported the addition of efatutazone to paclitaxel in patients with advanced ATC [160]. A further clinical trial is ongoing, but not yet recruiting (Table 2).

3.4. Histone Deacetylase Inhibitors—Valproic Acid

Several studies have shown that histone deacetylase (HDAC) inhibitors display promising effects for the treatment of several malignancies as they inhibit tumor proliferation, induce apoptosis, cell cycle arrest, and cancer differentiation [161,162,163].
In a phase II study valproic acid (VA) was administered to 13 patients with RAIR thyroid cancer of follicular origin. The primary endpoint of the study was to determine VA antitumor activity by evaluating measurable tumor response and/or decreased thyroglobulin levels. VA did not decrease tumor size and only generated a modest decrease in serum thyroglobulin levels. The secondary endpoint was to determine if VA could increase RAI uptake by the tumor cells, but results were disappointing [164].

4. Immunotherapy Landscape in Thyroid Cancer

In the last decade, the detailed understanding of the mechanism employed by cancer cells to elude the immune system have fostered a renewed interest for immune based-therapies [165]. Although thyroid carcinomas are not deemed to be “immunogenic” and display a low median tumor mutational burden (about 0.4 mutations/Mb) [166], several observations indicate a possible rationale for the use of immune checkpoint inhibitors in these tumors [167]. Indeed, immune cell infiltration, including natural killer cells, macrophages, mast cells, dendritic cells and T regulatory cells (Tregs) [168,169,170] has been reported in DTCs. Importantly, the relative amount of tumor-infiltrating lymphocytes and T-regs in primary thyroid cancer seems to correlate with prognosis [168,171]. Furthermore, the expression levels of programmed death 1 (PD-1) and programmed death ligand (PD-L1) correlate with higher risk of disease recurrence and reduced DFS [172,173]. To date, PD-L1 expression represents the most useful predictive biomarker to determine immunotherapy efficacy in thyroid tumors [168,174,175], with PD-L1 positivity ranging from 6.1 to 82.5% in PTCs and from 22.2% to 81.2% in ATCs [176].

Immune Checkpoint Inhibitors

Several clinical trials are investigating immune checkpoint inhibitors in thyroid cancer both alone or in combination with other drugs. A phase Ib trial tested the anti PD-1 antibody pembrolizumab on 22 PD-L1-positive patients with RAIR thyroid tumors. Clinical benefit rate was 50% (95% CI 28–72%), even though seven patients experienced early progression (32%; 95% CI 14–55%). Median PFS was seven months (95% CI 2–14 months), whereas median OS was not reached at the time of data cut-off (95% CI 22 months to NR) [177]. Based on these promising results, a phase II basket trial is currently ongoing (NCT02628067). Another study is testing the combination of the anti PD-1 nivolumab and the anti CTLA-4 ipilimumab in RAIR DTCs (NCT03246958).
Immunotherapy is also being evaluated in ATC. However, while the results of a phase II trial employing pembrolizumab are awaited for October 2019 (NCT02688608), a recently presented study failed to demonstrate any efficacy for the combination of the anti-PD-L1 durvalumab with the anti CTLA-4 tremelimumab and stereotactic radiation therapy in 12 patients with ATC [178]. Additional clinical trials are ongoing to establish if immune checkpoint inhibitors may prove of clinical benefit for thyroid cancer (Table 4).

5. Conclusions

In the last few years, a rapid advance in the knowledge of the molecular mechanisms underlying thyroid tumorigenesis along with the identification of pivotal driver genes contributing to disease progression has led to the introduction of several biological therapies, including TKIs, monoclonal antibodies and antibody-drug conjugates [179,180,181,182,183].
In this wide landscape of potentially targetable genomic alterations, RTKs modulating angiogenesis, proliferation and differentiation have represented the most easily druggable targets. Indeed, small molecules blocking these receptors have provided significant survival benefits for both RAIR DTCs and MTCs. However, these benefits have come at the cost of meaningful clinical and financial toxicities [184,185].
While additional studies are currently investigating other RTK-directed TKIs in DTCs and MTCs, the use of these drugs in ATC has been largely unsatisfactory. On the contrary, the combination of BRAF and MEK inhibitors has generated unprecedented response rates in patients diagnosed with these aggressive thyroid carcinomas, and validation of the published preliminary results is eagerly awaited with the hope that it may provide durable benefits comparable to those reported in BRAF-mutant melanoma.
Finally, despite not being classified as a highly immunogenic, preliminary findings suggest a possible benefit from immune checkpoint inhibitors in thyroid cancer, although these data are still immature. As in other types of solid tumors, it remains to be established which patients will derive meaningful benefits from this therapeutic approach [186].

Author Contributions

Conceptualization, E.T., F.M. and P.V.; formal analysis, M.M., M.S.P., S.S; data curation, E.T., F.M., C.R., S.R.V., G.M., S.D.G.; writing-original draft preparation, E.T., F.M., C.R., S.R.V., G.M., S.D.G.; writing-review and editing, E.T., F.M., C.R., S.R.V., G.M., S.D.G., M.M., M.S.P., S.S., A.P., F.G., M.R., L.M., P.V.; supervision, P.V.

Funding

This research received no external funding.

Acknowledgments

This study was partially funded by the 2016/2018 Research Plan of the University of Catania, Department of Clinical and Experimental Medicine (Project #B “Valutazione a distanza degli effetti nocivi della chemioterapia e della radioterapia su vari organi in pazienti affetti da patologie neoplastiche insorte in età pediatrica”).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed]
  2. US Preventive Services Task Force; Bibbins-Domingo, K.; Grossman, D.C.; Curry, S.J.; Barry, M.J.; Davidson, K.W.; Doubeni, C.A.; Epling, J.W., Jr.; Kemper, A.R.; Krist, A.H.; et al. Screening for Thyroid Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2017, 317, 1882–1887. [Google Scholar] [CrossRef] [PubMed]
  3. Pellegriti, G.; Frasca, F.; Regalbuto, C.; Squatrito, S.; Vigneri, R. Worldwide increasing incidence of thyroid cancer: Update on epidemiology and risk factors. J. Cancer Epidemiol. 2013, 2013, 965212. [Google Scholar] [CrossRef] [PubMed]
  4. Vigneri, R.; Malandrino, P.; Giani, F.; Russo, M.; Vigneri, P. Heavy metals in the volcanic environment and thyroid cancer. Mol. Cell. Endocrinol. 2017, 457, 73–80. [Google Scholar] [CrossRef] [PubMed]
  5. Vigneri, R.; Malandrino, P.; Vigneri, P. The changing epidemiology of thyroid cancer: Why is incidence increasing? Curr. Opin. Oncol. 2015, 27, 7. [Google Scholar] [CrossRef] [PubMed]
  6. Peacock, M. Calcium metabolism in health and disease. Clin. J. Am. Soc. Nephrol. 2010, 5 (Suppl. S1), S23–S30. [Google Scholar] [CrossRef]
  7. Cabanillas, M.E.; McFadden, D.G.; Durante, C. Thyroid cancer. Lancet 2016, 388, 2783–2795. [Google Scholar] [CrossRef]
  8. De la Fouchardiere, C.; Decaussin-Petrucci, M.; Berthiller, J.; Descotes, F.; Lopez, J.; Lifante, J.C.; Peix, J.L.; Giraudet, A.L.; Delahaye, A.; Masson, S.; et al. Predictive factors of outcome in poorly differentiated thyroid carcinomas. Eur. J. Cancer 2018, 92, 40–47. [Google Scholar] [CrossRef]
  9. Saini, S.; Tulla, K.; Maker, A.V.; Burman, K.D.; Prabhakar, B.S. Therapeutic advances in anaplastic thyroid cancer: A current perspective. Mol. Cancer 2018, 17, 154. [Google Scholar] [CrossRef]
  10. Randle, R.W.; Balentine, C.J.; Leverson, G.E.; Havlena, J.A.; Sippel, R.S.; Schneider, D.F.; Pitt, S.C. Trends in the presentation, treatment, and survival of patients with medullary thyroid cancer over the past 30 years. Surgery 2017, 161, 137–146. [Google Scholar] [CrossRef]
  11. Schneider, D.F.; Chen, H. New developments in the diagnosis and treatment of thyroid cancer. CA Cancer J. Clin. 2013, 63, 374–394. [Google Scholar] [CrossRef] [PubMed]
  12. Wells, S.A., Jr.; Asa, S.L.; Dralle, H.; Elisei, R.; Evans, D.B.; Gagel, R.F.; Lee, N.; 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]
  13. Sherman, S.I. Thyroid carcinoma. Lancet 2003, 361, 501–511. [Google Scholar] [CrossRef]
  14. Sampson, E.; Brierley, J.D.; Le, L.W.; Rotstein, L.; Tsang, R.W. Clinical management and outcome of papillary and follicular (differentiated) thyroid cancer presenting with distant metastasis at diagnosis. Cancer 2007, 110, 1451–1456. [Google Scholar] [CrossRef] [PubMed]
  15. Durante, C.; Haddy, N.; Baudin, E.; Leboulleux, S.; Hartl, D.; Travagli, J.P.; Caillou, B.; Ricard, M.; Lumbroso, J.D.; De Vathaire, F.; et al. Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: Benefits and limits of radioiodine therapy. J. Clin. Endocrinol. Metab. 2006, 91, 2892–2899. [Google Scholar] [CrossRef] [PubMed]
  16. Schlumberger, M.; Brose, M.; Elisei, R.; Leboulleux, S.; Luster, M.; Pitoia, F.; Pacini, F. Definition and management of radioactive iodine-refractory differentiated thyroid cancer. Lancet Diabetes Endocrinol. 2014, 2, 356–358. [Google Scholar] [CrossRef]
  17. Faugeras, L.; Pirson, A.S.; Donckier, J.; Michel, L.; Lemaire, J.; Vandervorst, S.; D’Hondt, L. Refractory thyroid carcinoma: Which systemic treatment to use? Ther. Adv. Med. Oncol. 2018, 10, 1758834017752853. [Google Scholar] [CrossRef] [PubMed]
  18. Tumino, D.; Frasca, F.; Newbold, K. Updates on the Management of Advanced, Metastatic, and Radioiodine Refractory Differentiated Thyroid Cancer. Front. Endocrinol. (Lausanne) 2017, 8, 312. [Google Scholar] [CrossRef] [PubMed]
  19. Priya, S.R.; Dravid, C.S.; Digumarti, R.; Dandekar, M. Targeted Therapy for Medullary Thyroid Cancer: A Review. Front. Oncol. 2017, 7, 238. [Google Scholar] [CrossRef] [PubMed]
  20. Acquaviva, G.; Visani, M.; Repaci, A.; Rhoden, K.J.; de Biase, D.; Pession, A.; Giovanni, T. Molecular pathology of thyroid tumours of follicular cells: A review of genetic alterations and their clinicopathological relevance. Histopathology 2018, 72, 6–31. [Google Scholar] [CrossRef] [PubMed]
  21. Krajewska, J.; Gawlik, T.; Jarzab, B. Advances in small molecule therapy for treating metastatic thyroid cancer. Expert Opin. Pharmacother. 2017, 18, 1049–1060. [Google Scholar] [CrossRef] [PubMed]
  22. Rao, S.N.; Zafereo, M.; Dadu, R.; Busaidy, N.L.; Hess, K.; Cote, G.J.; Williams, M.D.; William, W.N.; Sandulache, V.; Gross, N.; et al. Patterns of Treatment Failure in Anaplastic Thyroid Carcinoma. Thyroid 2017, 27, 672–681. [Google Scholar] [CrossRef] [PubMed]
  23. Massimino, M.; Tirro, E.; Stella, S.; Frasca, F.; Vella, V.; Sciacca, L.; Pennisi, M.S.; Vitale, S.R.; Puma, A.; Romano, C.; et al. Effect of Combined Epigenetic Treatments and Ectopic NIS Expression on Undifferentiated Thyroid Cancer Cells. Anticancer Res. 2018, 38, 6653–6662. [Google Scholar] [CrossRef] [PubMed]
  24. Massimino, M.; Vigneri, P.; Fallica, M.; Fidilio, A.; Aloisi, A.; Frasca, F.; Manzella, L. IRF5 promotes the proliferation of human thyroid cancer cells. Mol. Cancer 2012, 11, 21. [Google Scholar] [CrossRef] [PubMed]
  25. Xing, M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat. Rev. Cancer 2013, 13, 184–199. [Google Scholar] [CrossRef] [PubMed]
  26. Vella, V.; Malaguarnera, R. The Emerging Role of Insulin Receptor Isoforms in Thyroid Cancer: Clinical Implications and New Perspectives. Int. J. Mol. Sci. 2018, 19. [Google Scholar] [CrossRef] [PubMed]
  27. Vella, V.; Puppin, C.; Damante, G.; Vigneri, R.; Sanfilippo, M.; Vigneri, P.; Tell, G.; Frasca, F. DeltaNp73alpha inhibits PTEN expression in thyroid cancer cells. Int. J. Cancer 2009, 124, 2539–2548. [Google Scholar] [CrossRef] [PubMed]
  28. Schagdarsurengin, U.; Gimm, O.; Dralle, H.; Hoang-Vu, C.; Dammann, R. CpG island methylation of tumor-related promoters occurs preferentially in undifferentiated carcinoma. Thyroid 2006, 16, 633–642. [Google Scholar] [CrossRef]
  29. Zarkesh, M.; Zadeh-Vakili, A.; Azizi, F.; Foroughi, F.; Akhavan, M.M.; Hedayati, M. Altered Epigenetic Mechanisms in Thyroid Cancer Subtypes. Mol. Diagn. Ther. 2018, 22, 41–56. [Google Scholar] [CrossRef]
  30. Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef]
  31. 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] [PubMed]
  32. Regad, T. Targeting RTK Signaling Pathways in Cancer. Cancers 2015, 7, 1758–1784. [Google Scholar] [CrossRef] [PubMed]
  33. Manzella, L.; Massimino, M.; Stella, S.; Tirro, E.; Pennisi, M.S.; Martorana, F.; Motta, G.; Vitale, S.R.; Puma, A.; Romano, C.; et al. Activation of the IGF Axis in Thyroid Cancer: Implications for Tumorigenesis and Treatment. Int. J. Mol. Sci. 2019, 20. [Google Scholar] [CrossRef] [PubMed]
  34. Yakushina, V.D.; Lerner, L.V.; Lavrov, A.V. Gene Fusions in Thyroid Cancer. Thyroid 2018, 28, 158–167. [Google Scholar] [CrossRef] [PubMed]
  35. Demeure, M.J.; Aziz, M.; Rosenberg, R.; Gurley, S.D.; Bussey, K.J.; Carpten, J.D. Whole-genome sequencing of an aggressive BRAF wild-type papillary thyroid cancer identified EML4-ALK translocation as a therapeutic target. World J. Surg. 2014, 38, 1296–1305. [Google Scholar] [CrossRef] [PubMed]
  36. McFadden, D.G.; Dias-Santagata, D.; Sadow, P.M.; Lynch, K.D.; Lubitz, C.; Donovan, S.E.; Zheng, Z.; Le, L.; Iafrate, A.J.; Daniels, G.H. Identification of oncogenic mutations and gene fusions in the follicular variant of papillary thyroid carcinoma. J. Clin. Endocrinol. Metab. 2014, 99, E2457–E2462. [Google Scholar] [CrossRef]
  37. Kelly, L.M.; Barila, G.; Liu, P.; Evdokimova, V.N.; Trivedi, S.; Panebianco, F.; Gandhi, M.; Carty, S.E.; Hodak, S.P.; Luo, J.; et al. Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 4233–4238. [Google Scholar] [CrossRef]
  38. Landa, I.; Ibrahimpasic, T.; Boucai, L.; Sinha, R.; Knauf, J.A.; Shah, R.H.; 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]
  39. 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]
  40. Perot, 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]
  41. Leeman-Neill, R.J.; Kelly, L.M.; Liu, P.; Brenner, A.V.; Little, M.P.; Bogdanova, T.I.; Evdokimova, V.N.; Hatch, M.; Zurnadzy, L.Y.; Nikiforova, M.N.; et al. ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer. Cancer 2014, 120, 799–807. [Google Scholar] [CrossRef] [PubMed]
  42. Prasad, M.L.; Vyas, M.; Horne, M.J.; Virk, R.K.; Morotti, R.; Liu, Z.; Tallini, G.; Nikiforova, M.N.; Christison-Lagay, E.R.; Udelsman, R.; et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer 2016, 122, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
  43. Greco, A.; Miranda, C.; Pierotti, M.A. Rearrangements of NTRK1 gene in papillary thyroid carcinoma. Mol. Cell. Endocrinol. 2010, 321, 44–49. [Google Scholar] [CrossRef] [PubMed]
  44. Romei, C.; Ciampi, R.; Elisei, R. A comprehensive overview of the role of the RET proto-oncogene in thyroid carcinoma. Nat. Rev. Endocrinol. 2016, 12, 192–202. [Google Scholar] [CrossRef] [PubMed]
  45. Santoro, M.; Melillo, R.M.; Fusco, A. RET/PTC activation in papillary thyroid carcinoma: European Journal of Endocrinology Prize Lecture. Eur. J. Endocrinol. 2006, 155, 645–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Prescott, J.D.; Zeiger, M.A. The RET oncogene in papillary thyroid carcinoma. Cancer 2015, 121, 2137–2146. [Google Scholar] [CrossRef] [PubMed]
  47. Accardo, G.; Conzo, G.; Esposito, D.; Gambardella, C.; Mazzella, M.; Castaldo, F.; Di Donna, C.; Polistena, A.; Avenia, N.; Colantuoni, V.; et al. Genetics of medullary thyroid cancer: An overview. Int. J. Surg. 2017, 41 (Suppl. S1), S2–S6. [Google Scholar] [CrossRef]
  48. Mohammadi, M.; Hedayati, M. A Brief Review on the Molecular Basis of Medullary Thyroid Carcinoma. Cell J. 2017, 18, 485–492. [Google Scholar] [PubMed]
  49. Mulligan, L.M.; Eng, C.; Healey, C.S.; Clayton, D.; Kwok, J.B.; Gardner, E.; Ponder, M.A.; Frilling, A.; Jackson, C.E.; Lehnert, H.; et al. Specific mutations of the RET proto-oncogene are related to disease phenotype in MEN 2A and FMTC. Nat. Genet. 1994, 6, 70–74. [Google Scholar] [CrossRef]
  50. Marx, S.J. Molecular genetics of multiple endocrine neoplasia types 1 and 2. Nat. Rev. Cancer 2005, 5, 367–375. [Google Scholar] [CrossRef]
  51. Elisei, R.; Cosci, B.; Romei, C.; Bottici, V.; Renzini, G.; Molinaro, E.; Agate, L.; Vivaldi, A.; Faviana, P.; Basolo, F.; et al. Prognostic significance of somatic RET oncogene mutations in sporadic medullary thyroid cancer: A 10-year follow-up study. J. Clin. Endocrinol. Metab. 2008, 93, 682–687. [Google Scholar] [CrossRef] [PubMed]
  52. St Bernard, R.; Zheng, L.; Liu, W.; Winer, D.; Asa, S.L.; Ezzat, S. Fibroblast growth factor receptors as molecular targets in thyroid carcinoma. Endocrinology 2005, 146, 1145–1153. [Google Scholar] [CrossRef] [PubMed]
  53. Murugan, A.K.; Dong, J.; Xie, J.; Xing, M. Uncommon GNAQ, MMP8, AKT3, EGFR, and PIK3R1 mutations in thyroid cancers. Endocr. Pathol. 2011, 22, 97–102. [Google Scholar] [CrossRef] [PubMed]
  54. Gerber, T.S.; Schad, A.; Hartmann, N.; Springer, E.; Zechner, U.; Musholt, T.J. Targeted next-generation sequencing of cancer genes in poorly differentiated thyroid cancer. Endocr. Connect. 2018, 7, 47–55. [Google Scholar] [CrossRef] [Green Version]
  55. 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]
  56. Ricarte-Filho, J.C.; Ryder, M.; Chitale, D.A.; Rivera, M.; Heguy, A.; Ladanyi, M.; Janakiraman, M.; Solit, D.; Knauf, J.A.; 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 AKT1. Cancer Res. 2009, 69, 4885–4893. [Google Scholar] [CrossRef]
  57. Wu, G.; Mambo, E.; Guo, Z.; Hu, S.; Huang, X.; Gollin, S.M.; Trink, B.; Ladenson, P.W.; Sidransky, D.; Xing, M. Uncommon mutation, but common amplifications, of the PIK3CA gene in thyroid tumors. J. Clin. Endocrinol. Metab. 2005, 90, 4688–4693. [Google Scholar] [CrossRef]
  58. Hou, P.; Liu, D.; Shan, Y.; Hu, S.; Studeman, K.; Condouris, S.; Wang, Y.; Trink, A.; El-Naggar, A.K.; Tallini, G.; et al. Genetic alterations and their relationship in the phosphatidylinositol 3-kinase/Akt pathway in thyroid cancer. Clin. Cancer Res. 2007, 13, 1161–1170. [Google Scholar] [CrossRef]
  59. Wang, Y.; Hou, P.; Yu, H.; Wang, W.; Ji, M.; Zhao, S.; Yan, S.; Sun, X.; Liu, D.; Shi, B.; et al. High prevalence and mutual exclusivity of genetic alterations in the phosphatidylinositol-3-kinase/akt pathway in thyroid tumors. J. Clin. Endocrinol. Metab. 2007, 92, 2387–2390. [Google Scholar] [CrossRef]
  60. Hou, P.; Ji, M.; Xing, M. Association of PTEN gene methylation with genetic alterations in the phosphatidylinositol 3-kinase/AKT signaling pathway in thyroid tumors. Cancer 2008, 113, 2440–2447. [Google Scholar] [CrossRef]
  61. Vasko, V.; Ferrand, M.; Di Cristofaro, J.; Carayon, P.; Henry, J.F.; de Micco, C. Specific pattern of RAS oncogene mutations in follicular thyroid tumors. J. Clin. Endocrinol. Metab. 2003, 88, 2745–2752. [Google Scholar] [CrossRef] [PubMed]
  62. Agrawal, N.; Jiao, Y.; Sausen, M.; Leary, R.; Bettegowda, C.; Roberts, N.J.; Bhan, S.; Ho, A.S.; Khan, Z.; Bishop, J.; et al. Exomic sequencing of medullary thyroid cancer reveals dominant and mutually exclusive oncogenic mutations in RET and RAS. J. Clin. Endocrinol. Metab. 2013, 98, E364–E369. [Google Scholar] [CrossRef] [PubMed]
  63. DeLellis, R.A. Pathology and genetics of thyroid carcinoma. J. Surg. Oncol. 2006, 94, 662–669. [Google Scholar] [CrossRef] [PubMed]
  64. Nikiforova, M.N.; Kimura, E.T.; Gandhi, M.; Biddinger, P.W.; Knauf, J.A.; Basolo, F.; Zhu, Z.; Giannini, R.; Salvatore, G.; Fusco, A.; et al. BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J. Clin. Endocrinol. Metab. 2003, 88, 5399–5404. [Google Scholar] [CrossRef] [PubMed]
  65. De Biase, D.; Cesari, V.; Visani, M.; Casadei, G.P.; Cremonini, N.; Gandolfi, G.; Sancisi, V.; Ragazzi, M.; Pession, A.; Ciarrocchi, A.; et al. High-sensitivity BRAF mutation analysis: BRAF V600E is acquired early during tumor development but is heterogeneously distributed in a subset of papillary thyroid carcinomas. J. Clin. Endocrinol. Metab. 2014, 99, E1530–E1538. [Google Scholar] [CrossRef] [PubMed]
  66. Knauf, J.A.; Sartor, M.A.; Medvedovic, M.; Lundsmith, E.; Ryder, M.; Salzano, M.; Nikiforov, Y.E.; Giordano, T.J.; Ghossein, R.A.; Fagin, J.A. Progression of BRAF-induced thyroid cancer is associated with epithelial-mesenchymal transition requiring concomitant MAP kinase and TGFbeta signaling. Oncogene 2011, 30, 3153–3162. [Google Scholar] [CrossRef] [PubMed]
  67. Torregrossa, L.; Viola, D.; Sensi, E.; Giordano, M.; Piaggi, P.; Romei, C.; Materazzi, G.; Miccoli, P.; Elisei, R.; Basolo, F. Papillary Thyroid Carcinoma With Rare Exon 15 BRAF Mutation Has Indolent Behavior: A Single-Institution Experience. J. Clin. Endocrinol. Metab. 2016, 101, 4413–4420. [Google Scholar] [CrossRef] [Green Version]
  68. Ciampi, R.; Knauf, J.A.; Kerler, R.; Gandhi, M.; Zhu, Z.; Nikiforova, M.N.; Rabes, H.M.; Fagin, J.A.; Nikiforov, Y.E. Oncogenic AKAP9-BRAF fusion is a novel mechanism of MAPK pathway activation in thyroid cancer. J. Clin. Investig. 2005, 115, 94–101. [Google Scholar] [CrossRef] [Green Version]
  69. Sastre-Perona, A.; Santisteban, P. Role of the wnt pathway in thyroid cancer. Front. Endocrinol (Lausanne) 2012, 3, 31. [Google Scholar] [CrossRef]
  70. Garcia-Rostan, G.; Camp, R.L.; Herrero, A.; Carcangiu, M.L.; Rimm, D.L.; Tallini, G. Beta-catenin dysregulation in thyroid neoplasms: Down-regulation, aberrant nuclear expression, and CTNNB1 exon 3 mutations are markers for aggressive tumor phenotypes and poor prognosis. Am. J. Pathol. 2001, 158, 987–996. [Google Scholar] [CrossRef]
  71. Garcia-Rostan, G.; Tallini, G.; Herrero, A.; D’Aquila, T.G.; Carcangiu, M.L.; Rimm, D.L. Frequent mutation and nuclear localization of beta-catenin in anaplastic thyroid carcinoma. Cancer Res. 1999, 59, 1811–1815. [Google Scholar] [PubMed]
  72. Levine, A.J.; Oren, M. The first 30 years of p53: Growing ever more complex. Nat. Rev. Cancer 2009, 9, 749–758. [Google Scholar] [CrossRef] [PubMed]
  73. Manzella, L.; Stella, S.; Pennisi, M.S.; Tirro, 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. [Google Scholar] [CrossRef] [PubMed]
  74. 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]
  75. 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]
  76. 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] [Green Version]
  77. Sauer, S. Ligands for the Nuclear Peroxisome Proliferator-Activated Receptor Gamma. Trends Pharmacol. Sci. 2015, 36, 688–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Theocharis, S.; Margeli, A.; Vielh, P.; Kouraklis, G. Peroxisome proliferator-activated receptor-gamma ligands as cell-cycle modulators. Cancer Treat. Rev. 2004, 30, 545–554. [Google Scholar] [CrossRef] [PubMed]
  79. Eberhardt, N.L.; Grebe, S.K.; McIver, B.; Reddi, H.V. The role of the PAX8/PPARgamma fusion oncogene in the pathogenesis of follicular thyroid cancer. Mol. Cell. Endocrinol. 2010, 321, 50–56. [Google Scholar] [CrossRef]
  80. Raman, P.; Koenig, R.J. Pax-8-PPAR-gamma fusion protein in thyroid carcinoma. Nat. Rev. Endocrinol. 2014, 10, 616–623. [Google Scholar] [CrossRef]
  81. Liu, R.; Xing, M. TERT promoter mutations in thyroid cancer. Endocr. Relat. Cancer 2016, 23, R143–R155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Melo, M.; da Rocha, A.G.; Vinagre, J.; Batista, R.; Peixoto, J.; Tavares, C.; Celestino, R.; Almeida, A.; Salgado, C.; Eloy, C.; et al. TERT promoter mutations are a major indicator of poor outcome in differentiated thyroid carcinomas. J. Clin. Endocrinol. Metab. 2014, 99, E754–E765. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, X.; Qu, S.; Liu, R.; Sheng, C.; Shi, X.; Zhu, G.; Murugan, A.K.; Guan, H.; Yu, H.; Wang, Y.; et al. TERT promoter mutations and their association with BRAF V600E mutation and aggressive clinicopathological characteristics of thyroid cancer. J. Clin. Endocrinol. Metab. 2014, 99, E1130–E1136. [Google Scholar] [CrossRef] [PubMed]
  84. Giaginis, C.; Alexandrou, P.; Delladetsima, I.; Giannopoulou, I.; Patsouris, E.; Theocharis, S. Clinical significance of histone deacetylase (HDAC)-1, HDAC-2, HDAC-4, and HDAC-6 expression in human malignant and benign thyroid lesions. Tumour Biol. 2014, 35, 61–71. [Google Scholar] [CrossRef] [PubMed]
  85. Sun, Y.; Niu, W.; Du, F.; Du, C.; Li, S.; Wang, J.; Li, L.; Wang, F.; Hao, Y.; Li, C.; et al. Safety, pharmacokinetics, and antitumor properties of anlotinib, an oral multi-target tyrosine kinase inhibitor, in patients with advanced refractory solid tumors. J. Hematol. Oncol. 2016, 9, 105. [Google Scholar] [CrossRef] [PubMed]
  86. Sun, Y.; Du, F.; Gao, M.; Ji, Q.; Li, Z.; Zhang, Y.; Guo, Z.; Wang, J.; Chen, X.; Wang, J.; et al. Anlotinib for the Treatment of Patients with Locally Advanced or Metastatic Medullary Thyroid Cancer. Thyroid 2018, 28, 1455–1461. [Google Scholar] [CrossRef] [PubMed]
  87. Li, D.; Tang, P.Z.; Chen, X.; Ge, M.; Zhang, Y.; Guo, Z.; Wang, J.; Shi, F.; Zhang, J.; Cheng, Y.; et al. Anlotinib treatment in locally advanced or metastatic medullary thyroid carcinoma: A multicenter, randomized, double-blind, placebo-controlled phase IIB trial. J. Clin. Oncol. 2019, 37, 6019. [Google Scholar] [CrossRef]
  88. Rugo, H.S.; Herbst, R.S.; Liu, G.; Park, J.W.; Kies, M.S.; Steinfeldt, H.M.; Pithavala, Y.K.; Reich, S.D.; Freddo, J.L.; Wilding, G. Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: Pharmacokinetic and clinical results. J. Clin. Oncol. 2005, 23, 5474–5483. [Google Scholar] [CrossRef] [PubMed]
  89. Cohen, E.E.; Rosen, L.S.; Vokes, E.E.; Kies, M.S.; Forastiere, A.A.; Worden, F.P.; Kane, M.A.; Sherman, E.; Kim, S.; Bycott, P.; et al. Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: Results from a phase II study. J. Clin. Oncol. 2008, 26, 4708–4713. [Google Scholar] [CrossRef] [PubMed]
  90. Locati, L.D.; Licitra, L.; Agate, L.; Ou, S.H.; Boucher, A.; Jarzab, B.; Qin, S.; Kane, M.A.; Wirth, L.J.; Chen, C.; et al. Treatment of advanced thyroid cancer with axitinib: Phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments. Cancer 2014, 120, 2694–2703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Yakes, F.M.; Chen, J.; Tan, J.; Yamaguchi, K.; Shi, Y.; Yu, P.; Qian, F.; Chu, F.; Bentzien, F.; Cancilla, B.; et al. Cabozantinib (XL184), a novel MET and VEGFR2 inhibitor, simultaneously suppresses metastasis, angiogenesis, and tumor growth. Mol. Cancer Ther. 2011, 10, 2298–2308. [Google Scholar] [CrossRef]
  92. Kurzrock, R.; Sherman, S.I.; Ball, D.W.; Forastiere, A.A.; Cohen, R.B.; Mehra, R.; Pfister, D.G.; Cohen, E.E.; Janisch, L.; Nauling, F.; et al. Activity of XL184 (Cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J. Clin. Oncol. 2011, 29, 2660–2666. [Google Scholar] [CrossRef]
  93. Elisei, R.; Schlumberger, M.J.; Muller, S.P.; Schoffski, P.; Brose, M.S.; Shah, M.H.; Licitra, L.; Jarzab, B.; Medvedev, V.; Kreissl, M.C.; et al. Cabozantinib in progressive medullary thyroid cancer. J. Clin. Oncol. 2013, 31, 3639–3646. [Google Scholar] [CrossRef]
  94. Schlumberger, M.; Elisei, R.; Muller, S.; Schoffski, P.; Brose, M.; Shah, M.; Licitra, L.; Krajewska, J.; Kreissl, M.C.; Niederle, B.; et al. Overall survival analysis of EXAM, a phase III trial of cabozantinib in patients with radiographically progressive medullary thyroid carcinoma. Ann. Oncol. 2017, 28, 2813–2819. [Google Scholar] [CrossRef]
  95. Traynor, K. Cabozantinib approved for advanced medullary thyroid cancer. Am. J. Health Syst. Pharm. 2013, 70, 88. [Google Scholar] [CrossRef]
  96. Cabanillas, M.E.; de Souza, J.A.; Geyer, S.; Wirth, L.J.; Menefee, M.E.; Liu, S.V.; Shah, K.; Wright, J.; Shah, M.H. Cabozantinib As Salvage Therapy for Patients With Tyrosine Kinase Inhibitor-Refractory Differentiated Thyroid Cancer: Results of a Multicenter Phase II International Thyroid Oncology Group Trial. J. Clin. Oncol. 2017, 35, 3315–3321. [Google Scholar] [CrossRef]
  97. Ha, H.T.; Lee, J.S.; Urba, S.; Koenig, R.J.; Sisson, J.; Giordano, T.; Worden, F.P. A phase II study of imatinib in patients with advanced anaplastic thyroid cancer. Thyroid 2010, 20, 975–980. [Google Scholar] [CrossRef]
  98. Frank-Raue, K.; Fabel, M.; Delorme, S.; Haberkorn, U.; Raue, F. Efficacy of imatinib mesylate in advanced medullary thyroid carcinoma. Eur. J. Endocrinol. 2007, 157, 215–220. [Google Scholar] [CrossRef] [Green Version]
  99. De Groot, J.W.; Zonnenberg, B.A.; van Ufford-Mannesse, P.Q.; de Vries, M.M.; Links, T.P.; Lips, C.J.; Voest, E.E. A phase II trial of imatinib therapy for metastatic medullary thyroid carcinoma. J. Clin. Endocrinol. Metab. 2007, 92, 3466–3469. [Google Scholar] [CrossRef]
  100. Okamoto, K.; Kodama, K.; Takase, K.; Sugi, N.H.; Yamamoto, Y.; Iwata, M.; Tsuruoka, A. Antitumor activities of the targeted multi-tyrosine kinase inhibitor lenvatinib (E7080) against RET gene fusion-driven tumor models. Cancer Lett. 2013, 340, 97–103. [Google Scholar] [CrossRef] [Green Version]
  101. Cabanillas, M.E.; Schlumberger, M.; Jarzab, B.; Martins, R.G.; Pacini, F.; Robinson, B.; McCaffrey, J.C.; Shah, M.H.; Bodenner, D.L.; Topliss, D.; et al. A phase 2 trial of lenvatinib (E7080) in advanced, progressive, radioiodine-refractory, differentiated thyroid cancer: A clinical outcomes and biomarker assessment. Cancer 2015, 121, 2749–2756. [Google Scholar] [CrossRef]
  102. Schlumberger, M.; Tahara, M.; Wirth, L.J.; Robinson, B.; Brose, M.S.; Elisei, R.; Habra, M.A.; Newbold, K.; Shah, M.H.; Hoff, A.O.; et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N. Engl. J. Med. 2015, 372, 621–630. [Google Scholar] [CrossRef]
  103. Haddad, R.I.; Nasr, C.; Bischoff, L.; Busaidy, N.L.; Byrd, D.; Callender, G.; Dickson, P.; Duh, Q.Y.; Ehya, H.; Goldner, W.; et al. NCCN Guidelines Insights: Thyroid Carcinoma, Version 2.2018. J. Natl. Compr. Cancer Netw. 2018, 16, 1429–1440. [Google Scholar] [CrossRef] [Green Version]
  104. Schlumberger, M.; Jarzab, B.; Cabanillas, M.E.; Robinson, B.; Pacini, F.; Ball, D.W.; McCaffrey, J.; Newbold, K.; Allison, R.; Martins, R.G.; et al. A Phase II Trial of the Multitargeted Tyrosine Kinase Inhibitor Lenvatinib (E7080) in Advanced Medullary Thyroid Cancer. Clin. Cancer Res. 2016, 22, 44–53. [Google Scholar] [CrossRef]
  105. Polverino, A.; Coxon, A.; Starnes, C.; Diaz, Z.; DeMelfi, T.; Wang, L.; Bready, J.; Estrada, J.; Cattley, R.; Kaufman, S.; et al. AMG 706, an oral, multikinase inhibitor that selectively targets vascular endothelial growth factor, platelet-derived growth factor, and kit receptors, potently inhibits angiogenesis and induces regression in tumor xenografts. Cancer Res. 2006, 66, 8715–8721. [Google Scholar] [CrossRef]
  106. Sherman, S.I.; Wirth, L.J.; Droz, J.P.; Hofmann, M.; Bastholt, L.; Martins, R.G.; Licitra, L.; Eschenberg, M.J.; Sun, Y.N.; Juan, T.; et al. Motesanib diphosphate in progressive differentiated thyroid cancer. N. Engl. J. Med. 2008, 359, 31–42. [Google Scholar] [CrossRef]
  107. Schlumberger, M.J.; Elisei, R.; Bastholt, L.; Wirth, L.J.; Martins, R.G.; Locati, L.D.; Jarzab, B.; Pacini, F.; Daumerie, C.; Droz, J.P.; et al. Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer. J. Clin. Oncol. 2009, 27, 3794–3801. [Google Scholar] [CrossRef]
  108. Kumar, R.; Knick, V.B.; Rudolph, S.K.; Johnson, J.H.; Crosby, R.M.; Crouthamel, M.C.; Hopper, T.M.; Miller, C.G.; Harrington, L.E.; Onori, J.A.; et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol. Cancer Ther. 2007, 6, 2012–2021. [Google Scholar] [CrossRef]
  109. Bible, K.C.; Suman, V.J.; Molina, J.R.; Smallridge, R.C.; Maples, W.J.; Menefee, M.E.; Rubin, J.; Sideras, K.; Morris, J.C., 3rd; McIver, B.; et al. Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: Results of a phase 2 consortium study. Lancet Oncol. 2010, 11, 962–972. [Google Scholar] [CrossRef]
  110. Bible, K.C.; Suman, V.J.; Menefee, M.E.; Smallridge, R.C.; Molina, J.R.; Maples, W.J.; Karlin, N.J.; Traynor, A.M.; Kumar, P.; Goh, B.C.; et al. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J. Clin. Endocrinol. Metab. 2012, 97, 3179–3184. [Google Scholar] [CrossRef]
  111. Bible, K.C.; Suman, V.J.; Molina, J.R.; Smallridge, R.C.; Maples, W.J.; Menefee, M.E.; Rubin, J.; Karlin, N.; Sideras, K.; Morris, J.C., 3rd; et al. A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: MC057H. J. Clin. Endocrinol. Metab. 2014, 99, 1687–1693. [Google Scholar] [CrossRef]
  112. Wilhelm, S.M.; Carter, C.; Tang, L.; Wilkie, D.; McNabola, A.; Rong, H.; Chen, C.; Zhang, X.; Vincent, P.; McHugh, M.; et al. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004, 64, 7099–7109. [Google Scholar] [CrossRef]
  113. Schneider, T.C.; Abdulrahman, R.M.; Corssmit, E.P.; Morreau, H.; Smit, J.W.; Kapiteijn, E. Long-term analysis of the efficacy and tolerability of sorafenib in advanced radio-iodine refractory differentiated thyroid carcinoma: Final results of a phase II trial. Eur. J. Endocrinol. 2012, 167, 643–650. [Google Scholar] [CrossRef]
  114. Brose, M.S.; Nutting, C.M.; Jarzab, B.; Elisei, R.; Siena, S.; Bastholt, L.; de la Fouchardiere, C.; Pacini, F.; Paschke, R.; Shong, Y.K.; et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: A randomised, double-blind, phase 3 trial. Lancet 2014, 384, 319–328. [Google Scholar] [CrossRef]
  115. Lam, E.T.; Ringel, M.D.; Kloos, R.T.; Prior, T.W.; Knopp, M.V.; Liang, J.; Sammet, S.; Hall, N.C.; Wakely, P.E., Jr.; Vasko, V.V.; et al. Phase II clinical trial of sorafenib in metastatic medullary thyroid cancer. J. Clin. Oncol. 2010, 28, 2323–2330. [Google Scholar] [CrossRef]
  116. Savvides, P.; Nagaiah, G.; Lavertu, P.; Fu, P.; Wright, J.J.; Chapman, R.; Wasman, J.; Dowlati, A.; Remick, S.C. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid 2013, 23, 600–604. [Google Scholar] [CrossRef]
  117. Chow, L.Q.; Eckhardt, S.G. Sunitinib: From rational design to clinical efficacy. J. Clin. Oncol. 2007, 25, 884–896. [Google Scholar] [CrossRef]
  118. Carr, L.L.; Mankoff, D.A.; Goulart, B.H.; Eaton, K.D.; Capell, P.T.; Kell, E.M.; Bauman, J.E.; Martins, R.G. Phase II study of daily sunitinib in FDG-PET-positive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation. Clin. Cancer Res. 2010, 16, 5260–5268. [Google Scholar] [CrossRef]
  119. Bikas, A.; Kundra, P.; Desale, S.; Mete, M.; O’Keefe, K.; Clark, B.G.; Wray, L.; Gandhi, R.; Barett, C.; Jelinek, J.S.; et al. Phase 2 clinical trial of sunitinib as adjunctive treatment in patients with advanced differentiated thyroid cancer. Eur. J. Endocrinol. 2016, 174, 373–380. [Google Scholar] [CrossRef] [Green Version]
  120. Ravaud, A.; de la Fouchardiere, C.; Caron, P.; Doussau, A.; Do Cao, C.; Asselineau, J.; Rodien, P.; Pouessel, D.; Nicolli-Sire, P.; Klein, M.; et al. A multicenter phase II study of sunitinib in patients with locally advanced or metastatic differentiated, anaplastic or medullary thyroid carcinomas: Mature data from the THYSU study. Eur. J. Cancer 2017, 76, 110–117. [Google Scholar] [CrossRef]
  121. Carlomagno, F.; Vitagliano, D.; Guida, T.; Ciardiello, F.; Tortora, G.; Vecchio, G.; Ryan, A.J.; Fontanini, G.; Fusco, A.; Santoro, M. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 2002, 62, 7284–7290. [Google Scholar] [PubMed]
  122. Wedge, S.R.; Ogilvie, D.J.; Dukes, M.; Kendrew, J.; Chester, R.; Jackson, J.A.; Boffey, S.J.; Valentine, P.J.; Curwen, J.O.; Musgrove, H.L.; et al. ZD6474 inhibits vascular endothelial growth factor signaling, angiogenesis, and tumor growth following oral administration. Cancer Res. 2002, 62, 4645–4655. [Google Scholar] [PubMed]
  123. Wells, S.A., Jr.; Gosnell, J.E.; Gagel, R.F.; Moley, J.; Pfister, D.; Sosa, J.A.; Skinner, M.; Krebs, A.; Vasselli, J.; Schlumberger, M. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J. Clin. Oncol. 2010, 28, 767–772. [Google Scholar] [CrossRef] [PubMed]
  124. Wells, S.A., Jr.; Robinson, B.G.; Gagel, R.F.; Dralle, H.; Fagin, J.A.; Santoro, M.; Baudin, E.; Elisei, R.; Jarzab, B.; Vasselli, J.R.; et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: A randomized, double-blind phase III trial. J. Clin. Oncol. 2012, 30, 134–141. [Google Scholar] [CrossRef] [PubMed]
  125. Thornton, K.; Kim, G.; Maher, V.E.; Chattopadhyay, S.; Tang, S.; Moon, Y.J.; Song, P.; Marathe, A.; Balakrishnan, S.; Zhu, H.; et al. Vandetanib for the treatment of symptomatic or progressive medullary thyroid cancer in patients with unresectable locally advanced or metastatic disease: U.S. Food and Drug Administration drug approval summary. Clin. Cancer Res. 2012, 18, 3722–3730. [Google Scholar] [CrossRef] [PubMed]
  126. Leboulleux, S.; Bastholt, L.; Krause, T.; de la Fouchardiere, C.; Tennvall, J.; Awada, A.; Gomez, J.M.; Bonichon, F.; Leenhardt, L.; Soufflet, C.; et al. Vandetanib in locally advanced or metastatic differentiated thyroid cancer: A randomised, double-blind, phase 2 trial. Lancet Oncol. 2012, 13, 897–905. [Google Scholar] [CrossRef]
  127. Lin, Y.; Wang, C.; Gao, W.; Cui, R.; Liang, J. Overwhelming rapid metabolic and structural response to apatinib in radioiodine refractory differentiated thyroid cancer. Oncotarget 2017, 8, 42252–42261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Zhang, X.; Wang, C.; Lin, Y. Pilot Dose Comparison of Apatinib in Chinese Patients With Progressive Radioiodine-Refractory Differentiated Thyroid Cancer. J. Clin. Endocrinol. Metab. 2018, 103, 3640–3646. [Google Scholar] [CrossRef]
  129. Falchook, G.S.; Millward, M.; Hong, D.; Naing, A.; Piha-Paul, S.; Waguespack, S.G.; Cabanillas, M.E.; Sherman, S.I.; Ma, B.; Curtis, M.; et al. BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer. Thyroid 2015, 25, 71–77. [Google Scholar] [CrossRef]
  130. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.; Cabanillas, M.E.; Urbanowitz, G.; et al. Dabrafenib and Trametinib Treatment in Patients With Locally Advanced or Metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer. J. Clin. Oncol. 2018, 36, 7–13. [Google Scholar] [CrossRef]
  131. Ho, A.L.; Grewal, R.K.; Leboeuf, R.; Sherman, E.J.; Pfister, D.G.; Deandreis, D.; Pentlow, K.S.; Zanzonico, P.B.; Haque, S.; Gavane, S.; et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. N. Engl. J. Med. 2013, 368, 623–632. [Google Scholar] [CrossRef] [PubMed]
  132. Brose, M.S.; Cabanillas, M.E.; Cohen, E.E.; Wirth, L.J.; Riehl, T.; Yue, H.; Sherman, S.I.; Sherman, E.J. Vemurafenib in patients with BRAF(V600E)-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: A non-randomised, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 1272–1282. [Google Scholar] [CrossRef]
  133. Bilsland, J.G.; Wheeldon, A.; Mead, A.; Znamenskiy, P.; Almond, S.; Waters, K.A.; Thakur, M.; Beaumont, V.; Bonnert, T.P.; Heavens, R.; et al. Behavioral and neurochemical alterations in mice deficient in anaplastic lymphoma kinase suggest therapeutic potential for psychiatric indications. Neuropsychopharmacology 2008, 33, 685–700. [Google Scholar] [CrossRef] [PubMed]
  134. Cao, Z.; Gao, Q.; Fu, M.; Ni, N.; Pei, Y.; Ou, W.B. Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives. Oncol. Lett. 2019, 17, 2020–2030. [Google Scholar] [CrossRef] [PubMed]
  135. Crystal, A.S.; Shaw, A.T.; Sequist, L.V.; Friboulet, L.; Niederst, M.J.; Lockerman, E.L.; Frias, R.L.; Gainor, J.F.; Amzallag, A.; Greninger, P.; et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science 2014, 346, 1480–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Doebele, R.C.; Pilling, A.B.; Aisner, D.L.; Kutateladze, T.G.; Le, A.T.; Weickhardt, A.J.; Kondo, K.L.; Linderman, D.J.; Heasley, L.E.; Franklin, W.A.; et al. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin. Cancer Res. 2012, 18, 1472–1482. [Google Scholar] [CrossRef] [PubMed]
  137. Guan, J.; Wolfstetter, G.; Siaw, J.; Chand, D.; Hugosson, F.; Palmer, R.H.; Hallberg, B. Anaplastic lymphoma kinase L1198F and G1201E mutations identified in anaplastic thyroid cancer patients are not ligand-independent. Oncotarget 2017, 8, 11566–11578. [Google Scholar] [CrossRef] [PubMed]
  138. Gambacorti-Passerini, C.; Orlov, S.; Zhang, L.; Braiteh, F.; Huang, H.; Esaki, T.; Horibe, K.; Ahn, J.S.; Beck, J.T.; Edenfield, W.J.; et al. Long-term effects of crizotinib in ALK-positive tumors (excluding NSCLC): A phase 1b open-label study. Am. J. Hematol. 2018, 93, 607–614. [Google Scholar] [CrossRef] [PubMed]
  139. Godbert, Y.; Henriques de Figueiredo, B.; Bonichon, F.; Chibon, F.; Hostein, I.; Perot, G.; Dupin, C.; Daubech, A.; Belleannee, G.; Gros, A.; et al. Remarkable Response to Crizotinib in Woman With Anaplastic Lymphoma Kinase-Rearranged Anaplastic Thyroid Carcinoma. J. Clin. Oncol. 2015, 33, e84–e87. [Google Scholar] [CrossRef] [PubMed]
  140. Ji, J.H.; Oh, Y.L.; Hong, M.; Yun, J.W.; Lee, H.W.; Kim, D.; Ji, Y.; Kim, D.H.; Park, W.Y.; Shin, H.T.; et al. Identification of Driving ALK Fusion Genes and Genomic Landscape of Medullary Thyroid Cancer. PLoS Genet. 2015, 11, e1005467. [Google Scholar] [CrossRef] [PubMed]
  141. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef] [PubMed]
  142. Khotskaya, Y.B.; Holla, V.R.; Farago, A.F.; Mills Shaw, K.R.; Meric-Bernstam, F.; Hong, D.S. Targeting TRK family proteins in cancer. Pharmacol. Ther. 2017, 173, 58–66. [Google Scholar] [CrossRef] [PubMed]
  143. Rolfo, C.; Ruiz, R.; Giovannetti, E.; Gil-Bazo, I.; Russo, A.; Passiglia, F.; Giallombardo, M.; Peeters, M.; Raez, L. Entrectinib: A potent new TRK, ROS1, and ALK inhibitor. Expert Opin. Investig. Drugs 2015, 24, 1493–1500. [Google Scholar] [CrossRef] [PubMed]
  144. Hong, D.S.; Bauer, T.M.; Lee, J.J.; Dowlati, A.; Brose, M.S.; Farago, A.F.; Taylor, M.; Shaw, A.T.; Montez, S.; Meric-Bernstam, F.; et al. Larotrectinib in adult patients with solid tumours: A multi-centre, open-label, phase I dose-escalation study. Ann. Oncol. 2019, 30, 325–331. [Google Scholar] [CrossRef] [PubMed]
  145. Drilon, A.; Nagasubramanian, R.; Blake, J.F.; Ku, N.; Tuch, B.B.; Ebata, K.; Smith, S.; Lauriault, V.; Kolakowski, G.R.; Brandhuber, B.J.; et al. A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov. 2017, 7, 963–972. [Google Scholar] [CrossRef] [PubMed]
  146. Yeager, N.; Brewer, C.; Cai, K.Q.; Xu, X.X.; Di Cristofano, A. Mammalian target of rapamycin is the key effector of phosphatidylinositol-3-OH-initiated proliferative signals in the thyroid follicular epithelium. Cancer Res. 2008, 68, 444–449. [Google Scholar] [CrossRef] [PubMed]
  147. Saji, M.; Ringel, M.D. The PI3K-Akt-mTOR pathway in initiation and progression of thyroid tumors. Mol. Cell. Endocrinol. 2010, 321, 20–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Jin, N.; Jiang, T.; Rosen, D.M.; Nelkin, B.D.; Ball, D.W. Synergistic action of a RAF inhibitor and a dual PI3K/mTOR inhibitor in thyroid cancer. Clin. Cancer Res. 2011, 17, 6482–6489. [Google Scholar] [CrossRef] [PubMed]
  149. Borson-Chazot, F.; Dantony, E.; Illouz, F.; Lopez, J.; Niccoli, P.; Wassermann, J.; Do Cao, C.; Leboulleux, S.; Klein, M.; Tabarin, A.; et al. Effect of Buparlisib, a Pan-Class I PI3K Inhibitor, in Refractory Follicular and Poorly Differentiated Thyroid Cancer. Thyroid 2018, 28, 1174–1179. [Google Scholar] [CrossRef]
  150. Lim, S.M.; Chang, H.; Yoon, M.J.; Hong, Y.K.; Kim, H.; Chung, W.Y.; Park, C.S.; Nam, K.H.; Kang, S.W.; Kim, M.K.; et al. A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Ann. Oncol. 2013, 24, 3089–3094. [Google Scholar] [CrossRef]
  151. Schneider, T.C.; de Wit, D.; Links, T.P.; van Erp, N.P.; van der Hoeven, J.J.; Gelderblom, H.; van Wezel, T.; van Eijk, R.; Morreau, H.; Guchelaar, H.J.; et al. Beneficial Effects of the mTOR Inhibitor Everolimus in Patients with Advanced Medullary Thyroid Carcinoma: Subgroup Results of a Phase II Trial. Int. J. Endocrinol. 2015, 2015, 348124. [Google Scholar] [CrossRef] [PubMed]
  152. Schneider, T.C.; de Wit, D.; Links, T.P.; van Erp, N.P.; van der Hoeven, J.J.; Gelderblom, H.; Roozen, I.C.; Bos, M.; Corver, W.E.; van Wezel, T.; et al. Everolimus in Patients With Advanced Follicular-Derived Thyroid Cancer: Results of a Phase II Clinical Trial. J. Clin. Endocrinol. Metab. 2017, 102, 698–707. [Google Scholar] [CrossRef] [PubMed]
  153. Hanna, G.J.; Busaidy, N.L.; Chau, N.G.; Wirth, L.J.; Barletta, J.A.; Calles, A.; Haddad, R.I.; Kraft, S.; Cabanillas, M.E.; Rabinowits, G.; et al. Genomic Correlates of Response to Everolimus in Aggressive Radioiodine-refractory Thyroid Cancer: A Phase II Study. Clin. Cancer Res. 2018, 24, 1546–1553. [Google Scholar] [CrossRef] [Green Version]
  154. Owonikoko, T.K.; Zhang, G.; Lallani, S.B.; Chen, Z.; Martinson, D.E.; Khuri, F.R.; Lonial, S.; Marcus, A.; Sun, S.Y. Evaluation of preclinical efficacy of everolimus and pasireotide in thyroid cancer cell lines and xenograft models. PLoS ONE 2019, 14, e0206309. [Google Scholar] [CrossRef] [PubMed]
  155. Atkinson, H.; England, J.A.; Rafferty, A.; Jesudason, V.; Bedford, K.; Karsai, L.; Atkin, S.L. Somatostatin receptor expression in thyroid disease. Int. J. Exp. Pathol. 2013, 94, 226–229. [Google Scholar] [CrossRef] [PubMed]
  156. Faggiano, A.; Modica, R.; Severino, R.; Camera, L.; Fonti, R.; Del Prete, M.; Chiofalo, M.G.; Aria, M.; Ferolla, P.; Vitale, G.; et al. The antiproliferative effect of pasireotide LAR alone and in combination with everolimus in patients with medullary thyroid cancer: A single-center, open-label, phase II, proof-of-concept study. Endocrine 2018, 62, 46–56. [Google Scholar] [CrossRef] [PubMed]
  157. Manohar, P.M.; Beesley, L.J.; Taylor, J.M.; Hesseltine, E.; Haymart, M.R.; Esfandiari, N.H.; Hanauer, D.A.; Worden, F.P. Retrospective Study of Sirolimus and Cyclophosphamide in Patients with Advanced Differentiated Thyroid Cancers. J. Thyroid Disord. Ther. 2015, 4. [Google Scholar] [CrossRef] [Green Version]
  158. Sherman, E.J.; Dunn, L.A.; Ho, A.L.; Baxi, S.S.; Ghossein, R.A.; Fury, M.G.; Haque, S.; Sima, C.S.; Cullen, G.; Fagin, J.A.; et al. Phase 2 study evaluating the combination of sorafenib and temsirolimus in the treatment of radioactive iodine-refractory thyroid cancer. Cancer 2017, 123, 4114–4121. [Google Scholar] [CrossRef] [Green Version]
  159. Grommes, C.; Landreth, G.E.; Heneka, M.T. Antineoplastic effects of peroxisome proliferator-activated receptor gamma agonists. Lancet Oncol. 2004, 5, 419–429. [Google Scholar] [CrossRef]
  160. Smallridge, R.C.; Copland, J.A.; Brose, M.S.; Wadsworth, J.T.; Houvras, Y.; Menefee, M.E.; Bible, K.C.; Shah, M.H.; Gramza, A.W.; Klopper, J.P.; et al. Efatutazone, an oral PPAR-gamma agonist, in combination with paclitaxel in anaplastic thyroid cancer: Results of a multicenter phase 1 trial. J. Clin. Endocrinol. Metab. 2013, 98, 2392–2400. [Google Scholar] [CrossRef]
  161. Li, Y.; Seto, E. HDACs and HDAC Inhibitors in Cancer Development and Therapy. Cold Spring Harb. Perspect. Med. 2016, 6. [Google Scholar] [CrossRef] [PubMed]
  162. Haghpanah, V.; Malehmir, M.; Larijani, B.; Ahmadian, S.; Alimoghaddam, K.; Heshmat, R.; Ghavamzadeh, A.; Adabi, K.; Ghaffari, S.H. The Beneficial Effects of Valproic Acid in Thyroid Cancer Are Mediated through Promoting Redifferentiation and Reducing Stemness Level: An In Vitro Study. J. Thyroid Res. 2014, 2014, 218763. [Google Scholar] [CrossRef] [PubMed]
  163. Fu, Y.T.; Zheng, H.B.; Zhou, L.; Zhang, D.Q.; Liu, X.L.; Sun, H. Valproic acid, targets papillary thyroid cancer through inhibition of c-Met signalling pathway. Am. J. Transl. Res. 2017, 9, 3138–3147. [Google Scholar] [PubMed]
  164. Nilubol, N.; Merkel, R.; Yang, L.; Patel, D.; Reynolds, J.C.; Sadowski, S.M.; Neychev, V.; Kebebew, E. A phase II trial of valproic acid in patients with advanced, radioiodine-resistant thyroid cancers of follicular cell origin. Clin. Endocrinol. 2017, 86, 128–133. [Google Scholar] [CrossRef] [PubMed]
  165. Hu, Z.; Ott, P.A.; Wu, C.J. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat. Rev. Immunol. 2018, 18, 168–182. [Google Scholar] [CrossRef] [PubMed]
  166. Sanchez-Vega, F.; Mina, M.; Armenia, J.; Chatila, W.K.; Luna, A.; La, K.C.; Dimitriadoy, S.; Liu, D.L.; Kantheti, H.S.; Saghafinia, S.; et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell 2018, 173, 321–337.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Thorsson, V.; Gibbs, D.L.; Brown, S.D.; Wolf, D.; Bortone, D.S.; Ou Yang, T.H.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The Immune Landscape of Cancer. Immunity 2018, 48, 812–830.e14. [Google Scholar] [CrossRef] [Green Version]
  168. French, J.D.; Weber, Z.J.; Fretwell, D.L.; Said, S.; Klopper, J.P.; Haugen, B.R. Tumor-associated lymphocytes and increased FoxP3+ regulatory T cell frequency correlate with more aggressive papillary thyroid cancer. J. Clin. Endocrinol. Metab. 2010, 95, 2325–2333. [Google Scholar] [CrossRef] [PubMed]
  169. Mougiakakos, D.; Choudhury, A.; Lladser, A.; Kiessling, R.; Johansson, C.C. Regulatory T cells in cancer. Adv. Cancer Res. 2010, 107, 57–117. [Google Scholar] [CrossRef]
  170. Ostrand-Rosenberg, S.; Sinha, P. Myeloid-derived suppressor cells: Linking inflammation and cancer. J. Immunol. 2009, 182, 4499–4506. [Google Scholar] [CrossRef]
  171. Gogali, F.; Paterakis, G.; Rassidakis, G.Z.; Kaltsas, G.; Liakou, C.I.; Gousis, P.; Neonakis, E.; Manoussakis, M.N.; Liapi, C. Phenotypical analysis of lymphocytes with suppressive and regulatory properties (Tregs) and NK cells in the papillary carcinoma of thyroid. J. Clin. Endocrinol. Metab. 2012, 97, 1474–1482. [Google Scholar] [CrossRef] [PubMed]
  172. Chowdhury, S.; Veyhl, J.; Jessa, F.; Polyakova, O.; Alenzi, A.; MacMillan, C.; Ralhan, R.; Walfish, P.G. Programmed death-ligand 1 overexpression is a prognostic marker for aggressive papillary thyroid cancer and its variants. Oncotarget 2016, 7, 32318–32328. [Google Scholar] [CrossRef] [PubMed]
  173. Rosenbaum, M.W.; Gigliotti, B.J.; Pai, S.I.; Parangi, S.; Wachtel, H.; Mino-Kenudson, M.; Gunda, V.; Faquin, W.C. PD-L1 and IDO1 Are Expressed in Poorly Differentiated Thyroid Carcinoma. Endocr. Pathol. 2018, 29, 59–67. [Google Scholar] [CrossRef] [PubMed]
  174. Bai, Y.; Guo, T.; Huang, X.; Wu, Q.; Niu, D.; Ji, X.; Feng, Q.; Li, Z.; Kakudo, K. In papillary thyroid carcinoma, expression by immunohistochemistry of BRAF V600E, PD-L1, and PD-1 is closely related. Virchows Arch. 2018, 472, 779–787. [Google Scholar] [CrossRef] [PubMed]
  175. Shi, R.L.; Qu, N.; Luo, T.X.; Xiang, J.; Liao, T.; Sun, G.H.; Wang, Y.; Wang, Y.L.; Huang, C.P.; Ji, Q.H. Programmed Death-Ligand 1 Expression in Papillary Thyroid Cancer and Its Correlation with Clinicopathologic Factors and Recurrence. Thyroid 2017, 27, 537–545. [Google Scholar] [CrossRef] [PubMed]
  176. Zhang, G.Q.; Wei, W.J.; Song, H.J.; Sun, Z.K.; Shen, C.T.; Zhang, X.Y.; Chen, X.Y.; Qiu, Z.L.; Luo, Q.Y. Programmed Cell Death-Ligand 1 Overexpression in Thyroid Cancer. Endocr. Pract. 2019, 25, 279–286. [Google Scholar] [CrossRef] [PubMed]
  177. Mehnert, J.M.; Varga, A.; Brose, M.S.; Aggarwal, R.R.; Lin, C.C.; Prawira, A.; de Braud, F.; Tamura, K.; Doi, T.; Piha-Paul, S.A.; et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced, PD-L1-positive papillary or follicular thyroid cancer. BMC Cancer 2019, 19, 196. [Google Scholar] [CrossRef]
  178. Sherman, E.J.; Tsai, C.J.; Zhi, W.I.; Fetten, J.V.; Wu, V.; Ho, A.L.; Riaz, N.; Pfister, D.G.; Lee, N.Y. Pilot study combining PD-L1 antibody durvalumab (D) with CTLA-4 antibody tremelimumab (T) and stereotactic body radiotherapy (SBRT) to treat metastatic anaplastic thyroid cancer (ATC). J. Clin. Oncol. 2019, 37, 6088. [Google Scholar] [CrossRef]
  179. Lopez, J.S.; Banerji, U. Combine and conquer: Challenges for targeted therapy combinations in early phase trials. Nat. Rev. Clin. Oncol. 2017, 14, 57–66. [Google Scholar] [CrossRef]
  180. Massimino, M.; Stella, S.; Tirro, E.; Romano, C.; Pennisi, M.S.; Puma, A.; Manzella, L.; Zanghi, A.; Stagno, F.; Di Raimondo, F.; et al. Non ABL-directed inhibitors as alternative treatment strategies for chronic myeloid leukemia. Mol. Cancer 2018, 17, 56. [Google Scholar] [CrossRef]
  181. Pirosa, M.C.; Leotta, S.; Cupri, A.; Stella, S.; Martino, E.A.; Scalise, L.; Sapienza, G.; Calafiore, V.; Mauro, E.; Spadaro, A.; et al. Long-Term Molecular Remission Achieved by Antibody Anti-CD22 and Ponatinib in a Patient Affected by Ph’+ Acute Lymphoblastic Leukemia Relapsed after Second Allogeneic Hematopoietic Stem Cell Transplantation: A Case Report. Chemotherapy 2018, 63, 220–224. [Google Scholar] [CrossRef] [PubMed]
  182. Tirro, E.; Massimino, M.; Romano, C.; Pennisi, M.S.; Stella, S.; Vitale, S.R.; Fidilio, A.; Manzella, L.; Parrinello, N.L.; Stagno, F.; et al. Chk1 Inhibition Restores Inotuzumab Ozogamicin Citotoxicity in CD22-Positive Cells Expressing Mutant p53. Front. Oncol. 2019, 9, 57. [Google Scholar] [CrossRef] [PubMed]
  183. Stagno, F.; Vigneri, P.; Del Fabro, V.; Stella, S.; Restuccia, N.; Giallongo, C.; Massimino, M.; Berretta, S.; Pennisi, M.S.; Tibullo, D.; et al. Concomitant and feasible treatment with dasatinib and the anti-EGFR antibody cetuximab plus radiotherapy in a CML patient with multiple squamous neoplasias. Acta Oncol. 2010, 49, 109–110. [Google Scholar] [CrossRef] [PubMed]
  184. Brose, M.S.; Bible, K.C.; Chow, L.Q.M.; 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] [PubMed]
  185. Costa, R.; Carneiro, B.A.; Chandra, S.; Pai, S.G.; Chae, Y.K.; Kaplan, J.B.; Garrett, H.B.; Agulnik, M.; Kopp, P.A.; Giles, F.J. Spotlight on lenvatinib in the treatment of thyroid cancer: Patient selection and perspectives. Drug Des. Dev. Ther. 2016, 10, 873–884. [Google Scholar] [CrossRef] [PubMed]
  186. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 165. [Google Scholar] [CrossRef]
Figure 1. Genetic events involved in thyroid carcinogenesis. Papillary thyroid carcinomas (PTC), follicular thyroid carcinomas (FTC) and anaplastic thyroid carcinomas (ATC) originate from thyroid follicular cells and are characterized by molecular alterations (mutations, deletions, gene fusions) involving genes and proteins impinging upon different cellular pathways. The transition from PTC/FTC to poorly differentiated (PDTCs) and ATCs is attributed to additional molecular alterations. Medullary thyroid carcinoma (MTC) originates from para-follicular C-cells and is prevalently characterized by RET or RAS mutations.
Figure 1. Genetic events involved in thyroid carcinogenesis. Papillary thyroid carcinomas (PTC), follicular thyroid carcinomas (FTC) and anaplastic thyroid carcinomas (ATC) originate from thyroid follicular cells and are characterized by molecular alterations (mutations, deletions, gene fusions) involving genes and proteins impinging upon different cellular pathways. The transition from PTC/FTC to poorly differentiated (PDTCs) and ATCs is attributed to additional molecular alterations. Medullary thyroid carcinoma (MTC) originates from para-follicular C-cells and is prevalently characterized by RET or RAS mutations.
Genes 10 00709 g001
Figure 2. Schematic overview of ALK, NTRK and RET fusion proteins signaling. The indicated fusion proteins activate the JAK/STAT, PI3K, MAPK and PLCγ pathways involved in cell cycle progression, survival and differentiation. ALK, anaplastic lymphoma kinase; NTRK, neurotropic tropomyosin receptor kinase; RET, rearranged during transfection; RAS, Rat Sarcoma; JAK, Janus kinase; STAT, signal transducers and activators of transcription; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase; PLCγ, phospholipase C-γ; DAG, Diacylglycerol; PKC, protein kinase C.
Figure 2. Schematic overview of ALK, NTRK and RET fusion proteins signaling. The indicated fusion proteins activate the JAK/STAT, PI3K, MAPK and PLCγ pathways involved in cell cycle progression, survival and differentiation. ALK, anaplastic lymphoma kinase; NTRK, neurotropic tropomyosin receptor kinase; RET, rearranged during transfection; RAS, Rat Sarcoma; JAK, Janus kinase; STAT, signal transducers and activators of transcription; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase; PLCγ, phospholipase C-γ; DAG, Diacylglycerol; PKC, protein kinase C.
Genes 10 00709 g002
Figure 3. Molecules and cellular pathways that contribute to thyroid cancer development. Alterations in ALK, RET or other RTKs up-regulate RAS thereby pathologically modulating the MAPK and PI3K pathways that favor thyroid cell survival, de-differentiation and improper gene transcription. Mutations in the WNT pathway (CTNNB1, AXIN) and in other molecules (TERT, PPARγ, HMT, SW1-SNF, TP53) promote oncogenic activity, reduce apoptosis and compromise DNA repair (MMR). IDH1 or EIF1AX mutations alter epigenetic mechanisms or cause a defective assembly of the 43S-complex, respectively. Additional mutations in specific genes (BRAF, PTEN) promote the transition from differentiated to undifferentiated thyroid cancer. * indicates genes directly involved in thyroid carcinogenesis
Figure 3. Molecules and cellular pathways that contribute to thyroid cancer development. Alterations in ALK, RET or other RTKs up-regulate RAS thereby pathologically modulating the MAPK and PI3K pathways that favor thyroid cell survival, de-differentiation and improper gene transcription. Mutations in the WNT pathway (CTNNB1, AXIN) and in other molecules (TERT, PPARγ, HMT, SW1-SNF, TP53) promote oncogenic activity, reduce apoptosis and compromise DNA repair (MMR). IDH1 or EIF1AX mutations alter epigenetic mechanisms or cause a defective assembly of the 43S-complex, respectively. Additional mutations in specific genes (BRAF, PTEN) promote the transition from differentiated to undifferentiated thyroid cancer. * indicates genes directly involved in thyroid carcinogenesis
Genes 10 00709 g003
Figure 4. A working model describing vertical inhibition of cell cycle progression, survival and differentiation by agents targeting tyrosine kinase receptors. Multi-targeted kinase inhibitors included in each box have been investigated in thyroid cancer and are cataloged according to their receptor kinase specificity and selectivity. VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; c-KIT, Mast/Stem Cell Growth Factor Receptor Kit; FLT3, FMS-like tyrosine kinase 3; PDGFR, platelet-derived growth factor receptor; MET, proto-oncogene, receptor tyrosine kinase; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase.
Figure 4. A working model describing vertical inhibition of cell cycle progression, survival and differentiation by agents targeting tyrosine kinase receptors. Multi-targeted kinase inhibitors included in each box have been investigated in thyroid cancer and are cataloged according to their receptor kinase specificity and selectivity. VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; c-KIT, Mast/Stem Cell Growth Factor Receptor Kit; FLT3, FMS-like tyrosine kinase 3; PDGFR, platelet-derived growth factor receptor; MET, proto-oncogene, receptor tyrosine kinase; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase.
Genes 10 00709 g004
Figure 5. Mechanism of action of ALK, NTRK and RET fusion proteins inhibitors. Compounds targeting the ALK, NTRK and RET fusion proteins are listed in boxes. These drugs inhibit multiple cellular processes blocking cell cycle progression, survival and differentiation. ALK, anaplastic lymphoma kinase; NTRK, neurotropic tropomyosin receptor kinase; RET, rearranged during transfection; RAS, Rat Sarcoma; JAK, Janus kinase; STAT, signal transducers and activators of transcription; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase; PLCγ, phospholipase C-γ; DAG, Diacylglycerol; PKC, protein kinase C.
Figure 5. Mechanism of action of ALK, NTRK and RET fusion proteins inhibitors. Compounds targeting the ALK, NTRK and RET fusion proteins are listed in boxes. These drugs inhibit multiple cellular processes blocking cell cycle progression, survival and differentiation. ALK, anaplastic lymphoma kinase; NTRK, neurotropic tropomyosin receptor kinase; RET, rearranged during transfection; RAS, Rat Sarcoma; JAK, Janus kinase; STAT, signal transducers and activators of transcription; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase; PLCγ, phospholipase C-γ; DAG, Diacylglycerol; PKC, protein kinase C.
Genes 10 00709 g005
Figure 6. Strategies that target downstream mediators of receptor tyrosine kinases. RTK stimulation causes signaling activation that is blocked by inhibition of downstream mediators decreasing cell cycle progression, survival and differentiation. The panels report the downstream mediators inhibitors used in thyroid cancer stratified according to their molecular target specificity and selectivity. VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; c-KIT, Mast/Stem Cell Growth Factor Receptor Kit; FLT3, FMS-like tyrosine kinase 3; PDGFR, platelet-derived growth factor receptor; MET, proto-oncogene, receptor tyrosine kinase; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase.
Figure 6. Strategies that target downstream mediators of receptor tyrosine kinases. RTK stimulation causes signaling activation that is blocked by inhibition of downstream mediators decreasing cell cycle progression, survival and differentiation. The panels report the downstream mediators inhibitors used in thyroid cancer stratified according to their molecular target specificity and selectivity. VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; c-KIT, Mast/Stem Cell Growth Factor Receptor Kit; FLT3, FMS-like tyrosine kinase 3; PDGFR, platelet-derived growth factor receptor; MET, proto-oncogene, receptor tyrosine kinase; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; PI3K, phosphoinositide 3-kinase; AKT, V-Akt Murine Thymoma Viral Oncogene Homolog; mTOR, Mammalian Target of Rapamycin; BRAF, B-Raf proto-oncogene; MEK, Mitogen-Activated Protein Kinase; ERK, Extracellular Signal-Regulated Kinase.
Genes 10 00709 g006
Table 1. Clinical trials with multi-kinase inhibitors.
Table 1. Clinical trials with multi-kinase inhibitors.
AGENTCOMBINATIONSTUDY
POPULATION
DESIGNPATIENTSPRIMARY END POINTSTATUSIDENTIFIER
Cabozantinib
Lenvatinib
Sorafenib
Vandetanib
Advanced TCNonrandomized,
Open label, phase II
45PFS, OS, DLTsRecruitingNCT03630120
Cabozantinib RAIR DTC pretreated with anti-VEGFRRandomized,
Double blind, phase III
300PFS, ORRRecruitingNCT03690388
Cabozantinib RAIR DTCNonrandomized,
Open label, phase II
43SPActive, not recruitingNCT02041260
CabozantinibIpilimumab
Nivolumab
RAIR DTC pretreated with anti-VEGFRNonrandomized,
Open label, phase II
24ORRNot yet recruitingNCT03914300
CabozantinibAtezolizumabLA or M+ Solid TumorsNonrandomized,
Open label, phase Ib
1000DLTs, ORRRecruitingNCT03170960
Cabozantinib Recurrent or Refractory Solid TumorsNonrandomized,
Open label, phase I
41DLTsActive, not recruitingNCT01709435
Imatinib RAIR PTCNonrandomized,
Open label, phase I
18EPRecruitingNCT03469011
Lenvatinib Advanced RAIR TCRandomized,
Double blind, phase II
152ORR, SPRecruitingNCT02702388
Lenvatinib ATCNonrandomized,
Open label, phase II
34ORRTerminatedNCT02657369
Lenvatinib ATCNonrandomized,
Open label, phase II
39 *OSActive, not recruitingNCT02726503
LenvatinibDenosumabBone M+ RAIR DTCNonrandomized,
Open label, phase II
35EPNot yet recruitingNCT03732495
LenvatinibRAIRAI-sensitive DTCNonrandomized,
Open label, phase II
30PFSRecruitingNCT03506048
LenvatinibPembrolizumabRAIR DTCNonrandomized,
Open label, phase II
60ORRRecruitingNCT02973997
Pazopanib TCRandomized,
Open label, phase II
168PFSRecruitingNCT01813136
Pazopanib Advanced TCNonrandomized,
Open label, phase II
152ORRActive, not recruitingNCT00625846
PazopanibPaclitaxel
RT
ATCRandomized,
Open label, phase II
121 *OS, DLTsActive, not recruitingNCT01236547
Sorafenib Adjuvant after RAINonrandomized,
Open label, phase II
32ORRCompletedNCT00887107
Sorafenib Advanced.TCNonrandomized,
Open label, phase II
61ORRCompletedNCT00654238
Sorafenib Advanced.TCNonrandomized,
Open label, phase II
25ORRTerminatedNCT00095693
Vandetanib Hereditary MTCNonrandomized,
Open label, phase I/II
17SPActive, not recruitingNCT00514046
Vandetanib Advanced MTCRandomized,
Double blind, phase III
437PFSActive, not recruitingNCT00410761
Anaplastic thyroid cancer (ATC); Dose-limiting toxicities (DLTs); Differentiated thyroid cancer (DTC); Efficacy profile (EP); Locally advanced (LA); Medullary thyroid cancer (MTC); Metastatic (M+); Objective response rate (ORR); Overall survival (OS); Pharmacodynamic (PD); Pharmakinetics (PK); Progression free survival (PFS); Papillary Thyroid Cancer (PTC); Radioactive iodine (RAI); Radioactive iodine resistance (RAIR); Radio therapy (RT); Safety profile (SP); Thyroid cancer (TC). * number of estimated patients.
Table 2. Clinical trials with single target agents.
Table 2. Clinical trials with single target agents.
TARGETAGENTCOMBINATIONSTUDY
POPULATION
DESIGNPATIENTSPRIMARY END POINTSTATUSIDENTIFIER
ALKAlectinib RET-rearranged NSCLC or RET-mut TCNon-Randomized, Open Label, Phase I/II78*MTD, ORRActive, not recruitingNCT03131206
Ceritinib M+ or LA ATCSingle Group Assignment, Open Label, Phase II100*Development of progressionRecruitingNCT02289144
BRAFDabrafenibTrametinibRecurrent TCRandomized,
Open label, phase II
53ORRActive, not recruitingNCT01723202
DabrafenibTrametinib
RAI
M+ RAIR with RAS or BRAF mutationNonrandomized,
Open label, phase II
87ORRRecruitingNCT03244956
Dabrafenib LapatinibTC with BRAF mutationNonrandomized,
Open label, phase I
18DLTsActive, not recruitingNCT01947023
Vemurafenib Neoadjuvant-Advanced TCNonrandomized,
Open label, phase II
24EPActive, not recruitingNCT01709292
MEKSelumetinib OlaparibSolid tumors with Ras pathway alterations, and ovarian tumors with PARP resistanceNonrandomized,
Open label, phase I
90DLTsRecruitingNCT03162627
Selumetinib 131IRecurrent or M+ TCRandomized,
Double blind, phase II
60ORRRecruitingNCT02393690
Trametinib PaclitaxelAdvanced ATCNonrandomized,
Open label, phase I
12PFSRecruitingNCT03085056
Trametinib PazopanibAdvanced Solid Tumors (DTC, STS and Chol)Nonrandomized,
Open label, phase I
89DLTs, SPCompletedNCT01438554
Trametinib RAIRAS mutant or RAS/RAF wild-type, RAIR and/or M+ TCNonrandomized,
Open label, phase II
35PFS, ORRRecruitingNCT02152995
NTRKEntrectinib LA or M+ Solid Tumors harboring NTRK1/2/3, ROS1, or ALK RearrangementsNon-Randomized,
Open Label, Phase II
300*ORRRecruitingNCT02568267
Entrectinib Solid tumors with or without TRK, ROS1 or ALK FusionsNon-Randomized,
Open label, Phase I
65*MTD, RP2D, ORRRecruitingNCT02650401
Larotrectinib Solid Tumors Harboring NTRK FusionNon-Randomized, Open Label, Phase II320*ORR RecruitingNCT02576431
LOXO-195 Patients with previously treated NTRK Fusion cancersSingle Group Assignment, Open label, Phase I/II93*MTD, recommended dose, PR, CRRecruitingNCT03215511
PPAR-γEfatutazonePaclitaxelAdvanced ATCNonrandomized,
Open label, phase II
19ORRActive, not RecruitingNCT02152137
VEGFR-2Apatinib RTInoperable or RAIR TCNonrandomized,
Open label, phase II
20PFSRecruitingNCT03300765
Apatinib RAIR DTCRandomized,
Double blind, phase III
118PFSRecruitingNCT03048877
Apatinib Locally Advanced/M+ DTCNonrandomized,
Open label, phase II
20EPRecruitingNCT03167385
Apatinib Local Progressive/M+ RAIRNonrandomized,
Open label, phase II
40ORRRecruitingNCT03199677
Anaplastic thyroid cancer (ATC); Cholangiocarcinoma (Chol); Complete response (CR); Dose-limiting toxicities (DLTs); Differentiated thyroid cancer (DTC); Efficacy profile (EP); Locally advanced (LA); Maximum Tolerated Dose (MTD); Medullary thyroid cancer (MTC); Metastatic (M+); Non small cell lung cancer (NSCLC); Objective response rate (ORR); Overall survival (OS); Pharmacodynamic (PD); Pharmacokinetics (PK); Progression-free survival (PFS); Partial response (PR); Radioactive iodine (RAI); Radioactive iodine resistance (RAIR); Radiation therapy (RT); Recommended phase 2 dose (RP2D); Safety profile (SP); Soft tissue sarcoma (STS); Thyroid cancer (TC). * number of estimated patients.
Table 3. Clinical trials with mTOR inhibitors.
Table 3. Clinical trials with mTOR inhibitors.
AGENTCOMBINATIONSTUDY
POPULATION
DESIGNPATIENTSPRIMARY END POINTSTATUSIDENTIFIER
Everolimus LA or M+ TCNonrandomized,
Open label, phase II
40ORRCompletedNCT01164176
Everolimus RAIR TCNonrandomized,
Open label, phase II
33PFSActive, not RecruitingNCT00936858
EverolimusLenvatinibM+ DTC progressed on Lenvatinib aloneNonrandomized,
Open label, phase II
40PFSRecruitingNCT03139747
Everolimus NeratinibAdvanced Cancer with EGFR/HER2 Mut/Ampl, HER3/4 MutNonrandomized,
Open label, phase I
120DLTsRecruitingNCT03065387
Everolimus PasireotideRAIR DTC and MTCRandomized,
Open label, phase II
42ORRCompletedNCT01270321
Everolimus SorefenibM+ DTC progressed on Sorafenib aloneNonrandomized,
Open label, phase II
40ORR, PFSActive, not RecruitingNCT01263951
EverolimusSorefenibAdvanced TC never treated with m-TOR inhibitor or SorafenibNonrandomized,
Open label, phase II
41ORRActive, not RecruitingNCT01141309
Everolimus SorefenibAdvanced RAIR Hurthle Cell TCRandomized, Open label, phase II34lPFSRecruitingNCT02143726
Everolimus VatalinibAdvanced Solid TumorsNonrandomized,
Open label, phase I
96DLTs, SPCompletedNCT00655655
SirolimusCiclofosfamideM+ or RAIR DTCNonrandomized,
Open label, phase II
19ORRRecruiting
NCT03099356
SirolimusGrapefruit juiceAdvanced MalignanciesNonrandomized,
Open label, phase Ib
41PKCompletedNCT00375245
Temsirolimus Bevacizumab
Valproic Acid
Advanced or M+ Malignancy or Other Benign DiseaseNonrandomized,
Open label, phase I
216DLTsRecruitingNCT01552434
Temsirolimus VinorelbineUnresectable or M+ Solid TumorsNonrandomized,
Open label, phase I
19DLTs, ORRCompletedNCT01155258
Dose-limiting toxicities (DLTs); Differentiated thyroid cancer (DTC); Locally advanced (LA); Medullary thyroid cancer (MTC); Metastatic (M+); Objective response rate (ORR); Pharmacokinetics (PK); Progression-free survival (PFS); Radioactive iodine resistance (RAIR); Safety profile (SP); Thyroid cancer (TC).
Table 4. Clinical trials with immune checkpoint inhibitors.
Table 4. Clinical trials with immune checkpoint inhibitors.
TARGETAGENTCOMBINATIONSTUDY
POPULATION
DESIGNPATIENTSPRIMARY ENDPOINTSTATUSIDENTIFIER
PD-1Pembrolizumab M+ or LA ATCSingle Group Assignment
Open label, phase II
20RRRecruitingNCT02688608
Pembrolizumab Recurrent or M+ MTCNonrandomized
Parallel Assignment
Open label, phase II
32
DLTsRecruitingNCT03072160
Pembrolizumab Patients with rare cancer typesSingle Group Assignment,
Open label, phase II
350ORRRecruitingNCT03012620
Pembrolizumab Advanced Solid Tumors Single Group Assignment,
Open label, phase II
1350ORRRecruitingNCT02628067
PembrolizumabDocetaxelPoorly Chemo-responsive Thyroid and Salivary Gland TumorsNonrandomized,
Parallel Assignment Open label, phase I
46RRRecruitingNCT03360890
Pembrolizumab Docetaxel Doxorubicin ATCNonrandomized,
Open label, phase II
3*OSRActive, not RecruitingNCT03211117
PembrolizumabLenvatinibRAIR DTCSingle Group Assignment,
Open label, phase II
60CRR RecruitingNCT02973997
PD-1 and
CTLA-4
Nivolumab Ipilimumab RAIR DTC, ATC, MTCRandomized,
Parallel Assignment
Open label, phase II
54RRRRecruitingNCT03246958
PD-L1AtezolizumabBevacizumab
Cobimetinib
Paclitaxel
Vemurafenib
ATC, PDTCNonrandomized,
Parallel Assignment
Open label, phase II
50OSRecruitingNCT03181100
AtezolizumabCabozantinibM+ ATCNonrandomized
Sequential Assignment Open label, phase I-II
1000DLTsRecruitingNCT03170960
Durvalumab TCSingle Group Assignment Open label, phase I11DLTsRecruitingNCT03215095
PD-L1 and
CTLA-4
Durvalumab+
Tremelimumab
M+ ATCSingle Group Assignment Open label, phase 13OS Active, not RecruitingNCT03122496
Anaplastic thyroid cancer (ATC); Complete remission rate (CRR); Dose-limiting toxicities (DLTs); Differentiated thyroid cancer (DTC); Locally advanced (LA); Medullary thyroid cancer (MTC); Metastatic (M+); Objective response rate (ORR); Overall survival (OS); Overall survival rate (OSR); Poorly differentiated thyroid cancer (PDTC); Radioactive iodine resistance (RAIR); Radiographic Response Rate (RRR); Response rate (RR). * number of actual patients.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/genes10090709

AMA Style

Tirrò E, Martorana F, Romano C, Vitale SR, Motta G, Di Gregorio S, Massimino M, Pennisi MS, Stella S, Puma A, et al. Molecular Alterations in Thyroid Cancer: From Bench to Clinical Practice. Genes. 2019; 10(9):709. https://doi.org/10.3390/genes10090709

Chicago/Turabian Style

Tirrò, Elena, Federica Martorana, Chiara Romano, Silvia Rita Vitale, Gianmarco Motta, Sandra Di Gregorio, Michele Massimino, Maria Stella Pennisi, Stefania Stella, Adriana Puma, and et al. 2019. "Molecular Alterations in Thyroid Cancer: From Bench to Clinical Practice" Genes 10, no. 9: 709. https://doi.org/10.3390/genes10090709

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