Molecular mechanisms of resistance to kinase inhibitors and redifferentiation in thyroid cancers

in Endocrine-Related Cancer
Authors:
Marie-Claude Hofmann Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Marie-Claude Hofmann in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0002-4899-8039
,
Muthusamy Kunnimalaiyaan Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Muthusamy Kunnimalaiyaan in
Current site
Google Scholar
PubMed
Close
,
Jennifer R Wang Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Jennifer R Wang in
Current site
Google Scholar
PubMed
Close
,
Naifa L Busaidy Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Naifa L Busaidy in
Current site
Google Scholar
PubMed
Close
,
Steven I Sherman Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Steven I Sherman in
Current site
Google Scholar
PubMed
Close
,
Stephen Y Lai Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Stephen Y Lai in
Current site
Google Scholar
PubMed
Close
,
Mark Zafereo Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Mark Zafereo in
Current site
Google Scholar
PubMed
Close
, and
Maria E Cabanillas Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas, USA

Search for other papers by Maria E Cabanillas in
Current site
Google Scholar
PubMed
Close

Correspondence should be addressed to M-C Hofmann: mhofmann@mdanderson.org
Free access

Sign up for journal news

Protein kinases play critical roles in cell survival, proliferation, and motility. Their dysregulation is therefore a common feature in the pathogenesis of a number of solid tumors, including thyroid cancers. Inhibiting activated protein kinases has revolutionized thyroid cancer therapy, offering a promising strategy in treating tumors refractory to radioactive iodine treatment or cytotoxic chemotherapies. However, despite satisfactory early responses, these drugs are not curative and most patients inevitably progress due to drug resistance. This review summarizes up-to-date knowledge on various mechanisms that thyroid cancer cells develop to bypass protein kinase inhibition and outlines strategies that are being explored to overcome drug resistance. Understanding how cancer cells respond to drugs and identifying novel molecular targets for therapy still represents a major challenge for the treatment of these patients.

Abstract

Protein kinases play critical roles in cell survival, proliferation, and motility. Their dysregulation is therefore a common feature in the pathogenesis of a number of solid tumors, including thyroid cancers. Inhibiting activated protein kinases has revolutionized thyroid cancer therapy, offering a promising strategy in treating tumors refractory to radioactive iodine treatment or cytotoxic chemotherapies. However, despite satisfactory early responses, these drugs are not curative and most patients inevitably progress due to drug resistance. This review summarizes up-to-date knowledge on various mechanisms that thyroid cancer cells develop to bypass protein kinase inhibition and outlines strategies that are being explored to overcome drug resistance. Understanding how cancer cells respond to drugs and identifying novel molecular targets for therapy still represents a major challenge for the treatment of these patients.

Introduction

Thyroid cancer is the most common endocrine malignancy, with an estimated 44,280 cases in the United States in 2021 (Siegel et al. 2021). They originate from two types of endocrine cells: the thyrocytes, or follicular cells, or the parafollicular C cells. Our understanding of the molecular pathways involved in thyroid tumorigenesis has greatly improved due to the identification of several oncogenic alterations and the availability of agents to target them (Nikiforov & Nikiforova 2011). These alterations include point mutations as well as gene translocations and their chimeric fusion products, gene amplifications, and deletions. Many of these alterations result in oncogenic drivers that promote cell proliferation, motility, and metastasis (Fagin & Wells 2016). They often directly or indirectly increase the activity of protein kinases, which are critical for many cellular functions, including regulation of the cell cycle. The identification of these mutations has led to the development of new targeted therapies, mostly protein kinase inhibitors (PKIs), for advanced thyroid cancers (Sipos & Ringel 2022). So far, four of the FDA-approved PKIs for thyroid cancers are antiangiogenic multi-kinase inhibitors and five are mutation-selective. These novel drugs have revolutionized the treatment of patients with advanced thyroid cancers (Cabanillas et al. 2019). Kinase inhibitors have shown good efficacy in controlling the disease and are better tolerated than cytotoxic therapies. However, despite an early response, many patients will show transient benefit and progress under treatment, and tumor-specific mortality is nearly universal. The mechanisms leading to acquired drug resistance are far from being completely understood. In this review, we summarize the properties and function of protein kinases and kinase inhibitors, and how these drugs are used to treat thyroid cancers. We then describe some mechanisms of resistance recently identified and which strategies were used to overcome the challenges.

Protein kinases

Protein kinases and their dysregulation

Protein kinases are enzymes that covalently add phosphate groups to selected proteins. They catalyze the transfer of phosphate from ATP to specific amino acid residues of target proteins in a process called phosphorylation. Phosphorylation is considered one of the most abundant post-translational modifications of proteins and usually modulates their function by adjusting their activity, modifying their cellular location, or altering their association with other proteins (Seok 2021). The majority of protein kinases are serine/threonine kinases, which phosphorylate the hydroxyl groups of serines and threonines in their target proteins. Most other kinases are tyrosine kinases, which catalyze the addition of a phosphate group to tyrosine residues. Up to 30% of all human proteins may be modified by kinase activity, and kinases are known to regulate many cellular processes and pathways, especially those involved in signal transduction. Protein kinases may be located at the cell membrane and function as receptors, such as growth factor receptors (EGFR, MET, KIT). They may also reside in the cytoplasm, where their activation is dependent on upstream signaling molecules such as RAS and protein kinases functioning in signaling cascades (Fig. 1). Phosphorylation through protein kinases regulates key cellular processes that include gene expression, proliferation, differentiation, motility, membrane transport, metabolism, and apoptosis. Therefore, the predominance of dysregulation of protein kinases activity in many cancers is not surprising (Brognard & Hunter 2011).

Figure 1
Figure 1

Overview of the MAPK and AKT signaling pathways. The main protein kinases driving these pathways are depicted. Kinase inhibitors that are presently FDA-approved for thyroid cancers are highlighted.

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0129

Alterations of protein kinase activities are indeed a frequent component of the pathogenesis of solid and hematologic malignancies. These alterations are caused by a variety of mechanisms, such as genomic rearrangements (chromosomal translocations and fusions) (Nikiforov 2002, Hallberg & Palmer 2013), gene amplification or deletions (Lopez-Gines et al. 2010, Rodriguez-Antona et al. 2010, Cicenas et al. 2018), and missense point mutations (Lahiry et al. 2010). Missense point mutations often arise in the kinase domain of receptor tyrosine kinases or in cytoplasmic serine/threonine protein kinases. For example, a sporadic point mutation in the BRAF gene, which belongs to the MAPK pathway (Fig. 1), can constitutively activate the kinase to drive cell proliferation continually, without the presence of growth factors (Wan et al. 2004, Xing 2005). Most BRAF-activating missense point mutations are found in exons 11 and 15 of the gene, within the kinase domain of the protein. In particular, the c.T1799A mutation is present in over 80% of all BRAF-mutated cancers (Davies et al. 2002). The protein resulting from this mutation is a substitution of valine for glutamic acid at codon 600 (V600E), which leads to constitutive activation of BRAF and its downstream targets MEK and ERK. Activating BRAF mutations are found in 40–60% of melanoma and 50–60% of papillary thyroid carcinoma (PTC) (Fig. 2B) (Xing et al.2005, Long et al. 2011, Cancer Genome Atlas Research Network 2014). BRAF mutations have also been frequently reported in colorectal cancers (Barras 2015). An example of gene rearrangement/fusion in thyroid cancers is illustrated by somatic juxtapositions of 5’ activating sequences from other genes with 3’ sequences of the RET or NTRK genes encoding the tyrosine kinase domain (Fig. 1). Sporadic RET and NTRK rearrangements occur in approximately 16% of thyroid cancers (Nikiforov 2002, Park et al. 2022, Ullmann et al. 2022) (Fig. 2B). In addition to mutations affecting DNA sequences, epigenetic events can also increase or modify the expression of many receptor tyrosine kinases such as EGFR, ERBB2, and MET in many solid tumors (Hoque et al. 2010, Ogunwobi et al. 2013) and untimely activation of the kinases p38, MEK, ERK, AURKA, and AKT due to phosphorylation is well known (Cicenas et al. 2018).

Figure 2
Figure 2

Clinical and molecular spectrum of thyroid cancers. (A) Incidence of the main types of thyroid cancers (data from Fagin & Wells 2016). (B) Distribution of the top eight mutated genes in thyroid cancers (from COSMIC 2022).

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0129

Targeting protein kinases

Small molecule kinase inhibitors have been very efficient in targeting specific mutations driving tumorigenesis. These inhibitors are generally categorized according to their mechanism of action. They are classified as type I, II, III (allosteric), IV (substrate directed), V (bivalent), and VI (covalent) inhibitors (Bhullar et al. 2018, Cicenas et al. 2018, Martinez et al. 2020) (Table 1). Type I inhibitors compete for the substrate and bind the DFG (Asp-Phe-Gly) motif in the ATP-binding pocket of the active kinase when the Asp residue is oriented toward bound ATP (‘in’ conformation) (Peng et al. 2013). Such is the case for crizotinib, dasatinib, erlotinib, lapatinib, pazopanib, sunitinib, vemurafenib, and dabrafenib. Type II inhibitors bind to the ATP pocket of the inactive kinase (DFG-Asp ‘out’ conformation) (imatinib, sorafenib, axitinib, nilotinib). The disadvantages of type I and II inhibitors reside in their binding to the ATP-binding site, which is conserved in protein kinases. Therefore, developing inhibitors that are specific is particularly challenging (Table 1). This lack of specificity increases the potential for off-target side effects, in particular cardiotoxicity. In contrast, type III inhibitors such as the MEK1/2 inhibitors trametinib and cobimetinib occupy highly specific allosteric sites close to the ATP-binding pocket and therefore are thought to be more selective than type I and II inhibitors (Zhao et al. 2017). This design might also prevent the occurrence of ‘gate-keeper’ mutations, which can obstruct drug binding by modulating access to the kinase ATP-binding site. More recently, efforts have been made to develop inhibitors that bind to unique structural domains outside the ATP-binding pocket. This led to the synthesis of type IV inhibitors, which usually induce a conformational change disrupting interactions of the kinases with their downstream targets (JNK, ERK1/2 inhibitors) (Martinez et al. 2020). This approach again offers more selectivity by blocking only the kinase function associated with a particular disease, while preserving other kinase functions that have potential benefits. To further increase selectivity, type V inhibitors, or bivalent inhibitors, were developed that can target both the ATP-binding site and a unique structural feature found on the kinase (lenvatinib and specific c-SRC, ERK5 and PLK1 inhibitors) (Okamoto et al. 2015, Lee et al. 2021) (Table 1). Finally, type VI inhibitors bind covalently and irreversibly to their target for a longer duration of the effect (Weisner et al. 2019, Martinez et al. 2020). This is the case for afatinib (an EGFR inhibitor), ibrutinib (a BTK inhibitor), and neratinib (a HER1-HER4 inhibitor).

Table 1

Classification of some small molecule kinase inhibitors (data from Bhullar et al. 2018, Kannaiyan & Mahadevan 2018, Martinez et al. 2020 and Lee et al. 2021).

Class Mechanism of action Names Target protein kinases
Type I inhibitors Compete for the substrate and binds in the ATP-binding pocket of the active kinase conformation (DFG-Asp ‘in’)

Bosutinib

BCR-ABL, Src

Cabozantinib

VEGFR, PDGFR, KIT, MET, FLT3, RET

Ceritinib

ALK-EML4

Crizotinib

ALK-EML4, MET, ROS-1

Dabrafenib

BRAFV600E mutant kinase

Gefitinib

EGFR

Papozanib

VEGFR, PDGFR, KIT, FGFR3

Vandetanib

VEGFR, RET, MET

Vemurafenib

BRAFV600E mutant kinase

Encorafenib

BRAFV600E mutant kinase

Larotrectinib

NTRK fusions

Entrectinib

NTRK fusions

Type II inhibitors Compete for the substrate and binds in the ATP-binding pocket of the inactive kinase conformation (DFG-Asp ‘out’)

Imatinib

BCR-ABL, KIT, PDGFR

Sorafenib

VEGFR, PDGFR, BRAF, FTL3, RET, KIT

Axitinib

VEGFR

Nilotinib

BCR-ABL, KIT, PDGFR

Type III (allosteric inhibitors) Occupy a site next to the ATP-binding pocket so that both ATP and the allosteric inhibitor simultaneously bind to the protein kinase. Offer more selectivity against the targeted kinases and avoid the development of ATP-binding site gatekeeper mutations

Trametinib

MEK1/2

Cobimetinib

MEK1/2

Selumetinib

MEK1/2

Binimetinib

MEK1/2

GnF2

BCR-ABL

Type IV (substrate-directed inhibitors) Bind to a site distal from the ATP pocket and induce a conformational change. Offer more selectivity against the targeted kinases ONO12380 BCR-ABL
Type V (bivalent inhibitors) Consist of a small molecule that targets the ATP-binding site coupled to a peptide representing the substrate targeted by the specific kinase

Lenvatinib

VEGFR1–3, FGFR1–4, PDGFRα, KIT, RET

ARC-1411

PKA

ARC-3140

CK2

ERK5.1

ERK5

Rapalink-1

mTORC1

BI 2536

PLK1

BI 53

c-SRC

Type VI (covalent inhibitors) Irreversibly bind the targeted kinase

Afatinib

EGFR

Ibrutinib

BTK

Neratinib

HER1, HER2, HER3, HER4

Thyroid cancers

The thyroid lies within the neck and consists of two connected lobes. The functional unit of the gland is the spherical thyroid follicle lined with thyrocytes, or follicular cells, that secrete triiodothyronine (T3) and thyroxine (T4). T3 and T4 have a wide range of effects including control of metabolism, regulation of the cardiac rate, and control of growth during development. Parafollicular cells, or C cells, are located adjacent to the thyroid follicles and secrete calcitonin, which contributes to the control of blood calcium levels. The most common primary thyroid cancers originate from thyrocytes and are classified as differentiated thyroid cancers (DTCs). DTCs include PTC, follicular thyroid carcinoma (FTC), and Hürthle cell thyroid carcinoma (HTC). DTCs usually harbor single driver mutations in the RAS or BRAF gene, which are mutually exclusive. Poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) are rare and aggressive tumors that also originate from thyrocytes (Fig. 2A). They both show a de-differentiated phenotype; however, ATCs harbor a higher number of mutations than PDTCs and PTCs (Landa et al. 2016). Medullary thyroid carcinomas (MTCs) are tumors derived from neuroendocrine parafollicular C-cells and are often driven by a RET receptor mutation. In thyroid cancers, most somatic driver mutations are found in receptor tyrosine kinases or in serine/threonine kinases belonging to the MAPK signaling pathway, regardless of the histologic subtype (Fig. 1).

Molecular pathology of thyroid cancers

PTCs represent about 80% of thyroid cancers and often harbor somatic mutations in the MAPK pathway (Fig. 2A and B). Activation of the pathway is mainly due to BRAF or RAS mutations, or re-arrangements of the RET or NTRK genes. Primary PTCs often display single mutations, such as the BRAF V600E mutation mentioned earlier (60% of the cases), and these tumors harbor one of the lowest mutation densities of cancers that have been studied according to whole-exome sequencing (WES) data (Fagin & Wells 2016). About 10–20% of thyroid cancers are due to RET mutations or rearrangements, and another 10–20% are due to RAS point mutations (Fig. 2B). In classical PTCs, RET fusions account for 6–8% of the cases (RET/PTC rearrangements) and the presence of RAS mutations is low (1–6%) (Cancer Genome Atlas Research Network 2014, Song & Park 2019). If the PTC tumor is small and localized within the thyroid, the prognosis is excellent, especially in younger patients. However, larger tumors that may extend outside the thyroid and have invaded lymph nodes and/or distant organs carry a poorer prognosis.

FTCs account for ~12% of thyroid cancers (Fig. 2A and B). FTCs usually harbor NRAS, HRAS, or KRAS gene mutations (40–50% of the cases). Another common mutation is the PAX8/PPARG rearrangement (30–35% of the cases), while point mutations in the PTEN and PIK3CA genes are observed in about 5–10% of the cases. The prognosis of FTC depends on the age of the patients, the size and stage of the tumors, the completeness of surgery, the presence of distance metastases, and the responsiveness to radioactive iodine therapy (Sobrinho-Simoes et al. 2011).

HTCs amount to 2–3% of thyroid cancers (Fig. 2A and B). HTCs exhibit a more aggressive behavior compared with PTC and FTC and the frequency of distant metastases is high (McFadden & Sadow 2021). A fraction of HTCs harbor mutations in the RAS, TP53, PTEN, E1F1AX, and PAX8-PPARG genes, which are often found in other thyroid cancers. Mutations in the TERT promoter, as well as the DAXX and ATRX genes that cause alternate telomere elongation, are found in more aggressive forms of HTCs (Ganly et al. 2018). In addition, an interesting feature of the HTCs is their accumulation of abnormally shaped mitochondria, probably due to mutations in the mitochondrial DNA leading to impairments in proteins of the electron transport chain, in particular subunits of the complex I and III (Bonora et al. 2006). Further, chromosome gains and losses are often observed, with near haploidization in certain cases, or selective gains of chromosomes 7, 12, and 17 (Ganly et al. 2018).

PDTCs are more aggressive than PTC and FTC tumors and represent approximately 2–3% of thyroid cancers. Radioiodine therapy is of limited benefit and these tumors can readily metastasize to the neck lymph nodes, lungs, and bones. They are associated with a mean survival of 3.2 years (Landa et al. 2016). Molecular pathology data indicate that these tumors retain the driver mutations seen in differentiated PTCs or FTCs (BRAFV600E or RAS) and that they have acquired additional alterations such as PTEN loss of function (Volante et al. 2021). In addition, while TERT promoter mutations are found in about 5–15% of PTCs and FTCs, this mutation is significantly more frequent in PDTCs (20–50%) (Volante et al. 2021). Thus, the acquisition of a TERT promoter mutation appears to be a key transitional step in the microevolution of these tumors (Landa et al. 2016).

ATCs are rare but are among the most aggressive tumors known, with historic median survival of 3–5 months, although more recent studies have demonstrated improved survival (Maniakas et al. 2020). The current accepted model considers that ATCs derive from PTCs, FTCs, or HTCs through the acquisition of additional genomic alterations (Fig. 3). BRAF, RAS, and RET mutations are prevalent; however, de novo acquisition of TERT promoter, PIK3CA, and TP53 mutations (70% of the cases) are a hallmark of these malignancies (Landa et al. 2016, Pozdeyev et al. 2018, Qin et al. 2021). Mutations in the PTEN and NF1 genes are also occasionally seen. In general, ATC tumors harbor a higher mutation burden than PTC. ATC tumors easily invade neck structures, metastasize to distant organs, and are resistant to conventional chemotherapy and radiation therapy. Capdevilla and colleagues sought to understand more in detail how, and if, ATCs truly derive from PTCs by performing WES of 14 ATC samples, including from 3 patients with concomitant ATC and PTC (Capdevila et al. 2019). Interestingly, in the cases of concomitant ATC and PTC, most mutations identified in the ATC component differed from the ones in PTC. Phylogenic tree analyses indicated that ATC and PTC might diverge early in tumor development and evolve independently. Mutational analysis of 44 ATCs and 401 PTCs/FTCs from public databases confirmed that most of the high incidence lesions were not the same between ATCs and PTCs or FTCs. For example, despite the fact that BRAF and NRAS are frequently mutated in these tumor types, BRAF mutations have a higher incidence in PTC (60%) than ATC (37%), and NRAS mutations are more frequent in FTC (30–45%) than ATC (21%), supporting early divergence.

Figure 3
Figure 3

Genetic evolution of thyroid cancers. The diagram postulates that transformation from normal thyroid cells is associated with the accumulation of an increasing number of mutated genes, starting with early driver mutations in the BRAF, RAS, or RET genes. Tumors can then evolve to a less differentiated morphology and more aggressive behavior in poorly differentiated thyroid cancers (PDTC) with the acquisition of TERT promoter and/or PTEN mutations. Ultimately the tumors can develop into ATC through the acquisition of additional mutations in TP53, NF1, and PIK3CA. Data from Pozdeyev et al. (2018).

Citation: Endocrine-Related Cancer 29, 11; 10.1530/ERC-22-0129

MTCs originate from the interstitial C cells of the thyroid and account for 2–3% of thyroid cancers. Most cases are sporadic but familial cases exist that are inherited in an autosomal dominant manner and account for approximately 20% of the cases. Familial MTCs are most of the time associated with gain of function germline RET receptor mutations. Somatic RET mutations occur often in sporadic MTCs (30–60%), but RAS mutations can also be found. Evidence suggests that RET and RAS mutations very rarely overlap (Ciampi et al. 2019). Further, RET mutations are generally associated with an elevated risk for metastasis, tumor recurrence, and patient mortality while the association of RAS mutations with tumor aggressiveness in sporadic MTC is unclear (Vuong et al. 2018).

Molecular targeted therapies in thyroid cancers

While more than 90% of localized, well-differentiated carcinomas of the thyroid such as PTC and FTC can be cured by surgery combined with thyroid-stimulating hormone (TSH)-suppressive thyroid hormone therapy and occasional use of radioactive iodine, cancers that persist or recur following these regimens have a poorer prognosis. Cytotoxic chemotherapy or external beam radiotherapy shows poor efficacy in these patients. In addition, since MTC tumor cells have a neuroendocrine origin, they do not respond to radioiodine or TSH suppression. Therefore, the advent of therapies targeting growth factor receptors, or constitutively activated protein kinases, represented a welcomed additional option. This led to significant improvements in the treatment of advanced cases of radioiodine refractory PTC and FTC, as well as PDTC and ATC. In the United States, there are currently eight drugs and one drug combination that are FDA-approved for advanced thyroid cancers. Three antiangiogenic drugs, sorafenib, lenvatinib, and cabozantinib, are FDA-approved for DTCs (Brose et al. 2014, 2021, Schlumberger et al. 2015). The combination of dabrafenib and trametinib is FDA-approved for BRAFV600E-mutated ATC (Subbiah et al. 2018b, 2022, Wang et al. 2019a). The NTRK inhibitors larotrectinib and entrectinib are approved for NTRK fusion solid tumors, regardless of tumor histology. In MTCs, two antiangiogenic drugs, vandetanib and cabozantinib, are FDA-approved. Finally, the selective RET inhibitors pralsetinib and selpercatinib are approved for RET-altered thyroid cancers (Wells et al. 2012, Elisei et al. 2013, Subbiah et al. 2018a, (Fig. 1). All these drugs have resulted in remarkable improvement in progression-free survival, in particular in ATC. The anti-angiogenic drugs inhibit VEGFR activity, but they are not specific; whereas some secondary targets may also be anti-angiogenic, such as FGFR, others can lead to ‘off-target’ toxicities (Table 1). This lack of target specificity might cause side effects, such as fatigue, diarrhea, and weight loss, as well as poor wound healing, hypertension, and congestive heart failure. However, the selective RET inhibitors pralsetinib and selpercatinib are new-generation drugs that show a safer toxicity profile probably due to lower inhibition of other tyrosine kinases, including VEGFR (Subbiah et al. 2020, Wirth et al. 2020).

The majority of mutations found in thyroid cancers affect components of the MAPK pathway, with most alterations found in the RAS and BRAF genes (Fig. 2B). Targeting RAS mutations in cancers has proven challenging, but recent advances in targeting KRASG12C with inhibitors such as sotorasib and adagrasib have been promising (Huang et al. 2021). These drugs form an irreversible, covalent bond with the cysteine residue of KRASG12C, holding the protein in its inactive form and blocking downstream signaling. Unfortunately, KRASG12C mutations are very rare in thyroid cancers. As mentioned earlier, 60% of PTCs and 37% of ATCs harbor a BRAF mutation, most often the BRAFV600E mutation. Dabrafenib is a BRAFV600E-specific drug, which is FDA-approved alone or in combination with an MEK inhibitor such as trametinib for melanoma (Long et al. 2014). This combination was recently approved for most solid tumors in patients who have progressed following prior treatment and have no satisfactory alternative treatment options. In thyroid cancers, dabrafenib is only approved for BRAFV600E-mutated ATCs in combination with trametinib (Subbiah et al. 2018b). The combination dabrafenib–trametinib is also used as neoadjuvant therapy followed by surgery in cases of advanced unresectable disease (Wang et al. 2019a). This regimen represents significant progress in the treatment of ATC as long-term survival of these patients is now significantly increased (Maniakas et al. 2020, Subbiah et al. 2022).

Resistance to kinase inhibitors

Despite a large amount of promising data and early therapeutic successes, blocking signaling pathways with kinase inhibitors appears to be ultimately ineffective and most patients progress. To overcome this issue, second-generation BRAF and MEK inhibitors are presently studied in thyroid cancers, such as encorafenib (type I BRAF inhibitor) and binimetinib (type III MEK inhibitor), which showed significant benefit in metastatic colorectal cancer and advanced melanoma (Table 1) (Dummer et al. 2018, Tabernero et al. 2021). Many tumors display multiple coexisting cellular subclones that confer a high degree of heterogeneity; therefore, single-targeting approaches are limited because new clones may be selected that become dominant. Inherent genomic instability, such as found in ATC, predisposes the tumor cells to rapidly acquire additional alterations that increase their fitness in a changing microenvironment. Efforts have been made to combine targeted therapies with immune checkpoint inhibitors. In DTCs, treatments with immunotherapy are associated with low response rates (Mehnert et al. 2019). Side effects of immunotherapy have led to treatment discontinuations in RET-mutated tumors (Hegde et al. 2020). In ATC, where both PD-L1 positive cells and tumor-infiltrating lymphocytes are present in high frequency, kinase inhibitors combined with immunotherapy are useful only in a subset of patients, but the reasons are unclear (Iyer et al. 2018, Wang et al. 2019a, Capdevila et al. 2020).

Resistance to VEGFR inhibitors

Molecular-targeted therapies inhibiting the VEGFR tyrosine kinase have dominated the field of DTC therapeutics. Because this approach also targets other receptor tyrosine kinases such as RET, FGFR, PDGFR, KIT, and MET, it is likely to remain important in the treatment of this disease. Although some patient subsets show prolonged overall survival (Elisei et al. 2013, Brose et al. 2017), rates of cure are low, responses are only partial, and patients remain on drug for prolonged periods of time causing long-term toxicity. Acquired resistance to anti-VEGF therapy often involves escape mechanisms that activate parallel signaling pathways. For example, upregulation of fibroblast growth factor (FGF2) signaling is often observed in anti-VEGF-resistant tumors, especially in tumors that are exposed to a hypoxic environment (Casanovas et al. 2005). While preclinical studies showed that dual blockade of VEGF and FGF signaling pathways was beneficial, this did not translate in clinical studies (Semrad et al. 2017). Acquired resistance to sorafenib can also be due to activation of the PI3K/AKT pathway or reactivation of the JAK-STAT pathway and STAT3 phosphorylation (Zhu et al. 2017). Further, epigenetic mechanisms might also explain sorafenib resistance. For example, the group of Wang and colleagues demonstrated that miR-124 and miR-506 were significantly reduced in the serum of sorafenib-resistant thyroid cancer patients and that their common target, EZH2, was upregulated in these tumors and thyroid cancer cell lines in comparison with controls (Wang et al. 2019b). EZH2 is a well-known histone modification factor that represses the transcription of specific genes. Its upregulation is associated with poor clinical outcomes in many cancer types and EZH2 inhibitors are presently tested in clinical trials (Duan et al. 2020, Li et al. 2021a). Acquired resistance to lenvatinib in thyroid cancers has been also studied, mainly in preclinical models (Khan et al. 2019, Bertol et al. 2022). Khan and colleagues performed long-term cultures of ATC cells exposed to lenvatinib for 72 days. They demonstrated significant changes in lenvatinib-resistant cells toward a mesenchymal morphology. Transcriptomic and proteomic analyses uncovered upregulation of SNAIL and ZEB1 genes/proteins, activation of pro-survival signaling pathways, activation of the nuclear exporter protein exportin 1 (XPO1), and activation of the RAC/RHO GTPase effector p21 activated kinases (PAK). When combined with lenvatinib, XPO1 and PAK4 inhibitors showed antitumor activity that was superior to lenvatinib alone, both in vitro and in subcutaneous xenografts (Khan et al. 2019). This and similar studies may pave the way for future novel therapies.

Resistance to BRAFV600E inhibitors

Acquired (or secondary) resistance to BRAFV600E inhibitors presents a significant therapeutic challenge in thyroid cancer patients. Vemurafenib and dabrafenib monotherapies increase progression-free survival, but responses are incomplete (Kim et al. 2013, Dadu et al. 2015, Falchook et al. 2015, Brose et al. 2016). Resistance mechanisms have been extensively studied in melanoma and colon cancers. Sequence analysis of the mutated BRAF gene in vemurafenib-resistant melanoma samples demonstrated that secondary mutations within this gene are rare (Nazarian et al. 2010). Only one case of an additional mutation in the kinase domain was found (Wagenaar et al. 2014). Therefore, other mechanisms, including the activation of parallel signaling pathways, may be responsible for acquired resistance.

Overexpression of membrane receptors

Several studies have now demonstrated that cancer cells adapt to BRAF inhibitor therapies by overexpressing growth factor receptors at their surface. This is often the case for KIT, MET, EGFR, and PDGFRβ. This increase in expression may be epigenetically directed since no secondary activating mutation nor DNA amplification of these genes were detected (Nazarian et al. 2010, Corcoran et al. 2012, Prahallad et al. 2012). Corcoran and colleagues showed that in colorectal cancers, resistance to vemurafenib was mediated by the epigenetic upregulation of EGFR, which reactivated RAS and CRAF, and the downstream phosphorylation of ERK1/2 (Corcoran et al. 2012). Similarly, Montero-Conde and colleagues demonstrated that BRAF-mutated PTC cell lines acquire resistance to vemurafenib by overexpressing the ERBB or FGF receptor families, therefore reactivating the MAPK pathway downstream of activated RAS (Montero-Conde et al. 2013). Resistance to vemurafenib in melanoma can also be caused by overexpression of EPHA2, a member of the Ephrin receptor family of tyrosine kinases (Miao et al. 2015). These mechanisms of resistance driven by membrane receptor overexpression imply that the corresponding ligands must be present in sufficient quantity in the cellular microenvironment for their activation. Often, ligand production is indeed upregulated in an autocrine manner in cancer cells, and experimental data have shown that most cells can bypass receptor tyrosine kinase inhibitors by simply exposing them to one or more receptor tyrosine kinase ligands (Wilson et al. 2012). For example, exposure to hepatocyte growth factor from the microenvironment will promote intrinsic resistance to BRAF inhibitors in melanoma, colorectal cancer, and glioblastoma cells overexpressing the MET receptor (Straussman et al. 2012). These results underscore the importance of the cellular microenvironment for drug resistance.

Gene amplification

Gain of WT gene copy numbers has been associated with resistance to BRAF inhibitors. For example, several copies of the gene MCL1, a BCL2 family member and apoptosis modulator, have been associated with resistance to vemurafenib treatment in PTC (Duquette et al. 2015). Our group recently demonstrated that resistance to BRAF inhibitors can also be caused by acquired chromosomal polyploidy. In one case of PTC that dedifferentiated into ATC during dabrafenib treatment, we observed triploidy of chromosome 7, which harbors the EGFR, RAC1, MET, and BRAF genes. Copy numbers and expression of these protooncogenes were consequently increased in the dedifferentiated sample, probably causing tumor progression (Bagheri-Yarmand et al. 2021). It is now evident that overexpression of any non-mutated receptor tyrosine kinase has the potential to reactivate the MAPK pathway (Yadav et al. 2012) or the PI3K pathway (Atefi et al. 2011, Chapman et al. 2011, Greger et al. 2012, Delmas et al. 2015, Byeon et al. 2017) provided that the ligand is present in sufficient quantity.

Point mutations

Acquired resistance to BRAF inhibitors in melanoma and colorectal cancers has often been attributed to secondary mutations in the NRAS or KRAS genes (Nazarian et al. 2010, Kopetz et al. 2015, Monsma et al. 2015). Our group sought to identify the acquisition of secondary resistance mechanisms in thyroid cancers by exposing BRAFV600E -mutated PTC cells to long-term treatment with vemurafenib for a period of 5 months (Danysh et al. 2016). This treatment induced upregulation of the MET, EGF, and ERBB receptors and activation of the PI3K/AKT and MAPK pathways, as previously described by others (Montero-Conde et al. 2013, Byeon et al. 2017). Using WES, we also identified a subpopulation of cells harboring a secondary KRAS mutation (KRASG12D), conferring them significant proliferative and invasive advantages. These data, obtained with a cell line, were later validated by the observation that about 50% of our BRAFV600E-mutated patients progressing under vemurafenib or dabrafenib treatment (with/without MEK inhibitors) harbored a secondary RAS mutation (NRAS or KRAS) after WES analysis (Table 2) (Owen et al. 2019, Cabanillas et al. 2020). These studies were the first to establish that acquired resistance to kinase inhibitors in thyroid cancers can be RAS-driven. In addition to RAS point mutations, secondary mutations that possibly conferred drug resistance were often found in the PTEN, NF1, NF2, TP53, and CDKN2A genes (Table 2). We recently analyzed a secondary mutation in the RAC1 gene, RAC1P34R, found in a lymph node metastasis of a PTC patient who progressed under dabrafenib treatment. A cell line was established that showed dabrafenib resistance in vitro, and sensitivity to the drug was restored with a RAC1 activity inhibitor (Bagheri-Yarmand et al. 2021). RAC1 is a small GTPase involved in cell motility and cytoskeletal reorganization, as well as cell proliferation through its canonical targets, PAK1-3 (Mack et al. 2011). The RAC1P29S mutation, which affects the same protein domain as RAC1P34R, is a known driver mutation in primary and drug-resistant melanoma (Watson et al. 2014, Cannon et al. 2020). Similar to RAS, efforts to target RAC1 itself have been challenging; therefore, approaches to target downstream effectors such as PAK1-3 are underway. Understanding tumor heterogeneity and mutational patterns emerging under drug pressure is therefore fundamental to improving therapies.

Table 2

Mutations in PTC, PDTC, and ATC patients at baseline and at progression.

Patient Histology Mutation(s) at baseline Kinase inhibitor(s) Immune checkpoint inhibitor Additional mutations at progression
1 PTC BRAFV600E D+T ND BRCAD237A

PTENE40*
2 ATC BRAFV600E

PIK3CAE525K

TP53Q331*
D+T ND NF1R1276Q
3 PDTC BRAFV600E D ND CDKN2AD84N

TP53R273H
4 ATC BRAFV600E V ND KITM541L
5 ATC BRAFV600E D+T ND NF2K40fs*3
6 ATC BRAFV600E D+T ND NF2Y144*
7 ATC BRAFV600E

PIK3CAH1047R

TP53R273S
D+T ND METR1005Q

NOTCH1P2138R

NOTCH1E360K
8 ATC BRAFV600E

TP53R175H

EGFRG322S
D+T ND NRASQ61K
9 PDTC BRAFV600E

ATMI1986V
D+T ND KRASG12V
10 PDTC BRAFV600E

PIK3CAE545K
V+C A KRASG12D
11 ATC BRAFV600E

PIK3CAN345K

TP53R213*
V+C A NRASQ61K
12 PTC BRAFV600E V ND KRASG12V
13 PTC BRAFV600E D+T ND KRASG12V
14 PTC BRAFV600E D+T P NRASG13D

A, atezolizumab; C, cobimetinib; D, dabrafenib; ND, no immunotherapy; P, pembrolizumab; T, trametinib; V, vemurafenib.

Super-enhancers

Enhancers are short sequences of DNA that are located near or far from promoter regions and can be bound by specific transcription factors to increase transcription. In a broad range of human cell types, super-enhancers (SEs) are large clusters of enhancers with high levels of transcription factor binding (Tang et al. 2020). SEs are critical for driving the expression of genes that control cell identity. They also regulate and increase the expression of key oncogenes such as c-MYC in many tumor cells, and their development drives drug resistance (Li et al. 2021b). Therefore, disrupting SE structure or inhibiting SE protein co-factors can be utilized as therapeutic strategies. For example, BET bromodomain inhibitors can repress c-MYC expression by decreasing the binding of bromodomain-containing-protein 4 to c-MYC SE regions (Delmore et al. 2011). SE-dependent transcription also utilizes cyclin-dependent kinase 7 (CDK7) at the initiation complex (TFIIH), which can be inhibited by the covalent inhibitor THZ1 (Chipumuro et al. 2014). Inhibitors of BET and CDK7 were recently tested in preclinical studies and significantly reduced tumor growth in cancer cell lines and tumor-bearing mice (Mertz et al. 2011, Nakamura et al. 2017). The effects of selective CDK7 and BET inhibitors in advanced solid tumors are now studied in clinical trials. In thyroid cancer cells, BET inhibitors repressed MYC expression in ATC, and the CDK7 inhibitor THZ1 repressed RET expression in MTC cells (Valenciaga et al. 2018, Cao et al. 2019, Zhu et al. 2019).

Overcoming resistance

Additional kinase inhibitors

Overcoming acquired resistance in BRAF-mutated PTC and ATC has been difficult because targeting MEK downstream of BRAFV600E with MEK inhibitors such as selumetinib, cobimetinib, and trametinib, alone or in combination with BRAF inhibitors, rarely prevents progression (Table 2) (Wagle et al. 2014, Fofaria et al. 2015, Cabanillas et al. 2020). In addition, BRAF and/or MEK inhibitors might reactivate ERK1/2 activation/phosphorylation through parallel signaling pathways (Samatar & Poulikakos 2014). Therefore, recent efforts have focused on targeting ERK1/2/ itself (Germann et al. 2017, Roskoski 2019). Drugs showing promising preclinical activities such as BVD-523 (ulixertinib) are still in clinical trials, but the FDA recently granted expanded access to ulixertinib for treatment of stage IIb through Stage IV BRAF-mutant melanoma. Unfortunately, resistance to ERK1/2 inhibitors has already been identified, with activation of MEK5-ERK5 as a compensatory mechanism (Tubita et al. 2021).

In a recent study, Iyer and colleagues showed that ATC patients treated with lenvatinib, or dabrafenib combined with trametinib, showed additional response when given the PD-L1 inhibitor pembrolizumab after initial progression. Therefore, in certain cases, immunotherapy can be used as an effective therapy to improve the benefits of kinase inhibitors (Iyer et al. 2018).

Redifferentiation therapies

The uptake of iodine by thyroid follicular cells is mediated by a sodium iodide symporter (NIS) located in the basolateral membrane of follicular cells. NIS spatiotemporal expression and function depends on a number of modulators. TSH (thyrotropin) upregulates NIS expression and stimulates its transport to the plasma membrane (Kogai et al. 1997, Riedel et al. 2001). PIGU, a subunit of GPI transamidase that attaches GPI-anchors to proteins, ensures proper anchoring of NIS at the endoplasmic reticulum, and therefore at the plasma membrane after its vesicular transport, itself stimulated by ARF4 (Eisenhaber et al. 1998, Amit et al. 2017, Fletcher et al. 2020). These NIS effectors are counterbalanced by inhibitors of NIS function, expression, and localization. For example, at the plasma membrane, the NIS inhibitor PTTG-binding factor (PBF) inhibits the uptake of iodide by binding NIS (Smith et al. 2009). Further, β-catenin seems to control the availability of NIS since its overexpression leads to the decrease of NIS expression and its transfer from the plasma membrane to an intracellular location close to the nucleus (Lan et al. 2017a, b). In thyroid cancer cells, the balance between positive and negative NIS modulating factors is dysregulated. Cellular dedifferentiation, which increases with the accumulation of BRAF, RAS, TERT, or other gene mutations, correlates with the downregulation of NIS expression at the plasma membrane and its relocation in the cytoplasm, which is not its proper functional location (Tavares et al. 2018, Martin et al. 2019). Thus, redifferentiation therapies often aim at redistributing NIS properly at the plasma membrane to improve NIS-mediated RAI treatment.

Increasing the level of serum thyrotropin (TSH) before RAI therapy, either by inducing hypothyroidism or by administering recombinant human TSH, will significantly increase the uptake of RAI in differentiated tumors with intact iodine uptake mechanisms (Klubo-Gwiezdzinska et al. 2012, Haugen et al. 2016). However, additional modalities are required to re-differentiate tumors that have lost the capability to uptake iodine. Approaches have been attempted to increase NIS expression at the plasma membrane. For example, retinoic acid (RA) has been used to redifferentiate cancer cells for decades. RA is the bioactive metabolite of vitamin A and plays a critical role in cellular differentiation during mammalian development. RA indeed may increase the expression of NIS mRNA in human FTC cells (Schmutzler et al. 2002, Lan et al. 2017a); however a recent meta-analysis reported that only a minority of patients with RAI-refractory DTC responded to RA treatment (Pak et al. 2018). This poor response might be explained by the fact that RA treatment induces growth inhibition through retinoic acid receptor beta in cells where it is expressed, rather than through an increase of NIS at the plasma membrane (Elisei et al. 2005). Similarly, PPARG agonists and histone deacetylase inhibitors, which showed good effects in preclinical studies, offered disappointing results in clinical trials (Park et al. 2005, Woyach et al. 2009, Rosenbaum-Krumme et al. 2012, Sherman et al. 2013, Cheng et al. 2016, Nilubol et al. 2017). Further, PBF phosphorylation levels could be reduced using a c-SRC kinase inhibitor, at least in vitro (Smith et al. 2013). Other preclinical studies indicated that treatment with MEK inhibitors rescued PIGU expression in PTC cells (Amit et al. 2017). Recently, Fletcher and coworkers demonstrated that clotrimazole, an antifungal medication,and ebastine, an antihistaminic, also enhanced RAI uptake in normal mouse thyrocytes and human thyroid cancer cells due to their allosteric inhibition of valosin-containing protein (VCP/p97). They also demonstrated the importance of ARF4 in the trafficking of NIS to the plasma membrane (Fletcher et al. 2020). Further, clinically relevant redifferentiation was previously obtained in small cohorts of patients treated with MAPK pathway/BRAF inhibitors. In particular, dabrafenib, trametinib, and selumetinib seemed to improve the iodine uptake rate of thyroid cancer cells by improving NIS expression and function (Ho et al. 2013, Rothenberg et al. 2015, Jaber et al. 2018, Dunn et al. 2019, Leboulleux et al. 2021). However, a recently completed phase III clinical trial showed that the addition of selumetinib to adjuvant RAI did not significantly improve the complete remission rate in DTC patients with metastatic disease (Ho et al. 2022). Finally, redifferentiation of NTRK-rearranged, RAI-resistant thyroid tumors was possible with larotrectinib (Groussin et al. 2022), and selpercatinib restored iodine uptake in almost all metastases in a patient with a RET rearrangement/fusion as a sole mutation (Groussin et al. 2021). While these results are encouraging, they were obtained from case studies. Therefore, these drugs should not yet be used as routine short-term pretreatments before RAI in DTCs.

Conclusions

Protein kinases play critical roles in signaling pathways that transfer information from receptors located at the plasma membrane down to the nucleus, eventually modifying and adapting cell behavior to the environment. Mutational changes can dysregulate the function of many of these kinases, which become constitutively active and lead to cancer cell proliferation and metastases. Targeted therapies use kinase inhibitors that often target the ATP-binding pocket. These novel drugs are better tolerated than chemotherapies. However, resistance to these inhibitors is a challenge that will need to be overcome. Multiple molecular strategies will be required to thoroughly understand mechanisms of resistance and devise effective targeted treatments, including disruption of the transcriptional machinery of specific genes and cellular redifferentiation. Finally, there is also a need to identify biomarkers that will help predict response and resistance to improve the selection of an optimal targeted therapy for each patient.

Declaration of interest

Mark Zafereo has research (clinical trials) from Merck and Eli Lilly. Maria E Cabanillas has research grant funding from Merck, Eisai, Exelixis and Genentech, and has participated in advisory boards for Exelixis, Blueprint, Ignyta, Bayer and LOXO/Eli Lilly. Stephen Y Lai is a medical affairs consultant for Cardinal Health. The other authors have nothing to disclose.

Funding

This work was supported by the National Cancer Institute (NCI) Grants P30CA016672, R21CA259839, R21CA259839, R01CA227847 as well as the MD Anderson Cancer Center Petrick Thyroid Cancer Fund.

References

  • Amit M, Na’ara S, Francis D, Matanis W, Zolotov S, Eisenhaber B, Eisenhaber F, Weiler Sagie M, Malkin L & Billan S et al.2017 Post-translational regulation of radioactive iodine therapy response in papillary thyroid carcinoma. Journal of the National Cancer Institute 109 djx092. (https://doi.org/10.1093/jnci/djx092)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Atefi M, Von Euw E, Attar N, Ng C, Chu C, Guo D, Nazarian R, Chmielowski B, Glaspy JA & Comin-Anduix B et al.2011 Reversing melanoma cross-resistance to BRAF and MEK inhibitors by co-targeting the AKT/mTOR pathway. PLoS ONE 6 e28973. (https://doi.org/10.1371/journal.pone.0028973)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bagheri-Yarmand R, Busaidy NL, Mcbeath E, Danysh BP, Evans KW, Moss TJ, Akcakanat A, Ng PKS, Knippler CM & Golden JA et al.2021 RAC1 alterations induce acquired dabrafenib resistance in association with anaplastic transformation in a papillary thyroid cancer patient. Cancers 13 4950. (https://doi.org/10.3390/cancers13194950)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barras D 2015 BRAF mutation in colorectal cancer: an update. Biomarkers in Cancer 7 912. (https://doi.org/10.4137/BIC.S25248)

  • Bertol BC, Bales ES, Calhoun JD, Mayberry A, Ledezma ML, Sams SB, Orlicky DJ, Donadi EA, Haugen BR & French JD 2022 Lenvatinib plus anti-PD-1 combination therapy for advanced cancers: defining mechanisms of resistance in an inducible transgenic model of thyroid cancer. Thyroid 32 153163. (https://doi.org/10.1089/thy.2021.0371)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bhullar KS, Lagaron NO, Mcgowan EM, Parmar I, Jha A, Hubbard BP & Rupasinghe HPV 2018 Kinase-targeted cancer therapies: progress, challenges and future directions. Molecular Cancer 17 48. (https://doi.org/10.1186/s12943-018-0804-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bonora E, Porcelli AM, Gasparre G, Biondi A, Ghelli A, Carelli V, Baracca A, Tallini G, Martinuzzi A & Lenaz G et al.2006 Defective oxidative phosphorylation in thyroid oncocytic carcinoma is associated with pathogenic mitochondrial DNA mutations affecting complexes I and III. Cancer Research 66 60876096. (https://doi.org/10.1158/0008-5472.CAN-06-0171)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brognard J & Hunter T 2011 Protein kinase signaling networks in cancer. Current Opinion in Genetics and Development 21 411. (https://doi.org/10.1016/j.gde.2010.10.012)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, De La Fouchardiere C, Pacini F, Paschke R & Shong YK et al.2014 Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet 384 319328. (https://doi.org/10.1016/S0140-6736(1460421-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brose MS, Cabanillas ME, Cohen EE, Wirth LJ, Riehl T, Yue H, Sherman SI & Sherman EJ 2016 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: Oncology 17 12721282. (https://doi.org/10.1016/S1470-2045(1630166-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brose MS, Worden FP, Newbold KL, Guo M & Hurria A 2017 Effect of age on the efficacy and safety of lenvatinib in radioiodine-refractory differentiated thyroid cancer in the phase III SELECT trial. Journal of Clinical Oncology 35 26922699. (https://doi.org/10.1200/JCO.2016.71.6472)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brose MS, Robinson B, Sherman SI, Krajewska J, Lin CC, Vaisman F, Hoff AO, Hitre E, Bowles DW & Hernando J et al.2021 Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet: Oncology 22 11261138. (https://doi.org/10.1016/S1470-2045(2100332-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Byeon HK, Na HJ, Yang YJ, Ko S, Yoon SO, Ku M, Yang J, Kim JW, Ban MJ & Kim JH et al.2017 Acquired resistance to BRAF inhibition induces epithelial-to-mesenchymal transition in BRAF (V600E) mutant thyroid cancer by c-Met-mediated AKT activation. Oncotarget 8 596609. (https://doi.org/10.18632/oncotarget.13480)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cabanillas ME, Ryder M & Jimenez C 2019 Targeted therapy for advanced thyroid cancer: kinase inhibitors and beyond. Endocrine Reviews 40 15731604. (https://doi.org/10.1210/er.2019-00007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cabanillas ME, Dadu R, Iyer P, Wanland KB, Busaidy NL, Ying A, Gule-Monroe M, Wang JR, Zafereo M & Hofmann MC 2020 Acquired secondary RAS mutation in BRAF(V600E)-mutated thyroid cancer patients treated with BRAF inhibitors. Thyroid 30 12881296. (https://doi.org/10.1089/thy.2019.0514)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cancer Genome Atlas Research Network 2014 Integrated genomic characterization of papillary thyroid carcinoma. Cell 159 676690. (https://doi.org/10.1016/j.cell.2014.09.050)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cannon AC, Uribe-Alvarez C & Chernoff J 2020 RAC1 as a therapeutic target in malignant melanoma. Trends in Cancer 6 478488. (https://doi.org/10.1016/j.trecan.2020.02.021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cao X, Dang L, Zheng X, Lu Y, Lu Y, Ji R, Zhang T, Ruan X, Zhi J & Hou X et al.2019 Targeting super-enhancer-driven oncogenic transcription by CDK7 inhibition in anaplastic thyroid carcinoma. Thyroid 29 809823. (https://doi.org/10.1089/thy.2018.0550)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Capdevila J, Mayor R, Mancuso FM, Iglesias C, Caratu G, Matos I, Zafon C, Hernando J, Petit A & Nuciforo P et al.2019 Early evolutionary divergence between papillary and anaplastic thyroid cancers. Annals of Oncology 30 1843. (https://doi.org/10.1093/annonc/mdz216)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Capdevila J, Wirth LJ, Ernst T, Ponce Aix S, Lin CC, Ramlau R, Butler MO, Delord JP, Gelderblom H & Ascierto PA et al.2020 PD-1 blockade in anaplastic thyroid carcinoma. Journal of Clinical Oncology 38 26202627. (https://doi.org/10.1200/JCO.19.02727)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Casanovas O, Hicklin DJ, Bergers G & Hanahan D 2005 Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8 299309. (https://doi.org/10.1016/j.ccr.2005.09.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A & Maio M et al.2011 Improved survival with vemurafenib in melanoma with BRAF V600E mutation. New England Journal of Medicine 364 25072516. (https://doi.org/10.1056/NEJMoa1103782)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng W, Liu R, Zhu G, Wang H & Xing M 2016 Robust thyroid gene expression and radioiodine uptake induced by simultaneous suppression of BRAF V600E and histone deacetylase in thyroid cancer cells. Journal of Clinical Endocrinology and Metabolism 101 962971. (https://doi.org/10.1210/jc.2015-3433)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chipumuro E, Marco E, Christensen CL, Kwiatkowski N, Zhang T, Hatheway CM, Abraham BJ, Sharma B, Yeung C & Altabef A et al.2014 CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription in MYCN-driven cancer. Cell 159 11261139. (https://doi.org/10.1016/j.cell.2014.10.024)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ciampi R, Romei C, Ramone T, Prete A, Tacito A, Cappagli V, Bottici V, Viola D, Torregrossa L & Ugolini C et al.2019 Genetic landscape of somatic mutations in a large cohort of sporadic medullary thyroid carcinomas studied by next-generation targeted sequencing. iScience 20 324336. (https://doi.org/10.1016/j.isci.2019.09.030)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cicenas J, Zalyte E, Bairoch A & Gaudet P 2018 Kinases and cancer. Cancers 10 63. (https://doi.org/10.3390/cancers10030063)

  • Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, Brown RD, Della Pelle P, Dias-Santagata D & Hung KE et al.2012 EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discovery 2 227235. (https://doi.org/10.1158/2159-8290.CD-11-0341)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dadu R, Shah K, Busaidy NL, Waguespack SG, Habra MA, Ying AK, Hu MI, Bassett R, Jimenez C & Sherman SI et al.2015 Efficacy and tolerability of vemurafenib in patients with BRAF(V600E)-positive papillary thyroid cancer: M.D. Anderson Cancer Center off label experience. Journal of Clinical Endocrinology and Metabolism 100 E77E81. (https://doi.org/10.1210/jc.2014-2246)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Danysh BP, Rieger EY, Sinha DK, Evers CV, Cote GJ, Cabanillas ME & Hofmann MC 2016 Long-term vemurafenib treatment drives inhibitor resistance through a spontaneous KRAS G12D mutation in a BRAF V600E papillary thyroid carcinoma model. Oncotarget 7 3090730923. (https://doi.org/10.18632/oncotarget.9023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ & Bottomley W et al.2002 Mutations of the BRAF gene in human cancer. Nature 417 949954. (https://doi.org/10.1038/nature00766)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delmas A, Cherier J, Pohorecka M, Medale-Giamarchi C, Meyer N, Casanova A, Sordet O, Lamant L, Savina A & Pradines A et al.2015 The c-Jun/RHOB/AKT pathway confers resistance of BRAF-mutant melanoma cells to MAPK inhibitors. Oncotarget 6 1525015264. (https://doi.org/10.18632/oncotarget.3888)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM & Qi J et al.2011 BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146 904917. (https://doi.org/10.1016/j.cell.2011.08.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duan R, Du W & Guo W 2020 EZH2: a novel target for cancer treatment. Journal of Hematology and Oncology 13 104. (https://doi.org/10.1186/s13045-020-00937-8)

  • Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, Garbe C, Schadendorf D, Krajsova I & Gutzmer R et al.2018 Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial. Lancet: Oncology 19 603615. (https://doi.org/10.1016/S1470-2045(1830142-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dunn LA, Sherman EJ, Baxi SS, Tchekmedyian V, Grewal RK, Larson SM, Pentlow KS, Haque S, Tuttle RM & Sabra MM et al.2019 Vemurafenib redifferentiation of BRAF mutant, RAI-refractory thyroid cancers. Journal of Clinical Endocrinology and Metabolism 104 14171428. (https://doi.org/10.1210/jc.2018-01478)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Duquette M, Sadow PM, Husain A, Sims JN, Antonello ZA, Fischer AH, Song C, Castellanos-Rizaldos E, Makrigiorgos GM & Kurebayashi J et al.2015 Metastasis-associated MCL1 and P16 copy number alterations dictate resistance to vemurafenib in a BRAFV600E patient-derived papillary thyroid carcinoma preclinical model. Oncotarget 6 4244542467. (https://doi.org/10.18632/oncotarget.6442)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eisenhaber B, Bork P & Eisenhaber F 1998 Sequence properties of GPI-anchored proteins near the omega-site: constraints for the polypeptide binding site of the putative transamidase. Protein Engineering 11 11551161. (https://doi.org/10.1093/protein/11.12.1155)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elisei R, Vivaldi A, Agate L, Ciampi R, Molinaro E, Piampiani P, Romei C, Faviana P, Basolo F & Miccoli P et al.2005 All-trans-retinoic acid treatment inhibits the growth of retinoic acid receptor beta messenger ribonucleic acid expressing thyroid cancer cell lines but does not reinduce the expression of thyroid-specific genes. Journal of Clinical Endocrinology and Metabolism 90 24032411. (https://doi.org/10.1210/jc.2004-0969)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Elisei R, Schlumberger MJ, Muller SP, Schoffski P, Brose MS, Shah MH, Licitra L, Jarzab B, Medvedev V & Kreissl MC et al.2013 Cabozantinib in progressive medullary thyroid cancer. Journal of Clinical Oncology 31 36393646. (https://doi.org/10.1200/JCO.2012.48.4659)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fagin JA & Wells Jr SA 2016 Biologic and clinical perspectives on thyroid cancer. New England Journal of Medicine 375 10541067. (https://doi.org/10.1056/NEJMra1501993)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Falchook GS, Millward M, Hong D, Naing A, Piha-Paul S, Waguespack SG, Cabanillas ME, Sherman SI, Ma B & Curtis M et al.2015 BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer. Thyroid 25 7177. (https://doi.org/10.1089/thy.2014.0123)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fletcher A, Read ML, Thornton CEM, Larner DP, Poole VL, Brookes K, Nieto HR, Alshahrani M, Thompson RJ & Lavery GG et al.2020 Targeting novel sodium iodide symporter interactors ADP-ribosylation factor 4 and valosin-containing protein enhances radioiodine uptake. Cancer Research 80 102115. (https://doi.org/10.1158/0008-5472.CAN-19-1957)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fofaria NM, Frederick DT, Sullivan RJ, Flaherty KT & Srivastava SK 2015 Overexpression of Mcl-1 confers resistance to BRAFV600E inhibitors alone and in combination with MEK1/2 inhibitors in melanoma. Oncotarget 6 4053540556. (https://doi.org/10.18632/oncotarget.5755)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ganly I, Makarov V, Deraje S, Dong Y, Reznik E, Seshan V, Nanjangud G, Eng S, Bose P & Kuo F et al.2018 Integrated genomic analysis of Hurthle cell cancer reveals oncogenic drivers, recurrent mitochondrial mutations, and unique chromosomal landscapes. Cancer Cell 34 256 .e5270.e5. (https://doi.org/10.1016/j.ccell.2018.07.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Germann UA, Furey BF, Markland W, Hoover RR, Aronov AM, Roix JJ, Hale M, Boucher DM, Sorrell DA & Martinez-Botella G et al.2017 Targeting the MAPK signaling pathway in cancer: promising preclinical activity with the novel selective ERK1/2 inhibitor BVD-523 (ulixertinib). Molecular Cancer Therapeutics 16 23512363. (https://doi.org/10.1158/1535-7163.MCT-17-0456)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN, Dickerson SH, Laquerre SG, Liu L & Gilmer TM 2012 Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Molecular Cancer Therapeutics 11 909920. (https://doi.org/10.1158/1535-7163.MCT-11-0989)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Groussin L, Bessiene L, Arrondeau J, Garinet S, Cochand-Priollet B, Lupo A, Zerbit J, Clerc J & Huillard O 2021 Letter to the editor: selpercatinib-enhanced radioiodine uptake in RET-rearranged thyroid cancer. Thyroid 31 16031604. (https://doi.org/10.1089/thy.2021.0144)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Groussin L, Theodon H, Bessiene L, Bricaire L, Bonnet-Serrano F, Cochand-Priollet B, Leroy K, Garinet S, Pasmant E & Zerbit J et al.2022 Redifferentiating effect of larotrectinib in NTRK-rearranged advanced radioactive-iodine refractory thyroid cancer. Thyroid 32 594598. (https://doi.org/10.1089/thy.2021.0524)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hallberg B & Palmer RH 2013 Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology. Nature Reviews: Cancer 13 685700. (https://doi.org/10.1038/nrc3580)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM & Schlumberger M et al.2016 2015 American Thyroid Association management guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: the American Thyroid Association guidelines task force on thyroid nodules and differentiated thyroid cancer. Thyroid 26 1133. (https://doi.org/10.1089/thy.2015.0020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hegde A, Andreev-Drakhlin AY, Roszik J, Huang L, Liu S, Hess K, Cabanillas M, Hu MI, Busaidy NL & Sherman SI et al.2020 Responsiveness to immune checkpoint inhibitors versus other systemic therapies in RET-aberrant malignancies. ESMO Open 5 e000799. (https://doi.org/10.1136/esmoopen-2020-000799)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, Pentlow KS, Zanzonico PB, Haque S & Gavane S et al.2013 Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. New England Journal of Medicine 368 623632. (https://doi.org/10.1056/NEJMoa1209288)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ho AL, Dedecjus M, Wirth LJ, Tuttle RM, Inabnet WB 3rd, Tennvall J, Vaisman F, Bastholt L, Gianoukakis AG & Rodien P et al.2022 Selumetinib plus adjuvant radioactive iodine in patients with high-risk differentiated thyroid cancer: a phase III, randomized, placebo-controlled trial (Astra). Journal of Clinical Oncology 40 18701878. (https://doi.org/10.1200/JCO.21.00714)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hoque MO, Brait M, Rosenbaum E, Poeta ML, Pal P, Begum S, Dasgupta S, Carvalho AL, Ahrendt SA & Westra WH et al.2010 Genetic and epigenetic analysis of erbB signaling pathway genes in lung cancer. Journal of Thoracic Oncology 5 18871893. (https://doi.org/10.1097/JTO.0b013e3181f77a53)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang L, Guo Z, Wang F & Fu L 2021 KRAS mutation: from undruggable to druggable in cancer. Signal Transduction and Targeted Therapy 6 386. (https://doi.org/10.1038/s41392-021-00780-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Iyer PC, Dadu R, Gule-Monroe M, Busaidy NL, Ferrarotto R, Habra MA, Zafereo M, Williams MD, Gunn GB & Grosu H et al.2018 Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. Journal for ImmunoTherapy of Cancer 6 68. (https://doi.org/10.1186/s40425-018-0378-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jaber T, Waguespack SG, Cabanillas ME, Elbanan M, Vu T, Dadu R, Sherman SI, Amit M, Santos EB & Zafereo M et al.2018 Targeted therapy in advanced thyroid cancer to resensitize tumors to radioactive iodine. Journal of Clinical Endocrinology and Metabolism 103 36983705. (https://doi.org/10.1210/jc.2018-00612)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kannaiyan R & Mahadevan D 2018 A comprehensive review of protein kinase inhibitors for cancer therapy. Expert Review of Anticancer Therapy 18 12491270. (https://doi.org/10.1080/14737140.2018.1527688)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Khan HY, Ge J, Nagasaka M, Aboukameel A, Mpilla G, Muqbil I, Szlaczky M, Chaker M, Baloglu E & Landesman Y et al.2019 Targeting XPO1 and PAK4 in 8505C anaplastic thyroid cancer cells: putative implications for overcoming lenvatinib therapy resistance. International Journal of Molecular Sciences 21 237. (https://doi.org/10.3390/ijms21010237)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim KB, Cabanillas ME, Lazar AJ, Williams MD, Sanders DL, Ilagan JL, Nolop K, Lee RJ & Sherman SI 2013 Clinical responses to vemurafenib in patients with metastatic papillary thyroid cancer harboring BRAF(V600E) mutation. Thyroid 23 12771283. (https://doi.org/10.1089/thy.2013.0057)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Klubo-Gwiezdzinska J, Burman KD, Van Nostrand D, Mete M, Jonklaas J & Wartofsky L 2012 Radioiodine treatment of metastatic thyroid cancer: relative efficacy and side effect profile of preparation by thyroid hormone withdrawal versus recombinant human thyrotropin. Thyroid 22 310317. (https://doi.org/10.1089/thy.2011.0235)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kogai T, Endo T, Saito T, Miyazaki A, Kawaguchi A & Onaya T 1997 Regulation by thyroid-stimulating hormone of sodium/iodide symporter gene expression and protein levels in FRTL-5 cells. Endocrinology 138 22272232. (https://doi.org/10.1210/endo.138.6.5189)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kopetz S, Desai J, Chan E, Hecht JR, O’dwyer PJ, Maru D, Morris V, Janku F, Dasari A & Chung W et al.2015 Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. Journal of Clinical Oncology 33 40324038. (https://doi.org/10.1200/JCO.2015.63.2497)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lahiry P, Torkamani A, Schork NJ & Hegele RA 2010 Kinase mutations in human disease: interpreting genotype-phenotype relationships. Nature Reviews: Genetics 11 6074. (https://doi.org/10.1038/nrg2707)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lan L, Basourakos S, Cui D, Zuo X, Deng W, Huo L, Chen H, Zhang G, Deng L & Shi B et al.2017a ATRA increases iodine uptake and inhibits the proliferation and invasiveness of human anaplastic thyroid carcinoma SW1736 cells: involvement of beta-catenin phosphorylation inhibition. Oncology Letters 14 77337738. (https://doi.org/10.3892/ol.2017.7225)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lan L, Basourakos S, Cui D, Zuo X, Deng W, Huo L, Chen L, Zhang G, Deng L & Shi B et al.2017b Inhibiting beta-catenin expression promotes efficiency of radioiodine treatment in aggressive follicular thyroid cancer cells probably through mediating NIS localization. Oncology Reports 37 426434. (https://doi.org/10.3892/or.2016.5228)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah RH, Dogan S, Ricarte-Filho JC, Krishnamoorthy GP & Xu B et al.2016 Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. Journal of Clinical Investigation 126 10521066. (https://doi.org/10.1172/JCI85271)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leboulleux S, Do Cao CD, Zerdoud S, Attard M, Bournaud C, Benisvy D, Taieb D, Bardet S, Terroir-Cassou-Mounat M & Betrian S et al.2021 MERAIODE: a redifferentiation phase II trial with trametinib and dabrafenib followed by radioactive iodine administration for metastatic radioactive iodine refractory differentiated thyroid cancer patients with a BRAFV600E mutation (NCT 03244956). Journal of the Endocrine Society 5 A876. (https://doi.org/10.1210/jendso/bvab048.1789)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lee S, Kim J, Jo J, Chang JW, Sim J & Yun H 2021 Recent advances in development of hetero-bivalent kinase inhibitors. European Journal of Medicinal Chemistry 216 113318. (https://doi.org/10.1016/j.ejmech.2021.113318)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li C, Wang Y, Gong Y, Zhang T, Huang J, Tan Z & Xue L 2021a Finding an easy way to harmonize: a review of advances in clinical research and combination strategies of EZH2 inhibitors. Clinical Epigenetics 13 62. (https://doi.org/10.1186/s13148-021-01045-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li GH, Qu Q, Qi TT, Teng XQ, Zhu HH, Wang JJ, Lu Q & Qu J 2021b Super-enhancers: a new frontier for epigenetic modifiers in cancer chemoresistance. Journal of Experimental and Clinical Cancer Research 40 174. (https://doi.org/10.1186/s13046-021-01974-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Long GV, Menzies AM, Nagrial AM, Haydu LE, Hamilton AL, Mann GJ, Hughes TM, Thompson JF, Scolyer RA & Kefford RF 2011 Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. Journal of Clinical Oncology 29 12391246. (https://doi.org/10.1200/JCO.2010.32.4327)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Long GV, Stroyakovskiy D, Gogas H, Levchenko E, De Braud F, Larkin J, Garbe C, Jouary T, Hauschild A & Grob JJ et al.2014 Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. New England Journal of Medicine 371 18771888. (https://doi.org/10.1056/NEJMoa1406037)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lopez-Gines C, Gil-Benso R, Ferrer-Luna R, Benito R, Serna E, Gonzalez-Darder J, Quilis V, Monleon D, Celda B & Cerda-Nicolas M 2010 New pattern of EGFR amplification in glioblastoma and the relationship of gene copy number with gene expression profile. Modern Pathology 23 856865. (https://doi.org/10.1038/modpathol.2010.62)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mack NA, Whalley HJ, Castillo-Lluva S & Malliri A 2011 The diverse roles of Rac signaling in tumorigenesis. Cell Cycle 10 15711581. (https://doi.org/10.4161/cc.10.10.15612)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maniakas A, Dadu R, Busaidy NL, Wang JR, Ferrarotto R, Lu C, Williams MD, Gunn GB, Hofmann MC & Cote G et al.2020 Evaluation of overall survival in patients with anaplastic thyroid carcinoma, 2000–2019. JAMA Oncology 6 13971404. (https://doi.org/10.1001/jamaoncol.2020.3362)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martin M, Geysels RC, Peyret V, Bernal Barquero CE, Masini-Repiso AM & Nicola JP 2019 Implications of Na(+)/I(-) symporter transport to the plasma membrane for thyroid hormonogenesis and radioiodide therapy. Journal of the Endocrine Society 3 222234. (https://doi.org/10.1210/js.2018-00100)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martinez R, Defnet A & Shapiro P 2020 Avoiding or co-opting ATP inhibition: overview of type III, IV, V, and VI kinase inhibitors. In Next Generation Kinase Inhibitors. Ed Shapiro P Cham: Springer. (https://doi.org/10.1007/978-3-030-48283-1_3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McFadden DG & Sadow PM 2021 Genetics, diagnosis, and management of Hurthle cell thyroid neoplasms. Frontiers in Endocrinology 12 696386. (https://doi.org/10.3389/fendo.2021.696386)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mehnert JM, Varga A, Brose MS, Aggarwal RR, Lin CC, Prawira A, De Braud F, Tamura K, Doi T & Piha-Paul SA et al.2019 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 19 196. (https://doi.org/10.1186/s12885-019-5380-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mertz JA, Conery AR, Bryant BM, Sandy P, Balasubramanian S, Mele DA, Bergeron L & Sims 3rd RJ 2011 Targeting MYC dependence in cancer by inhibiting BET bromodomains. PNAS 108 1666916674. (https://doi.org/10.1073/pnas.1108190108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Miao B, Ji Z, Tan L, Taylor M, Zhang J, Choi HG, Frederick DT, Kumar R, Wargo JA & Flaherty KT et al.2015 EPHA2 is a mediator of vemurafenib resistance and a novel therapeutic target in melanoma. Cancer Discovery 5 274287. (https://doi.org/10.1158/2159-8290.CD-14-0295)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Monsma DJ, Cherba DM, Eugster EE, Dylewski DL, Davidson PT, Peterson CA, Borgman AS, Winn ME, Dykema KJ & Webb CP et al.2015 Melanoma patient derived xenografts acquire distinct vemurafenib resistance mechanisms. American Journal of Cancer Research 5 15071518.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Montero-Conde C, Ruiz-Llorente S, Dominguez JM, Knauf JA, Viale A, Sherman EJ, Ryder M, Ghossein RA, Rosen N & Fagin JA 2013 Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discovery 3 520533. (https://doi.org/10.1158/2159-8290.CD-12-0531)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nakamura Y, Hattori N, Iida N, Yamashita S, Mori A, Kimura K, Yoshino T & Ushijima T 2017 Targeting of super-enhancers and mutant BRAF can suppress growth of BRAF-mutant colon cancer cells via repression of MAPK signaling pathway. Cancer Letters 402 100109. (https://doi.org/10.1016/j.canlet.2017.05.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N & Sazegar H et al.2010 Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 468 973977. (https://doi.org/10.1038/nature09626)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nikiforov YE 2002 RET/PTC rearrangement in thyroid tumors. Endocrine Pathology 13 316. (https://doi.org/10.1385/ep:13:1:03)

  • Nikiforov YE & Nikiforova MN 2011 Molecular genetics and diagnosis of thyroid cancer. Nature Reviews: Endocrinology 7 569580. (https://doi.org/10.1038/nrendo.2011.142)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nilubol N, Merkel R, Yang L, Patel D, Reynolds JC, Sadowski SM, Neychev V & Kebebew E 2017 A phase II trial of valproic acid in patients with advanced, radioiodine-resistant thyroid cancers of follicular cell origin. Clinical Endocrinology 86 128133. (https://doi.org/10.1111/cen.13154)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ogunwobi OO, Puszyk W, Dong HJ & Liu C 2013 Epigenetic upregulation of HGF and c-Met drives metastasis in hepatocellular carcinoma. PLoS ONE 8 e63765. (https://doi.org/10.1371/journal.pone.0063765)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Okamoto K, Ikemori-Kawada M, Jestel A, Von Konig K, Funahashi Y, Matsushima T, Tsuruoka A, Inoue A & Matsui J 2015 Distinct binding mode of multikinase inhibitor lenvatinib revealed by biochemical characterization. ACS Medicinal Chemistry Letters 6 8994. (https://doi.org/10.1021/ml500394m)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Owen DH, Konda B, Sipos J, Liu T, Webb A, Ringel MD, Timmers CD & Shah MH 2019 KRAS G12V mutation in acquired resistance to combined BRAF and MEK inhibition in papillary thyroid cancer. Journal of the National Comprehensive Cancer Network 17 409413. (https://doi.org/10.6004/jnccn.2019.7292)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pak K, Shin S, Kim SJ, Kim IJ, Chang S, Koo P, Kwak J & Kim JH 2018 Response of retinoic acid in patients with radioactive iodine-refractory thyroid cancer: a meta-analysis. Oncology Research and Treatment 41 100104. (https://doi.org/10.1159/000484206)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park JW, Zarnegar R, Kanauchi H, Wong MG, Hyun WC, Ginzinger DG, Lobo M, Cotter P, Duh QY & Clark OH 2005 Troglitazone, the peroxisome proliferator-activated receptor-gamma agonist, induces antiproliferation and redifferentiation in human thyroid cancer cell lines. Thyroid 15 222231. (https://doi.org/10.1089/thy.2005.15.222)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Park JC, Ashok A, Liu C & Kang H 2022 Real-world experience of NTRK fusion-positive thyroid cancer. JCO Precision Oncology 6 e2100442. (https://doi.org/10.1200/PO.21.00442)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Peng YH, Shiao HY, Tu CH, Liu PM, Hsu JT, Amancha PK, Wu JS, Coumar MS, Chen CH & Wang SY et al.2013 Protein kinase inhibitor design by targeting the Asp-Phe-Gly (DFG) motif: the role of the DFG motif in the design of epidermal growth factor receptor inhibitors. Journal of Medicinal Chemistry 56 38893903. (https://doi.org/10.1021/jm400072p)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pozdeyev N, Gay LM, Sokol ES, Hartmaier R, Deaver KE, Davis S, French JD, Borre PV, Labarbera DV & Tan AC et al.2018 Genetic analysis of 779 advanced differentiated and anaplastic thyroid cancers. Clinical Cancer Research 24 30593068. (https://doi.org/10.1158/1078-0432.CCR-18-0373)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A & Bernards R 2012 Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 483 100103. (https://doi.org/10.1038/nature10868)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Qin Y, Wang JR, Wang Y, Iyer P, Cote GJ, Busaidy NL, Dadu R, Zafereo M, Williams MD & Ferrarotto R et al.2021 Clinical utility of circulating cell-free DNA mutations in anaplastic thyroid carcinoma. Thyroid 31 12351243. (https://doi.org/10.1089/thy.2020.0296)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Riedel C, Levy O & Carrasco N 2001 Post-transcriptional regulation of the sodium/iodide symporter by thyrotropin. Journal of Biological Chemistry 276 2145821463. (https://doi.org/10.1074/jbc.M100561200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rodriguez-Antona C, Pallares J, Montero-Conde C, Inglada-Perez L, Castelblanco E, Landa I, Leskela S, Leandro-Garcia LJ, Lopez-Jimenez E & Leton R et al.2010 Overexpression and activation of EGFR and VEGFR2 in medullary thyroid carcinomas is related to metastasis. Endocrine-Related Cancer 17 716. (https://doi.org/10.1677/ERC-08-0304)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rosenbaum-Krumme SJ, Freudenberg LS, Jentzen W, Bockisch A & Nagarajah J 2012 Effects of rosiglitazone on radioiodine negative and progressive differentiated thyroid carcinoma as assessed by (1)(2)(4)I PET/CT imaging. Clinical Nuclear Medicine 37 e47e52. (https://doi.org/10.1097/RLU.0b013e3182443ca6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roskoski Jr R 2019 Targeting ERK1/2 protein-serine/threonine kinases in human cancers. Pharmacological Research 142 151168. (https://doi.org/10.1016/j.phrs.2019.01.039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rothenberg SM, Mcfadden DG, Palmer EL, Daniels GH & Wirth LJ 2015 Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clinical Cancer Research 21 10281035. (https://doi.org/10.1158/1078-0432.CCR-14-2915)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Samatar AA & Poulikakos PI 2014 Targeting RAS-ERK signalling in cancer: promises and challenges. Nature Reviews: Drug Discovery 13 928942. (https://doi.org/10.1038/nrd4281)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, Habra MA, Newbold K, Shah MH & Hoff AO et al.2015 Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. New England Journal of Medicine 372 621630. (https://doi.org/10.1056/NEJMoa1406470)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schmutzler C, Schmitt TL, Glaser F, Loos U & Kohrle J 2002 The promoter of the human sodium/iodide-symporter gene responds to retinoic acid. Molecular and Cellular Endocrinology 189 145155. (https://doi.org/10.1016/s0303-7207(0100735-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Semrad TJ, Kim EJ, Tanaka MS, Sands J, Roberts C, Burich RA, Li Y, Gandara DR, Lara Jr P & Mack PC 2017 Phase II study of dovitinib in patients progressing on anti-vascular endothelial growth factor therapy. Cancer Treatment and Research Communications 10 2126. (https://doi.org/10.1016/j.ctarc.2016.12.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Seok SH 2021 Structural insights into protein regulation by phosphorylation and substrate recognition of protein kinases/phosphatases. Life 11 957. (https://doi.org/10.3390/life11090957)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sherman EJ, Su YB, Lyall A, Schoder H, Fury MG, Ghossein RA, Haque S, Lisa D, Shaha AR & Tuttle RM et al.2013 Evaluation of romidepsin for clinical activity and radioactive iodine reuptake in radioactive iodine-refractory thyroid carcinoma. Thyroid 23 593599. (https://doi.org/10.1089/thy.2012.0393)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Siegel RL, Miller KD, Fuchs HE & Jemal A 2021 Cancer statistics, 2021. CA: A Cancer Journal for Clinicians 71 733. (https://doi.org/10.3322/caac.21654)

  • Sipos JA & Ringel MD 2022 Molecular testing in thyroid cancer diagnosis and management. Best Practice and Research: Clinical Endocrinology and Metabolism 101680. (https://doi.org/10.1016/j.beem.2022.101680)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Smith VE, Read ML, Turnell AS, Watkins RJ, Watkinson JC, Lewy GD, Fong JC, James SR, Eggo MC & Boelaert K et al.2009 A novel mechanism of sodium iodide symporter repression in differentiated thyroid cancer. Journal of Cell Science 122 33933402. (https://doi.org/10.1242/jcs.045427)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Smith VE, Sharma N, Watkins RJ, Read ML, Ryan GA, Kwan PP, Martin A, Watkinson JC, Boelaert K & Franklyn JA et al.2013 Manipulation of PBF/PTTG1IP phosphorylation status; a potential new therapeutic strategy for improving radioiodine uptake in thyroid and other tumors. Journal of Clinical Endocrinology and Metabolism 98 28762886. (https://doi.org/10.1210/jc.2012-3640)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sobrinho-Simoes M, Eloy C, Magalhaes J, Lobo C & Amaro T 2011 Follicular thyroid carcinoma. Modern Pathology 24 (Supplement 2) S10S18. (https://doi.org/10.1038/modpathol.2010.133)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Song YS & Park YJ 2019 Genomic characterization of differentiated thyroid carcinoma. Endocrinology and Metabolism 34 110. (https://doi.org/10.3803/EnM.2019.34.1.1)

  • Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J & Frederick DT et al.2012 Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487 500504. (https://doi.org/10.1038/nature11183)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subbiah V, Gainor JF, Rahal R, Brubaker JD, Kim JL, Maynard M, Hu W, Cao Q, Sheets MP & Wilson D et al.2018a Precision targeted therapy with BLU-667 for RET-driven cancers. Cancer Discovery 8 836849. (https://doi.org/10.1158/2159-8290.CD-18-0338)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski C, Cabanillas ME & Urbanowitz G et al.2018b Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid cancer. Journal of Clinical Oncology 36 713. (https://doi.org/10.1200/JCO.2017.73.6785)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subbiah V, Velcheti V, Tuch BB, Ebata K, Busaidy NL, Cabanillas ME, Wirth LJ, Stock S, Smith S & Lauriault V et al.2018c Selective RET kinase inhibition for patients with RET-altered cancers. Annals of Oncology 29 18691876. (https://doi.org/10.1093/annonc/mdy137)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subbiah V, Yang D, Velcheti V, Drilon A & Meric-Bernstam F 2020 State-of-the-art strategies for targeting RET-dependent cancers. Journal of Clinical Oncology 38 12091221. (https://doi.org/10.1200/JCO.19.02551)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski CC, Cabanillas ME & Boran A et al.2022 Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: updated analysis from the phase II ROAR basket study. Annals of Oncology 33 406415. (https://doi.org/10.1016/j.annonc.2021.12.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tabernero J, Grothey A, Van Cutsem E, Yaeger R, Wasan H, Yoshino T, Desai J, Ciardiello F, Loupakis F & Hong YS et al.2021 Encorafenib plus cetuximab as a new standard of care for previously treated BRAF V600E-mutant metastatic colorectal cancer: updated survival results and subgroup analyses from the Beacon study. Journal of Clinical Oncology 39 273284. (https://doi.org/10.1200/JCO.20.02088)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tang F, Yang Z, Tan Y & Li Y 2020 Super-enhancer function and its application in cancer targeted therapy. NPJ Precision Oncology 4 2. (https://doi.org/10.1038/s41698-020-0108-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tavares C, Coelho MJ, Eloy C, Melo M, Da Rocha AG, Pestana A, Batista R, Ferreira LB, Rios E & Selmi-Ruby S et al.2018 NIS expression in thyroid tumors, relation with prognosis clinicopathological and molecular features. Endocrine Connections 7 7890. (https://doi.org/10.1530/EC-17-0302)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tubita A, Tusa I & Rovida E 2021 Playing the whack-A-mole game: ERK5 activation emerges among the resistance mechanisms to RAF-MEK1/2-ERK1/2-targeted therapy. Frontiers in Cell and Developmental Biology 9 647311. (https://doi.org/10.3389/fcell.2021.647311)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ullmann TM, Thiesmeyer JW, Lee YJ, Beg S, Mosquera JM, Elemento O, Fahey TJ 3rd, Scognamiglio T & Houvras Y 2022 RET fusion-positive papillary thyroid cancers are associated with a more aggressive phenotype. Annals of Surgical Oncology 29 42664273. (https://doi.org/10.1245/s10434-022-11418-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Valenciaga A, Saji M, Yu L, Zhang X, Bumrah C, Yilmaz AS, Knippler CM, Miles W, Giordano TJ & Cote GJ et al.2018 Transcriptional targeting of oncogene addiction in medullary thyroid cancer. JCI Insight 3 e122225. (https://doi.org/10.1172/jci.insight.122225)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Volante M, Lam AK, Papotti M & Tallini G 2021 Molecular pathology of poorly differentiated and anaplastic thyroid cancer: what do pathologists need to know? Endocrine Pathology 32 6376. (https://doi.org/10.1007/s12022-021-09665-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vuong HG, Odate T, Ngo HTT, Pham TQ, Tran TTK, Mochizuki K, Nakazawa T, Katoh R & Kondo T 2018 Clinical significance of RET and RAS mutations in sporadic medullary thyroid carcinoma: a meta-analysis. Endocrine-Related Cancer 25 633641. (https://doi.org/10.1530/ERC-18-0056)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wagenaar TR, Ma L, Roscoe B, Park SM, Bolon DN & Green MR 2014 Resistance to vemurafenib resulting from a novel mutation in the BRAFV600E kinase domain. Pigment Cell and Melanoma Research 27 124133. (https://doi.org/10.1111/pcmr.12171)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wagle N, Van Allen EM, Treacy DJ, Frederick DT, Cooper ZA, Taylor-Weiner A, Rosenberg M, Goetz EM, Sullivan RJ & Farlow DN et al.2014 MAP kinase pathway alterations in BRAF-mutant melanoma patients with acquired resistance to combined RAF/MEK inhibition. Cancer Discovery 4 6168. (https://doi.org/10.1158/2159-8290.CD-13-0631)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Jones CM, Marshall CJ, Springer CJ & Barford D et al.2004 Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 116 855867. (https://doi.org/10.1016/s0092-8674(0400215-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang JR, Zafereo ME, Dadu R, Ferrarotto R, Busaidy NL, Lu C, Ahmed S, Gule-Monroe MK, Williams MD & Sturgis EM et al.2019a Complete surgical resection following neoadjuvant dabrafenib plus trametinib in BRAF(V600E)-mutated anaplastic thyroid carcinoma. Thyroid 29 10361043. (https://doi.org/10.1089/thy.2019.0133)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang Z, Dai J, Yan J, Zhang Y & Yin Z 2019b Targeting EZH2 as a novel therapeutic strategy for sorafenib-resistant thyroid carcinoma. Journal of Cellular and Molecular Medicine 23 47704778. (https://doi.org/10.1111/jcmm.14365)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Watson IR, Li L, Cabeceiras PK, Mahdavi M, Gutschner T, Genovese G, Wang G, Fang Z, Tepper JM & Stemke-Hale K et al.2014 The RAC1 P29S hotspot mutation in melanoma confers resistance to pharmacological inhibition of RAF. Cancer Research 74 48454852. (https://doi.org/10.1158/0008-5472.CAN-14-1232-T)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Weisner J, Landel I, Reintjes C, Uhlenbrock N, Trajkovic-Arsic M, Dienstbier N, Hardick J, Ladigan S, Lindemann M & Smith S et al.2019 Preclinical efficacy of covalent-allosteric AKT inhibitor borussertib in combination with trametinib in KRAS-mutant pancreatic and colorectal cancer. Cancer Research 79 23672378. (https://doi.org/10.1158/0008-5472.CAN-18-2861)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wells Jr SA, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, Baudin E, Elisei R, Jarzab B & Vasselli JR et al.2012 Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. Journal of Clinical Oncology 30 134141. (https://doi.org/10.1200/JCO.2011.35.5040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y & Sosman J et al.2012 Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 487 505509. (https://doi.org/10.1038/nature11249)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, Worden F, Brose M, Patel J & Leboulleux S et al.2020 Efficacy of selpercatinib in RET-altered thyroid cancers. New England Journal of Medicine 383 825835. (https://doi.org/10.1056/NEJMoa2005651)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Woyach JA, Kloos RT, Ringel MD, Arbogast D, Collamore M, Zwiebel JA, Grever M, Villalona-Calero M & Shah MH 2009 Lack of therapeutic effect of the histone deacetylase inhibitor vorinostat in patients with metastatic radioiodine-refractory thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 94 164170. (https://doi.org/10.1210/jc.2008-1631)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xing M 2005 BRAF mutation in thyroid cancer. Endocrine Related Cancer 12 245262. (https://doi.org/10.1677/erc.1.0978)

  • Xing M, Westra WH, Tufano RP, Cohen Y, Rosenbaum E, Rhoden KJ, Carson KA, Vasko V, Larin A & Tallini G et al.2005 BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. Journal of Clinical Endocrinology and Metabolism 90 63736379. (https://doi.org/10.1210/jc.2005-0987)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yadav V, Zhang X, Liu J, Estrem S, Li S, Gong XQ, Buchanan S, Henry JR, Starling JJ & Peng SB 2012 Reactivation of mitogen-activated protein kinase (MAPK) pathway by FGF receptor 3 (FGFR3)/Ras mediates resistance to vemurafenib in human B-RAF V600E mutant melanoma. Journal of Biological Chemistry 287 2808728098. (https://doi.org/10.1074/jbc.M112.377218)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhao Z, Xie L & Bourne PE 2017 Insights into the binding mode of MEK type-III inhibitors. A step towards discovering and designing allosteric kinase inhibitors across the human kinome. PLoS ONE 12 e0179936. (https://doi.org/10.1371/journal.pone.0179936)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu YJ, Zheng B, Wang HY & Chen L 2017 New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacologica Sinica 38 614622. (https://doi.org/10.1038/aps.2017.5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Zhu X, Park S, Lee WK & Cheng SY 2019 Potentiated anti-tumor effects of BETi by MEKi in anaplastic thyroid cancer. Endocrine-Related Cancer 26 739750. (https://doi.org/10.1530/ERC-19-0107)

    • PubMed
    • Search Google Scholar
    • Export Citation