Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jan 3.
Published in final edited form as: Nat Rev Cancer. 2023 Jul 12;23(9):631–650. doi: 10.1038/s41568-023-00598-y

Pathogenesis of cancers derived from thyroid follicular cells

James A Fagin 1,, Gnana P Krishnamoorthy 1, Iñigo Landa 1
PMCID: PMC10763075  NIHMSID: NIHMS1954703  PMID: 37438605

Abstract

The genomic simplicity of differentiated cancers derived from thyroid follicular cells offers unique insights into how oncogenic drivers impact tumour phenotype. Essentially, the main oncoproteins in thyroid cancer activate nodes in the receptor tyrosine kinase–RAS–BRAF pathway, which constitutively induces MAPK signalling to varying degrees consistent with their specific biochemical mechanisms of action. The magnitude of the flux through the MAPK signalling pathway determines key elements of thyroid cancer biology, including differentiation state, invasive properties and the cellular composition of the tumour microenvironment. Progression of disease results from genomic lesions that drive immortalization, disrupt chromatin accessibility and cause cell cycle checkpoint dysfunction, in conjunction with a tumour microenvironment characterized by progressive immunosuppression. This Review charts the genomic trajectories of these common endocrine tumours, while connecting them to the biological states that they confer.

Introduction

Cancers derived from thyroid follicular cells are classified into five main types: papillary thyroid carcinoma (PTC) (representing 65–93% of all thyroid cancers worldwide)1, follicular thyroid carcinoma (FTC) (6–10%)24, oncocytic thyroid carcinoma (OC) (3–7%)5, poorly differentiated thyroid carcinoma (PDTC) (0.5–2%)6 and anaplastic thyroid carcinoma (ATC) (1%)7. PTC, FTC and OC are generically termed differentiated thyroid carcinomas (DTCs), most patients with which present with localized disease and have a 5-year survival rate of >98% (ref. 8), by contrast to PDTC, which has a 5-year survival rate of 76% (ref. 9). The 2022 WHO classification of thyroid tumours introduced a new intermediate clinical entity — differentiated high-grade thyroid carcinoma — to define DTCs with a high mitotic rate and/or tumour necrosis, as these have a 5-year survival rate comparable with that of PDTC9,10. ATC is an extremely aggressive form of the disease. Until recently, patients with ATC had a dismal median overall survival of 4 months, although this has improved markedly since 2018 with the FDA approval of new oncoprotein-targeted treatments coupled to evidence that immunotherapies may confer additional benefit1113. Thyroid parafollicular or C cells are a neuroendocrine lineage that gives rise to medullary thyroid cancers, which account for <5% of all thyroid cancers. This is a distinct entity that has been recently reviewed elsewhere14,15 and will not be discussed further in this Review.

The incidence of thyroid cancer in the USA was approximately 14.6 per 100,000 in 2022, with an estimated total of 43,800 cases8. Furthermore, there had been a steep rise in thyroid cancer incidence in the past three decades mostly owing to overdiagnosis of small PTCs, part of a large subclinical reservoir of tumours that would have otherwise mostly not caused disease or mortality16. However, incidence rates since 2013 have stabilized and may have even begun to fall8, probably owing to greater awareness of the dangers of overdiagnosis, as well as a recent reassessment by pathologists of the histological features that merit a diagnosis of thyroid cancer1719. Active surveillance of patients with PTCs measuring <1 cm, termed papillary microcarcinomas, shows that most do not grow after prolonged monitoring, particularly in older subjects20,21. The genomic and transcriptomic characterization of papillary microcarcinomas has so far not identified distinct differences with larger tumours that progress or metastasize to regional lymph nodes22. Whether the trend towards quiescence of many PTCs is due to cell autonomous events, such as oncogene-induced senescence23,24, or to host effects, is unknown.

Constitutively active mutant effectors in the MAPK pathway are the major drivers of thyroid cancers across the entire spectrum of the disease with the notable exception of OCs (Box 1). Since the topic of thyroid cancer pathogenesis was last reviewed in depth7,25,26, there has been an explosion of new information on the genomics of all forms of sporadic2 as well as radiation-induced thyroid cancer. Comprehensive next-generation sequencing studies of PDTC and ATC performed in the past decade show that these aggressive tumours arise from DTCs through the acquisition of mutations that drive tumour immortalization, activate the PI3K pathway, disable key cell cycle checkpoints and/or disrupt chromatin2730. This is supported by evidence that genomic lesions that are clonal in advanced disease can be found in rare subsets of DTCs as subclonal events28,31. How each of these ‘disease progression’ pathways alter tumour biology and, in some cases, therapeutic dependencies are a major focus of this Review.

Box 1. Mitochondrial defects and near haploidization of the nuclear genome in oncocytic carcinomas.

Until 2018, oncocytic carcinomas (OCs) of the thyroid, formerly known as Hürthle cell neoplasms, were considered to be variants of follicular adenoma or carcinoma. However, they are now recognized as a distinct entity that accounts for about 3–7% of all malignancies arising from thyroid follicular cells5. They are characterized histologically by a follicular-patterned architecture of oxyphil cells containing abundant mitochondria, a large nucleus and a prominent nucleolus271, whereas the presence of capsular invasion or vascular invasion distinguishes advanced (widely invasive OC) from indolent (minimally invasive OC) tumours272. Through genomic analysis, it was revealed that OCs are a distinct entity, characterized by mitochondrial DNA mutations that disrupt the electron transport chain, particularly in genes encoding complex I subunits273,274. By contrast to the mitochondrial DNA mutations observed in other cancer types, which are usually present in only a subset of the mitochondria, and are thus termed heteroplasmic mutations275, those in OC tend to be present in virtually all mitochondria — that is, they are homoplasmic — indicating that they must confer OC cells with a fitness advantage276,277. These mutations render cells defective in mitochondrial respiration and reliant on aerobic glycolysis, a vulnerability that could be exploited therapeutically in the future278. Aside from the mitochondrial defects, comprehensive whole-exome studies of OCs identified global loss of heterozygosity (LOH), either through near-haploid genomes (depicted as chromosomes with blue shading in the figure) or by genome duplication with uniparental disomy (UPD), which spares only chromosomes 5, 7 and 12 (refs. 276,277) (see the figure). The functional implications of the global LOH are not fully understood. However, a recent integrative analysis of genomic, transcriptomic and metabolomic data on OCs showed that global LOH or UPD may account for the presence of an immunosuppressive tumour microenvironment, primarily via depletion of T helper cells279. Recurrent mutations in genes such as NRAS, death domain-associated protein 6 (DAXX), cyclin-dependent kinase inhibitor 1A (CDKN1A), NF1, TP53 and eukaryotic translation initiation factor 1A X isoform (EIF1AX) and the telomerase reverse transcriptase (TERT) promoter are also relatively frequent events in OCs276,277. ATP, ATP synthase protein; CO, cytochrome c oxidase subunit; CYB, cytochrome B; MT-TL1, mitochondrially encoded tRNA leucine 1; ND, NADH dehydrogenase subunit.

Box 1

Thyroid cells are highly specialized, with their primary function being the biosynthesis and secretion of the thyroid hormones. This exquisitely regulated process is crucial for organismal homeostasis and, in essence, endows thyroid cells with the capacity to actively transport and incorporate iodide into thyroid hormones32. As a consequence, radioactive iodine (RAI) therapy is the most prescribed treatment for thyroid cancer, and its efficacy is contingent on the cancer cells retaining some ability to metabolize iodine. The genetic drivers of thyroid cancer impinge on this process in distinct ways, which is elaborated on in this Review. Although we do not address the recent therapeutic advances in detail, we discuss key therapeutic vulnerabilities and how thyroid cancer cells adapt to the selective inhibition of the common truncal drivers of the disease, as well as present new information on the transcriptional and compositional heterogeneity of these tumours, which is central to understand the complex interactions that drive thyroid cancer pathogenesis.

Genetic predisposition to thyroid cancer

Although more than 90% of thyroid cancers are sporadic33, genome-wide association studies of large cohorts identified low penetrance genetic variants associated with non-medullary thyroid cancers, most with moderate effect sizes34,35. Among all cancer types, those of the thyroid have among the highest genetic risk, which in Icelandic and European populations extends beyond the nuclear family36. Genetic variants on chromosomes 9q22.33 and 14q13.3 show the highest association in Europeans34,37 and were also linked with thyroid cancer in Korean populations38. The gene encoding the thyroid-specific transcription factor forkhead box E1 (FOXE1) is positioned close to the variants on 9q22.33. Single nucleotide polymorphisms (SNPs) in and around the FOXE1 gene were also strongly associated with thyroid cancer in Spanish and Italian populations37, as well as in patients with radiation-induced thyroid cancer39. Notably, a risk variant within the FOXE15′-untranslated region impacts FOXE1 transcription by recruiting a transcriptional complex that is distinct from that which binds to the wild-type allele37. Moreover, NK2 homeobox 1 (NKX2-1), which also encodes a thyroid lineage transcription factor, is within a gene block lying in proximity to the other main susceptibility locus on 14q13.3. The 9q22.33 and 14q13.3 SNPs are also associated with low serum levels of thyroid-stimulating hormone (TSH; also known as thyrotropin)34. Hence, these thyroid cancer susceptibility alleles may impact the TSH signalling axis in thyroid cells through perturbations in the regulatory control of key lineage transcription factors.

The strongest association with thyroid cancer in Korean populations has been identified with variants of the neuregulin-1 (NRG1) gene on 8p12 (ref. 38), also identified in Europeans35. Consistent with this observation, NRG1 expression is increased in thyroid cancer tissues harbouring the risk variant alleles38. NRG1 is the ligand for human epidermal growth factor receptor 3 (HER3; also known as ERBB3), a member of the epidermal growth factor family of receptor tyrosine kinases (RTKs), which, as discussed later in this Review, has a central role in adaptive resistance of thyroid cancers to MAPK pathway inhibitors40,41.

Predisposition to familial non-medullary thyroid carcinoma is common, with up to 9% of patients having a first-degree relative with the disease33. Infrequently, they may arise as components of recognized cancer predisposition syndromes conferred by pathogenic germline mutations of PTEN, adenomatous polyposis coli (APC), DICER1, which encodes an endoribonuclease, PRKAR1A, which encodes the regulatory R1α subunit of the cAMP-dependent protein kinase A (PKA) or WRN, which encodes the Werner syndrome helicase (Table 1) (reviewed elsewhere42,43). More commonly, the familial susceptibility occurs as an isolated event only impacting risk of thyroid cancer. However, owing to the high frequency of microcarcinomas in the general population, co-occurrence within families may be a fortuitous event rather than one determined by genetic predisposition. Numerous candidate low penetrance susceptibility variants have been identified in families with familial non-medullary thyroid carcinoma33,42,44. This information is not yet clinically applicable for screening purposes because most variants have not been fully validated and the segregation of variant alleles within families is confounded by the low penetrance of the disease42.

Table 1 |.

Molecular and clinical features of familial syndromes causing follicular cell-derived thyroid neoplasms

Familial syndrome Genes Inheritance Thyroid phenotype penetrance Thyroid phenotype distribution Specific PTC subtype Other phenotypes
PTEN hamartoma tumour syndrome PTEN Autosomal dominant ~80% MNG/FA: ~50%
PTC: ~30%
FTC: ~20%
ATC: ~1%
Both classic and FV-PTC Mucocutaneous lesions, genital pigmentation, breast cancer, endometrial carcinoma, macrocephaly, autism spectrum disorder
Familial adenomatous polyposis APC Autosomal dominant ~16% PTC: 100% Exclusively cribriform Colorectal adenomatous polyps, colorectal cancer, desmoid tumours, hepatoblastoma, medulloblastoma
Carney complex PRKAR1A Autosomal dominant ~60% MNG/FA: ~60%
PTC: ~15%
FTC: ~25%
Predominantly FV-PTC Pigmentation in skin and mucosa, multiple myxomas, primary pigmented nodular adrenocortical disease, Sertoli cell tumours, growth hormone-secreting pituitary adenoma, breast ductal adenoma, osteochondromyxoma
Werner’s syndrome WRN Autosomal recessive ~18% PTC: ~40%
FTC: ~50%
ATC: ~10%
Predominantly FV-PTC Premature greying and/or thinning of scalp hair, atherosclerosis, type 2 diabetes, cataracts, short stature, melanoma, meningioma, soft-tissue sarcomas, leukaemia, osteosarcomas
DICER1 syndrome DICER1 Autosomal dominant ~45% MNG/FA: ~90%
PTC: ~5%
FTC: ~5%
Both classic and FV-PTC Pleuropulmonary blastoma, pulmonary cysts, cystic nephroma, ovarian tumours, ciliary body medullo-epithelioma, nasal chondro-mesenchymal hamartoma, embryonal rhabdo-myosarcoma, pituitary blastoma, pineoblastoma, central nervous system sarcoma, presacral malignant teratoid tumour42,43

Distributions of pathogenic mutations in all syndromes are typically loss-of-function events consistent with the tumour suppressor roles of the affected gene products. Thyroid lesions in patients with DICER1 syndrome typically display a germline loss of function in the DICER1 gene and a somatic second-hit missense mutation affecting the RNase IIIb domain of the protein. ATC, anaplastic thyroid carcinoma; FA, follicular adenoma; FTC, follicular thyroid carcinoma; FV-PTC, follicular variant of papillary thyroid carcinoma; MNG, multinodular goitre; PTC, papillary thyroid carcinoma.

Radiation-induced thyroid cancer

Exposure to ionizing radiation during childhood increases the risk of developing PTC later in life, as demonstrated by the strong dose–response relationship between radiation dose to the thyroid and thyroid cancer incidence in individuals exposed to radioiodine isotopes during childhood following the Chernobyl nuclear reactor accident45. Whole-genome sequencing of a large series of post-Chernobyl thyroid cancers showed strong radiation dose-dependent increases in clonal small deletions and simple and balanced structural variants bearing genomic hallmarks of DNA repair by non-homologous end joining. By contrast, there was no impact of radiation on the frequency of single nucleotide substitutions46. Consistent with this, there was a radiation dose-dependent increase in the frequency of fusions versus driver mutations46, primarily of RET, neurotrophic tyrosine kinase receptor 1 (NTRK1), NTRK3, BRAF and anaplastic lymphoma kinase (ALK), as previously reported in smaller cohorts of radiation-exposed patients4750. Indeed, RET fusions can be induced by exposure of human thyroid cells to ionizing radiation in vitro51. The propensity for radiation to induce RET rearrangements may be facilitated by the close spatial proximity of the chromosomal regions involved in the recombination events during interphase in normal human thyroid cells52. Contrary to previous reports53,54, there is no evidence for a transcriptomic signature of radiation-induced thyroid cancer. Gene expression and epigenomic changes show strong associations with the tumour driver but not with radiation-dose exposure46.

Cell of origin of thyroid cancers

Two major competing hypotheses have been advanced to define the subpopulation of cells that can give rise to the different histological types of thyroid cancer. A multistep carcinogenesis model suggests that undifferentiated thyroid carcinomas derive from well-differentiated tumours via the sequential accumulation of genetic mutations55. Alternatively, thyroid cancer cells are proposed to originate from transformation of fetal thyroid cells at various stages of differentiation, with ATC, PTC and FTC arising from thyroid stem cells, thyroblasts and pro-thyrocytes, respectively56. A recent study investigated the transformation efficiency of thyroid cancer oncogenes introduced by CRISPR–Cas9 editing into either thyroid progenitor cells (TPCs) or mature thyrocytes derived from human embryonic stem cells and found that oncoproteins transformed TPCs with much greater efficiency than mature thyrocytes57. Although these data suggest that thyroid progenitors may be more susceptible to transformation, they do not negate the need for sequential genetic events for disease progression, as ATCs developed only in the TPCs when TP53 was co-mutated with either BRAF or RAS57.

Drivers of differentiated thyroid cancers

PTC and FTC are prototypical cancers driven by oncoproteins that signal in part through the canonical MAPK pathway. Mutations of genes encoding proteins in this pathway are almost invariably clonal and drive the majority of DTCs, including 89% of PTCs58,59. However, a subset of tumours harbour oncoproteins that induce tumorigenesis through distinct mechanisms. Although they are comparatively infrequent, they provide unique insights into the biology of thyroid cancer and are also discussed.

Thyroid tumorigenesis by mutant effectors of the MAPK pathway

The Cancer Genome Atlas (TCGA) study of PTC showed that 59% of the 496 samples harboured activating BRAF mutations, almost all resulting in the V600E substitution58. Consistent with previous reports60,61, these were mutually exclusive with RAS mutations, which were present in 13% of tumours (NRASHRAS > KRAS) and with the 16% of cases harbouring fusion oncogenes, primarily of the RTKs RET, NTRK3, NTRK1 and ALK58. Generically, these recombinant fusion oncoproteins consist of N-terminal domains contributed by the upstream gene partner, coupled to the kinase domain of the respective RTK62. The N-terminal fragments usually contain coiled-coil motifs that drive dimerization and autophosphorylation of tyrosine residues in the kinase domain, leading to constitutive downstream signalling. BRAF was also activated through gene fusions in 2.3% of tumours, some of which were confirmed to selectively overexpress the BRAF kinase domain58. RTK fusions are particularly prevalent in sporadic paediatric PTCs63.

PTCs have a low overall frequency of non-synonymous somatic mutations (0.41 per Mb) when compared with other cancer types64,65. Moreover, >70% of PTCs lack copy number gains or losses58,66. The overall genomic simplicity of PTCs allows for remarkably clear associations between driver mutations and gene expression profiles, signalling pathway transcriptional outputs, proteomic changes, differentiation states and histological characteristics. Although both RAS mutants and BRAF-V600E activate the MAPK pathway, they do so to different degrees. Mutant RAS constitutively signals through RAF dimers, which are subject to negative feedback by ERK, resulting in an attenuation of the flux through the pathway. By contrast, BRAF-V600E signals as a monomer and is partially unresponsive to negative feedback by ERK, resulting in a higher MAPK signalling pathway output67. Consistent with this, BRAF-mutant PTCs are transcriptionally distinct from their RAS-mutant counterparts58. This prompted the creation of a transcriptional score to distinguish them from each other – the BRAF–RAS score (BRS), which is a 71-gene signature that quantifies the extent to which a particular tumour transcriptionally resembles either a BRAFV600E-mutant or a RAS-mutant tumour58. There is a highly significant correlation between the BRS and a thyroid differentiation score (TDS), which is a set of 16 genes that encode proteins that regulate iodine metabolism: that is, RAS-like tumours have a higher TDS than BRAFV600E-like tumours58 (Fig. 1). Furthermore, the application of the BRS and the TDS to thyroid cancers driven by other oncoproteins can be instructive, as tumours with a high TDS tend to respond to RAI therapy68. Genetically engineered mouse models of Braf V600E-induced thyroid cancer closely phenocopy the histological characteristics of human PTC, have a high MAPK pathway transcriptional output and are associated with a marked inhibition of differentiated thyroid function, manifesting as a decrease in iodine uptake and thyroid hormone biosynthesis6971. RAS-like PTCs mostly retain the structure of the thyroid follicle, a characteristic feature of the non-infiltrative follicular variant of PTC (FV-PTC), which tend to be encapsulated and rarely develop nodal metastases72. By contrast, BRAF V600E-like tumours have the classical or tall cell variant (TCV) histology, are frequently invasive and have a tropism to regional lymph nodes58.

Fig. 1 |. Characteristics of BRAF-like and RAS-like differentiated thyroid cancers.

Fig. 1 |

Differentiated thyroid carcinomas can be categorized transcriptomically as either BRAF-like or RAS-like on the basis of their similarity to papillary thyroid carcinomas (PTCs) harbouring either BRAFV600E or RAS mutations, respectively. The BRAFV600E mutation occurs in 62% of PTCs, particularly in the classical and tall cell variants of the disease. By contrast NRAS, HRAS and KRAS mutations are frequent events in follicular thyroid carcinoma (FTC) (45%) and in the follicular variant of PTC29,58,94,152,267269. The characteristic histological feature of BRAF-like tumours is the presence of papillae, consisting of epithelial cells arranged around a fibrovascular core, whereas RAS-like tumours tend to retain the structure of the thyroid follicle. Mutant BRAF and RAS activate MAPK signalling but do so to different degrees (the asterisk in the figure represents the mutational activation of RAS). The MAPK signalling flux induced by mutant RAS is dampened by negative feedback of activated ERK on CRAF, which disrupts RAF dimer signalling. BRAF-V600E signals as a monomer and is therefore unresponsive to the negative feedback by activated ERK on CRAF and hence has a higher MAPK pathway output. The expression of thyroid lineage transcription factors and of genes required for iodide uptake and metabolism is inhibited by MAPK signalling. The former is quantified by the thyroid differentiation score (TDS), which is lower in BRAF-like than in RAS-like tumours. Expression of the thyroid transcription factor paired box 8 (PAX8) and cadherin 16 (CDH16), which control correct apical–basal polarization of thyroid cells, is lower in BRAF-like than in RAS-like differentiated thyroid carcinomas. The BRAF-V600E mutation also induces expression of matrix metalloproteinases, rendering BRAF-mutant tumours more invasive than their RAS-mutant counterparts. RTK, receptor tyrosine kinase. Histological images courtesy of Bin Xu and Ronald Ghossein.

The association of BRAF-V600E with tumour invasiveness may also be a consequence of its higher MAPK signalling flux, which leads to transcriptional activation of genes encoding proteins involved in extracellular matrix (ECM) remodelling, including matrix metalloproteinase 3 (MMP3), MMP9 and MMP13 (ref. 73). Similar relationships among BRAF-V600E, ERK signalling, invasiveness and expression of MMP1 have been shown in human melanomas74,75. It is also plausible that the degree of pathway flux may be a determining factor in cell transformation. Activation of high output MAPK signalling through endogenous expression of BrafV600E or overexpression of RET and NTRK1 fusions is sufficient to drive thyroid tumorigenesis in mice69,70,7678 and zebrafish79. By contrast, thyrocyte-specific endogenous expression of HrasG12V (ref. 80) or KrasG12D (ref. 81) in mice induces cell hyperplasia and requires cooperative events to induce transformation8084.

TCV-PTC is a distinct subtype of DTC that has a disease-specific survival rate of 82%, when compared with a population of stage-matched and treatment-matched classical PTC, which has a disease-specific survival rate of 98% (ref. 85). This subtype almost universally harbours the BRAF-V600E mutant, but by contrast to classical PTC, they have abundant mitochondria with a high frequency of homoplasmic or heteroplasmic mutations in mitochondrial DNA-encoded genes for oxidative phosphorylation (OXPHOS) complex I subunits, analogous to those present in OC86 (Box 1). How putative OXPHOS dysfunction may modify BRAF-V600E-induced PTC tumorigenesis is unknown.

Drivers in the TSH receptor–cAMP signalling pathway

Thyroid cells are under the regulatory control of TSH for growth and expression of genes required for thyroid hormone biosynthesis87. The TSH receptor (TSHR) is a G protein-coupled seven-transmembrane receptor, which upon binding to its ligand engages with multiple G protein subtypes88. Signalling through Gsα has a dominant role in mediating TSH action on cell growth and gene expression by activating adenylyl cyclase and the cAMP signalling pathway89. This is best exemplified by activating mutations of TSHR90,91 or GNAS92, the gene encoding Gsα, which gives rise to autonomously functioning thyroid nodules (AFTNs) that, when they reach a critical size, cause hyperthyroidism. Furthermore, these gain-of-function mutations in TSHR and GNAS, which activate adenylyl cyclase, are genetic hallmarks of AFTN, consistent with the stimulatory effects of cAMP on cell growth and thyroid hormone biosynthesis. AFTNs are usually benign and rarely progress to thyroid cancer93. However, activating TSHR mutations are present in 0.5% of PTCs58 and 10% of FTCs94. TSHR-mutant FTCs may cause hyperthyroidism, particularly those with a high metastatic burden95,96. Preliminary evidence from a small series of patients suggests that a high mutant TSHR allelic ratio may favour malignant transformation95. Interestingly, a recurrent activating mutation of the enhancer of zeste homologue 1 (EZH1) gene, which encodes a catalytic subunit of the chromatin remodelling polycomb repressor complex 2 (PRC2), is co-mutated with TSHR or GNAS in a subset of AFTNs97. Expression of the EZH1-G571R mutant promotes histone H3 lysine 27 (H3K27) trimethylation and induces cyclin D1 expression and ultimately thyroid cell growth. It is unclear, however, whether the co-mutation pattern is enriched in FTCs97.

Germline loss-of-function mutations of PRKAR1A confer predisposition to benign and malignant thyroid tumours, as well as tumours of other endocrine cell types as part of the Carney complex98 (Table 1). Loss of function of this regulatory protein results in decreased basal activity of PKA, but markedly increases cAMP-stimulated PKA levels98. Thyroid-specific homozygous deletion of Prkar1a in mice results in a high penetrance of FTC and hyperthyroidism99. Despite this, to our knowledge, no somatic mutations of PRKAR1A have been reported in PTC58 or FTC94 to date. Isolated cases of PDTC and ATC with somatic truncation mutations or deep deletions of PRKAR1A have been reported, but their role in transformation has not been firmly established100,101.

Interactions between the MAPK and cAMP signalling pathways in thyroid cancer

Constitutive MAPK pathway activation through RET fusions or BRAF-V600E disrupts the TSHR–adenylyl cyclase–PKA signalling axis at multiple nodes, primarily by decreasing the expression of TSHR and inhibiting the catalytic activity of adenylyl cyclase24,102 (Fig. 2). This is the simplest explanation for the significant inverse correlation between TDS and BRS in PTCs from the TCGA study58. The functional corollary of this reciprocal relationship is that it can be reversed by RAF and/or MEK inhibitors in cell lines103,104, Braf V600E-induced PTCs in mice31,71,105 and in subsets of patients with BRAFV600E or RAS-mutant thyroid cancers106108, leading to a rescue of expression of differentiated thyroid genes. This includes genes encoding the thyroid lineage transcription factors paired box 8 (PAX8) and forkhead box E1 (FOXE1) and the key genes they regulate, that is, sodium iodide symporter (NIS), dual oxidase 1 (DUOX1) and DUOX2 and thyroid peroxidase (TPO), the protein product of which is required for iodine oxidation and incorporation into tyrosine residues of thyroglobulin (TG). As this gene expression programme governs the ability of thyroid cancer cells to incorporate radioactive iodine, treatment of RET-mutant, NTRK-mutant, RAF-mutant and RAS-mutant thyroid cancers with selective MAPK pathway inhibitors has been shown to enhance clinical responses to iodine-131 in pilot phase II clinical trials and in index cases106112.

Fig. 2 |. Interactions between MAPK and cAMP signalling in thyroid cancer cells.

Fig. 2 |

Thyroid cells are under the regulatory control of thyroid-stimulating hormone (TSH), acting through the TSH receptor (TSHR) to signal primarily through Gsα and adenylyl cyclase to increase intracellular cAMP levels. cAMP in turn activates protein kinase A (PKA), which phosphorylates substrates to ultimately regulate the growth and differentiated function of thyroid cells in part through inducing expression of the lineage transcription factors paired box 8 (PAX8) and forkhead box E1 (FOXE1), which regulate the expression of genes required for thyroid hormone biosynthesis (for example, sodium iodide symporter (NIS), thyroglobulin (TG), thyroid peroxidase (TPO) and TSHR). Activation of the MAPK pathway, through receptor tyrosine kinase (RTK) fusions, or RAS or BRAF mutations, impairs TSH–cAMP signalling at various nodes in the pathway24,102,105,270. This takes place by repressing expression of TSHR and inhibiting the catalytic activity of adenylyl cyclase, thus impairing PAX8-mediated and FOXE1-mediated transcription and inhibiting differentiated function in thyroid cancer cells. The figure illustrates the functional consequences of the BRAF-V600E mutation, highlighted with an asterisk.

Other inputs may have important roles in mediating the effects of MAPK pathway activation on thyroid-specific functional properties. For instance, transforming growth factor β (TGFβ) also impairs TSH-induced expression of the thyroid differentiation genes Tg and Nis in rat thyroid cell lines113,114. Oncogenic BRAF indirectly leads to the engagement of TGFβ or activin signalling115, driven in part through activation of a TGFβ autocrine loop114. This results in phosphorylation of SMAD, which binds to PAX8 and impairs its transactivation of Nis, and presumably of other PAX8-regulated genes required for thyroid hormone biosynthesis116. This circuit is reversed by treatment with potent MEK inhibitors, whereas TGFβ and/or activin receptor antagonists are insufficient to rescue thyroid differentiation in BrafV600E-driven PTCs in mice115, indicating that the impairment of differentiated thyroid function by constitutive MAPK signalling is in part independent of SMAD activation.

Genomic fusions of nuclear receptors and lineage transcription factors

Approximately 1% of PTCs and 8–30% of FTCs are driven by PAX8– peroxisome proliferator-activated receptor – (PPARG) fusions94,117, which consist of the coding region of all but the extreme C terminus of the thyroid lineage transcription factor PAX8, fused to the entire coding sequence of PPARG, a nuclear receptor that among other functions governs the gene expression programme of adipogenesis118. Targeted expression of the PAX8PPARG fusion to thyroid cells of transgenic mice induces thyroid cell hyperplasia, whereas in the context of homozygous Pten loss, the PAX8–PPARG fusion drives locally invasive thyroid cancers with lung metastases119.

The PAX8 and PPARG components of the fusion retain their respective DNA-binding domains and appear to activate distinct transcriptional programmes. PAX8–PPARG chromatin immunoprecipitation-sequencing of mouse PAX8PPARGThy;PtenThy−/− cancers showed that binding sites were enriched for motifs of both PAX8 and PPARG, commonly in close vicinity to transcription start sites120. The PPARG component of PAX8–PPARG likely accounts for the regulation of genes involved in lipid metabolism, cell cycle and motility. Interestingly, treatment of tumour-bearing PAX8PPARGThy;PtenThy−/− mice with the PPARG agonist pioglitazone induced transdifferentiation of cancer cells into adipocytes and a profound reduction in tumour size119. Accordingly, a patient with PAX8–PPARG-driven metastatic thyroid cancer had an impressive response to this PPARG agonist121. Dysregulated expression of PPARG has also been described in a radiation-induced PTC and a sporadic FTC harbouring cAMP-responsive element-binding protein 3-like protein 2 (CREB3L2)–PPARG fusions49,122, suggesting that overexpression of this nuclear receptor may have a key role in driving tumorigenesis in this subset of tumours.

The PAX8 component of PAX8–PPARG likely accounts for its binding to motifs adjacent to genes regulating the cellular response to hypoxia and cytoskeletal structure120. In this regard, PAX8 has a critical role in the determination of thyroid cell polarity and assembly of the thyroid follicle, acting primarily through regulation of cadherin-16 (CDH16)123, which encodes an adhesion protein that is uniquely expressed in thyroid and kidney cells – both lineages in which PAX8 has an essential role in development124. Consistent with this, PAX8–PPARG-driven PTCs retain the follicular architecture of the normal thyroid gland125. Furthermore, the expression of PAX8 and CDH16 is higher in FV-PTCs, which retain follicular structures, than in classical and TCV-PTCs, which do not58, suggesting that this pathway may be an important determinant of the distinctive histological architecture of the major variants of DTCs (Fig. 1).

Genomic dysregulation of mRNA translation

A subset of PTCs from the TCGA study were found to have recurrent mutations of the X chromosome copy of the gene eukaryotic translation initiation factor 1A (EIF1AX), a component of the 43S translation preinitiation complex58. They have also been described in benign thyroid adenomas and FTC126,127. In PTC, they are frequently the sole tumour driver, whereas in PDTC and ATC, EIF1AX mutations are almost invariably associated with RAS mutations28,84,127. EIF1AX mutations in thyroid cancer primarily affect the N-terminal domain of the encoded protein, as well as a splice site within the C terminus28. The EIF1AX-A113 splice mutation is found almost exclusively in thyroid cancer and is enriched in more advanced disease28. EIF1A mutants promote preferential translation of activating transcription factor 4 (ATF4), which orchestrates adaptive responses to cellular stress. ATF4 in turn induces the expression of the GADD34 phosphatase (also known as protein phosphatase 1 regulatory subunit 15A), which dephosphorylates eIF2A and leads to a global increase in protein synthesis (Fig. 3). ATF4 also induces MYC, which together promotes the expression of amino acid transporters, thus increasing amino acid supply and driving activation of mTOR84. Mutant RAS, which as mentioned is co-mutated with EIF1AX in advanced thyroid cancers, stabilizes the MYC protein, likely explaining the vulnerability of RAS and EIF1AX co-mutated cancers to bromodomain-containing protein 4 (BRD4) inhibitors, which target MYC transcription84. Interestingly, EIF1AX mutations in uveal melanomas, in which they were originally described, show mutual exclusivity with MYC amplifications, consistent with a common requirement for transformation128.

Fig. 3 |. Mechanisms of mutant eIF1A and RAS cooperation in thyroid tumorigenesis.

Fig. 3 |

Regulation of translation determines the levels of proteins synthesized by their corresponding mRNAs. Once the small 40S ribosomal subunit binds to the 5′-untranslated region of mRNAs, the preinitiation complex (PIC) containing the initiator methionyl tRNAi (tRNA1Met) initiates the scanning process for the AUG initiation codon. The eukaryotic translation initiation factor 1A (eIF1A) is a component of the PIC that is mutated in thyroid cancer (indicated by an asterisk). eIF1A mutants have a higher affinity for other subunits of the PIC, leading to its stabilization and to the illegitimate translation of activating transcription factor 4 (ATF4). ATF4 then induces dephosphorylation of the translation initiation factor eIF2A (which loads the tRNA1Met onto the 40S small ribosomal subunit). This takes place via a GADD34-mediated negative feedback pathway, leading to enhanced ternary complex loading to form the PIC and to an increase in protein synthesis. RAS mutants (indicated by an asterisk) in turn stabilize MYC, an effect augmented by the EIF1AX-A113 splice mutant, the most common somatic variant in thyroid cancer. ATF4 and MYC induce expression of amino acid (AA) transporters and cooperate to activate mTOR complex 1 (mTORC1), further augmenting global protein synthesis84. ASCT2, alanine/serine/cysteine/threonine transporter 2; EAA, essential amino acid; LAT1, l-type amino acid transporter 1; RTK, receptor tyrosine kinase; ternary complex: a complex of GTP-bound eIF2A and the tRNA1Met.

Thyroid cancer progression

The role of immortalization in thyroid cancer progression

The genetic drivers of DTCs described earlier likely have a role in tumour initiation, as they are clonal in early-stage DTCs and remain truncal during the evolution of the disease58,59,129. An important transitional step in disease progression includes genomic events that promote tumour cell immortalization, in particular the highly prevalent genetic alterations of telomerase reverse transcriptase (TERT)130. The telomerase complex is active during embryogenesis, but is silenced postnatally in somatic cells131. Continued cell replication requires a mechanism to repair critically shortened telomeres to protect chromosomes from undergoing end-to-end fusions, which in turn would cause telomere crisis and apoptosis132. Reactivation of telomerase is the most common strategy preventing this from happening in cancer cells133. Recurrent hotspot mutations of the TERT promoter were first described in patients with melanoma134,135, and later in numerous other cancer types, including those of the thyroid136139. The base substitutions in the TERT proximal gene promoter generate de novo consensus binding sites for the E-twenty-six (ETS) family of transcription factors, which have a substantial role in mediating the transcriptional output of the MAPK signalling pathway135. The ETS family members, GA-binding protein α (GABPA) and GABPB, transactivate the mutant TERT promoter and have been proposed to have a selective role in inducing TERT mRNA in thyroid cancers with coexisting BRAF and TERT promoter mutations140. However, GABP mRNA levels in PTC are low compared with other members of this family, and it may be that multiple ETS family members, including GABP, contribute to the transcriptional regulation of TERT141143. Hypermethylation of a CpG island in the distal TERT gene promoter region has been implicated as a mechanism of increased TERT expression in cancer, which is in contrast to the canonical role of CpG island hypermethylation as a repressor of gene transcription144. A recent analysis of 23 cancer lineages, including thyroid, found that hypomethylation at a CpG region flanking the TERT transcription start site is also strongly associated with active transcription and bound by the histone activation marks acetylated H3K9 (H3K19ac) and H3K14ac145,146.

Recurrent TERT promoter mutations, aberrant methylation and possible copy number gains are associated with worse clinical outcomes in patients with DTC130,147. TERT promoter mutations are subclonal in DTC with a prevalence of 9% and become clonal in PDTC and ATC, where they are found in 40% and 73% of cases, respectively, consistent with a key role in tumour microevolution28. The combination of BRAFV600E and TERT has been highlighted as being particularly deleterious, although TERT defects are also enriched in PDTCs and ATCs harbouring RAS or RTK fusion drivers28,29,148. Other components of the telomerase complex, such as the telomerase RNA component, the non-coding RNA template for telomere replication, are also expressed at higher levels in advanced disease130, consistent with the assumption that the catalytic activity of TERT is primarily involved in telomere elongation. Whether one of the proposed non-canonical effects of TERT149 is also involved in disease progression remains to be demonstrated. There is no evidence that alternate mechanisms of telomere elongation have a major role in thyroid cancer, or that mutations that can favour this process, such as those of the α-thalassemia/mental retardation syndrome X-linked (ATRX)–death domain-associated protein 6 complex (DAXX) complex and the histone variant H3.3, are prevalent in this disease.

Genetic events associated with progression to PDTCs and ATCs

There have, to date, been no large-scale, TCGA-like efforts to investigate the integrated genomic and transcriptomic landscapes of human PDTC and ATC. Despite this, a picture of the major categories of genetic lesions associated with progression has emerged on the basis of smaller whole-genome, whole-exome or cancer-exome series2730,150152.

PDTCs are characterized by a solid (sheets of neoplastic epithelial cells), trabecular (strands (‘beams’) of neoplastic epithelium separated by connective tissue) or insular (tightly packed islands or nests of neoplastic epithelial cells) histological growth pattern, a high mitotic rate and areas of necrosis and vascular invasion. They metastasize at a distance in 40–70% of the cases, and by contrast they infrequently invade regional lymph nodes when compared with classical or TCV-PTCs153. RAS mutations are the primary oncogenic driver, present in 30–45% of the cases, with RET and ALK fusions accounting for an additional 15% (refs. 28,154). Although the precise sequence of genetic events is unclear, TERT promoter mutations likely enable their more aggressive phenotype, presumably by allowing the accumulation of additional genetic lesions. High-grade BRAF-mutant PTC with a high mitotic rate and areas of necrosis have a comparable disease-specific mortality to RAS-mutant PDTC9. They also have a high frequency of TERT genetic defects and acquire mutations in the same category of genes found in RAS-mutant PDTC, but retain histological features of PTC, tend to be locally invasive and have a tropism for regional lymph nodes.

ATCs are the most aggressive forms of thyroid cancer. Although there has been meaningful progress in treatments for patients with ATC, the 5-year survival of patients with ATC presenting with metastatic disease is less than 10% (ref. 8). This subtype are rapidly growing, locally invasive tumours that frequently have lymph node and distant metastases at presentation or that develop during the course of the disease. The histology is pleomorphic (mixtures of neoplastic epithelial and myoepithelial/stromal components in various patterns) with cells of sarcomatoid, giant cell and epithelial appearance present at varying degrees. ATCs also have a high mitotic rate and areas of necrosis10. Similar to DTCs, they have a high frequency of MAPK pathway driver alterations28. Loss-of-function mutations of TP53 are a hallmark of the disease, such that ATC can be recapitulated with high penetrance in genetically engineered mouse models with thyroid targeted deletions of Trp53 combined with either activating mutations of Braf and Hras or expression of RET fusions83,155157. The major pathways involved in disease progression are discussed subsequently.

The PI3K pathway in advanced thyroid cancer

Germline mutations of PTEN, which encodes a phosphatase for phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3), and less frequently of PIK3CA (which encodes the PI3K catalytic subunit, p110α) and AKT1, can predispose affected individuals to develop thyroid neoplasms158,159. However, somatic mutations of these genes are not commonly involved in sporadic thyroid cancer initiation58,94. Thyrocyte-specific expression of PIK3CAH1074R is insufficient to induce thyroid cell transformation in mice160. Furthermore, homozygous loss of Pten increases cell proliferation and leads to goitre and development of benign thyroid adenomas in mice161. Interestingly, induction of TSH by treatment with methimazole and sodium perchlorate (which increase TSH secretion by decreasing thyroid hormone synthesis) failed to stimulate further growth of Pten null thyrocytes in vivo161, suggesting that the proliferative effects of TSH may be mediated in part through activation of the PI3K–AKT pathway, possibly through autocrine stimulation by insulin-like growth factor 1 (IGF1)162,163.

Nevertheless, mutations of effectors in the PI3K–AKT–mTOR pathway are more prevalent in advanced thyroid cancers (ATC > PDTC > PTC)28,164,165. Loss-of-function PTEN mutations tend to co-occur with RAS or NF1 mutations in PDTC and ATC, whereas PIK3CA mutations are more commonly seen in concert with BRAFV600E. Although these combinatorial relationships are not absolute, their role in thyroid cell transformation is supported by experiments in genetically engineered mouse models. Thyroid-specific expressions of KrasG12D or Pten loss alone are insufficient to induce thyroid cancer, but their combined effects lead to metastatic FTCs and decreased survival166. By contrast, the combination of BrafV600E with either Pten loss or expression of PIK3CAH1074R induces a more severe phenotype, including early transition from PTC to ATC160. BRAF-mutant and PIK3CA-mutant human ATC cell lines show enhanced sensitivity to the combination of MEK and pan-class 1 PI3K inhibitors compared with the respective mono-therapies, indicative of novel dependencies developing during tumour progression160. However, combined blockade of the MAPK and PI3K pathways with selective inhibitors in patients with cancer causes substantial toxicities167,168. The constitutive activation of MAPK and PI3K pathways converges to inhibit the heterotrimeric tuberous sclerosis complex (TSC), the GTPase activating protein that lies upstream of the small GTPase RHEB, leading to mTOR complex 1 (mTORC1) activation, a key node controlling cell growth through its effects on protein, lipid and nucleotide biosynthesis169. A small subset of ATCs harbour inactivating mutations of STK11 (also known as LKB1)28, a serine/threonine kinase. When STK11 is inactivated, it fails to phosphorylate AMP-activated protein kinase α, leading to inactivation of TSC2 and de-repression of mTORC1 (ref. 170). Therefore, mTORC1 inhibition with rapamycin analogues may be particularly beneficial for some patients with tumours harbouring mutant MTOR, RHEB, TSC1 or TSC2 (refs. 171,172), but this may not be the case for those that harbour oncoproteins that lie upstream of TSC173.

Mice harbouring a germline inactivating mutation of the ubiquitously expressed nuclear receptor thyroid hormone receptor β (TRβ) (known as the TRβPV/PV mouse) develop metastatic FTC174. Loss-of-function mutations of TRβ result in a state of thyroid hormone resistance in humans175 and in mice. Although mutations of nuclear thyroid hormone receptors do not have a role in human thyroid cancer pathogenesis, this is one of the only mouse models in which a single engineered mutant gene causes metastatic thyroid cancer, and as such it has attracted considerable interest, particularly as a vehicle to investigate the role of the PI3K pathway in promoting tumour metastases. TRβPV/PV mice develop unrestrained elevated levels of TSH because of resistance to the negative feedback of thyroid hormone on pituitary thyrotrophs, which is required but not sufficient for thyroid cell transformation176. The unbound wild-type TRβ was previously shown to stimulate AKT–mTOR signalling by binding to the p85 regulatory subunit of PI3K in fibroblasts177. This non-canonical effect may account in part for the transforming effects of mutant TRβ in thyroid cells, as it was found to have higher binding affinity to p85 than wild-type TRβ, leading to constitutive phosphorylation of AKT and a therapeutic vulnerability to pan-PI3K inhibitors178,179. Germline deletion of Akt1 attenuates both tumour growth and metastatic spread in TRβPV/PV mice, whereas Akt3 and to a lesser extent Akt2 knockouts have a more selective inhibitory effect on the development of lung metastases180,181. A caveat is that the biological consequences of expression of mutant TRβ and deletion of Akt isoforms cannot be attributed with certainty to cell autonomous effects in thyroid cells as they are mutated in the germline in these aforementioned genetically engineered mouse models and therefore impact numerous cell lineages.

Loss of cell cycle and DNA damage response checkpoints

Mutations of TP53 are the genetic hallmark of ATC. They are extraordinarily rare in PTC (0.7%)58, where they tend to be subclonal28, uncommon in PDTC (9%) but present in 63–73% of ATCs28,29,182. TP53 mutant ATC foci have been seen to arise adjacent to PTCs that are wild-type for this gene, further documenting its importance in tumour microevolution129. A pan-cancer study identified TP53 mutations as the genomic event that is most strongly associated with copy number changes183. Potential explanations for this include loss of the regulatory role of p53 in maintaining the integrity of the spindle assembly checkpoint, which when disrupted leads to an increased rate of tetraploidy and chromosome mis-segregation184. Thus, TP53 loss promotes aneuploidy because it facilitates the generation of chromosome abnormalities and prevents the culling of cells, which have undergone abnormal mitoses185.

Genomic instability caused by Trp53 loss in mice does not follow a random, chaotic process. Instead, it causes a series of evolutionary changes that begin with deletion events, which are later followed by copy number gains and amplifications186. Although the sequence of events after TP53 loss in ATC is not known, these tumours are commonly aneuploid and harbour recurrent specific copy number losses and gains, some of which predict for worse disease outcomes, such as gain of chromosome 1q and 20q, or loss of 13q28,187. In addition, through the loss of the critical role of p53 in the response to DNA damage, where it helps mediate DNA repair or removal of damaged cells by apoptosis, TP53 mutations also likely favour the accumulation of oncogenic mutations. p53 is also critical in constraining the activation of the MAPK pathway by BRAF or RAS oncoproteins, by triggering cell cycle arrest and senescence if the pathway output exceeds a critical threshold188190. Removal of this constraint by TP53 loss in ATCs is associated with a greater MAPK pathway transcriptional output155,156. In addition, lack of p53 has been shown to activate the MAPK pathway in ‘RAS-less’ mouse embryonic fibroblasts, presumably through induction of RAF kinase activity191. Regardless of the underlying mechanism, potent activation of MAPK signalling by enforced overexpression of BRAFV600E in thyroid cells in vivo has important phenotypic consequences, such as driving the production of inflammatory cytokines by tumour cells, which in turn promotes the recruitment of macrophages to the tumour microenvironment (TME)192. Accordingly, ATCs are characterized by their heavy immune infiltrate, primarily consisting of tumour-associated macrophages that form a dense interconnected network that is in close contact with the cancer cells, and likely has a critical role in their biology193,194.

In an analysis of a cancer-exome sequencing panel of 196 ATCs, Pozdeyev et al.29 categorized this tumour subtype into four main mutational clusters. Cluster 2 was characterized by loss-of-function alterations of cyclin-dependent kinase inhibitor 2A (CDKN2A) and CDKN2B, which were primarily deep deletions, as well as driver mutations associated with either PTC or FTC development, and cluster 4 by mutations of RB1. Hence, besides the defects of TP53, which impact the integrity of the DNA damage checkpoint, loss of function of genes that lead to impairment of the G1-to-S phase transition is a common feature of ATC182,195.

Chromatin remodelling gene defects in advanced thyroid cancers

A forward genetic screen using Sleeping Beauty (SB) transposon mutagenesis to introduce insertional disruptions randomly into the genome of HrasG12V mutant mouse thyroid follicular cells identified genes that cooperate with RAS to induce PDTC, which primarily clustered in genes associated with chromatin remodelling functional nodes and the PI3K pathway80. Consistent with this, mutations of genes encoding subunits of the SWI/SNF chromatin remodelling complexes, the histone lysine methyltransferases KMT2A, KMT2C and KMT2D and the histone acetyltransferases CREB-binding protein (CBP) and P300 are enriched in PDTCs and ATCs compared with PTCs28,29,196. SWI/SNF complexes are preferentially targeted to enhancers distal to the transcription start sites of genes, many of which are linked to developmental processes and lineage specification197. Recurrent mutations in many of the subunits of the SWI/SNF complexes occur in ~20% of all human cancers across multiple tumour lineages198,199. SWI/SNF complexes are antagonistic with the activity of PRC2. Therefore, loss of SWI/SNF complex function can promote oncogenesis via derepression of PRC2 and upregulation of a stem cell-associated programme200. Thyroid-specific homozygous loss of the SWI/SNF complex subunit genes AT-rich interactive domain-containing 1A (Arid1α), Arid2 or SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (Smarcb1) in mouse BrafV600E-driven tumours promotes disease progression to PDTC and ATC. Loss of function of each of these SWI/SNF complex subunit genes leads to distinct and more compact chromatin states and loss of thyroid differentiation. Although treatment of mouse Braf V600E-mutant PTC with RAF or MEK inhibitors can partially restore differentiation, this is prevented by coexisting SWI/SNF complex subunit mutations, rendering them insensitive to redifferentiation therapies aimed to restore radioiodine responsiveness. This was also noted in a small series of patients in clinical trials of redifferentiation therapy, in which metastatic DTC or PDTC tumours with SMARCB1, ARID1A or ARID2 mutations failed to regain RAI avidity in response to RAF or MEK inhibitors31 (Fig. 4). However, these patient studies were based on a small number of cases and hence the findings will need to be confirmed in larger trials. The functional consequences of other epigenetic lesions, including those of the KMT2 gene family, have not been investigated so far in thyroid cancer.

Fig. 4 |. Epigenetic reprogramming in thyroid cancer progression and differentiation.

Fig. 4 |

Loss-of-function recurrent mutations of chromatin-modifying genes, including those encoding subunits of the SWI/SNF complexes, are enriched in advanced thyroid cancers28. Left: BRAFV600E-mutant metastatic thyroid cancer cells have decreased expression of the lineage transcription factors forkhead box E1 (FOXE1) and paired box 8 (PAX8), and impaired accessibility to their respective DNA-binding sites required to induce expression of thyroid differentiation genes. RAF and/or MEK inhibitors (RAFi and MEKi, respectively) restore expression of PAX8 and FOXE1, which in concert with SWI/SNF complexes promote an open chromatin state enabling expression of iodine metabolism genes and restoration of response to radioactive iodine (RAI) therapy. Right: BRAF-mutant tumours with coexisting SWI/SNF complex subunit mutations fail to remodel chromatin and enable thyroid transcription factor accessibility to DNA to regulate expression of thyroid differentiation genes following treatment with MAPK pathway inhibitors, rendering metastases refractory to RAI. NIS, sodium iodide symporter; TDS, thyroid differentiation score; TG, thyroglobulin; TPO, thyroid peroxidase; TSHR, thyroid-stimulating hormone receptor.

Heterogeneity of thyroid cancers

DTCs can be clonally heterogeneous, whereby mutations co-occurring with a primary driver can lead to the emergence of a dominant subclone that outcompetes other cells201. High depth genomic sequencing studies show that many of the mutations leading to disease progression are subclonal in PTCs, including mutations in TERT, TP53 and those of genes encoding subunits of SWI/SNF complexes, and become clonal in PDTCs or ATCs28,31,196. Clonal diversity predicts an increased risk of subsequent disease progression201. Cancer cells can also diverge from each other through development of distinct transcriptional states, which are not necessarily governed by genomic events. A single-cell RNA-sequencing (scRNA-seq) study of 11 tissue samples from patients with PTC at different stages of disease progression defined three transcriptional states of malignant thyrocytes along the course of disease progression202: follicular-like, partial epithelial-to-mesenchymal (EMT)-like and dedifferentiation-like. The follicular-like state represents normal and premalignant thyrocytes that have retained a high degree of thyroid differentiation (high TDS) and are RAS-like. Partial EMT-like cells, which derived from primary tumours and lymph node metastases, exhibited partial upregulation of transcriptional drivers of EMT, induction of genes encoding proteins related to the ECM, downregulation of thyroid-specific epithelial genes and preserved expression of other epithelial markers. Finally, cells in a dedifferentiated-like state derived from samples with progressive disease. They expressed very low levels of thyroid-specific epithelial markers, had high levels of SOX4, GATA2 and MYC mRNAs and were enriched for oncogenic and immune-related signalling pathways. A separate scRNA-seq study explored the evolutionary trajectory during progression from PTC to ATC and in the process identified a subset of PTC cells with gain of transcriptional signatures of EMT, mTORC1 activation and negative regulation of p53 that represented an intermediate state between PTC and ATC203. The drivers of this transcriptional microevolution in thyroid cancer are unknown. Although key genetic subclones could contribute to transcriptional heterogeneity, it can also be driven by alternate mechanisms, including distinct metabolic and epigenetic states, as well as interactions with other cell types in the TME204.

The immune tumour microenvironment

The TME is a complex ecosystem composed of tumour cells, immune cells, fibroblasts, endothelial cells and non-cellular components, primarily consisting of ECM and soluble mediators (cytokines, chemokines and growth factors)205. A detailed review of the impact of the immune TME on thyroid cancer biology is beyond the scope of this Review. However, it should be noted that the dynamic interplay between tumour and immune cells has a crucial role in tumour initiation and progression in several cancer types205,206, including those of the thyroid207, in which cancer cells generate an immunosuppressive and tolerogenic microenvironment. The immune landscapes of DTCs, PDTCs and ATCs are very distinct, although there is limited information on their functional implications208. CD4+ T helper cells are enriched in low-grade DTC208, whereas an abundance of regulatory T (Treg) cells correlates with recurrent disease and the presence of nodal metastases209,210. Consistent with this, CD8+ cytotoxic T lymphocytes (CTLs) are enriched in those DTCs with favourable prognostic features, whereas CD8+, granzyme B T cells, a marker of T cell exhaustion, are associated with tumour recurrence209,211. T cells in advanced DTCs also show activation of immune checkpoints, including the PD1–PDL1 axis, cytotoxic T lymphocyte-associated antigen 4 (CTLA4), T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) and T cell immunoglobulin mucin receptor 3 (TIM3)212214. Moreover, ATCs are heavily infiltrated with immune cells, including higher levels of Treg cells, CD4+ T cells, CD8+ T cells and natural killer (NK) cells compared with DTCs, with many of these cells expressing markers of dysfunction203. Overall, the evolution of the immune landscape from PTC to ATC is marked by progressive immunosuppression203. However, caution is needed in the interpretation of the studies reporting these findings, as most were based on limited numbers of tissue samples or on RNA-based analyses.

Myeloid cell infiltration likely has an important role in driving the immunosuppressive TME. Following conditional activation of BRAFV600E in mouse thyroid glands, there is an increased expression of the myeloid cell chemoattractants macrophage colony-stimulating factor 1 (CSF1) and CC-chemokine ligand 2 (CCL2). This is followed by the development of PTCs that are densely infiltrated with tumour-associated macrophages expressing CSF1 receptor (CSF1R) and CC-chemokine receptor 2 (CCR2), the respective receptors for these cytokines. Targeting CCR2-expressing cells during mutant BRAF induction reduced macrophage density and impaired PTC development192. A switch from pro-inflammatory M1-like macrophages in PTCs to pro-tumorigenic M2-like macrophages in ATCs suggests that the latter may have a particularly prominent role in thyroid cancer progression193,203,215. BRAF-V600E-expressing PTCs may also drive recruitment of myeloid-derived suppressor cells (MDSCs) through activation of the developmental transcription factor TBX3 in the cancer cells and induction of CXC-chemokine receptor 2 (CXCR2) ligands216,217. MDSCs are also enriched in ATCs218 and can dampen antitumour immunity by expressing immune checkpoint molecules, depleting amino acids required for T cell responses, and by producing deleterious nitric oxide and reactive oxygen species219. MDSCs can also favour tumour progression via non-immunological mechanisms, such as supporting angiogenesis, promoting tumour stemness and aiding the formation of the pre-metastatic niche220. Consistent with the latter, circulating levels of MDSCs are increased in patients with ATC221.

The production of interferon γ is largely restricted to T lymphocytes and NK cells, and its downstream signalling induces expression of major histocompatibility complex I (MHCI) and MHCII in immune and non-immune cells222, which is required to initiate adaptive immune responses223. Tumour antigens are taken up by dendritic cells and cross-presented to prime CD8+ T cells. In a second step, antigens are presented by tumour cells to the primed CD8+ T cells, leading to tumour cell killing224. Tumours invoke multiple mechanisms to evade immune recognition via both these steps, for instance, by modulating antigen expression or disrupting the antigen processing and presentation pathway. In this regard, MHCI expression is decreased by MAPK activation in cancer cells225, thus repressing a central component in the activation of adaptive immune responses to tumour cells. This is also true for MHCII expression in human BRAF-mutant PTC cells226. Moreover, RAF kinase inhibitors restored MHCII expression and CD4+ T cell infiltration in mouse BRAF-mutant PTCs and sensitized tumours to treatment with an anti-PD1 antibody226.

Genomic events associated with metastatic thyroid cancer

As is the case for most cancer types, mortality in thyroid cancer is primarily due to metastatic disease. The metastatic process arises from subsets of cancer cells endowed with features that allow them to undergo intravasation, survive in the circulation, extravasate and successfully home at a distant site, where they may remain dormant for extended periods227,228. Although many of the biological requirements for cancer cells to successfully undergo this process have been identified227,228, how these apply to different lineages and how oncogenic drivers affect them is poorly understood. The properties that confer thyroid cancer cells with an ability to metastasize are mostly unknown, as are the signals that allow them to home at different sites, exit dormancy and develop clinical metastatic disease. The MSK-MET pan-cancer genomic profiling of 25,000 patients, which included a representative number of patients with PTC and PDTC, identified tumour cell lineage-specific genetic mutations associated with metastatic disease229. PTC metastases were more chromosomally unstable than primary tumours, as determined by the fraction of the genome altered, had a higher tumour mutation burden and more frequent TERT promoter mutations. In addition to these features, CDKN2A deletions and mutations of the RNA splicing factor RNA-binding 10 (RBM10) were more common in PTC with a high metastatic burden. Consistent with this, RBM10 loss in thyroid cancer has been shown to alter the ratio of cassette exon inclusion events in a subset of transcripts that encode proteins that regulate interactions between the ECM and the cytoskeleton, leading to RAC activation, which in turn favours increased cell movement and metastatic competence230. In terms of metastatic tropism, BRAFV600E-mutant tumours are less likely to metastasize to bone, whereas those with chromosome 22q loss, which is commonly associated with RAS mutations, more frequently metastasize to this site229. Chromosome 22q loss of heterozygosity (LOH) is present in 14% of PTCs, mostly in FV-PTC, consistent with the association with RAS mutations58. Candidate cancer genes on 22q include checkpoint kinase 2 (CHEK2), NF2 and SMARCB1. In the TCGA study of PTCs, CHEK2 mutations were significantly associated with chromosome 22q LOH58. Arm-level chromosome 22q LOH is enriched in PDTC and ATC, in which it is found in 35% and 25% of tumours, respectively28.

Therapeutic strategies for thyroid cancer

Radioiodine therapy

RAI is the most commonly prescribed treatment for thyroid cancer. The rationale for this approach is predicated on its relative selectivity for thyroid cells, as incorporation of iodide into cells is mediated through the sodium iodide symporter, the expression of which is restricted to very few sites other than the thyroid32. It is used in the adjuvant setting and to treat metastatic disease231. However, there are currently no data from prospective randomized clinical trials supporting the efficacy of RAI therapy. Despite this, it has been used empirically for several decades, particularly following surgery for thyroid cancers presenting with extensive lymph node metastases, on the basis of data from prospective observational trials and retrospective registry studies232,233. However, this approach needs to be interpreted in the context of the genetics of DTC, most of which are driven by BRAFV600E, which have a high MAPK pathway transcriptional output and a low TDS, which in most cases predicts for refractoriness to RAI. This has been demonstrated in mouse models71,105 and in patient series234. Consistent with this, a recent study of patients with metastatic thyroid cancer who had major responses to RAI showed that these responses were universally associated with RAS-like tumours, whose MAPK pathway output is attenuated by negative feedback and which have a high TDS68. By contrast, RAI-refractory cancers in this study were enriched for BRAFV600E, chromosome 1q amplification and concomitant mutations of genes encoding PI3K pathway effectors and RNA splicing factors68. Several phase II, open-label clinical trials have demonstrated that a short course of RAF and/or MEK inhibitors can restore RAI avidity and response to RAI therapy in approximately 30% of patients with RAI-refractory metastatic thyroid cancer driven by mutant BRAF or RAS or RTK fusions106112. The long-term impact of these redifferentiation therapies on progression-free survival and overall survival has not been established.

Angiogenesis inhibitors

First-line systemic therapies for metastatic DTC, PDTC and OC are the multikinase inhibitors lenvatinib and sorafenib235,236. The efficacy of these drugs is mostly due to their antiangiogenic activity through inhibition of the kinase activity of vascular endothelial growth factor receptors (VEGFRs) and other endothelial cell receptors237,238, rather than through inhibitory effects on mutant oncoproteins235,239,240. Angiogenesis has an important role in tumour initiation, progression and metastasis241, and yet inhibitors of this process are only effective in a limited number of tumour types242. Their efficacy is believed to be due in part to restricting the supply of oxygen and nutrients to the tumour. The neovasculature can also secrete angiocrine factors that regulate tumour growth243. Which of these mechanisms drive the remarkable responses of DTC to lenvatinib, in particular, is unknown235. Notably, normal thyroid follicular cells are closely juxtaposed to an endothelial capillary network, which is required for thyroid follicle formation through production of exosomes244,245, conceivably generating a dependency that is maintained during tumorigenesis.

Therapeutic targeting of tumour drivers

As discussed throughout this Review, the drivers of recurrent and metastatic RAI-refractory thyroid cancer are primarily oncogenic kinases and small GTPases. Tumours are dependent on these truncal driver mutations for viability as genetic or pharmacological inhibition of their activity has been shown to suppress cancer cell growth in preclinical models. Furthermore, many of them can be targeted effectively with selective small molecule inhibitors71,83,155,246. Highly selective RET and NTRK inhibitors induce durable responses in patients with metastatic thyroid cancers harbouring the respective fusions and are FDA-approved for this indication247,248. On the basis of studies in RET and NTRK fusion-driven non-small-cell lung cancers, the primary mechanisms of acquired resistance are due to on-target solvent front mutations in the fusion kinase domains249,250. Furthermore, adaptive resistance to RET kinase inhibitors through off-target fibroblast growth factor receptor activation has been shown in a zebrafish model of RET fusion-driven thyroid cancer79, and it is possible that analogous mechanisms may also operate in human tumours. Although considerable progress has been made with small molecule targeting of specific KRAS mutants251 and of HRAS252, this is of marginal relevance to RAI-refractory thyroid cancers as there are as yet no drugs specifically targeting NRASQ61R, the most prevalent small GTPase mutation in this disease.

Lineage-specific adaptation of BRAF-mutant cancers to MAPK pathway inhibition

BRAF-V600E mutations are highly prevalent in melanomas and thyroid cancers and are also present in a subset of colorectal cancers60,253. On the basis of the premise that the viability of all three lineages would be dependent on this tumour driver, there was an initial expectation that all of these tumour types might respond to an equivalent degree to RAF and/or MEK kinase inhibitors. Much to the contrary, responses differ markedly between these tumour types, with thyroid and in particular colorectal cancers showing lower overall response rates compared with melanomas254256. This is likely because RAF and MEK inhibitors relieve distinct negative feedback inputs upstream of BRAF-V600E in these cancer cell types, leading to reactivation of MAPK signalling. In colorectal cancer cells, activation of the epidermal growth factor receptor (EGFR) ensues after MAPK pathway inhibitor treatment, whereas in thyroid cancer cells, the resistance to the RAF inhibitor vemurafenib is primarily due to activation of NRG1–HER3 or HER2 (also known as ERBB2) signalling through relief of negative feedback40. The clinical relevance of this adaptive resistance mechanism in thyroid cancer is supported by data from a phase I trial showing a 69% response rate to the combination of the RAF kinase inhibitor dabrafenib and the HER kinase inhibitor lapatinib in patients with BRAF-mutant metastatic DTC257. It is intriguing to consider whether germline thyroid cancer predisposition alleles mapping close to the NRG1 gene locus and associated with higher NRG1 expression may underpin this lineage-specific response38. A recent study showed that dysregulated activation of Yes-associated protein (YAP), a transcriptional co-activator that executes the growth-promoting effects of HIPPO pathway inhibition, governs the expression of genes in the NRG1–HER3 or HER2 pathway and amplifies resistance to MAPK pathway blockade in BRAF-mutant thyroid cancers and melanomas41 (Fig. 5).

Fig. 5 |. Adaptive responses of BRAF-mutant thyroid cancers to RAF kinase inhibitors.

Fig. 5 |

Left: BRAF-V600E hyperactivates the MAPK pathway because it constitutively signals as a monomer and is unresponsive to negative feedback by ERK on RAF dimers. In this context, expression of the receptor tyrosine kinases (RTKs) human epidermal growth factor receptor 2 (HER2) and HER3 is attenuated and RAS-GTP levels are low. Right: RAF kinase inhibitors suppress MEK and ERK phosphorylation and induce Yes-associated protein (YAP) nuclear translocation and activation. The mechanism by which RAF kinase inhibitors promote YAP nuclear translocation is unknown. Under this therapeutic pressure, YAP activates the transcription of genes encoding neuregulin-1 (NRG1), HER2, HER3 and RAS. NRG1 is the ligand for HER3, and upon its activation of the HER3/HER2 heterodimer, downstream signalling via Src homology region 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2), growth factor receptor-bound protein 2 (GRB2) and SOS induces RAS activation, resulting in adaptive resistance to RAF kinase inhibition.

Therapy for ATC

Surgery, when feasible, followed by chemoradiotherapy is the current standard of care for these rapidly growing inflammatory cancers258. The prognosis of patients with ATC with distant metastases (stage IVc) is poor. Approximately 45% of ATCs have BRAFV600E mutations28,29, and fortunately, BRAF-mutant ATCs show significant responses to the RAF kinase inhibitor vemurafenib259. As thyroid cancers develop strong adaptive resistance to RAF kinase inhibition through relief of negative feedback40, a combination of RAF and MEK kinase inhibitors is a logical choice to potently inhibit the MAPK pathway. An open-label phase II study of patients with BRAFV600E-mutant ATC treated with a combination of dabrafenib and the MEK inhibitor trametinib showed a remarkable overall response rate of 69% (refs. 12,260). By contrast, the overall response rate of BRAFV600E-driven DTC to the same combination was only 30% (ref. 261). This was unexpected as ATCs have a more aggressive biological behaviour. In addition, compared with DTCs, ATCs are heavily infiltrated with macrophages and MDSCs193,194 and enriched for NK cells and CD4+ and CD8+ T cells, although these latter cell types express markers of exhaustion203. How MAPK pathway inhibitors modulate this complex cellular network to drive robust antitumour responses is a subject of active investigation. However, there is encouraging evidence that relief of T cell suppression with the PD1 inhibitor spartalizumab induces clinical responses in patients with ATC expressing high levels of PDL1 (ref. 262).

Conclusions

As outlined in this Review, the main genomic defects involved in the evolution of thyroid cancers have now been largely identified. The oncoproteins that drive tumour initiation are mostly either oncogenic kinases or small GTPases, each of which is associated with distinct phenotypic features that are recapitulated quite faithfully in genetically engineered mouse models. Despite their apparent genomic simplicity, DTC cells within a tumour exhibit distinct transcriptional states. Whether these are stable or transitional and whether they result from cell autonomous events or arise in response to cues from the TME is unknown. Comprehensive integrated genomic, epigenomic and transcriptomic analyses of PDTC and ATC are still lacking, as most information has been derived from relatively small series studies.

Current understanding of the genomic characteristics of the different types of thyroid cancer has yielded major improvements in our ability to diagnose the disease with a high degree of accuracy in fine needle biopsies of thyroid nodules, resulting in a meaningful decrease in unnecessary surgeries263,264. The evidence that constitutive activation of MAPK signalling suppresses differentiated thyroid function has clinical implications, as small molecule MAPK pathway inhibitors sensitize a subset of patients with metastatic thyroid cancer to RAI therapy106112. Although therapeutic targeting of BRAF or RTK-mutant ATCs is effective in treating the disease110,260, patients ultimately develop resistance265,266. Advanced thyroid cancers are complex and heterogeneous. Further progress will require a deeper understanding of their biology. Single-cell and spatial integrated ‘omic’ approaches in patient samples and preclinical models may help devise new mechanism-based approaches to activate the antitumour immune response and target specific cancer cell states or other elements of the TME.

Acknowledgements

J.A.F. acknowledges funding from the NIH, Cycle for Survival and from the Frank Cohen, Jaymie and Peter Flowers and Salvatore Ranieri funds. I.L. acknowledges funding from the NIH. The authors are also grateful to B. Xu and R. Ghossein for providing histological images for Fig. 1.

Glossary

Adaptive resistance

MAPK pathway oncoproteins activate downstream signalling networks, which in turn elicit negative regulatory events designed to dampen pathway output. Upon targeted inhibition of the oncoprotein with small molecules, the network adapts by relieving this negative feedback, causing intrinsic resistance to their action

Adipogenesis

Differentiation process through which mesenchymal cells commit to preadipocytes, and these in turn differentiate into adipocytes

Angiocrine factors

Growth factors, trophogens or membrane-bound paracrine factors supplied by endothelial cells to regulate neighbouring cell growth and homeostasis

Capsular invasion

Invasion of tumour through the entire capsule distinguishes malignant from benign follicular-patterned thyroid neoplasms

Cassette exon

Intervening exon between two other exons from the mature mRNA sequence that can be either included or skipped to generate two distinct protein isoforms

Goitre

Abnormally enlarged thyroid gland

Hyperthyroidism

Syndrome associated with excessive production of thyroid hormones by thyroid cells

Non-homologous end joining

Pathway that repairs DNA double-stranded breaks without the need for a homologous template to ligate the break ends

Non-synonymous somatic mutations

Mutations acquired postnatally that change the amino acid sequence of a protein

Oncogene-induced senescence

Antiproliferative effects of oncoproteins mediated by a DNA damage response to DNA hyper-replication

Open-label clinical trials

A type of study in which health providers and study subjects are aware of the treatment or drug being given

Oxyphil cells

Cells with an eosinophilic cytoplasm containing abundant, abnormally large, mitochondria and large centrally located nuclei

Radioactive iodine (RAI) therapy

Administration of iodine-131 to ablate thyroid tissue. In patients with thyroid cancer, it is used to destroy the remnants of thyroid after thyroidectomy as adjuvant therapy or to treat RAI-avid metastases

RAI avidity

Refers to the cellular property of incorporating and retaining radioactive iodine isotopes

Sleeping beauty (SB) transposon

Composed of a Sleeping Beauty transposase and a synthetic DNA transposon designed to integrate into the genome of vertebrates to introduce new phenotypes and identify the genes responsible for them

SWI/SNF chromatin remodelling complexes

Multisubunit protein complexes that elicit a DNA-stimulated ATPase activity that destabilizes histone–DNA interactions to mobilize nucleosomes, which increases accessibility of transcription factors to chromatin to activate or repress gene expression

Telomere crisis

When telomeres become critically short, they are unable to protect chromosome ends from the DNA damage response and repair pathways. Telomere shortening can enable cancer growth through telomere crisis, a state of extensive genomic instability causing translocations, amplifications and deletions

Tetraploidy

A cell containing four homologous copies of all chromosomes

Thyroglobulin

A glycoprotein produced by thyroid cells that is secreted into the lumen of the follicle. Tyrosine residues in thyroglobulin incorporate iodine through the action of thyroid peroxidase, which upon cleavage and coupling in lysosomes gives rise to thyroid hormones

Thyroid follicular cells

The major cell type of the thyroid gland, derived from the endoderm, and responsible for the production and secretion of the thyroid hormones thyroxine and triiodothyronine

Thyroid parafollicular or C cells

Neuroendocrine cells of the thyroid, which secrete calcitonin, a hormone that helps control the level of calcium in blood

Thyroid-stimulating hormone

(TSH). Glycoprotein hormone secreted by the pituitary gland that binds to the TSH receptor on thyroid cells to stimulate cell growth and expression of iodine metabolism genes

Thyrotrophs

Pituitary cells that secrete TSH

Tumour microevolution

Cancer is commonly understood to develop as a microevolutionary process, whereby a mutation initially confers a cell with a growth advantage allowing it to clonally expand. Sequential acquisition of new mutations in turn provides further stepwise fitness to the emerging clones

Uniparental disomy

(UPD). Refers to the presence of two copies of a chromosome (chromosomes) derived from a single parent. In cancer, it manifests as large blocks of homozygosity with normal copy number

Footnotes

Competing interests

The authors declare no competing interests.

References

  • 1.IARC. Cancer Incidence in Five Continents Volume X (IARC Scientific Publication, 2014). [PubMed] [Google Scholar]
  • 2.LiVolsi VA & Asa SL The demise of follicular carcinoma of the thyroid gland. Thyroid 4, 233–236 (1994). [DOI] [PubMed] [Google Scholar]
  • 3.Kakudo K, Kameyama K, Hirokawa M, Katoh R & Nakamura H Subclassification of follicular neoplasms recommended by the Japan thyroid association reporting system of thyroid cytology. Int. J. Endocrinol 2015, 938305 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Alevizaki M et al. Increasing prevalence of papillary thyroid carcinoma in recent years in Greece: the majority are incidental. Thyroid 19, 749–754 (2009). [DOI] [PubMed] [Google Scholar]
  • 5.Goffredo P, Roman SA & Sosa JA Hürthle cell carcinoma: a population-level analysis of 3311 patients. Cancer 119, 504–511 (2013). [DOI] [PubMed] [Google Scholar]
  • 6.Asioli S et al. Poorly differentiated carcinoma of the thyroid: validation of the Turin proposal and analysis of IMP3 expression. Mod. Pathol 23, 1269–1278 (2010). [DOI] [PubMed] [Google Scholar]
  • 7.Fagin JA & Wells SA Jr. Biologic and clinical perspectives on thyroid cancer 6. N. Engl. J. Med 375, 1054–1067 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.National Cancer Institute. Surveillance, Epidemiology and End Results Program. Cancer Stat Facts: Thyroid Cancer; 2022. https://seer.cancer.gov/statfacts/html/thyro.html (2022).
  • 9.Xu B et al. Primary high-grade non-anaplastic thyroid carcinoma: a retrospective study of 364 cases. Histopathology 80, 322–337 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Baloch ZW et al. Overview of the 2022 WHO classification of thyroid neoplasms. Endocr. Pathol 33, 27–63 (2022). [DOI] [PubMed] [Google Scholar]
  • 11.Maniakas A et al. Evaluation of overall survival in patients with anaplastic thyroid carcinoma, 2000–2019. JAMA Oncol. 6, 1397–1404 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Subbiah V et al. Dabrafenib and trametinib treatment in patients with locally advanced or metastatic BRAF V600-mutant anaplastic thyroid cancer. J. Clin. Oncol 36, 7–13 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cabanillas ME et al. Neoadjuvant BRAF- and immune-directed therapy for anaplastic thyroid carcinoma. Thyroid 28, 945–951 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Jaber T, Dadu R & Hu MI Medullary thyroid carcinoma. Curr. Opin. Endocrinol. Diabetes Obes 28, 540–546 (2021). [DOI] [PubMed] [Google Scholar]
  • 15.Fugazzola L Medullary thyroid cancer — an update. Best. Pract. Res. Clin. Endocrinol. Metab 37, 101655 (2023). [DOI] [PubMed] [Google Scholar]
  • 16.Davies L & Welch HG Increasing incidence of thyroid cancer in the United States, 1973–2002. J. Am. Med. Assoc 295, 2164–2167 (2006). [DOI] [PubMed] [Google Scholar]; This study shows that the steep rise in thyroid cancer incidence in the USA over the indicated period was predominantly due to the increased detection of PTCs <2 cm in size.
  • 17.Davies L & Hoang JK Thyroid cancer in the USA: current trends and outstanding questions. Lancet Diabetes Endocrinol. 9, 11–12 (2021). [DOI] [PubMed] [Google Scholar]
  • 18.Roman BR, Morris LG & Davies L The thyroid cancer epidemic, 2017 perspective. Curr. Opin. Endocrinol. Diabetes Obes 24, 332–336 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Nikiforov YE et al. Nomenclature revision for encapsulated follicular variant of papillary thyroid carcinoma: a paradigm shift to reduce overtreatment of indolent tumors. JAMA Oncol. 2, 1023–1029 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ito Y et al. An observational trial for papillary thyroid microcarcinoma in Japanese patients. World J. Surg 34, 28–35 (2010). [DOI] [PubMed] [Google Scholar]
  • 21.Tuttle RM et al. Natural history and tumor volume kinetics of papillary thyroid cancers during active surveillance. JAMA Otolaryngol. Head Neck Surg 143, 1015–1020 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]; Together with Ito et al. (2010), this study provides evidence that most small PTCs show minimal growth rates during active surveillance.
  • 22.Song YS et al. Genomic and transcriptomic characteristics according to size of papillary thyroid microcarcinoma. Cancers 10.3390/cancers12051345 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bellelli R et al. Oncogene-induced senescence and its evasion in a mouse model of thyroid neoplasia. Mol. Cell Endocrinol 460, 24–35 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wang J et al. Conditional expression of RET/PTC induces a weak oncogenic drive in thyroid PCCL3 cells and inhibits thyrotropin action at multiple levels. Mol. Endocrinol 17, 1425–1436 (2003). [DOI] [PubMed] [Google Scholar]
  • 25.Xing M Molecular pathogenesis and mechanisms of thyroid cancer. Nat. Rev. Cancer 13, 184–199 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cabanillas ME, McFadden DG & Durante C Thyroid cancer. Lancet 388, 2783–2795 (2016). [DOI] [PubMed] [Google Scholar]
  • 27.Kunstman JW et al. Characterization of the mutational landscape of anaplastic thyroid cancer via whole-exome sequencing. Hum. Mol. Genet 24, 2318–2329 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Landa I et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J. Clin. Invest 126, 1052–1066 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]; Genomic and transcriptomic analysis of advanced thyroid cancers that shows the major categories of lesions associated with disease progression, many of which are present in well-differentiated tumours as subclonal events.
  • 29.Pozdeyev N et al. Genetic analysis of 779 advanced differentiated and anaplastic thyroid cancers. Clin. Cancer Res 24, 3059–3068 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yoo SK et al. Integrative analysis of genomic and transcriptomic characteristics associated with progression of aggressive thyroid cancer. Nat. Commun 10, 2764 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Saqcena M et al. SWI/SNF complex mutations promote thyroid tumor progression and insensitivity to redifferentiation therapies. Cancer Discov. 11, 1158–1175 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper shows that loss-of-function mutations in genes encoding SWI/SNF complex subunits render BRAF-mutant cancers refractory to redifferentiation therapies with MAPK pathway inhibitors aimed at restoring RAI avidity.
  • 32.Portulano C, Paroder-Belenitsky M & Carrasco N The Na+/I symporter (NIS): mechanism and medical impact. Endocr. Rev 35, 106–149 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Peiling Yang S & Ngeow J Familial non-medullary thyroid cancer: unraveling the genetic maze. Endocr. Relat. Cancer 23, R577–R595 (2016). [DOI] [PubMed] [Google Scholar]
  • 34.Gudmundsson J et al. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nat. Genet 41, 460–464 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]; A genome-wide association study identifying variants, adjacent to thyroid transcription factor genes NKX2–1 and FOXE1, as risk factors for thyroid cancer.
  • 35.Gudmundsson J et al. Discovery of common variants associated with low TSH levels and thyroid cancer risk. Nat. Genet 44, 319–322 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Amundadottir LT et al. Cancer as a complex phenotype: pattern of cancer distribution within and beyond the nuclear family. PLoS Med. 1, e65 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Landa I et al. The variant rs1867277 in FOXE1 gene confers thyroid cancer susceptibility through the recruitment of USF1/USF2 transcription factors. PLoS Genet. 5, e1000637 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Son HY et al. Genome-wide association and expression quantitative trait loci studies identify multiple susceptibility loci for thyroid cancer. Nat. Commun 8, 15966 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study identifies SNPs in the NRG1 gene as one of the strongest variants predisposing to PTC, which are associated with elevated NRG1 expression in thyroid tissues.
  • 39.Takahashi M et al. The FOXE1 locus is a major genetic determinant for radiation-related thyroid carcinoma in Chernobyl. Hum. Mol. Genet 19, 2516–2523 (2010). [DOI] [PubMed] [Google Scholar]
  • 40.Montero-Conde C et al. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discov. 3, 520–533 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Garcia-Rendueles MER et al. Yap governs a lineage-specific neuregulin1 pathway-driven adaptive resistance to RAF kinase inhibitors. Mol. Cancer 21, 213 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; Together with Montero-Conde et al. (2013), this study demonstrates that BRAF-mutant cancers are intrinsically resistant to MAPK pathway inhibitors through increased expression and activation of genes encoding proteins in the NRG1–HER3 or HER2 pathway, an effect governed in part by the HIPPO pathway transcriptional co-activator YAP.
  • 42.Cameselle-Teijeiro JM, Mete O, Asa SL & LiVolsi V Inherited follicular epithelial-derived thyroid carcinomas: from molecular biology to histological correlates. Endocr. Pathol 32, 77–101 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kamani T, Charkhchi P, Zahedi A & Akbari MR Genetic susceptibility to hereditary non-medullary thyroid cancer. Hered. Cancer Clin. Pract 20, 9 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang Y et al. Identification of rare variants predisposing to thyroid cancer. Thyroid 29, 946–955 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Cardis E et al. Risk of thyroid cancer after exposure to 131I in childhood. J. Natl Cancer Inst 97, 724–732 (2005). [DOI] [PubMed] [Google Scholar]
  • 46.Morton LM et al. Radiation-related genomic profile of papillary thyroid carcinoma after the Chernobyl accident. Science 10.1126/science.abg2538 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; Integrated genomic and transcriptomic characterization of post-Chernobyl thyroid cancers that shows radiation dose-dependent increases in small clonal deletions and balanced translocations bearing genomic hallmarks of the DNA repair pathway non-homologous end joining.
  • 47.Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H & Fagin JA Distinct pattern of ret oncogene rearrangements in morphological variants of radiation-induced and sporadic thyroid papillary carcinomas in children. Cancer Res. 57, 1690–1694 (1997). [PubMed] [Google Scholar]
  • 48.Rabes HM et al. Pattern of radiation-induced RET and NTRK1 rearrangements in 191 post-Chernobyl papillary thyroid carcinomas: biological, phenotypic, and clinical implications. Clin. Cancer Res 6, 1093–1103 (2000). [PubMed] [Google Scholar]
  • 49.Ricarte-Filho JC et al. Identification of kinase fusion oncogenes in post-Chernobyl radiation-induced thyroid cancers. J. Clin. Invest 123, 4935–4944 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Efanov AA et al. Investigation of the relationship between radiation dose and gene mutations and fusions in post-Chernobyl thyroid cancer. J. Natl Cancer Inst 110, 371–378 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mizuno T et al. Preferential induction of RET/PTC1 rearrangement by X-ray irradiation. Oncogene 19, 438–443 (2000). [DOI] [PubMed] [Google Scholar]
  • 52.Nikiforova MN et al. Proximity of chromosomal loci that participate in radiation-induced rearrangements in human cells. Science 290, 138–141 (2000). [DOI] [PubMed] [Google Scholar]
  • 53.Selmansberger M et al. Dose-dependent expression of CLIP2 in post-Chernobyl papillary thyroid carcinomas. Carcinogenesis 36, 748–756 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Selmansberger M et al. CLIP2 as radiation biomarker in papillary thyroid carcinoma. Oncogene 34, 3917–3925 (2015). [DOI] [PubMed] [Google Scholar]
  • 55.Riesco-Eizaguirre G & Santisteban P Endocrine tumours: advances in the molecular pathogenesis of thyroid cancer: lessons from the cancer genome. Eur. J. Endocrinol 175, R203–R217 (2016). [DOI] [PubMed] [Google Scholar]
  • 56.Zane M et al. Normal vs cancer thyroid stem cells: the road to transformation. Oncogene 35, 805–815 (2016). [DOI] [PubMed] [Google Scholar]
  • 57.Veschi V et al. Recapitulating thyroid cancer histotypes through engineering embryonic stem cells. Nat. Commun 14, 1351 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper demonstrates the greater transformation efficiency of thyroid progenitors compared with mature differentiated thyroid cells with thyroid cancer oncoproteins.
  • 58.Network C. G. A. R. Integrated genomic characterization of papillary thyroid carcinoma. Cell 159, 676–690 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper demonstrates the genomic and transcriptomic study from the TCGA establishing the role of PTC drivers in governing the transcriptomic and phenotypic features of the disease leading to a new molecular classification of DTCs.
  • 59.Ghossein RA, Katabi N & Fagin JA Immunohistochemical detection of mutated BRAF V600E supports the clonal origin of BRAF-induced thyroid cancers along the spectrum of disease progression. J. Clin. Endocrinol. Metab 98, E1414–E1421 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kimura ET et al. High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 63, 1454–1457 (2003). [PubMed] [Google Scholar]
  • 61.Frattini M et al. Alternative mutations of BRAF, RET and NTRK1 are associated with similar but distinct gene expression patterns in papillary thyroid cancer 99. Oncogene 23, 7436–7440 (2004). [DOI] [PubMed] [Google Scholar]
  • 62.Santoro M, Moccia M, Federico G & Carlomagno F RET gene fusions in malignancies of the thyroid and other tissues. Genes 10.3390/genes11040424 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Franco AT et al. Fusion oncogenes are associated with increased metastatic capacity and persistent disease in pediatric thyroid cancers. J. Clin. Oncol 40, 1081–1090 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lawrence MS et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Martinez-Jimenez F et al. A compendium of mutational cancer driver genes. Nat. Rev. Cancer 20, 555–572 (2020). [DOI] [PubMed] [Google Scholar]
  • 66.Taylor AM et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689.e3 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lito P et al. Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. Cancer Cell 22, 668–682 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Boucai L et al. Genomic and transcriptomic characteristics of metastatic thyroid cancers with exceptional responses to radioactive iodine therapy. Clin. Cancer Res 29, 1620–1630 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Franco AT et al. Thyrotrophin receptor signaling dependence of Braf-induced thyroid tumor initiation in mice. Proc. Natl Acad. Sci. USA 108, 1615–1620 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Charles RP, Iezza G, Amendola E, Dankort D & McMahon M Mutationally activated BRAFV600E elicits papillary thyroid cancer in the adult mouse. Cancer Res. 71, 3863–3871 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chakravarty D et al. molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. J. Clin. Invest 121, 4700–4711 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that genetic or pharmacological inhibition of the MAPK pathway rescues expression of thyroid differentiation genes and RAI avidity of BRAFV600E-induced PTC in mice.
  • 72.Rivera M et al. Encapsulated papillary thyroid carcinoma: a clinico-pathologic study of 106 cases with emphasis on its morphologic subtypes (histologic growth pattern). Thyroid 19, 119–127 (2009). [DOI] [PubMed] [Google Scholar]
  • 73.Mesa C Jr. et al. Conditional activation of RET/PTC3 and BRAFV600E in thyroid cells is associated with gene expression profiles that predict a preferential role of BRAF in extracellular matrix remodeling. Cancer Res. 66, 6521–6529 (2006). [DOI] [PubMed] [Google Scholar]
  • 74.Huntington JT et al. Overexpression of collagenase 1 (MMP-1) is mediated by the ERK pathway in invasive melanoma cells: role of BRAF mutation and fibroblast growth factor signaling. J. Biol. Chem 279, 33168–33176 (2004). [DOI] [PubMed] [Google Scholar]
  • 75.Weiss MB et al. TWIST1 is an ERK1/2 effector that promotes invasion and regulates MMP-1 expression in human melanoma cells. Cancer Res. 72, 6382–6392 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Santoro M et al. Development of thyroid papillary carcinomas secondary to tissue- specific expression of the RET/PTC1 oncogene in transgenic mice. Oncogene 12, 1821–1826 (1996). [PubMed] [Google Scholar]
  • 77.Russell JP et al. The TRK-T1 fusion protein induces neoplastic transformation of thyroid epithelium. Oncogene 19, 5729–5735 (2000). [DOI] [PubMed] [Google Scholar]
  • 78.Jhiang SM et al. Targeted expression of the ret/PTC1 oncogene induces papillary thyroid carcinomas. Endocrinology 137, 375–378 (1996). [DOI] [PubMed] [Google Scholar]
  • 79.Raman R et al. Inhibition of FGF receptor blocks adaptive resistance to RET inhibition in CCDC6-RET-rearranged thyroid cancer. J. Exp. Med 10.1084/jem.20210390 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Montero-Conde C et al. Transposon mutagenesis identifies chromatin modifiers cooperating with Ras in thyroid tumorigenesis and detects ATXN7 as a cancer gene. Proc. Natl Acad. Sci. USA 114, E4951–E4960 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zou MJ et al. KRASG12D-mediated oncogenic transformation of thyroid follicular cells requires long-term TSH stimulation and is regulated by SPRY1. Lab. Invest 95, 1269–1277 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Garcia-Rendueles ME et al. NF2 loss promotes oncogenic RAS-induced thyroid cancers via YAP-dependent transactivation of RAS proteins and sensitizes them to MEK inhibition2. Cancer Discov. 5, 1178–1193 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Untch BR et al. Tipifarnib inhibits HRAS-driven dedifferentiated thyroid cancers. Cancer Res. 10.1158/0008-5472.CAN-17-1925 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Krishnamoorthy GP et al. EIF1AX and RAS mutations cooperate to drive thyroid tumorigenesis through ATF4 and c-MYC. Cancer Discov. 9, 264–281 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Morris LG, Shaha AR, Tuttle RM, Sikora AG & Ganly I Tall-cell variant of papillary thyroid carcinoma: a matched-pair analysis of survival. Thyroid 20, 153–158 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tsybrovskyy O et al. Papillary thyroid carcinoma tall cell variant shares accumulation of mitochondria, mitochondrial DNA mutations, and loss of oxidative phosphorylation complex I integrity with oncocytic tumors. J. Pathol. Clin. Res 8, 155–168 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Maenhaut C et al. in Endotext (eds Feingold KR et al. ) (MDText.com, 2000). [Google Scholar]
  • 88.Laugwitz KL et al. The human thyrotropin receptor: a heptahelical receptor capable of stimulating members of all four G protein families. Proc. Natl Acad. Sci. USA 93, 116–120 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Meinkoth JL, Goldsmith PK, Spiegel AM, Feramisco JR & Burrow GN Inhibition of thyrotropin-induced DNA synthesis in thyroid follicular cells by microinjection of an antibody to the stimulatory G protein of adenylate cyclase, Gs. J. Biol. Chem 267, 13239–13245 (1992). [PubMed] [Google Scholar]
  • 90.Parma J et al. Somatic mutations in the thyrotropin receptor gene cause hyperfunctioning thyroid adenomas. Nature 365, 649–651 (1993). [DOI] [PubMed] [Google Scholar]; This paper shows that gain-of-function mutations of TSHR activate cAMP signalling and drive development of benign hyperfunctioning thyroid tumours.
  • 91.Parma J et al. Somatic mutations causing constitutive activity of the thyrotropin receptor are the major cause of hyperfunctioning thyroid adenomas: identification of additional mutations activating both the cyclic adenosine 3’,5’-monophosphate and inositol phosphate-Ca2+ cascades. Mol. Endocrinol 9, 725–733 (1995). [DOI] [PubMed] [Google Scholar]
  • 92.Parma J et al. Diversity and prevalence of somatic mutations in the thyrotropin receptor and Gsα genes as a cause of toxic thyroid adenomas. J. Clin. Endocrinol. Metab 82, 2695–2701 (1997). [DOI] [PubMed] [Google Scholar]
  • 93.Mirfakhraee S, Mathews D, Peng L, Woodruff S & Zigman JM A solitary hyperfunctioning thyroid nodule harboring thyroid carcinoma: review of the literature. Thyroid Res. 6, 7 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Nicolson NG et al. Comprehensive genetic analysis of follicular thyroid carcinoma predicts prognosis independent of histology. J. Clin. Endocrinol. Metab 103, 2640–2650 (2018). [DOI] [PubMed] [Google Scholar]
  • 95.Mon SY et al. Cancer risk and clinicopathological characteristics of thyroid nodules harboring thyroid-stimulating hormone receptor gene mutations. Diagn. Cytopathol 46, 369–377 (2018). [DOI] [PubMed] [Google Scholar]
  • 96.Russo D et al. A Val 677 activating mutation of the thyrotropin receptor in a Hürthle cell thyroid carcinoma associated with thyrotoxicosis. Thyroid 9, 13–17 (1999). [DOI] [PubMed] [Google Scholar]
  • 97.Calebiro D et al. Recurrent EZH1 mutations are a second hit in autonomous thyroid adenomas. J. Clin. Invest 126, 3383–3388 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kirschner LS et al. Mutations of the gene encoding the protein kinase A type I-α regulatory subunit in patients with the Carney complex. Nat. Genet 26, 89–92 (2000). [DOI] [PubMed] [Google Scholar]
  • 99.Pringle DR et al. Thyroid-specific ablation of the Carney complex gene, PRKAR1A, results in hyperthyroidism and follicular thyroid cancer. Endocr. Relat. Cancer 19, 435–446 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Sandrini F et al. Regulatory subunit type I-α of protein kinase A (PRKAR1A): a tumor-suppressor gene for sporadic thyroid cancer. Genes Chromosom. Cancer 35, 182–192 (2002). [DOI] [PubMed] [Google Scholar]
  • 101.Gao J et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal 10.1126/scisignal.2004088 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mitsutake N et al. Conditional BRAFV600E expression induces DNA synthesis, apoptosis, dedifferentiation, and chromosomal instability in thyroid PCCL3 cells. Cancer Res. 65, 2465–2473 (2005). [DOI] [PubMed] [Google Scholar]
  • 103.Knauf JA, Kuroda H, Basu S & Fagin JA RET/PTC-induced dedifferentiation of thyroid cells is mediated through Y1062 signaling through SHC-RAS-MAP kinase. Oncogene 22, 4406–4412 (2003). [DOI] [PubMed] [Google Scholar]
  • 104.Liu D et al. Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant. Clin. Cancer Res 13, 1341–1349 (2007). [DOI] [PubMed] [Google Scholar]
  • 105.Nagarajah J et al. Sustained ERK inhibition maximizes responses of BrafV600E thyroid cancers to radioiodine. J. Clin. Invest 126, 4119–4124 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ho AL et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. N. Engl. J. Med 368, 623–632 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]; Pilot clinical trial showing that inhibition of MEK promotes response to RAI therapy in RAI-refractory metastatic thyroid cancers, particularly those with RAS mutations.
  • 107.Dunn LA et al. Vemurafenib redifferentiation of BRAF mutant, RAI-refractory thyroid cancers. J. Clin. Endocrinol. Metab 104, 1417–1428 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rothenberg SM, McFadden DG, Palmer EL, Daniels GH & Wirth LJ Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clin. Cancer Res 21, 1028–1035 (2015). [DOI] [PubMed] [Google Scholar]
  • 109.Groussin L et al. Redifferentiating effect of larotrectinib in NTRK-rearranged advanced radioactive-iodine refractory thyroid cancer. Thyroid 10.1089/thy.2021.0524 (2022). [DOI] [PubMed] [Google Scholar]
  • 110.Lee YA et al. NTRK and RET fusion-directed therapy in pediatric thyroid cancer yields a tumor response and radioiodine uptake. J. Clin. Invest 10.1172/JCI144847 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tchekmedyian V et al. Enhancing radioiodine incorporation in BRAF-mutant, radioiodine-refractory thyroid cancers with vemurafenib and the anti-ErbB3 monoclonal antibody CDX-3379: results of a pilot clinical trial. Thyroid 32, 273–282 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Weber M et al. Enhancing radioiodine incorporation into radioiodine-refractory thyroid cancer with MAPK inhibition (ERRITI): a single-center prospective two-arm study. Clin. Cancer Res 28, 4194–4202 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Colletta G, Cirafici AM & Di Carlo A Dual effect of transforming growth factor beta on rat thyroid cells: inhibition of thyrotropin-induced proliferation and reduction of thyroid-specific differentiation markers. Cancer Res. 49, 3457–3462 (1989). [PubMed] [Google Scholar]
  • 114.Riesco-Eizaguirre G et al. The BRAFV600E oncogene induces transforming growth factor β secretion leading to sodium iodide symporter repression and increased malignancy in thyroid cancer. Cancer Res. 69, 8317–8325 (2009). [DOI] [PubMed] [Google Scholar]
  • 115.Luckett KA et al. Co-inhibition of SMAD and MAPK signaling enhances 124I uptake in BRAF-mutant thyroid cancers. Endocr. Relat. Cancer 28, 391–402 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Costamagna E, Garcia B & Santisteban P The functional interaction between the paired domain transcription factor Pax8 and Smad3 is involved in transforming growth factor-β repression of the sodium/iodide symporter gene. J. Biol. Chem 279, 3439–3446 (2004). [DOI] [PubMed] [Google Scholar]
  • 117.Kroll TG et al. PAX8-PPARγ1 fusion oncogene in human thyroid carcinoma [corrected]. Science 289, 1357–1360 (2000). [DOI] [PubMed] [Google Scholar]
  • 118.Tontonoz P, Hu E & Spiegelman BM Stimulation of adipogenesis in fibroblasts by PPARγ2, a lipid-activated transcription factor. Cell 79, 1147–1156 (1994). [DOI] [PubMed] [Google Scholar]
  • 119.Dobson ME et al. Pioglitazone induces a proadipogenic antitumor response in mice with PAX8–PPARγ fusion protein thyroid carcinoma. Endocrinology 152, 4455–4465 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhang Y et al. Genomic binding of PAX8–PPARG fusion protein regulates cancer-related pathways and alters the immune landscape of thyroid cancer. Oncotarget 8, 5761–5773 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Giordano TJ et al. Pioglitazone therapy of PAX8–PPARγ fusion protein thyroid carcinoma. J. Clin. Endocrinol. Metab 103, 1277–1281 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lui WO et al. CREB3L2PPARγ fusion mutation identifies a thyroid signaling pathway regulated by intramembrane proteolysis. Cancer Res. 68, 7156–7164 (2008). [DOI] [PubMed] [Google Scholar]
  • 123.Koumarianou P, Gomez-Lopez G & Santisteban P Pax8 controls thyroid follicular polarity through cadherin-16. J. Cell Sci 130, 219–231 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that the thyroid transcription factor PAX8 regulates thyroid cell polarity through expression of the adhesion protein cadherin 16 and by facilitating epithelial cell interactions with the extracellular matrix.
  • 124.Plachov D et al. Pax8, a murine paired box gene expressed in the developing excretory system and thyroid gland. Development 110, 643–651 (1990). [DOI] [PubMed] [Google Scholar]
  • 125.Raman P & Koenig RJ Pax-8–PPAR-γ fusion protein in thyroid carcinoma. Nat. Rev. Endocrinol 10, 616–623 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Karunamurthy A et al. Prevalence and phenotypic correlations of EIF1AX mutations in thyroid nodules. Endocr. Relat. Cancer 23, 295–301 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Paulsson JO et al. Whole-genome sequencing of follicular thyroid carcinomas reveal recurrent mutations in microRNA processing subunit DGCR8. J. Clin. Endocrinol. Metab 106, 3265–3282 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Robertson AG et al. Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell 32, 204–220 e215 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Dong W et al. Clonal evolution analysis of paired anaplastic and well-differentiated thyroid carcinomas reveals shared common ancestor. Genes Chromosom. Cancer 57, 645–652 (2018). [DOI] [PubMed] [Google Scholar]
  • 130.Montero-Conde C et al. Comprehensive molecular analysis of immortalization hallmarks in thyroid cancer reveals new prognostic markers. Clin. Transl. Med 12, e1001 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wright WE, Piatyszek MA, Rainey WE, Byrd W & Shay JW Telomerase activity in human germline and embryonic tissues and cells. Dev. Genet 18, 173–179 (1996). [DOI] [PubMed] [Google Scholar]
  • 132.Blasco MA et al. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 91, 25–34 (1997). [DOI] [PubMed] [Google Scholar]
  • 133.Counter CM et al. Telomere shortening associated with chromosome instability is arrested in immortal cells which express telomerase activity. EMBO J. 11, 1921–1929 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Horn S et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959–961 (2013). [DOI] [PubMed] [Google Scholar]
  • 135.Huang FW et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]; Together with Horn et al. (2013), this study first demonstrates recurrent somatic non-coding mutations driving tumorigenesis. The mutations generate de novo consensus binding sites for the ETS family of transcription factors in the TERT gene promoter.
  • 136.Liu X et al. Highly prevalent TERT promoter mutations in aggressive thyroid cancers. Endocr. Relat. Cancer 20, 603–610 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Landa I et al. Frequent somatic TERT promoter mutations in thyroid cancer: higher prevalence in advanced forms of the disease. J. Clin. Endocrinol. Metab 98, E1562–E1566 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Vinagre J et al. Frequency of TERT promoter mutations in human cancers. Nat. Commun 4, 2185 (2013). [DOI] [PubMed] [Google Scholar]
  • 139.Killela PJ et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc. Natl Acad. Sci. USA 110, 6021–6026 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Liu R, Zhang T, Zhu G & Xing M Regulation of mutant TERT by BRAF V600E/MAP kinase pathway through FOS/GABP in human cancer. Nat. Commun 9, 579 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Thornton CEM, Hao J, Tamarapu PP & Landa I Multiple ETS factors participate in the transcriptional control of TERT mutant promoter in thyroid cancers. Cancers 10.3390/cancers14020357 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Bullock M et al. ETS factor ETV5 activates the mutant telomerase reverse transcriptase promoter in thyroid cancer. Thyroid 29, 1623–1633 (2019). [DOI] [PubMed] [Google Scholar]
  • 143.Song YS et al. Interaction of BRAF-induced ETS factors with mutant TERT promoter in papillary thyroid cancer. Endocr. Relat. Cancer 26, 629–641 (2019). [DOI] [PubMed] [Google Scholar]
  • 144.Lee DD et al. DNA hypermethylation within TERT promoter upregulates TERT expression in cancer. J. Clin. Invest 129, 223–229 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Rowland TJ, Bonham AJ & Cech TR Allele-specific proximal promoter hypomethylation of the telomerase reverse transcriptase gene (TERT) associates with TERT expression in multiple cancers. Mol. Oncol 14, 2358–2374 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.McKelvey BA et al. Characterization of allele-specific regulation of telomerase reverse transcriptase in promoter mutant thyroid cancer cell lines. Thyroid 30, 1470–1481 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Xing MZ et al. BRAF V600E and TERT promoter mutations cooperatively identify the most aggressive papillary thyroid cancer with highest recurrence. J. Clin. Oncol 32, 2718 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper demonstrates that concurrent BRAF and TERT promoter mutations mark PTCs with worse clinical outcomes.
  • 148.Park H et al. Molecular classification of follicular thyroid carcinoma based on TERT promoter mutations. Mod. Pathol 35, 186–192 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Thompson CAH & Wong JMY Non-canonical functions of telomerase reverse transcriptase: emerging roles and biological relevance. Curr. Top. Med. Chem 20, 498–507 (2020). [DOI] [PubMed] [Google Scholar]
  • 150.Zhang L et al. Novel recurrent altered genes in Chinese patients with anaplastic thyroid cancer. J. Clin. Endocrinol. Metab 106, 988–998 (2021). [DOI] [PubMed] [Google Scholar]
  • 151.Wang JR et al. Impact of somatic mutations on survival outcomes in patients with anaplastic thyroid carcinoma. JCO Precis. Oncol 6, e2100504 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Chen H et al. Molecular profile of advanced thyroid carcinomas by next-generation sequencing: characterizing tumors beyond diagnosis for targeted therapy. Mol. Cancer Ther 17, 1575–1584 (2018). [DOI] [PubMed] [Google Scholar]
  • 153.Ibrahimpasic T et al. Outcomes in patients with poorly differentiated thyroid carcinoma. J. Clin. Endocrinol. Metab 99, 1245–1252 (2014). [DOI] [PubMed] [Google Scholar]
  • 154.Volante M et al. RAS mutations are the predominant molecular alteration in poorly differentiated thyroid carcinomas and bear prognostic impact. J. Clin. Endocrinol. Metab 94, 4735–4741 (2009). [DOI] [PubMed] [Google Scholar]
  • 155.McFadden DG et al. p53 constrains progression to anaplastic thyroid carcinoma in a Braf-mutant mouse model of papillary thyroid cancer. Proc. Natl Acad. Sci. USA 111, E1600–E1609 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that loss of p53 function promotes progression of BRAF mutant-induced tumours to ATCs.
  • 156.Knauf JA et al. Hgf/Met activation mediates resistance to BRAF inhibition in murine anaplastic thyroid cancers. J. Clin. Invest 10.1172/JCI120966 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.La Perle KM, Jhiang SM & Capen CC Loss of p53 promotes anaplasia and local invasion in ret/PTC1-induced thyroid carcinomas. Am. J. Pathol 157, 671–677 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Orloff MS et al. Germline PIK3CA and AKT1 mutations in Cowden and Cowden-like syndromes. Am. J. Hum. Genet 92, 76–80 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Yehia L, Ngeow J & Eng C PTEN-opathies: from biological insights to evidence-based precision medicine. J. Clin. Invest 129, 452–464 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Charles RP, Silva J, Iezza G, Phillips WA & McMahon M Activating BRAF and PIK3CA mutations cooperate to promote anaplastic thyroid carcinogenesis. Mol. Cancer Res 12, 979–986 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Yeager N, Klein-Szanto A, Kimura S & Di CA Pten loss in the mouse thyroid causes goiter and follicular adenomas: insights into thyroid function and Cowden disease pathogenesis. Cancer Res. 67, 959–966 (2007). [DOI] [PubMed] [Google Scholar]
  • 162.Kimura T et al. Regulation of thyroid cell proliferation by TSH and other factors: a critical evaluation of in vitro models. Endocr. Rev 22, 631–656 (2001). [DOI] [PubMed] [Google Scholar]
  • 163.Tode B et al. Insulin-like growth factor-I: autocrine secretion by human thyroid follicular cells in primary culture. J. Clin. Endocrinol. Metab 69, 639–647 (1989). [DOI] [PubMed] [Google Scholar]
  • 164.Pappa T et al. Oncogenic mutations in PI3K/AKT/mTOR pathway effectors associate with worse prognosis in BRAFV600E-driven papillary thyroid cancer patients. Clin. Cancer Res 27, 4256–4264 (2021). [DOI] [PubMed] [Google Scholar]
  • 165.Ricarte-Filho JC et al. Mutational profile of advanced primary and metastatic radioactive iodine-refractory thyroid cancers reveals distinct pathogenetic roles for BRAF, PIK3CA, and AKT1. Cancer Res. 69, 4885–4893 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Miller KA et al. Oncogenic Kras requires simultaneous PI3K signaling to induce ERK activation and transform thyroid epithelial cells in vivo. Cancer Res. 69, 3689–3694 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Bedard PL et al. A phase Ib dose-escalation study of the oral pan-PI3K inhibitor buparlisib (BKM120) in combination with the oral MEK1/2 inhibitor trametinib (GSK1120212) in patients with selected advanced solid tumors. Clin. Cancer Res 21, 730–738 (2015). [DOI] [PubMed] [Google Scholar]
  • 168.Algazi AP et al. A phase I trial of BKM120 combined with vemurafenib in BRAFV600E/k mutant advanced melanoma. J. Clin. Oncol 32, 9101–9101 (2014). [Google Scholar]
  • 169.Liu GY & Sabatini DM mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol 21, 183–203 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Alessi DR, Sakamoto K & Bayascas JR LKB1-dependent signaling pathways. Annu. Rev. Biochem 75, 137–163 (2006). [DOI] [PubMed] [Google Scholar]
  • 171.Grabiner BC et al. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov. 4, 554–563 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Wagle N et al. Activating mTOR mutations in a patient with an extraordinary response on a phase I trial of everolimus and pazopanib. Cancer Discov. 4, 546–553 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hanna GJ et al. Genomic correlates of response to everolimus in aggressive radioiodine-refractory thyroid cancer: a phase II study. Clin. Cancer Res 24, 1546–1553 (2018). [DOI] [PubMed] [Google Scholar]
  • 174.Zhu XG, Zhao L, Willingham MC & Cheng SY Thyroid hormone receptors are tumor suppressors in a mouse model of metastatic follicular thyroid carcinoma. Oncogene 29, 1909–1919 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Sakurai A et al. Generalized resistance to thyroid hormone associated with a mutation in the ligand-binding domain of the human thyroid hormone receptor beta. Proc. Natl Acad. Sci. USA 86, 8977–8981 (1989). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Lu C, Zhao L, Ying H, Willingham MC & Cheng SY Growth activation alone is not sufficient to cause metastatic thyroid cancer in a mouse model of follicular thyroid carcinoma. Endocrinology 151, 1929–1939 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Cao X, Kambe F, Moeller LC, Refetoff S & Seo H Thyroid hormone induces rapid activation of Akt/protein kinase B-mammalian target of rapamycin-p70S6K cascade through phosphatidylinositol 3-kinase in human fibroblasts. Mol. Endocrinol 19, 102–112 (2005). [DOI] [PubMed] [Google Scholar]
  • 178.Furuya F, Hanover JA & Cheng SY Activation of phosphatidylinositol 3-kinase signaling by a mutant thyroid hormone β receptor. Proc. Natl Acad. Sci. USA 103, 1780–1785 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Furuya F, Lu C, Willingham MC & Cheng SY Inhibition of phosphatidylinositol 3-kinase delays tumor progression and blocks metastatic spread in a mouse model of thyroid cancer. Carcinogenesis 28, 2451–2458 (2007). [DOI] [PubMed] [Google Scholar]
  • 180.Saji M et al. Akt1 deficiency delays tumor progression, vascular invasion, and distant metastasis in a murine model of thyroid cancer. Oncogene 30, 4307–4315 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Saji M et al. Akt isoform-specific effects on thyroid cancer development and progression in a murine thyroid cancer model. Sci. Rep 10, 18316 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Khan SA et al. Unique mutation patterns in anaplastic thyroid cancer identified by comprehensive genomic profiling. Head Neck 41, 1928–1934 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Ciriello G et al. Emerging landscape of oncogenic signatures across human cancers. Nat. Genet 45, 1127–1133 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Schvartzman JM, Duijf PH, Sotillo R, Coker C & Benezra R Mad2 is a critical mediator of the chromosome instability observed upon Rb and p53 pathway inhibition. Cancer Cell 19, 701–714 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Kastenhuber ER & Lowe SW Putting p53 in Context. Cell 170, 1062–1078 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Baslan T et al. Ordered and deterministic cancer genome evolution after p53 loss. Nature 608, 795–802 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper shows that Trp53 loss in mouse tumours leads to a non-random sequence of disruptions characterized by Trp53 LOH, and gene deletions followed by genome doubling and copy number gains and gene amplifications.
  • 187.Hemmer S, Wasenius VM, Knuutila S, Franssila K & Joensuu H DNA copy number changes in thyroid carcinoma. Am. J. Pathol 154, 1539–1547 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Bartkova J et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006). [DOI] [PubMed] [Google Scholar]
  • 189.Sarkisian CJ et al. Dose-dependent oncogene-induced senescence in vivo and its evasion during mammary tumorigenesis. Nat. Cell Biol 9, 493–505 (2007). [DOI] [PubMed] [Google Scholar]
  • 190.Di Micco R et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006). [DOI] [PubMed] [Google Scholar]
  • 191.Drosten M et al. Loss of p53 induces cell proliferation via Ras-independent activation of the Raf/Mek/Erk signaling pathway. Proc. Natl Acad. Sci. USA 111, 15155–15160 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Ryder M et al. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid cancer progression. PLoS ONE 8, e54302 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Ryder M, Ghossein RA, Ricarte-Filho JC, Knauf JA & Fagin JA Increased density of tumor-associated macrophages is associated with decreased survival in advanced thyroid cancer. Endocr. Relat. Cancer 15, 1069–1074 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Caillou B et al. Tumor-associated macrophages (TAMs) form an interconnected cellular supportive network in anaplastic thyroid carcinoma. PLoS ONE 6, e22567 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]; Together with Ryder et al. (2008), this study presents evidence that ATCs are enveloped by an interconnected network of tumour-associated macrophages.
  • 195.Stenman A et al. Pan-genomic sequencing reveals actionable CDKN2A/2B deletions and kataegis in anaplastic thyroid carcinoma. Cancers 10.3390/cancers13246340 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Landa I et al. Comprehensive genetic characterization of human thyroid cancer cell lines: a validated panel for preclinical studies. Clin. Cancer Res 25, 3141–3151 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Alver BH et al. The SWI/SNF chromatin remodelling complex is required for maintenance of lineage specific enhancers. Nat. Commun 8, 14648 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Kadoch C et al. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nat. Genet 45, 592–601 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Wang X, Haswell JR & Roberts CW Molecular pathways: SWI/SNF (BAF) complexes are frequently mutated in cancer-mechanisms and potential therapeutic insights. Clin. Cancer Res 20, 21–27 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Wilson BG et al. Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation 16. Cancer Cell 18, 316–328 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Masoodi T et al. Evolution and impact of subclonal mutations in papillary thyroid cancer. Am. J. Hum. Genet 105, 959–973 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Pu W et al. Single-cell transcriptomic analysis of the tumor ecosystems underlying initiation and progression of papillary thyroid carcinoma. Nat. Commun 12, 6058 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; This scRNA-seq study of human PTCs shows distinct transcriptional states of thyroid cells along the course of disease progression.
  • 203.Luo H et al. Characterizing dedifferentiation of thyroid cancer by integrated analysis. Sci. Adv 10.1126/sciadv.abf3657 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; This single-cell analysis of progression from PTC to ATC links CREB3L1 overexpression arising from chromosome 11 copy number gain to EMT and mTOR pathway activation.
  • 204.Fan J et al. Linking transcriptional and genetic tumor heterogeneity through allele analysis of single-cell RNA-seq data. Genome Res. 28, 1217–1227 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Hornburg M et al. Single-cell dissection of cellular components and interactions shaping the tumor immune phenotypes in ovarian cancer. Cancer Cell 39, 928–944.e6 (2021). [DOI] [PubMed] [Google Scholar]
  • 206.Braun DA et al. Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell 39, 632–648.e8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Wu P, Sun W & Zhang H An immune-related prognostic signature for thyroid carcinoma to predict survival and response to immune checkpoint inhibitors. Cancer Immunol. Immunother 71, 747–759 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Cunha LL & Ward LS Translating the immune microenvironment of thyroid cancer into clinical practice. Endocr. Relat. Cancer 29, R67–R83 (2022). [DOI] [PubMed] [Google Scholar]
  • 209.Yan T et al. Single-cell transcriptomic analysis of ecosystems in papillary thyroid carcinoma progression. Front. Endocrinol 12, 729565 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.French JD et al. Programmed death-1+ T cells and regulatory T cells are enriched in tumor-involved lymph nodes and associated with aggressive features in papillary thyroid cancer. J. Clin. Endocrinol. Metab 97, E934–E943 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that specific subpopulations of tumour-infiltrating immunosuppressive cells are associated with more aggressive PTCs.
  • 211.Cunha LL et al. CD8+ tumour-infiltrating lymphocytes and COX2 expression may predict relapse in differentiated thyroid cancer. Clin. Endocrinol 83, 246–253 |(2015). [DOI] [PubMed] [Google Scholar]
  • 212.Angell TE et al. BRAF V600E in papillary thyroid carcinoma is associated with increased programmed death ligand 1 expression and suppressive immune cell infiltration. Thyroid 24, 1385–1393 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Moretti S et al. Indoleamine 2,3-dioxygenase 1 (IDO1) is up-regulated in thyroid carcinoma and drives the development of an immunosuppressant tumor microenvironment. J. Clin. Endocrinol. Metab 99, E832–E840 (2014). [DOI] [PubMed] [Google Scholar]
  • 214.Cunha LL et al. Differentiated thyroid carcinomas may elude the immune system by B7H1 upregulation. Endocr. Relat. Cancer 20, 103–110 (2013). [DOI] [PubMed] [Google Scholar]
  • 215.Giannini R et al. Immune profiling of thyroid carcinomas suggests the existence of two major phenotypes: an ATC-like and a PDTC-like. J. Clin. Endocrinol. Metab 104, 3557–3575 (2019). [DOI] [PubMed] [Google Scholar]
  • 216.Zhang P et al. Targeting myeloid derived suppressor cells reverts immune suppression and sensitizes BRAF-mutant papillary thyroid cancer to MAPK inhibitors. Nat. Commun 13, 1588 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study connects BRAF-V600E with recruitment of MDSCs and points to combinatorial MAPK pathway inhibition and MDSC depletion as a therapeutic strategy for thyroid cancer.
  • 217.Bertol BC et al. Lenvatinib plus anti-PD-1 combination therapy for advanced cancers: defining mechanisms of resistance in an inducible transgenic model of thyroid cancer. Thyroid 32, 153–163 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Xu B et al. Prolonged survival of anaplastic thyroid carcinoma is associated with resectability, low tumor-infiltrating neutrophils/myeloid-derived suppressor cells, and low peripheral neutrophil-to-lymphocyte ratio. Endocrine 76, 612–619 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Veglia F, Sanseviero E & Gabrilovich DI Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol 21, 485–498 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Li K et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal. Transduct. Target. Ther 6, 362 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Suzuki S et al. Immunosuppression involving increased myeloid-derived suppressor cell levels, systemic inflammation and hypoalbuminemia are present in patients with anaplastic thyroid cancer. Mol. Clin. Oncol 1, 959–964 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Alspach E, Lussier DM & Schreiber RD Interferon γ and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harb. Perspect. Biol 10.1101/cshperspect.a028480 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Dhatchinamoorthy K, Colbert JD & Rock KL Cancer immune evasion through loss of MHC class I antigen presentation. Front. Immunol 12, 636568 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Jhunjhunwala S, Hammer C & Delamarre L Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 21, 298–312 (2021). [DOI] [PubMed] [Google Scholar]
  • 225.Brea EJ et al. Kinase regulation of human MHC class I molecule expression on cancer cells. Cancer Immunol. Res 4, 936–947 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Zhi J et al. Inhibition of BRAF sensitizes thyroid carcinoma to immunotherapy by enhancing tsMHCII-mediated immune recognition. J. Clin. Endocrinol. Metab 106, 91–107 (2021). [DOI] [PubMed] [Google Scholar]
  • 227.Massague J & Ganesh K Metastasis-initiating cells and ecosystems. Cancer Discov. 11, 971–994 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Gao Y et al. Metastasis organotropism: redefining the congenial soil. Dev. Cell 49, 375–391 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Nguyen B et al. Genomic characterization of metastatic patterns from prospective clinical sequencing of 25,000 patients. Cell 185, 563–575.e11 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; Pan-cancer analysis of somatic mutations associated with distant metastases and metastatic tropism.
  • 230.Krishnamoorthy GP et al. Abstract 986: RBM10 loss in thyroid cancer leads to aberrant splicing of cytoskeletal and extracellular matrix mRNAs and increased metastatic fitness. Cancer Res. 82, 986–986 (2022).35074755 [Google Scholar]
  • 231.Haugen BR et al. 2015 American Thyroid Association Management Guidelines for adult patients with thyroid nodules and differentiated thyroid cancer: the American Thyroid Association Guidelines Task Force on thyroid nodules and differentiated thyroid cancer. Thyroid 26, 1–133 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Jonklaas J et al. Outcomes of patients with differentiated thyroid carcinoma following initial therapy. Thyroid 16, 1229–1242 (2006). [DOI] [PubMed] [Google Scholar]
  • 233.Ruel E et al. Adjuvant radioactive iodine therapy is associated with improved survival for patients with intermediate-risk papillary thyroid cancer. J. Clin. Endocrinol. Metab 100, 1529–1536 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Liu J et al. The genetic duet of BRAF V600E and TERT promoter mutations robustly predicts loss of radioiodine avidity in recurrent papillary thyroid cancer. J. Nucl. Med 61, 177–182 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Schlumberger M et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N. Engl. J. Med 372, 621–630 (2015). [DOI] [PubMed] [Google Scholar]
  • 236.Brose MS et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet 384, 319–328 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Matsui J et al. E7080, a novel inhibitor that targets multiple kinases, has potent antitumor activities against stem cell factor producing human small cell lung cancer H146, based on angiogenesis inhibition. Int. J. Cancer 122, 664–671 (2008). [DOI] [PubMed] [Google Scholar]
  • 238.Kumar R et al. Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors. Br. J. Cancer 101, 1717–1723 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Tahara M et al. Exploratory analysis of biomarkers associated with clinical outcomes from the study of lenvatinib in differentiated cancer of the thyroid. Eur. J. Cancer 75, 213–221 (2017). [DOI] [PubMed] [Google Scholar]
  • 240.Brose MS et al. Analysis of biomarkers and association with clinical outcomes in patients with differentiated thyroid cancer: subanalysis of the sorafenib phase III DECISION trial. Clin. Cancer Res 25, 7370–7380 (2019). [DOI] [PubMed] [Google Scholar]; Together with Schlumberger et al. (2015), this clinical trial of multikinase inhibitors in thyroid cancer led to the first FDA-approved small molecule systemic therapy for the disease.
  • 241.Carmeliet P & Jain RK Angiogenesis in cancer and other diseases. Nature 407, 249–257 (2000). [DOI] [PubMed] [Google Scholar]
  • 242.Jayson GC, Kerbel R, Ellis LM & Harris AL Antiangiogenic therapy in oncology: current status and future directions. Lancet 388, 518–529 (2016). [DOI] [PubMed] [Google Scholar]
  • 243.Butler JM, Kobayashi H & Rafii S Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer 10, 138–146 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Hick AC et al. Reciprocal epithelial: endothelial paracrine interactions during thyroid development govern follicular organization and C-cells differentiation. Dev. Biol 381, 227–240 (2013). [DOI] [PubMed] [Google Scholar]
  • 245.Degosserie J et al. Extracellular vesicles from endothelial progenitor cells promote thyroid follicle formation. J. Extracell. Vesicles 7, 1487250 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Carlomagno F et al. ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res. 62, 7284–7290 (2002). [PubMed] [Google Scholar]
  • 247.Subbiah V et al. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): a multi-cohort, open-label, registrational, phase 1/2 study. Lancet Diabetes Endocrinol. 10.1016/S2213-8587(21)00120-0 (2021). [DOI] [PubMed] [Google Scholar]
  • 248.Wirth LJ et al. Efficacy of selpercatinib in RET-altered thyroid cancers. N. Engl. J. Med 383, 825–835 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Addeo A et al. RET aberrant cancers and RET inhibitor therapies: current state-of-the-art and future perspectives. Pharmacol. Ther 242, 108344 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Cocco E, Scaltriti M & Drilon A NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol 15, 731–747 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Hong DS et al. KRASG12C inhibition with sotorasib in advanced solid tumors. N. Engl. J. Med 383, 1207–1217 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Ho AL et al. Tipifarnib in head and neck squamous cell carcinoma with HRAS mutations. J. Clin. Oncol 39, 1856–1864 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Davies H et al. Mutations of the BRAF gene in human cancer. Nature 417, 949–954 (2002). [DOI] [PubMed] [Google Scholar]
  • 254.Sosman JA et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N. Engl. J. Med 366, 707–714 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Kopetz S et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF-mutated colorectal cancer. J. Clin. Oncol 33, 4032–4038 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Brose MS et al. Vemurafenib in patients with BRAFV600E-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: a non-randomised, multicentre, open-label, phase 2 trial. Lancet Oncol. 17, 1272–1282 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Sherman EJ et al. Combination of dabrafenib and lapatinib for the treatment of BRAF-mutant thyroid cancer: a phase I study. J. Clin. Oncol 35, 6085 (2017). [Google Scholar]
  • 258.Bible KC et al. 2021 American Thyroid Association Guidelines for management of patients with anaplastic thyroid cancer. Thyroid 31, 337–386 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Hyman DM et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N. Engl. J. Med 373, 726–736 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Subbiah V et al. Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: updated analysis from the phase II ROAR basket study. Ann. Oncol 33, 406–415 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; Together with Subbiah et al. (2018), this study provides evidence that combined RAF and MEK inhibition leads to major responses and prolonged survival in patients with BRAF-mutant ATC.
  • 261.Busaidy NL et al. Dabrafenib versus dabrafenib + trametinib in BRAF-mutated radioactive iodine refractory differentiated thyroid cancer: results of a randomized, phase 2, open-label multicenter trial. Thyroid 32, 1184–1192 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Capdevila J et al. PD-1 blockade in anaplastic thyroid carcinoma. J. Clin. Oncol 38, 2620–2627 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Chiosea S et al. Molecular profiling of 50,734 Bethesda III–VI thyroid nodules by ThyroSeq v3: implications for personalized management. J. Clin. Endocrinol. Metab 10.1210/clinem/dgad220 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Zafereo M et al. A thyroid genetic classifier correctly predicts benign nodules with indeterminate cytology: two independent, multicenter, prospective validation trials. Thyroid 30, 704–712 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Lee M et al. Genomic and transcriptomic correlates of thyroid carcinoma evolution after BRAF inhibitor therapy. Mol. Cancer Res 20, 45–55 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Hofmann MC et al. Molecular mechanisms of resistance to kinase inhibitors and redifferentiation in thyroid cancers. Endocr. Relat. Cancer 29, R173–R190 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Andre F et al. AACR project GENIE: powering precision medicine through an international consortium. Cancer Discov. 7, 818–831 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Yoo SK et al. Comprehensive analysis of the transcriptional and mutational landscape of follicular and papillary thyroid cancers. PLoS Genet. 12, e1006239 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Song E et al. Genetic profile of advanced thyroid cancers in relation to distant metastasis. Endocr. Relat. Cancer 27, 285–293 (2020). [DOI] [PubMed] [Google Scholar]
  • 270.Lopez-Marquez A, Carrasco-Lopez C, Fernandez-Mendez C & Santisteban P Unraveling the complex interplay between transcription factors and signaling molecules in thyroid differentiation and function, from embryos to adults. Front. Endocrinol 12, 654569 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.McFadden DG & Sadow PM Genetics, diagnosis, and management of Hürthle cell thyroid neoplasms. Front. Endocrinol 12, 696386 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Chindris AM et al. Clinical and molecular features of Hürthle cell carcinoma of the thyroid. J. Clin. Endocrinol. Metab 100, 55–62 (2015). [DOI] [PubMed] [Google Scholar]
  • 273.Yeh JJ et al. Somatic mitochondrial DNA (mtDNA) mutations in papillary thyroid carcinomas and differential mtDNA sequence variants in cases with thyroid tumours. Oncogene 19, 2060–2066 (2000). [DOI] [PubMed] [Google Scholar]
  • 274.Gasparre G et al. Disruptive mitochondrial DNA mutations in complex I subunits are markers of oncocytic phenotype in thyroid tumors. Proc. Natl Acad. Sci. USA 104, 9001–9006 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.He Y et al. Heteroplasmic mitochondrial DNA mutations in normal and tumour cells. Nature 464, 610–614 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Ganly I et al. Integrated genomic analysis of Hürthle cell cancer reveals oncogenic drivers, recurrent mitochondrial mutations, and unique chromosomal landscapes. Cancer Cell 34, 256–270.e5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Gopal RK et al. Widespread chromosomal losses and mitochondrial DNA alterations as genetic drivers in Hürthle cell carcinoma. Cancer Cell 34, 242–255.e5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]; Together with Ganly et al. (2018), this study provides a comprehensive genomic analysis of OCs showing near haploidization of the nuclear genome coupled to mitochondrial DNA mutations of complex I subunits of the OXPHOS pathway.
  • 278.Frank AR et al. Mitochondrial-encoded complex I impairment induces a targetable dependency on aerobic fermentation in Hürthle cell carcinoma of the thyroid. Cancer Discov. 10.1158/2159-8290.CD-22-0982 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Ganly I et al. Mitonuclear genotype remodels the metabolic and microenvironmental landscape of Hürthle cell carcinoma. Sci. Adv 8, eabn9699 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES