You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/235690333

Molecular pathogenesis and mechanism of thyroid cancer

Article  in  Nature Reviews Cancer · March 2013


DOI: 10.1038/nrc3431 · Source: PubMed

CITATIONS READS

887 1,932

1 author:

Mingzhao Xing
Johns Hopkins Medicine
183 PUBLICATIONS   16,081 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

arachidonic acid release View project

Thyroid genomics View project

All content following this page was uploaded by Mingzhao Xing on 06 January 2015.

The user has requested enhancement of the downloaded file.


REVIEWS

Molecular pathogenesis and


mechanisms of thyroid cancer
Mingzhao Xing
Abstract | Thyroid cancer is a common endocrine malignancy. There has been exciting
progress in understanding its molecular pathogenesis in recent years, as best exemplified by
the elucidation of the fundamental role of several major signalling pathways and related
molecular derangements. Central to these mechanisms are the genetic and epigenetic
alterations in these pathways, such as mutation, gene copy-number gain and aberrant gene
methylation. Many of these molecular alterations represent novel diagnostic and prognostic
molecular markers and therapeutic targets for thyroid cancer, which provide unprecedented
opportunities for further research and clinical development of novel treatment strategies for
this cancer.

Differentiated thyroid
Thyroid cancer is a common endocrine malignancy that but it is often not curative. The recent progress in under-
cancer has rapidly increased in global incidence in recent dec- standing the molecular pathogenesis of thyroid cancer
(DTC). Collectively includes ades1,2. In the United States, the average annual increase has shown great promise for the development of more-
both papillary thyroid cancer in thyroid cancer incidence of 6.6% between 2000 and effective treatment strategies for thyroid cancer. This
(PTC) and follicular thyroid
2009 is the highest among all cancers2. Although the has mainly resulted from the identification of molecular
cancer (FTC), which are
histologically differentiated, by
death rate of thyroid cancer is relatively low, the rate of alterations, including the genetic and epigenetic altera-
comparison with poorly disease recurrence or persistence is high, which is asso- tions of signalling pathways — such as the RAS–RAF–
differentiated thyroid cancer ciated with increased incurability and patient morbidity MEK–MAPK–ERK pathway (MAPK pathway) and the
(PDTC) and undifferentiated and mortality 3. PI3K–AKT pathway — which is reshaping thyroid cancer
anaplastic thyroid cancer (ATC).
There are several histological types and subtypes of medicine. This Review discusses the recent remarkable
Radioiodine treatment thyroid cancer with different cellular origins, charac- progress in understanding the molecular pathogenesis
A classical treatment for teristics and prognoses4 (TABLE 1). There are two types and mechanisms of thyroid cancer.
patients with thyroid cancer. of endocrine thyroid cells — follicular thyroid cells and
It is used after total
parafollicular C cells — from which thyroid cancers Common genetic alterations in thyroid cancer
thyroidectomy and consists of
treating patients with the
are derived. Follicular thyroid cell-derived tumours, Gene mutations. Numerous genetic alterations that
radioiodine isotope 131I, which including papillary thyroid cancer (PTC), follicular thy- have a fundamental role in the tumorigenesis of vari-
emits high-energy β-particles roid cancer (FTC), poorly differentiated thyroid cancer ous thyroid tumours have been identified (TABLE 2).
and takes advantage of the (PDTC) and anaplastic thyroid cancer (ATC), account A prominent example is the T1799A transverse point
unique function of thyroid
follicular cells to accumulate
for the majority of thyroid malignancies. PTC and FTC mutation of BRAF, which results in the expression of
iodide as a substrate for the are collectively classified as differentiated thyroid cancer BRAF‑V600E mutant protein and causes constitutive
synthesis of thyroid hormone. (DTC). Parafollicular C cell-derived medullary thy- activation of this serine/threonine kinase6–11. BRAFV600E
roid cancer (MTC) accounts for a small proportion of mutation occurs in approximately 45% of PTCs12. There
thyroid malignancies2. The primary molecular mecha- are also a few rare types of BRAF mutations identified
Laboratory for Cellular and nism underlying MTC tumorigenesis is the aberrant in PTC, which mostly affect nucleotides around codon
Molecular Thyroid Research,
activation of RET signalling (which is caused by RET 600 and constitutively activate the BRAF kinase13,14. The
Division of Endocrinology and
Metabolism, Johns Hopkins mutations5), which are not present in follicular thy- requirement for BRAF‑V600E to maintain tumour growth
University School of Medicine, roid cell-derived tumours. The molecular pathogenesis was initially demonstrated in a xenograft tumour model15.
1830 East Monument Street, of follicular thyroid cell-derived tumours is the focus of A previous comprehensive multicentre study dem-
Suite 333, Baltimore, this Review. onstrated a strong association of BRAFV600E with poor
Maryland 21287, USA.
Correspondence to M.X. 
Conventional surgical thyroidectomy with adjuvant clinicopathological outcomes of PTC, including aggres-
e‑mail: mxing1@jhmi.edu ablation by radioiodine treatment has been the mainstay sive pathological features, increased recurrence, loss of
doi:10.1038/nrc3431 of treatment for follicular thyroid cell-derived cancer, radioiodine avidity and treatment failures16. This was

184 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

At a glance
The common occurrence of RAS mutations in follicu-
lar thyroid adenoma (FTA), a presumed premalignant
• Thyroid cancer is a common endocrine malignancy, and exciting progress has lesion, suggests that activated RAS may have a role in
occurred in recent years in understanding its molecular pathogenesis. early follicular thyroid cell tumorigenesis. However,
• Genetic and epigenetic alterations are the driving forces of thyroid cancer. additional genetic alterations other than RAS muta-
Examples of these alterations include mutations in BRAF (BRAFV600E), RAS, PIK3CA, tion are apparently required to transform FTA into
PTEN, TP53, β‑catenin (CTNNB1), anaplastic lymphoma kinase (ALK) and isocitrate
thyroid cancer. This is consistent with studies in which
dehydrogenase 1 (IDH1), translocations (RET–PTC and paired box 8 (PAX8)–peroxisome
proliferator-activated receptor‑γ (PPARG)) and aberrant gene methylation.
the expression of mutant HRAS was induced in nor-
• At the core of the molecular pathogenesis of thyroid cancer is the uncontrolled
mal thyroid cells in vitro, which resulted in only well-
activity of various signalling pathways, including the MAPK, PI3K–AKT, nuclear demarcated and differentiated colonies with phenotypes
factor‑κB (NF‑κB), RASSF1–mammalian STE20‑like protein kinase 1 (MST1)–forkhead consistent with FTA27 and cell proliferation with normal
box O3 (FOXO3), WNT–β‑catenin, hypoxia-inducible factor 1α (HIF1α) and differentiation28. More direct evidence has come from
thyroid-stimulating hormone (TSH)–TSH receptor (TSHR) pathways. transgenic mouse studies in which conditional physio­
• The progression of thyroid cancer is a process of accumulation of genetic and logical expression of a KRAS mutant in the thyroid gland
epigenetic alterations with corresponding progressive derangements of signalling could not transform thyroid cells, but concurrent KRAS
pathways. These are accompanied by numerous secondary molecular alterations, mutant expression and Pten deletion induced a rapid
both in the cell and in the tumour microenvironment, which, acting in cooperation, occurrence of aggressive FTC29.
amplify and synergize their impacts on thyroid tumorigenesis. Mutations or deletions of the tumour suppressor
• Aberrant silencing of thyroid iodide-handling genes and consequent loss of gene PTEN are the classical genetic alterations that acti-
radioiodine avidity of thyroid cancer promoted by BRAF‑V600E is a unique molecular
vate the PI3K–AKT pathway and are the genetic basis
pathological process in thyroid cancer, which causes the failure of radioiodine
for follicular thyroid cell tumorigenesis in Cowden’s
treatment.
syndrome30. Mutations of PIK3CA, which encodes the
• The recent molecular findings provide unprecedented opportunities for further
research and clinical development of novel molecular-based treatment strategies for p110α catalytic subunit of PI3K, are also common in thy-
thyroid cancer. roid cancer, particularly FTC, PDTC and ATC25,26,31–35.
As in other human cancers36, activating PIK3CA muta-
tions in thyroid cancer occur in exon 9 and exon 20.
subsequently confirmed in numerous other studies, AKT1 mutations were found in metastatic thyroid can-
MAPK pathway
This pathway has a fundamental although, perhaps owing to variations in study design, cers in one study, and their functional relevance remains
role in the regulation of cell some studies showed inconsistent results, as summa- to be established35.
growth, proliferation, apoptosis rized and discussed in large meta-analyses that uni- Other important genes that are mutated in thyroid
and metabolic activities, formly support the aggressive role of BRAFV600E mutation tumorigenesis include β‑catenin (CTNNB1)37,38, TP5339,40,
through regulating the
expression of various genes.
in PTC 17,18. BRAF V600E-transgenic mice also devel- isocitrate dehydrogenase 1 (IDH1)41,42, anaplastic lym-
oped aggressive PTC19–21. Interestingly, some human phoma kinase (ALK)43 and epidermal growth factor
PI3K–AKT pathway PTC tumours have recently been found to exhibit intra­ receptor (EGFR)44. The preferential occurrence of these
This pathway also has a tumour heterogeneity in the BRAF genotype — with a mutations in PDTC and ATC, which are the most aggres-
fundamental role in the
minority of cells harbouring BRAFV600E and the majority sive thyroid cancers, suggests that they may have a role in
regulation of cell growth,
proliferation, apoptosis and harbouring wild-type BRAF22. This raises an interesting the progression and aggressiveness of thyroid cancer.
metabolic activities, through ‘chicken and egg’ puzzle of whether BRAF‑V600E initi- In Hürthle-cell thyroid carcinoma (HCTC), mutations of
regulating the expression of ates PTC tumorigenesis or instead BRAFV600E occurs fol- NADH dehydrogenase (ubiquinone) 1α subcomplex 13
various genes. lowing the development of a thyroid tumour23. Although (NDUFA13; also known as GRIM19) are fairly com-
Pten
it is possible that BRAFV600E could be a secondary genetic mon45. Unlike other types of thyroid cancers, HCTC does
A tumour suppressor gene, the event in PTC tumorigenesis as previously proposed24, not harbour classical genetic alterations, such as BRAF
protein product of which an alternative possibility is that once PTC is initiated by and RAS mutations or RET–PTC 46,47, but commonly
converts phosphatidylinositol BRAFV600E, secondary oncogenic alterations could take harbours DNA haploidization in recurrent disease47.
(3,4,5)-trisphosphate (PIP3) to
over to drive the tumorigenesis of PTC such that BRAF
phosphatidylinositol (4,5)-
trisphosphate (PIP2), mutation is no longer selected for 23. Gene amplifications and copy-number gains. Oncogenic
counteracting the conversion Second in prevalence to BRAF mutations in thyroid gene amplification or copy-number gain are additional
of PIP2 to PIP3 by PI3K and cancer are RAS mutations. RAS is in its active state prominent genetic mechanisms in thyroid tumori-
thus terminating PI3K–AKT when bound with GTP. The intrinsic GTPase of RAS genesis (TABLE 3). This is particularly the case for genes
signalling.
hydrolyses GTP and converts RAS into an inactive GDP- encoding receptor tyrosine kinases (RTKs) 26. Copy-
Cowden’s syndrome bound state, thus terminating RAS signalling. RAS muta- number gains are also common for the genes encoding
Also known as Cowden’s tions cause the loss of its GTPase activity, thus locking PI3K–AKT pathway members, including PIK3CA,
disease or multiple hamartoma RAS in a constitutively active GTP-bound state. There PIK3CB, 3‑phosphoinositide-dependent protein
syndrome. A rare, autosomally
are three isoforms of RAS: HRAS, KRAS and NRAS, and kinase 1 (PDPK1; also known as PDK1), AKT1 and
inherited disorder that is
caused by mutations or defects NRAS is predominantly mutated in thyroid tumours, AKT2 (REFS 25,26,32). Overall, genetic copy-number
in the tumour suppressor gene mostly involving codons 12 and 61. Although RAS is gains of these genes are more prevalent in ATC than in
PTEN and is characterized by a classical dual activator of the MAPK and PI3K–AKT DTC, suggesting that these genetic alterations are impor-
multiple tumour-like growths pathways, RAS mutations seem to preferentially activate tant for the progression and aggressiveness of thyroid
called hamartomas and an
increased risk of certain
the PI3K–AKT pathway in thyroid tumorigenesis, as cancer. Copy-number gain of these genes in ATC could
cancers, such as breast cancer suggested by the preferential association of RAS muta- occur either through genetic amplification or chromo-
and follicular thyroid cancer. tions with AKT phosphorylation in thyroid cancers25,26. somal instability and aneuploidy. An important and

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 185

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Copy-number gain expected consequence of these copy-number gains is Gene amplification or copy-number gain of the
A genetic abnormality in which the activation of downstream signalling pathways, as IQ-motif-containing GTPase-activating protein 1
the copy number of a is suggested by the association of copy-number gains of (IQGAP1) is another important genetic event that has
chromosomal region or gene is genes encoding RTKs with increased phosphorylation been recently discovered in thyroid cancer 48. IQGAP1
more than the normal two
copies (one paternal allele and
of AKT and ERK in thyroid cancer 26. is a multifunctional scaffold protein that has an impor-
one maternal allele), which Many of the genes with copy-number gains are tant role in human tumorigenesis49. IQGAP1 amplifica-
occurs through the proto-oncogenes. A mechanism for them to contribute tion was found to increase protein expression and to
amplification of a local region to thyroid tumorigenesis is through increased protein be associated with invasiveness of thyroid cancer cells.
of DNA within a chromosome,
expression and consequent aberrant activation of the Interestingly, coexistence of IQGAP1 copy-number gain
or through aneuploidy, in which
multiple copies or fragments of
signalling pathways in which they are involved25,26,33. and BRAFV600E mutation was particularly associated with
identical chromosomes are It is interesting to note that in DTCs, PIK3CA mutation a high incidence of recurrent PTC48.
present. is mutually exclusive with PIK3CA copy-number gain31,33,
suggesting that either of these genetic alterations is suf- Gene translocations. Gene translocation resulting in onco-
ficient to promote thyroid tumorigenesis through the genic rearrangements in thyroid cancer is best exem-
PI3K–AKT pathway. However in aggressive undifferen- plified by RET–PTC. There are more than 10 types of
tiated ATC, PIK3CA mutations and amplifications often RET–PTC translocation, as determined by the types
occur simultaneously in the same tumour 26,33, which is of partner genes, and the most common types are
a mechanism through which mutant PIK3CA can be RET–PTC1 and RET–PTC350–52. RET is a proto-oncogene
amplified to drive the progression and aggressiveness of encoding an RTK. RET–PTC occurs as a consequence
thyroid cancer. of genetic recombination between the 3′ tyrosine kinase

Table 1 | Thyroid tumours and their characteristics


Tumour type Cell of origin Prevalence (% of Standard care and Characteristics
thyroid cancers) prognosis
FTA Follicular thyroid This is a benign Conservative monitoring; Common benign thyroid tumour; similar
cells (which produce lesion thyroidectomy if architecture to FTC, but typically encapsulated;
thyroid hormone symptomatic lacking capsular or vascular invasion; lacking
and thyroglobulin) metastasis; lacking nuclear features of PTC
PTC* Follicular thyroid 80–85 Thyroidectomy and, in Well differentiated, with papillary architecture
cells selected cases, radioiodine and characteristic nuclear features that include
ablation (novel drugs for enlargement, oval shape, elongation, overlapping
resistant disease); good and clearing, inclusions and grooves; propensity
overall prognosis for lymphatic metastasis; PTC subtypes include
conventional PTC (CPTC), follicular-variant PTC
(FVPTC), tall-cell PTC (TCPTC) and a few rare
variants
FTC* Follicular thyroid 10–15 Thyroidectomy and Well differentiated, hypercellular, microfollicular
cells radioiodine ablation (novel patterns, lacking nuclear features of PTC; vascular
drugs for resistant cases); or capsular invasion; propensity for metastasis via
good overall prognosis the blood stream; Hürthle cell thyroid cancer is
a unique subtype of FTC that accounts for 2–3%
of thyroid cancers and is characterized by large,
mitochondria-rich oncocytic cells and dense
nuclei and nucleoli, as well as a high propensity for
metastasis and a poor prognosis
PDTC Follicular thyroid 5–10 Surgery, radioiodine Poorly differentiated, often overlapping with PTC
cells (in selected cases), and FTC; intermediate aggressiveness between
chemotherapy, radiotherapy, differentiated and undifferentiated thyroid
novel drugs; poor prognosis cancers
ATC Follicular thyroid 2–3 Surgery, chemotherapy, Undifferentiated; admixture of spindle,
cells radiotherapy, novel drugs, pleomorphic giant and epithelioid cells; extremely
palliative care; highly and invasive and metastatic; highly lethal; may occur
rapidly lethal de novo or derive from PTC, FTC or PDTC
Medullary thyroid Parafollicular C cells 2–3 Surgery, chemotherapy, Moderate aggressiveness, high propensity for
cancer (which produce radiotherapy, novel drugs (for lymphatic metastasis; RET mutation; occurring in
calcitonin) example, vandetanib) familial, MEN2 or sporadic forms
Primary lymphoma Lymphocytes <1 Chemotherapy Unusual and uncommon type of lymphoma
of the thyroid gland
Metastatic cancer Non-thyroid origin <1 Thyroidectomy in selected Most commonly metastasized from renal and
from other organs cases; treatment of original breast cancers; characteristics of original cancer
cancer
ATC, anaplastic thyroid cancer; FTA, follicular thyroid adenoma; FTC, follicular thyroid cancer; MEN2, multiple endocrine neoplasia type 2; PDTC, poorly
differentiated thyroid cancer; PTC, papillary thyroid cancer. *PTC and FTC are collectively classified as differentiated thyroid cancer (DTC).

186 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Receptor tyrosine kinases portion of RET and the 5′ portion of a partner gene, such variant PTC (FVPTC), but more commonly in classical
(RTKs). These are typically as the coiled-coil domain-containing gene 6 (CCDC6; PTC56. A recent study found a correlation between the
growth-promoting also known as H4) in RET–PTC1 and nuclear recep- presence of RET–PTC and a high growth rate of benign
transmembrane receptors tor co-activator 4 (NCOA4; also known as ELE1) in thyroid tumours57. However, the role of RET–PTC in
that transduce
extracellular signalling into
RET–PTC3 (REFS 53,54). Spatial contiguity of RET and early thyroid tumorigenesis is unclear. RET–PTC is
intracellular signalling through the partner gene in the nucleus is a structural basis a classical oncoprotein that activates the MAPK and
the activation of their for the formation of RET–PTC rearrangements55. The PI3K–AKT pathways58,59. RET–PTC activates both path-
cytoplasmic kinase domain in rearrangements result in ligand-independent dimeri- ways by recruiting signalling adaptors to phosphorylated
response to extracellular
zation and constitutive tyrosine kinase activity of RET. Tyr1062 on the intracellular domain of the RET fusion
signals such as ligand-binding.
RET–PTC can occur in benign FTA and follicular protein60. It is thus not surprising that RET–PTC occurs

Table 2 | Gene mutations in thyroid tumours


Mutations Types of thyroid Approximate Primary signalling pathways Functional impact on the protein Refs
tumours prevalence (%)* affected and tumour
BRAFV600E CPTC 45 MAPK Activating; promoting tumorigenesis, 6–12,
invasion, metastasis, recurrence and 16–18
FVPTC 15 mortality
TCPTC 80–100
ATC 25
BRAFK601E FVPTC 5 MAPK Activating; probably similar to BRAFV600E 14
HRAS, KRAS, FTA 20–25 MAPK and PI3K–AKT Activating; promoting tumorigenesis, 26,31,33,
NRAS invasion and metastasis of PDTC and 35,148,
FTC 30–45 FTC 184–187,
FVPTC 30–45 206–211
PDTC 20–40
ATC 20–30
PTEN (mutation) FTA 0 PI3K–AKT Inactivating the gene but activating the 26,31–33,
PI3K pathway; promoting tumorigenesis 212
FTC 10–15 and invasiveness
ATC 10–20
PTC 1–2
PTEN (deletion) FTC 30 PI3K–AKT Inactivating the gene but activating the 212
PI3K pathway; promoting tumorigenesis
and invasiveness
PIK3CA FTA 0–5 PI3K–AKT Activating; promoting tumorigenesis 25,26,
and invasiveness 31–35
FTC 5–15
ATC 15–25
PTC 1–2
AKT1 Metastatic cancer 15 PI3K–AKT Unclear; seems to favour metastasis 35
CTNNB1 PDTC 25 WNT–β‑catenin Activating; promoting tumour 37,38
progression
ATC 60–65
TP53 PDTC 25 p53‑coupled pathways Inactivating; promoting tumour 39,40
progression
ATC 70–80
IDH1 FTC 5–25 IDH1‑associated metabolic Inactivating; impact on tumours is 41,42
pathways unclear
FVPTC 20
CPTC 10
ATC 10–30
ALK ATC 10 MAPK and PI3K–AKT Activating; probably promoting tumour 43
progression
EGFR CPTC 5 MAPK and PI3K–AKT Activating; impact on tumours is unclear 44
NDUFA13 (also HCTC 15 Component of complex I of the Presumably inactivating; affecting 45
known as GRIM19) mitochondrial respiratory chain mitochondrial metabolism and cell death
ALK, anaplastic lymphoma kinase; ATC, anaplastic thyroid cancer; CPTC, conventional PTC; CTNNB1, β‑catenin; EGFR, epidermal growth factor receptor; FTA,
follicular thyroid adenoma; FTC, follicular thyroid cancer; FVPTC, follicular-variant PTC; HCTC, Hürthle cell thyroid cancer; IDH1, isocitrate dehydrogenase 1;
NDUFA13, NADH dehydrogenase (ubiquinone) 1α subcomplex 13; PDTC, poorly differentiated thyroid cancer; PTC, papillary thyroid cancer; TCPTC, tall-cell PTC.
*The values represent estimated overall prevalence of the indicated mutations.

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 187

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Gene translocation in FTA and FVPTC; these are follicular thyroid tumours hypomethylated genes have important established meta­
A genetic rearrangement in in which the PI3K–AKT pathway has a primary role in bolic and cellular functions. Thus, alterations in gene
which a chromosomal fragment tumorigenesis61. methylation coupled to BRAF‑V600E and probably to
is translocated to another The paired box 8 (PAX8)–peroxisome proliferator- other oncoproteins represent a prominent epigenetic
chromosome where it is not
normally located, which may
activated receptor‑γ (PPARG) fusion gene (PAX8–PPARG) mechanism in thyroid tumorigenesis.
create a recombinant gene is another prominent recombinant oncogene in thyroid Promoter methylation of PTEN is also common in
product with new function or cancer, occurring in up to 60% of FTC and FVPTC62–64. FTC and ATC72–74, which is consistent with the loss of
uncontrolled function PAX8–PPARG also occurs in FTA64, although, like expression of PTEN that is found in a subset of these
(compared with the original
RET–PTC, its prevalence is low in benign thyroid thyroid cancers73,75,76. PTEN methylation is associated
gene product).
tumours and its oncogenic role is unclear. PAX8–PPARγ with genetic alterations of the PI3K–AKT pathway in
Follicular thyroid tumours exerts a dominant-negative effect on the wild-type thyroid tumours, including mutations of various iso-
Collectively includes follicular tumour suppressor PPARγ and also transactivates forms of RAS, PIK3CA mutation and amplification, and
thyroid adenoma (FTA), certain PAX8‑responsive genes 65. Interestingly, the PTEN mutations72. This is consistent with a model in
follicular thyroid cancer (FTC)
and follicular variant papillary
expression of PPARγ was dramatically reduced in FTC which aberrant activation of the PI3K–AKT pathway,
thyroid cancer (FVPTC); they that developed in the TRβPV mouse model — which driven by activating genetic alterations, causes aber-
share common dense follicular expresses a knock‑in dominant-negative mutant of rant methylation and hence silencing of PTEN, which
cell architectures. human thyroid hormone receptor‑β (TRβ) — resulting in turn leads to failure to terminate the PI3K–AKT
in follicular thyroid tumorigenesis66. An AKAP9–BRAF signalling and creates a self-amplifying loop72.
Gene methylation
An epigenetic covalent
fusion gene resulting in activation of the BRAF kinase Despite the identification of these common genetic
addition of a methyl group to also occurs in ionizing-radiation-induced PTC67, but not alterations in thyroid cancers, it is important to note
the fifth carbon of the cytosine in sporadic PTC68. Unlike RET–PTC, the importance of that approximately 30–35% of DTCs do not harbour
residue in a CpG dinucleotide, AKAP9–BRAF in thyroid tumorigenesis is probably known genetic alterations, and so further investigation is
typically in CpG islands (that is,
CpG-rich regions) in the
limited owing to its rarity. required to identify the underlying genetic backgrounds.
5ʹ-flanking promoter regions of
genes. Such methylation is Aberrant gene methylation. Aberrant gene methylation is Altered signalling pathways in thyroid cancer
usually closely associated with an epigenetic hallmark of human cancer — which is also The MAPK signalling pathway. The MAPK pathway has
chromatin remodelling and
common in thyroid cancer 69 — that usually silences a a fundamental role in the regulation of cell proliferation
silencing of the corresponding
genes.
gene when occurring in the promoter regions. BRAFV600E and survival and in human tumorigenesis (FIG. 1). The
mutation was found to be associated with hypermethyla- importance of this pathway has been well established
tion of several tumour suppressor genes, including tis- in thyroid tumorigenesis, particularly for PTC 77,78.
sue inhibitor of metalloproteinases 3 (TIMP3), SLC5A8, In thyroid cancer, the MAPK pathway is driven by activat-
death-associated protein kinase 1 (DAPK1) and retinoic ing mutations, including BRAF and RAS mutations, by
acid receptor‑β (RARB)70. A recent DNA methylation RET–PTC and, in some cases, by the recently discovered
microarray study revealed broad hypermethylation of ALK mutations43.
genes throughout the genome driven by BRAF‑V600E MAPK-mediated thyroid tumorigenesis involves
signalling in PTC cells71. Interestingly, this study also a wide range of secondary molecular alterations that
revealed a large range of genes throughout the genome synergize and amplify the oncogenic activity of this
that, driven by BRAF‑V600E, became hypomethylated pathway, such as genome-wide hypermethylation and
and hence overexpressed. These hypermethylated or hypomethylation71. Additionally, upregulation of various

Table 3 | Copy-number gains in thyroid cancer*


Gene Prevalence in ATC cases (%) Prevalence in FTC cases (%)
EGFR 19/41 (46.3) 19/59 (32.2)
PDGFRA 11/46 (23.9) 4/52 (7.7)
PDGFRB 14/37 (37.8) 8/59 (13.6)
VEGFR1 20/44 (45.5) 26/59 (44.1)
VEGFR2 8/46 (17.4) 2/52 (3.8)
KIT 10/46 (21.7) 6/61 (9.8)
MET 5/42 (11.9) 5/58 (8.6)
PIK3CA 18/47 (38.3) 15/63 (23.8)
PIK3CB 16/42 (38.1) 25/55 (45.5)
PDPK1 (also known as PDK1) 8/40 (20) 14/58 (24.1)
AKT1 9/48 (18.8) 5/61 (8.2)
AKT2 0/44 (0) 13/58 (22.4)
ATC, anaplastic thyroid cancer; EGFR, epidermal growth factor receptor; FTC, follicular thyroid cancer; PDGFR, platelet-derived
growth factor receptor; PDPK1, 3‑phosphoinositide dependent protein kinase 1; VEGFR, vascular endothelial growth factor
receptor. *From REF. 26. Copy-number gains of some of these genes in thyroid cancers were also investigated in the studies
reported in REFS 25,31–33, which showed comparable prevalences.

188 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

well-established oncogenic proteins can occur. These vimentin 87, hypoxia-inducible factor 1α (HIF1α) 88,
include chemokines79,80, vascular endothelial growth fac- prokineticin 1 (PROK1; also known as EG‑VEGF)89,
tor A (VEGFA)81, MET82,83, nuclear factor‑κB (NF‑κB)84, urokinase plasminogen activator (uPA) and its receptor
matrix metalloproteinases (MMPs)79,84,85, prohibitin86, (uPAR)90,91, transforming growth factor‑β1 (TGFβ1)92–94

Cytokines Mitogenic stimuli Apoptotic signal

Cell RTK
membrane

RAS
IKK RASSF1A

BRAF-V600E

IκB NF-κB MST1


MEK Degradation

P
IκB NF-κB ERK FOXO3 14-3-3

• Ubiquitylation P P S207
• Proteasomal ERK FOXO3
degradation

P
P
Nucleus NF-κB ERK FOXO3

↑ Tumour-promoting and ↑ Tumour-promoting ↓ Tumour suppression


anti-apoptotic genes genes (e.g. VEGFA, MET, and thyroid genes ↓ Pro-apoptotic genes
(e.g. VEGFA, vimentin, HIF1A, UPA, UPAR, (e.g. TIMP3, SLC5A8, in FOXO pathway
UPA, UPAR, MMPs) TGFB1, TSP1) DAPK1, NIS, TSHR, TPO)

Thyroid cell growth and proliferation,


tumorigenesis and progression of thyroid cancer

Figure 1 | The MAPK and related pathways in thyroid cancer. Shown in the middle of the figure is the classical MAPK
pathway leading from an extracellular mitogenic stimulus that activates a receptor tyrosine kinaseNature Reviews | Cancer
(RTK) in the cell
membrane, to RAS, RAF (shown as BRAF‑V600E), MEK and ERK. ERK is activated by phosphorylation (P) and enters the
nucleus where it upregulates tumour-promoting genes and downregulates tumour suppressor genes and thyroid
iodide-handling genes. On the left side of the figure is the nuclear factor‑κB (NF‑κB) pathway, in which extracellular stimuli
activate the pathway by acting on receptors in the cell membrane, leading to activation of the inhibitor of κB (IκB) kinase
(IKK), resulting in the phosphorylation of IκB. Phosphorylated IκB becomes dissociated from NF‑κB, which is normally
bound with IκB in a complex and sequestered in the cytoplasm. Phosphorylated IκB undergoes ubiquitylation and
proteasomal degradation. Free NF‑κB then enters the nucleus to promote the expression of tumour-promoting genes.
Through an undefined mechanism that is independent of MEK signalling, BRAF‑V600E promotes the phosphorylation
of IκB and the release of NF‑κB, thus activating the NF‑κB pathway. Shown on the right side of the figure is the
RASSF1–mammalian STE20‑like protein kinase 1 (MST1)–forkhead box O3 (FOXO3) pathway. Activated by extracellular
pro-apoptotic stimuli through membrane receptors, RASSF1A activates MST1. Activated MST1 then phosphorylates
FOXO3 on Ser207. The resulting phosphorylated FOXO3 becomes dissociated from 14‑3‑3 proteins in the cytoplasm.
14‑3‑3 proteins undergo proteasomal degradation, and phosphorylated FOXO3 enters the nucleus to promote the
expression of pro-apoptotic genes in the FOXO pathway. BRAF‑V600E directly interacts with and inhibits MST1 and
prevents its activation by RASSF1A, thereby suppressing the pro-apoptotic signalling of the FOXO3 pathway. The
downward arrow for the FOXO activities shown in the nucleus indicates this negative effect of BRAF‑V600E on
pro-apoptotic genes, which are normally upregulated by the RASSF1A–MST1–FOXO3 pathway. The triple independent
coupling of BRAF‑V600E to the pathways shown here represents a unique and powerful mechanism of thyroid
tumorigenesis driven by BRAF‑V600E. DAPK1, death-associated protein kinase 1; HIF1A, hypoxia-inducible factor 1α;
MMP, matrix metalloproteinase; NIS, sodium–iodide symporter; TGFB1, transforming growth factor β1; TIMP3, tissue
inhibitor of metalloproteinases 3; TPO, thyroid peroxidase; TSHR, thyroid-stimulating hormone receptor; TSP1,
thrombospondin 1; UPA, urokinase plasminogen activator; UPAR, urokinase plasminogen activator receptor; VEGFA,
vascular endothelial growth factor A.

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 189

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

and thrombospondin 1 (TSP1)93. These proteins are all AKT in thyroid cancers, particularly FTC100–102. Among
established players in human tumorigenesis through a the three AKT isoforms, AKT1 and AKT2, but not
variety of mechanisms that drive cancer cell prolifera- AKT3, were found to be robustly expressed and activated
tion, growth, migration and survival, as well as tumour in thyroid cancer, suggesting a particularly important
angiogenesis, invasion and metastasis. role for these two isoforms in thyroid tumorigenesis101.
Many of these proteins are key constituents of the In the TRβPV transgenic mouse model — in which
unique extracellular matrix (ECM) microenvironments the PI3K–AKT pathway is activated, which involves the
that have been proposed to be important in the patho- interaction of TRβPV with the p85α regulatory subu-
genesis of thyroid cancer driven by oncoproteins such nit of PI3K — FTC spontaneously developed103,104, and
as BRAF‑V600E95. It is now clear that the ECM micro- the activation and nuclear localization of AKT1 was
environment does not only function as a structural sup- observed105. Human tumour studies suggested that the
port for the cellular elements of cancer, but it also has invasiveness and metastasis of FTC promoted by the
a profound impact on the behaviours of cancer cells, PI3K–AKT pathway particularly involved the activa-
including their viability, proliferation, adhesion and tion and nuclear localization of AKT1 (REF. 102). This
motility. Thyroid cancer cells and stromal cells, such as is consistent with the occurrence of AKT1 mutations in
fibroblasts and macrophages, produce proteins that form metastatic thyroid cancers35. Furthermore, Akt1 abla-
paracrine and autocrine signalling loops. For example, tion delayed or prevented tumour progression, vascular
BRAF‑V600E‑mediated MAPK pathway activation pro- intravasation and distant metastasis of FTC in TRβPV
motes the release of TSP1 into the ECM where it interacts mice106. The role of AKT2 in thyroid tumorigenesis has
with and modulates other proteins. These include integ- also been demonstrated in vivo, as the occurrence of thy-
rins and non-integrin cell-membrane receptors, matrix roid tumours was reduced in Pten+/−Akt2−/− mice107. Thus,
proteins, cytokines, VEGFA and MMPs, which in turn whereas the MAPK pathway has a central role primarily
activate signalling in thyroid cancer cells and promote in PTC, the PI3K–AKT pathway has such a role primar-
tumour progression and metastasis93,95. Another exam- ily in FTC and its invasion and metastasis. This is consist-
ple is the tumour-promoting cytokine TGFβ1, which ent with genetic alterations in the PI3K–AKT pathway
is released into the ECM owing to the activation of the occurring most frequently in FTC, whereas those in the
BRAF‑V600E–MAPK pathway 92,94. Cytokines can create MAPK pathway occur most frequently in PTC17,18,61,78,98.
an inflammatory microenvironment that may produce The inhibition of thyroid cancer cell proliferation by
reactive oxygen species and oxidative stress, which may PI3K–AKT pathway inhibitors was dependent on the
in turn stimulate the MAPK pathway and augment thy- presence of genetic alterations in the PI3K–AKT path-
roid tumorigenesis96. Thus, secondary molecular altera- way 108,109, suggesting that thyroid cancer cells harbouring
tions in the tumour ECM microenvironment, triggered such genetic alterations have become dependent on the
by the aberrant signalling of the MAPK pathway, have over-activation of this pathway.
an important role in the pathogenesis of thyroid cancer. Vascular invasiveness and capsular invasiveness are
In terms of stromal cells in the tumour micro­ defining histopathological features of FTC110. Genetic
environment, an important role of tumour-associated alterations in the PI3K–AKT pathway are far more
macrophages is that they produce cytokines such as common in FTC than in its precursor, FTA31,33. It is
TGFβ1. The role of stromal fibroblasts in thyroid tumori­ therefore conceivable that overactivated PI3K–AKT
genesis seems to be affected by the expression patterns of signalling promotes the conversion of FTA to FTC by
the two fibroblast growth factor receptor 2 (FGFR2) iso- conferring tumour cell invasiveness. There are also sec-
forms, FGFR2‑IIIb and FGFR2‑IIIc, in cancer cells and ondary molecular alterations, albeit limited in number
fibroblasts: co‑implantation of thyroid cancer cells so far, that are robustly induced by PI3K–AKT signalling
and fibroblasts expressing the same type of FGFR2 and that have an important role in the pathogenesis of
isoform did not affect xenograft tumour progression, FTC; these include alterations of the WNT–β-catenin111,
whereas co‑implantation of thyroid cancer cells and HIF1α112,113, FOXO3 (REF. 114) and NF‑κB114 pathways.
fibroblasts that respectively expressed FGFR2‑IIIb
and FGFR2‑IIIc resulted in increased tumour pro- The NF‑κB signalling pathway. The NF‑κB pathway
gression97. These results highlight the importance and has an important role in the regulation of inflammatory
complexity of cellular and molecular constituents in responses that are linked to tumorigenesis115 (FIG. 1).
determining the impact of the microenvironment on Increased NF‑κB activation in thyroid cancer cell lines
thyroid cancer cell behaviour. and tissues has long been documented116–118. Recent
studies demonstrate that the NF‑κB pathway controls
The PI3K–AKT signalling pathway. The PI3K–AKT proliferative and anti-apoptotic signalling pathways in
pathway also has a fundamental role in thyroid tumori­ thyroid cancer cells119,120. The NF‑κB pathway is involved
Capsular invasiveness genesis61,98 (FIG. 2). Early evidence to support the role of in upregulating the expression of several oncogenic
A phenomenon in which this pathway in thyroid tumorigenesis came from the proteins that are also upregulated by the MAPK path-
thyroid cancer invades the finding that Cowden’s syndrome (which is caused by way. Furthermore, RET–PTC, RAS and BRAF‑V600E
connective tissue capsule that germline mutations of PTEN) was associated with FTA — members of the MAPK pathway — can cause acti-
surrounds the tumour, which is
a defining feature of
and FTC99. A functional role for the PI3K–AKT pathway vation of the NF‑κB pathway in thyroid cancers119.
progression to malignancy in sporadic thyroid tumorigenesis was initially revealed Interestingly, NF‑κB signalling was upregulated by
from a benign thyroid tumour. by the finding of increased expression and activation of expression of BRAF‑V600E121,122, but not by expression

190 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

Extracellular signal of a constitutively active mutant of MEK in NIH3T3


Cell RTK cells123, suggesting a direct coupling of BRAF‑V600E
membrane to the NF‑κB pathway. Through a mechanism that is
not yet specifically defined, BRAF‑V600E was shown
RAS
to cause IκB degradation (and consequent activation of
Genetic alterations NF‑κB) independently of MEK–ERK signalling in thy-
roid cancer cells84. This dual coupling of BRAF‑V600E
PI3K–AKT pathway PI3K to the NF‑κB pathway and to the MEK–ERK pathway
is consistent with the finding that simultaneously sup-
pressing the two pathways using NF‑κB and MEK inhib-
PIP2 PIP3 PDK1 itors synergistically inhibited the proliferation of thyroid
PTEN PTEN P P
silencing methylation AKT 14-3-3 FOXO3 cancer cells harbouring a BRAF‑V600E mutation124.
PTEN
The RASSF1–MST1–FOXO3 signalling pathway.
P P RASSF1 is a member of the RAS association domain
GSK3β AKT mTOR 14-3-3
family, which, in response to apoptotic signalling, asso-
ciates with and activates mammalian STE20‑like pro-
Degradation β-catenin Translation tein kinase 1 (MST1; also known as STK4)125. Activated
MST1 phosphorylates Ser207 in the forkhead domain
of the forkhead transcription factor FOXO3. This phos-
Nucleus
P phorylation disrupts the interaction of FOXO3 with
FOXO3 (on AKT
P motif) 14‑3‑3 proteins in the cytoplasm and promotes FOXO3
β-catenin AKT translocation to the nucleus, where it promotes the tran-
FOXO3 scription of pro-apoptotic genes126 (FIG. 1). Thus, this
RASSF1–MST1–FOXO3 pathway has an important
tumour-suppressive role by promoting apoptosis.
Hypermethylation of the promoter of RASSF1A is
common and is associated with its silencing in thyroid
↑ Tumour-promoting ↑ Tumour-promoting ↓ Pro-apoptotic genes
genes genes in FOXO pathway cancers127,128. This occurs even in benign FTA, albeit
to a lesser extent, suggesting that impairment of the
RASSF1–MST1–FOXO3 pathway is involved in early
thyroid tumorigenesis128.
Thyroid cell growth and proliferation,
tumorigenesis and progression of thyroid cancer Activation of the RASSF1–MST1–FOXO3 signal-
ling pathway promoted thyroid cancer cell apoptosis21.
Interestingly, this study demonstrated that BRAF‑V600E,
Figure 2 | The PI3K–AKT and related pathways in thyroid cancer. Extracellular
Nature Reviews | Cancer but not wild-type BRAF, directly interacted with the car-
signals activate receptor tyrosine kinases (RTKs) in the cell membrane, leading to the
activation of RAS and subsequent activation of PI3K. Activated PI3K catalyses the boxyl terminus of MST1 and inhibited its kinase activities,
conversion of phosphatidylinositol (4,5)-bisphosphate (PIP2) to phosphatidylinositol resulting in decreased FOXO3 transactivation. This was
(3,4,5)-trisphosphate (PIP3). PIP3 activates 3‑phosphoinositide-dependent protein independent of MEK–MAPK signalling. This suggests
kinase 1 (PDK1; also known as PDPK1), which consequently associates with AKT that, in addition to the classical coupling to MEK–MAPK,
and leads to phosphorylation (P) and activation of AKT by PDK1. Phosphorylated negative regulation of RASSF1–MST1–FOXO3 is a
AKT, which is an activated form of AKT, enters the nucleus where it induces mechanism that is involved in BRAF‑V600E‑driven
tumour-promoting genes. In the cytoplasm, phospho-AKT also activates other thyroid tumorigenesis. This unique mechanistic model
signalling molecules or pathways, a prominent one being the mTOR pathway, which explains the finding that thyroid cancers that developed
has an important role in tumorigenesis by promoting translation. Phospho-AKT can in BRAFV600EMst1−/− transgenic mice were more aggres-
also directly phosphorylate glycogen synthase kinase 3β (GSK3β) and consequently
sive than those that developed in BRAFV600E mice21. The
inactivates it. GSK3β normally inhibits β‑catenin, thus the effect of phospho-AKT is to
relieve GSK3β‑mediated suppression of β‑catenin. Consequently, β‑catenin can enter direct interaction of BRAF‑V600E with MST1 to pre-
the nucleus where it promotes the expression of tumour-promoting genes. In the vent RASSF1‑mediated MST1 activation also provides
nucleus, phospho-AKT can phosphorylate forkhead box O3 (FOXO3) on its an explanation for the mutually exclusive occurrence
AKT-specific motif. Such phosphorylated FOXO3 is translocated out of the nucleus to of BRAFV600E mutation and RASSF1A hypermethylation
the cytoplasm where it binds 14‑3‑3 proteins to be sequestered in the cytoplasm, thus in thyroid cancer 128, as either event may be sufficient to
terminating the pro-apoptotic activities of the FOXO3 pathway. The downward arrow inactivate RASSF1–MST1–FOXO3 signalling. Therefore,
for the FOXO activities in the nucleus in the figure indicates this negative effect of BRAF‑V600E is coupled independently to three major
AKT on pro-apoptotic genes in the FOXO pathway, which would otherwise be pathways: MEK–MAPK, RASSF1–MST1–FOXO3 and
upregulated by the FOXO3 pathway. This unique coupling of phospho-AKT to the NF‑κB, and thus represents a unique and powerful
three pathways provides a powerful driving force for thyroid tumorigenesis. The
oncogenic mechanism in thyroid tumorigenesis (FIG. 1).
major negative regulatory mechanism of the PI3K–AKT pathway is PTEN, which is a
phosphatase that converts PIP3 to PIP2, thus terminating the activation of the The activation of the PI3K–AKT pathway, such as that
pathway. The inset shows the self enhancement mechanism of PI3K–AKT signalling in induced by PTEN inactivation, can also downregulate
which genetic-alteration-driven activation of the pathway causes PTEN methylation the activities of the RASSF1–MST1–FOXO3 pathway in
and silencing with consequent loss of termination of the signalling, thus maintaining FTC114. This involves AKT-mediated phosphorylation of
the pathway in full and constitutive activation. FOXO family members, which causes their translocation

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 191

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

from the nucleus to the cytoplasm, where they are seques- G11-mediated phospholipase Cβ–inositol 1,4,5‑trispho-
tered by 14‑3‑3 proteins, thus reducing the transcription sphate–intracellular Ca2+ signalling. FTC spontaneously
of pro-apoptotic genes129 (FIG. 2). developed in TRβPV mice, but when these mice were
crossed with Tshr−/− mice, no thyroid cancer devel-
The WNT–β‑catenin signalling pathway. The WNT– oped138. This study thus elegantly demonstrates the
β‑catenin pathway has a well established role in the regula- requirement of TSHR signalling in thyroid carcinogen-
tion of cell growth and proliferation, as well as in stem cell esis in this mouse model. Similar findings were reported
differentiation, and its constitutive activation is commonly in another mouse model in which thyroid-specific
found in human cancers130. β‑catenin, when upregulated knock‑in of BrafV600E (LSL-BrafV600Ethyroid peroxidase
by upstream WNT signalling, is translocated into the (Tpo)-Cre) caused the development of aggressive PTC,
nucleus and transcribes various tumour-promoting genes. and crossing these mice with Tshr−/− mice resulted in a
In thyroid cancer, activation of WNT–β‑catenin signal- failure to develop thyroid cancer 139. In this study, thy-
ling is often caused by activating mutations of CTNNB1 roid-specific deletion of Gnas (which encodes Gsα) in
(which encodes β‑catenin), particularly in PDTC and LSL-BrafV600ETpo-Cre mice attenuated thyroid tumour
ATC37,38. Furthermore, the expression of β‑catenin was formation. Interestingly, there is a clinical association
higher in ATC than in DTC131. Thus, the WNT–β‑catenin of higher serum levels of TSH with a higher risk for
pathway seems to have a particularly important role in malignancy of thyroid nodules in humans140–142.
thyroid tumour aggressiveness. However, it is not clear whether TSHR signalling
Aberrant activation of WNT–β‑catenin signalling is required directly for the initiation of thyroid cancer
often occurs as a consequence of the activation of the or whether it is simply required, as would be expected
PI3K–AKT pathway in thyroid cancers132,133. This occurs physiologically, for the TSHR-dependent generation of
through glycogen synthase kinase 3β (GSK3β), which thyroid cells, from which an oncogene-driven thyroid
is directly phosphorylated — and hence inactivated cancer would originate. Overactivation of TSHR sig-
— by AKT134. As GSK3β promotes the degradation of nalling, such as that achieved through activating muta-
β‑catenin, its inactivation results in the upregulation tions in TSHR or Gsα, is well known to cause benign
of WNT–β‑catenin signalling (FIG.  2). Interestingly, hyperfunctional FTA143,144. These tumours are almost
RET–PTC can activate the WNT–β‑catenin pathway by never malignant, which suggests that TSHR signal-
activating the PI3K–AKT pathway and also by directly ling may be protective against malignant transforma-
phosphorylating β‑catenin in thyroid cancer cells58. tion of thyroid cells. Consistently, low serum levels of
TSH are associated with common genetic variants that
The  HIF1α pathway. It has long been known that predispose to an increased risk of thyroid cancer 145,146.
hypoxia is a strong stimulus of cancer metabolism, It therefore seems that the TSH–TSHR system has a
growth and progression. HIF1α is a key mediator of dichotomous role in the development of thyroid can-
the response to hypoxia, in which it binds to HIF1β cer: it may suppress malignant transformation of thy-
(also known as ARNT) to form the HIF1 transcription roid cells and hence suppress the occurrence of thyroid
factor that induces the expression of various genes asso- cancer, but it may promote the growth and progression
ciated with cell metabolism and tumour angiogenesis135. of thyroid cancer once it has been initiated by oncogenic
Angiogenesis, which is a key step in the progression of alterations.
solid tumours, is a common response to intratumoural
hypoxia. This process is promoted by VEGFA, the Progressive molecular alterations
expression of which is strongly upregulated by HIF1. Progressive accumulation of genetic alterations. The
HIF1α is not expressed in normal thyroid tissues but accumulation of genetic alterations during thyroid
is expressed in thyroid cancers, particularly in aggres- tumour progression is probably best exemplified by
sive types, such as ATC, and this is consistent with a genetic alterations to members of the PI3K–AKT path-
role in thyroid cancer progression112,136. The oncogene way 61. Mutations in members of this pathway, including
MET, which is another target of HIF1, is also abun- those in RAS (particularly NRAS), PIK3CA and PTEN, as
dantly expressed in association with upregulated HIF1α well as PIK3CA amplifications, increasingly occur from
in thyroid cancer 136. Interestingly, HIF1 can be upregu- FTA to FTC and to ATC26,31–33,147. Hypermethylation
lated in thyroid cancer by both the PI3K–AKT112,113 and of the PTEN promoter also progressively increases in
the MAPK pathways88, thus contributing to the effects frequency from FTA to FTC and to ATC72. The coex-
of these two pathways on thyroid tumour progression. istence of these genetic and epigenetic alterations also
increasingly occurs from low-grade to high-grade thy-
The thyroid-stimulating hormone receptor signalling roid tumours31,33. Genetic or epigenetic defects of PTEN
pathway. Through activation by thyroid-stimulating can occur simultaneously with activating mutations of
hormone (TSH), the TSH receptor (TSHR) has a funda- other genes in the PI3K–AKT pathway; in such cases,
mental role in the regulation of thyroid cell proliferation, the PI3K–AKT pathway can presumably remain maxi-
differentiation and function, as well as in the development mally activated26,33,72. Indeed, deletion of Pten accelerated
of the thyroid gland137. TSHR is a guanine-nucleotide- the progression of FTC in TRβPV mice114.
binding G-protein-coupled receptor that triggers two The coexistence of multiple genetic alterations to
intracellular signalling pathways: Gsα-mediated adenylyl members of the MAPK pathway also occurs, as exem-
cyclase–cyclic AMP (cAMP) signalling and the Gq- or plified by the simultaneous presence of BRAF V600E

192 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

which is a process that may be further accelerated by


MAPK pathway ↑ PTC MAPK pathway ↑↑ additional or secondary genetic alterations, including
(e.g. via BRAFV600E)
mutations in TP53, CTNNB1 and ALK.
MAPK This model is supported by studies of transgenic mice
Follicular
MAPK pathway ↑↑ pathway ↑↑↑ in which the deletion of Pten and knock‑in of KrasG12D (to
thyroid cell PDTC ATC activate both MAPK and PI3K–AKT pathways), but not
PI3K–AKT ↑↑ PI3K–AKT ↑↑↑ either genetic manipulation alone, caused the develop-
PI3K–AKT ↑ ment of aggressive thyroid cancer 29. Similarly, this phe-
(e.g. via RAS, PI3K–AKT ↑↑↑ nomenon is observed in melanoma. Transgenic mice with
PTEN, PIK3CA induced expression of BRAFV600E in melanocytes to acti-
mutations) FTA FTC
vate the MAPK pathway alone developed only melano­
PI3K–AKT ↑↑ cytic hyperplasias, but the expression of BRAFV600E
Figure 3 | Model of the progression of thyroid tumorigenesis driven by the MAPK and with Pten deletion (to also activate the PI3K–AKT
PI3K–AKT pathways. Activation of the MAPK pathway by genetic alterations, such as the pathway) caused melanoma with a rapid onset of
Nature Reviews | Cancer
BRAFV600E mutation, primarily drives the development of papillary thyroid cancer (PTC) metastasis150. Thus, dual activation of the MAPK and
from follicular thyroid cells. By contrast, activation of the PI3K–AKT pathway by genetic PI3K–AKT pathways may be a common mechanism
alterations, such as mutations in RAS, PTEN and PIK3CA, primarily drives the development for promoting tumour progression.
of follicular thyroid adenoma (FTA) and follicular thyroid cancer (FTC) from follicular
thyroid cells. Conversion from FTA to FTC is largely due to increasing activation of the Impairment of the iodide-handling machinery
PI3K–AKT pathway. As genetic alterations accumulate and intensify the signalling of The main and unique function of follicular thy-
each of the two pathways, PTC and FTC can progress to poorly differentiated thyroid
roid cells is to use iodide to synthesize thyroid hor-
cancer (PDTC). When both pathways are fully activated through accumulated genetic
alterations, conversion from PDTC to anaplastic thyroid cancer (ATC) is strongly mone151,152 (FIG. 4). In this process, iodide is transported
facilitated. It is also possible that PDTC and ATC can both develop de novo directly from into the cell through the sodium–iodide symporter
follicular thyroid cells, and that ATC can develop from PTC or FTC if appropriate genetic (NIS) that is located in the basal membrane. At the
alterations occur. Many secondary molecular alterations also progressively accumulate apical membrane, pendrin transports iodide out of
and synergize with the two pathways in driving the progression of thyroid tumorigenesis, the cell into the lumen of the thyroid follicle. In the
as discussed in the text (not shown in the figure). The increasing number of vertical lumen of the thyroid follicle, iodide undergoes oxida-
arrows and colour intensity of the ovals symbolize the increasing genetic alterations and tion by TPO and is incorporated into tyrosine residues
signalling of the two pathways as thyroid tumorigenesis progresses. The figure is of thyroglobulin (TG), which is later cleaved through
modified from REF. 33 © (2007) American Association for Cancer Research. proteolysis to produce thyroid hormones. This process
is upregulated by TSH-mediated activation of TSHR.
mutation, RAS mutations and RET–PTC in aggres- This is the biological basis for the conventional radio­
sive recurrent PTC and ATC26,148,149; these alterations iodine treatment of thyroid cancer, but the iodide-
are otherwise mutually exclusive in well-differentiated handling machinery is often impaired, particularly in
thyroid cancers7,9. It is thus compelling to propose that advanced thyroid cancers, thus making radioiodine
the process of thyroid cancer progression is one of treatment ineffective.
progressive accumulation of multiple genetic altera- Aberrant activation of the MAPK pathway has a
tions, which synergistically cooperate to amplify their crucial role in the impairment of the iodide-handling
oncogenicity. machinery 17. BRAFV600E mutation is associated with the
loss of radioiodine avidity and with radioiodine treat-
Cooperation of the MAPK and PI3K–AKT pathways. ment failure in PTC16,35,153–155. Consistently, BRAFV600E
The MAPK and PI3K–AKT pathways are primarily mutation is highly prevalent (78–95% of cases) in
involved in differentiated PTC and FTC, respectively, recurrent radioiodine-refractory PTC16,35,149,155, in con-
and the simultaneous activation of both pathways trast with the lower prevalence of BRAFV600E mutation
becomes more frequent as the grade of thyroid tumours (45% of cases) in primary PTC12. Numerous studies
increases26,31–33,35. One study analysed 24 genetic altera- have reported an association of BRAFV600E mutation
tions in the major genes of the MAPK and PI3K–AKT with decreased or absent expression of thyroid iodide-
pathways in 48 ATC samples and found that the majority handling genes: NIS, TSHR, TPO, TG and SLC26A4
(81%) of the samples harboured genetic alterations that (which encodes pendrin) in thyroid cancer 82,153,154,156–160.
could potentially activate both pathways26. The coexist- A direct role of BRAFV600E in the downregulation of
ence of phosphorylated ERK and AKT was also com- thyroid hormone synthesis pathway genes was dem-
mon in ATC but not in DTC26. Thus, genetic alterations onstrated by showing that induced expression of
that activate the MAPK and PI3K–AKT pathways are an BRAF‑V600E in thyroid cells impaired the expression
important mechanism that drives the progression of thy- of almost all of these genes, which was restored by ceas-
roid cancer. In this model, as illustrated in FIG. 3, when a ing the expression of BRAF‑V600E or by suppressing
genetic alteration occurs in the MAPK pathway it drives the MAPK pathway with a MEK inhibitor 161. Similarly,
tumorigenesis of the thyroid cell towards PTC; when a in thyroid cells expressing RET–PTC1, NIS expression
genetic alteration occurs in the PI3K–AKT pathway it was increased following treatment with a MEK inhibi-
drives tumorigenesis towards FTA and FTC. As genetic tor 162. Suppression of BRAF‑V600E in mouse models
alterations accumulate and both pathways become of thyroid cancer also restored the expression of thy-
activated, the tumour progresses into PDTC and ATC, roid iodide-handling genes and radioiodine uptake163.

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 193

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

a Follicular thyroid cell b BRAF-V600E+ thyroid tumour cell


Basal Apical Follicular Basal Apical Follicular
lumen lumen
2K+ 2K+
ATPase I– I–
I– ATPase
3Na+ 3Na+
I– I– I– +TG
I– I– Pendrin TG-I
NIS I–
Na+ Na+ I– I– +TG TG-I
I–
TG-I BRAF-V600E
TSH TSHR cAMP
I– I– +TG
I– I– TPO TG-I
I– I–
NIS Pendrin TPO TG-I
Na+ Na+ TPO
I– I– I– +TG TG-I
TPO BRAF-
TSH TSHR cAMP TPO TG-I I– I–
I– V600E NIS I– I– +TG TPO
I– I– I– I– +TG TPO
TG-I Na+ Na+ Pendrin TG-I
NIS I– I– Pendrin TPO
Na+ Na+ TPO TG-I I– I– +TG TPO
I– I– +TG TSH TSHR cAMP
TSH TSHR cAMP I– TPO TG-I
I– I– I– I– +TG TPO TG-I
NIS I–
Pendrin TG-I
Na+ Na+
I– I– I– +TG TG-I
TSH TSHR cAMP TG-I
I– I– BRAF-V600E
I– I– I– I– +TG TG-I
NIS Pendrin
Na+ Na+ I–
I– I– +TG
I– 2K+
2K+ ATPase
ATPase I– 3Na+
3Na+

Figure 4 | Iodide-handling machinery in the thyroid cell and its silencing by BRAF‑V600E. a |Nature Reviews | Cancer
A follicular thyroid cell is
shown, which abundantly expresses the molecules involved in the uptake and metabolism of iodide (I−), including the
sodium–iodide symporter (NIS) in the basal membrane, which transports I− coupled with Na+ into the cell from the
extracellular compartment. I− is then transported into the follicular lumen via pendrin in the apical membrane where it is
oxidized by thyroid peroxidase (TPO) and incorporated into tyrosine amino-acid residues in thyroglobulin (TG) to form
iodinated TG (TG‑I) for the synthesis of thyroid hormone. The whole process is upregulated by cyclic AMP (cAMP)
signalling that is triggered by the binding of thyroid-stimulating hormone (TSH) to its receptor (TSHR) in the membrane.
With normal expression and function of this system, I− is abundantly taken up and accumulated in the follicular thyroid cell
and in the follicular lumen. b | BRAF‑V600E, through activating the MAPK pathway in thyroid cancer, causes the silencing
of thyroid-specific genes and shuts off the iodide-handling machinery. Consequently, I− uptake is reduced in the thyroid
cell and is sparsely accumulated in the follicular lumen. For simplicity, much molecular detail is omitted.

This aberrant gene silencing by BRAF‑V600E may Translational promises in thyroid cancer
involve alterations to histone acetylation at gene pro- Remarkable advances in the translation of molecular
moters 164,165. An autocrine loop involving TGFβ is findings in thyroid cancer to the clinic have occurred
another mechanism 92: BRAF‑V600E promotes the recently 170. For example, because of its cancer specific-
secretion of TGFβ, which, through the activation of ity, the detection of BRAFV600E mutation in fine-needle
SMADs and consequent impairment of the thyroid- aspiration biopsy (FNAB) samples was initially tested
gene transcription factor PAX8, is a potent repressor for the evaluation of thyroid nodules171,172, which has
of NIS in thyroid cells166. now found increasing clinical use170,173. The diagnos-
Activation of the PI3K–AKT pathway was also tic utility of detecting RET–PTC using FNAB has also
shown to downregulate the iodide-handling machinery been intensively investigated170. The combinatorial use
in thyroid cells both in vitro and in vivo167,168. Inhibition of genetic markers identified in FNAB samples, includ-
of the PI3K–AKT pathway induced NIS expression ing BRAF mutation, RAS mutations, RET–PTC and
and iodide uptake in human thyroid cancer cells164,169. PAX8–PPARG, has been shown to improve the diag-
In addition to NIS, suppression of the PI3K–AKT path- nostic accuracy for thyroid nodules that are otherwise
way also induced the expression of TSHR, TPO and diagnostically indeterminate by conventional cytology
TG in human thyroid cancer cells, which was enhanced assessment, although this diagnostic approach may need
by treatment with histone deacetylase inhibitors, and further improvement 174,175. Applying genomic informa-
radioiodine uptake was correspondingly robustly tion to address clinical issues of thyroid neoplasia has
induced164. This could be augmented by co‑treatment also shown promise. For example, a gene-expression
with TSH 164. The involvement of both MAPK and classifier (with high sensitivity and negative predictive
PI3K–AKT pathways in the silencing of thyroid iodide- power) is being used in the clinic to assist the diagnostic
handling genes is consistent with the accumulation of evaluation of indeterminate thyroid nodules176.
genetic alterations in both pathways as thyroid tumours The prognostic application of BRAFV600E mutation
progress, during which there is an increasing loss of has also become part of the clinical management of
radioiodine avidity. PTC170,173. A large, multicentre study demonstrated a

194 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

strong association of BRAFV600E mutation with patient pathway significantly increased the response rate of
mortality from PTC177. Large studies have demonstrated breast and gynaecological cancers to inhibitors of the
that BRAFV600E is strongly associated with poor clinico- PI3K–AKT pathway 203.
pathological outcomes even in conventionally low-risk The involvement of multiple signalling pathways in
patients16,178–181. BRAFV600E detected in FNAB samples aggressive thyroid cancer suggests that it may be neces-
preoperatively predicted poor clinicopathological out- sary to target them simultaneously for effective treat-
comes of PTC180,182,183 and even predicted lymph node ment. Indeed, single agent clinical trials of various kinase
metastasis (as determined by prophylactic central neck inhibitors in thyroid cancer have generally shown only
dissection in patients without preoperative evidence of partial responses. Several recent preclinical studies test-
lymph node metastasis)183. It is thus recommended that ing the combination of MEK or BRAF‑V600E inhibitors
preoperative testing of BRAFV600E in FNAB samples be with PI3K, AKT or mTOR inhibitors showed synergistic
used for assisting risk stratification and defining surgical effects in inhibiting the proliferation and promoting apop­
and medical treatments for patients with PTC170. Owing tosis of thyroid cancer cells124,199,204,205. Synergistic effects
to their association with a worse prognosis of PDTC, of simultaneously targeting the MAPK and PI3K–AKT
RAS mutations are also promising prognostic markers pathways were even more pronounced in cells that har-
for this type of thyroid cancer 184–186. It has been recently boured genetic alterations in both pathways199. It has been
shown that RAS mutations are also associated with poor recently demonstrated that simultaneously inhibiting the
clinicopathological outcomes of FTC187 but this needs to MAPK, PI3K–AKT and histone deacetylase pathways
be evaluated further. could more robustly induce the expression of thyroid
Many components of the MAPK and PI3K–AKT iodide-handling genes (and thus increase radioiodine
pathways, from RTKs in the cell membrane to the uptake) in thyroid cancer cells compared with inhibit-
various downstream signalling relay molecules, such ing each pathway alone, and this could be enhanced by
as BRAF, MEK, AKT and mTOR, are therapeutic tar- co‑treatment with TSH164. Therefore, to restore radioio-
gets that are being actively tested for the treatment of dine avidity by simultaneously targeting multiple signal-
thyroid cancer 170. Novel small-molecule protein-kinase ling pathways is another promising area of translational
inhibitors have shown promise in clinical trials on research on thyroid cancer.
thyroid cancer, including axitinib188, sorafenib189–191,
motesanib192 and pazopanib193. A promising therapeutic Concluding remarks
strategy is genetic-based targeting of thyroid cancer 194, Remarkable progress in understanding the molecular
as supported by many preclinical studies demonstrat- pathogenesis of thyroid cancer has been made in recent
ing the selective inhibition of BRAFV600E-mutant thy- years, as exemplified by the elucidation of the funda-
roid cancer cells by various MEK inhibitors 124,195–199 mental role of signalling pathways, such as the MAPK
and BRAF‑V600E‑specific inhibitors200,201. The lat- and PI3K–AKT pathways. Activation of these pathways,
ter include PLX4032 (also known as vemurafenib), often in close connection and cooperation, constitute
which has been recently approved by the US Food the primary oncogenic mechanism that promotes the
and Drug Administration (FDA) for the treatment of develop­ment and progression of thyroid cancer. Central
BRAFV600E-positive melanoma202. Some of these MEK to this mechanism are the many powerful oncogenic
and BRAF‑V600E inhibitors are currently being clini- alterations that drive these pathways. Important sec-
cally tested for thyroid cancer. Therapeutic targeting of ondary molecular derangements driven by the overac-
the PI3K–AKT pathway may also be genetically guided, tivation of these pathways have also been increasingly
as genetic alterations that activate this pathway confer uncovered, which synergize and amplify oncogenic sig-
thyroid cancer cells with remarkably increased sensitivi- nalling in thyroid tumorigenesis. This understanding of
ties to AKT and mTOR inhibitors108,109. This therapeutic the molecular pathogenesis of thyroid cancer has now
strategy for thyroid cancer may prove to be useful and opened unprecedented opportunities for the develop-
should be tested clinically, as encouraged by a recent ment of novel clinical strategies for the management of
clinical trial showing that mutations in the PI3K–AKT thyroid cancer.

1. Jemal, A. et al. Global cancer statistics. CA Cancer 6. Cohen, Y. et al. BRAF mutation in papillary thyroid References 7 and 9 demonstrate a mutual
J. Clin. 61, 69–90 (2011). carcinoma. J. Natl Cancer Inst. 95, 625–627 (2003). exclusivity between BRAF, RAS and RET–PTC
2. Howlader, N. et al. SEER Cancer Statistics Review This is the first submitted and accepted study that mutations, thus suggesting an independent role of
1975–2009 (Vintage 2009 Populations). National reported a high prevalence of BRAFV600E mutation in BRAF mutation in activating the MAPK pathway.
Cancer Institute [online], http://seer.cancer.gov/ thyroid cancer, which was awarded a United States 10. Xu, X., Quiros, R. M., Gattuso, P., Ain, K. B. &
csr/1975_2009_pops09 (2012). patent. Prinz, R. A. High prevalence of BRAF gene mutation in
3. Tuttle, R. M. et al. Thyroid carcinoma. J. Natl Compr. 7. Soares, P. et al. BRAF mutations and RET/PTC papillary thyroid carcinomas and thyroid tumor cell
Canc. Netw. 8, 1228–1274 (2010). rearrangements are alternative events in the lines. Cancer Res. 63, 4561–4567 (2003).
4. DeLellis, R. A., Lloyd, R. V., Heitz, P. U. & Eng, etiopathogenesis of PTC. Oncogene 22, 4578–4580 11. Fukushima, T. et al. BRAF mutations in papillary
C. World Health Organization Classification of (2003). carcinomas of the thyroid. Oncogene 22, 6455–6457
Tumours. Pathology And Genetics Of Tumors Of 8. Namba, H. et al. Clinical implication of hot spot BRAF (2003).
Endocrine Organs (IARC Press, 2004). mutation, V599E, in papillary thyroid cancers. J. Clin. 12. Xing, M. BRAF mutation in thyroid cancer. Endocr.
This book describes the most recent version of World Endocrinol. Metab. 88, 4393–4397 (2003). Relat. Cancer 12, 245–262 (2005).
Health Organization classification of thyroid tumours. 9. Kimura, E. T. et al. High prevalence of BRAF mutations This is the first comprehensive review of BRAF
5. Hofstra, R. M. et al. A mutation in the RET proto- in thyroid cancer: genetic evidence for constitutive mutation in thyroid cancer that provides a good
oncogene associated with multiple endocrine activation of the RET/PTC-RAS-BRAF signaling summary and analysis of the prevalence and other
neoplasia type 2B and sporadic medullary thyroid pathway in papillary thyroid carcinoma. Cancer Res. characteristics of this mutation in various types of
carcinoma. Nature 367, 375–376 (1994). 63, 1454–1457 (2003). thyroid cancer.

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 195

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

13. Hou, P., Liu, D. & Xing, M. Functional characterization 29. Miller, K. A. et al. Oncogenic Kras requires 49. White, C. D., Brown, M. D. & Sacks, D. B. IQGAPs in
of the T1799‑1801del and A1799‑1816ins BRAF simultaneous PI3K signaling to induce ERK activation cancer: a family of scaffold proteins underlying
mutations in papillary thyroid cancer. Cell Cycle 6, and transform thyroid epithelial cells in vivo. Cancer tumorigenesis. FEBS Lett. 583, 1817–1824
377–379 (2007). Res. 69, 3689–3694 (2009). (2009).
14. Trovisco, V. et al. Type and prevalence of BRAF 30. Gustafson, S., Zbuk, K. M., Scacheri, C. & Eng, C. 50. Rabes, H. M. et al. Pattern of radiation-induced RET
mutations are closely associated with papillary thyroid Cowden syndrome. Semin. Oncol. 34, 428–434 and NTRK1 rearrangements in 191 post-chernobyl
carcinoma histotype and patients’ age but not with (2007). papillary thyroid carcinomas: biological, phenotypic,
tumour aggressiveness. Virchows Arch. 446, 31. Wang, Y. et al. High prevalence and mutual exclusivity and clinical implications. Clin. Cancer Res. 6,
589–595 (2005). of genetic alterations in the phosphatidylinositol- 1093–1103 (2000).
15. Liu, D., Liu, Z., Condouris, S. & Xing, M. BRAF V600E 3‑kinase/akt pathway in thyroid tumors. J. Clin. 51. Santoro, M. et al. Gene rearrangement and Chernobyl
maintains proliferation, transformation, and Endocrinol. Metab. 92, 2387–2390 (2007). related thyroid cancers. Br. J. Cancer 82, 315–322
tumorigenicity of BRAF-mutant papillary thyroid 32. Santarpia, L., El Naggar, A. K., Cote, G. J., Myers, J. N. (2000).
cancer cells. J. Clin. Endocrinol. Metab. 92, & Sherman, S. I. Phosphatidylinositol 3‑kinase/akt and 52. Klugbauer, S., Demidchik, E. P., Lengfelder, E. &
2264–2271 (2007). ras/raf-mitogen-activated protein kinase pathway Rabes, H. M. Molecular analysis of new subtypes of
16. Xing, M. et al. BRAF mutation predicts a poorer mutations in anaplastic thyroid cancer. J. Clin. ELE/RET rearrangements, their reciprocal transcripts
clinical prognosis for papillary thyroid cancer. J. Clin. Endocrinol. Metab. 93, 278–284 (2008). and breakpoints in papillary thyroid carcinomas of
Endocrinol. Metab. 90, 6373–6379 (2005). 33. Hou, P. et al. Genetic alterations and their relationship children after Chernobyl. Oncogene 16, 671–675
This is the first comprehensive multicentre study in the phosphatidylinositol 3‑kinase/Akt pathway in (1998).
that shows the association of BRAF mutation with thyroid cancer. Clin. Cancer Res. 13, 1161–1170 53. Ciampi, R. & Nikiforov, Y. E. RET/PTC rearrangements
aggressive behaviours of PTC, including aggressive (2007). & BRAF mutations in thyroid tumorigenesis.
pathological features, disease recurrence, loss of This study demonstrates collectively common Endocrinology 148, 936–941 (2007).
radioiodine avidity of the tumour and treatment genetic alterations in the PI3K–AKT pathway in 54. Santoro, M., Melillo, R. M. & Fusco, A. RET/PTC
failure. thyroid cancer, thus providing a strong genetic activation in papillary thyroid carcinoma: European
17. Xing, M. BRAF mutation in papillary thyroid cancer: basis for the extensive role of this pathway in Journal of Endocrinology Prize Lecture. Eur.
pathogenic role, molecular bases, and clinical thyroid tumorigenesis. J. Endocrinol. 155, 645–653 (2006).
implications. Endocr. Rev. 28, 742–762 (2007). 34. Garcia-Rostan, G. et al. Mutation of the PIK3CA gene 55. Nikiforova, M. N. et al. Proximity of chromosomal loci
This is a large meta-analysis on the role of BRAF in anaplastic thyroid cancer. Cancer Res. 65, that participate in radiation-induced rearrangements
mutation in the aggressiveness of PTC. 10199–10207 (2005). in human cells. Science 290, 138–141 (2000).
18. Kim, T. H. et al. The association of the BRAF(V600E) This is the first study that demonstrates common This study demonstrates the proximity of
mutation with prognostic factors and poor clinical PIK3CA mutations in ATC. chromosomal loci that are involved in the formation
outcome in papillary thyroid cancer: a meta-analysis. 35. Ricarte-Filho, J. C. et al. Mutational profile of of RET–PTC translocations, thus providing a
Cancer 118, 1764–1773 (2012). advanced primary and metastatic radioactive iodine- structural basis and explanation for the formation
This is a large meta-analysis on the relationship of refractory thyroid cancers reveals distinct of genetic rearrangements.
BRAF mutation with clinicopathological behaviours pathogenetic roles for BRAF, PIK3CA, and AKT1. 56. Marotta, V., Guerra, A., Sapio, M. R. & Vitale, M. RET/
of PTC. Cancer Res. 69, 4885–4893 (2009). PTC rearrangement in benign and malignant thyroid
19. Charles, R. P., Iezza, G., Amendola, E., Dankort, D. & 36. Samuels, Y. et al. High frequency of mutations of the diseases: a clinical standpoint. Eur. J. Endocrinol. 165,
McMahon, M. Mutationally activated BRAF(V600E) PIK3CA gene in human cancers. Science 304, 554 499–507 (2011).
elicits papillary thyroid cancer in the adult mouse. (2004). 57. Sapio, M. R. et al. High growth rate of benign thyroid
Cancer Res. 71, 3863–3871 (2011). 37. Garcia-Rostan, G. et al. Frequent mutation and nodules bearing RET/PTC rearrangements. J. Clin.
20. Knauf, J. A. et al. Targeted expression of BRAFV600E nuclear localization of β-catenin in anaplastic thyroid Endocrinol. Metab. 96, e916–e919 (2011).
in thyroid cells of transgenic mice results in papillary carcinoma. Cancer Res. 59, 1811–1815 (1999). 58. Castellone, M. D. et al. The β-catenin axis integrates
thyroid cancers that undergo dedifferentiation. Cancer This is the first study to demonstrate common multiple signals downstream from RET/papillary
Res. 65, 4238–4245 (2005). mutations in β‑catenin in ATC. thyroid carcinoma leading to cell proliferation. Cancer
This is the first transgenic mouse study 38. Garcia-Rostan, G. et al. β-catenin dysregulation in Res. 69, 1867–1876 (2009).
demonstrating that BRAF‑V600E could induce the thyroid neoplasms: down-regulation, aberrant nuclear 59. Miyagi, E. et al. Chronic expression of RET/PTC 3
development of aggressive PTC. expression, and CTNNB1 exon 3 mutations are enhances basal and insulin-stimulated PI3 kinase/AKT
21. Lee, S. J. et al. Cross-regulation between oncogenic markers for aggressive tumor phenotypes and poor signaling and increases IRS‑2 expression in FRTL‑5
BRAF(V600E) kinase and the MST1 pathway in prognosis. Am. J. Pathol. 158, 987–996 (2001). thyroid cells. Mol. Carcinog. 41, 98–107 (2004).
papillary thyroid carcinoma. PLoS ONE 6, e16180 39. Fagin, J. A. et al. High prevalence of mutations of the 60. Hayashi, H. et al. Characterization of intracellular
(2011). p53 gene in poorly differentiated human thyroid signals via tyrosine 1062 in RET activated by glial cell
This study for the first time demonstrated a direct carcinomas. J. Clin. Invest. 91, 179–184 (1993). line-derived neurotrophic factor. Oncogene 19,
role for BRAF‑V600E in downregulating the 40. Donghi, R. et al. Gene p53 mutations are restricted to 4469–4475 (2000).
pro-apoptotic RASSF1–MST1–FOXO3 signalling poorly differentiated and undifferentiated carcinomas 61. Xing, M. Genetic alterations in the
pathway by demonstrating the direct interaction of of the thyroid gland. J. Clin. Invest. 91, 1753–1760 phosphatidylinositol‑3 kinase/Akt pathway in thyroid
BRAF‑V600E with MST1. (1993). cancer. Thyroid 20, 697–706 (2010).
22. Guerra, A. et al. The primary occurrence of References 39 and 40 demonstrate the high This is a comprehensive review on genetic
BRAF(V600E) is a rare clonal event in papillary thyroid prevalence of mutations in TP53 in ATC. alterations in the PI3K–AKT pathway in thyroid
carcinoma. J. Clin. Endocrinol. Metab. 97, 517–524 41. Murugan, A. K., Bojdani, E. & Xing, M. Identification cancer.
(2012). and functional characterization of isocitrate 62. Kroll, T. G. et al. PAX8‑PPARγ1 fusion oncogene in
23. Xing, M. BRAFV600E mutation and papillary thyroid dehydrogenase 1 (IDH1) mutations in thyroid cancer. human thyroid carcinoma. Science 289, 1357–1360
cancer: chicken or egg? J. Clin. Endocrinol. Metab. 97, Biochem. Biophys. Res. Commun. 393, 555–559 (2000).
2295–2298 (2012). (2010). 63. Dwight, T. et al. Involvement of the PAX8/peroxisome
24. Vasko, V. et al. High prevalence and possible de novo This is the first demonstration that IDH1 mutations proliferator-activated receptor γ rearrangement in
formation of BRAF mutation in metastasized papillary occur in thyroid cancer. follicular thyroid tumors. J. Clin. Endocrinol. Metab.
thyroid cancer in lymph nodes. J. Clin. Endocrinol. 42. Hemerly, J. P., Bastos, A. U. & Cerutti, J. M. 88, 4440–4445 (2003).
Metab. 90, 5265–5269 (2005). Identification of several novel non‑p.R132 IDH1 64. Eberhardt, N. L., Grebe, S. K., McIver, B. &
25. Abubaker, J. et al. Clinicopathological analysis of variants in thyroid carcinomas. Eur. J. Endocrinol. Reddi, H. V. The role of the PAX8/PPARγ fusion
papillary thyroid cancer with PIK3CA alterations in a 163, 747–755 (2010). oncogene in the pathogenesis of follicular thyroid
Middle Eastern population. J. Clin. Endocrinol. Metab. 43. Murugan, A. K. & Xing, M. Anaplastic thyroid cancers cancer. Mol. Cell Endocrinol. 321, 50–56 (2010).
93, 611–618 (2008). harbor novel oncogenic mutations of the ALK gene. 65. Placzkowski, K. A., Reddi, H. V., Grebe, S. K.,
26. Liu, Z. et al. Highly prevalent genetic alterations in Cancer Res. 71, 4403–4411 (2011). Eberhardt, N. L. & McIver, B. The role of the PAX8/
receptor tyrosine kinases and phosphatidylinositol This is the first report of novel ALK mutations that PPARγ fusion oncogene in thyroid cancer. PPAR Res.
3‑kinase/akt and mitogen-activated protein kinase can activate both the MAPK and PI3K–AKT 2008, 672829 (2008).
pathways in anaplastic and follicular thyroid pathways in ATC. 66. Ying, H. et al. Mutant thyroid hormone receptor β
cancers. J. Clin. Endocrinol. Metab. 93, 3106–3116 44. Murugan, A. K., Dong, J., Xie, J. & Xing, M. Uncommon represses the expression and transcriptional activity of
(2008). GNAQ, MMP8, AKT3, EGFR, and PIK3R1 mutations in peroxisome proliferator-activated receptor γ during
This study demonstrates a high prevalence of thyroid cancers. Endocr. Pathol. 22, 97–102 (2011). thyroid carcinogenesis. Cancer Res. 63, 5274–5280
genetic alterations that can activate both the 45. Maximo, V. et al. Somatic and germline mutation in (2003).
MAPK and PI3K–AKT pathways in aggressive GRIM‑19, a dual function gene involved in 67. Ciampi, R. et al. Oncogenic AKAP9‑BRAF fusion is a
thyroid cancer, thus providing a strong genetic mitochondrial metabolism and cell death, is linked to novel mechanism of MAPK pathway activation in
basis for the joint role of the two pathways in the mitochondrion-rich (Hurthle cell) tumours of the thyroid cancer. J. Clin. Invest. 115, 94–101 (2005).
progression and aggressiveness of thyroid thyroid. Br. J. Cancer 92, 1892–1898 (2005). 68. Lee, J. H., Lee, E. S., Kim, Y. S., Won, N. H. &
cancer. 46. Musholt, P. B. et al. Follicular histotypes of oncocytic Chae, Y. S. BRAF mutation and AKAP9 expression in
27. Bond, J. A., Wyllie, F. S., Rowson, J., Radulescu, A. & thyroid carcinomas do not carry mutations of the sporadic papillary thyroid carcinomas. Pathology 38,
Wynford-Thomas, D. In vitro reconstruction of tumour BRAF hot-spot. World J. Surg. 32, 722–728 (2008). 201–204 (2006).
initiation in a human epithelium. Oncogene 9, 47. Corver, W. E. et al. Genome haploidisation with 69. Xing, M. Gene methylation in thyroid tumorigenesis.
281–290 (1994). chromosome 7 retention in oncocytic follicular thyroid Endocrinology 148, 948–953 (2007).
28. Gire, V. & Wynford-Thomas, D. RAS oncogene carcinoma. PLoS ONE. 7, e38287 (2012). 70. Hu, S. et al. Association of aberrant methylation of
activation induces proliferation in normal human 48. Liu, Z. et al. IQGAP1 plays an important role in the tumor suppressor genes with tumor aggressiveness
thyroid epithelial cells without loss of differentiation. invasiveness of thyroid cancer. Clin. Cancer Res. 16, and BRAF mutation in papillary thyroid cancer. Int.
Oncogene 19, 737–744 (2000). 6009–6018 (2010). J. Cancer. 119, 2322–2329 (2006).

196 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

71. Hou, P., Liu, D. & Xing, M. Genome-wide alterations in 89. Pasquali, D. et al. Upregulation of endocrine gland- This study demonstrates that the efficacy of PI3K–
gene methylation by the BRAF V600E mutation in derived vascular endothelial growth factor in papillary AKT inhibitors in thyroid cancer cells depends on
papillary thyroid cancer cells. Endocr. Relat. Cancer thyroid cancers displaying infiltrative patterns, lymph genetic alterations in the pathway, thus providing
18, 687–697 (2011). node metastases, and BRAF mutation. Thyroid 21, the first preclinical evidence supporting a
This is a DNA methylation microarray study that 391–399 (2011). genetic-based therapeutic approach targeting the
revealed genome-wide hypomethylation and 90. Nowicki, T. S. et al. Inhibition of uPAR and uPA PI3K–AKT pathway in thyroid cancer.
hypermethylation and hence expression alterations reduces invasion in papillary thyroid carcinoma cells. 109. Liu, R. et al. The Akt-specific inhibitor MK2206
of genes driven by BRAFV600E, thus uncovering a Laryngoscope 120, 1383–1390 (2010). selectively inhibits thyroid cancer cells harboring
prominent epigenetic mechanism in thyroid 91. Nowicki, T. S. et al. Downregulation of uPAR inhibits mutations that can activate the PI3K/Akt pathway.
tumorigenesis driven by this oncogene. migration, invasion, proliferation, FAK/PI3K/Akt J. Clin. Endocrinol. Metab. 96, e577–e585 (2011).
72. Hou, P., Ji, M. & Xing, M. Association of PTEN gene signaling and induces senescence in papillary thyroid 110. LiVolsi, V. A. & Baloch, Z. W. Follicular-patterned
methylation with genetic alterations in the carcinoma cells. Cell Cycle 10, 100–107 (2011). tumors of the thyroid: the battle of benign versus
phosphatidylinositol 3‑kinase/AKT signaling 92. Riesco-Eizaguirre, G. et al. The BRAFV600E oncogene malignant versus so‑called uncertain. Endocr. Pathol.
pathway in thyroid tumors. Cancer 113, induces transforming growth factor β secretion leading 22, 184–189 (2011).
2440–2447 (2008). to sodium iodide symporter repression and increased This is an excellent discussion of the criteria and
This study proposes a model in which activation of malignancy in thyroid cancer. Cancer Res. 69, challenges for defining follicular thyroid tumours.
the PI3K–AKT pathway, driven by genetic 8317–8325 (2009). 111. Abbosh, P. H. & Nephew, K. P. Multiple signaling
alterations, causes aberrant methylation and hence This study demonstrates a unique mechanism in pathways converge on β-catenin in thyroid cancer.
silencing of PTEN, which in turn leads to the failure which BRAF‑V600E causes silencing of NIS by Thyroid 15, 551–561 (2005).
in terminating PI3K–AKT signalling, thus creating a promoting the secretion of TGFβ, which in turn 112. Burrows, N. et al. Expression of hypoxia-inducible
self-amplification loop that enhances and maintains activates SMAD to silence NIS. factor 1α in thyroid carcinomas. Endocr. Relat. Cancer
the constitutive activation of the PI3K–AKT 93. Nucera, C. et al. B‑Raf(V600E) and thrombospondin‑1 17, 61–72 (2010).
pathway in thyroid cancer. promote thyroid cancer progression. Proc. Natl Acad. 113. Burrows, N. et al. GDC‑0941 inhibits metastatic
73. Alvarez-Nunez, F. et al. PTEN promoter methylation in Sci. USA 107, 10649–10654 (2010). characteristics of thyroid carcinomas by targeting both
sporadic thyroid carcinomas. Thyroid 16, 17–23 This study demonstrates an important role of TSP1 the phosphoinositide‑3 kinase (PI3K) and hypoxia-
(2006). and its relationship with BRAF‑V600E, thus inducible factor‑1α (HIF‑1α) pathways. J. Clin.
74. Schagdarsurengin, U., Gimm, O., Dralle, H., providing an excellent example of the role of the Endocrinol. Metab. 96, e1934–e1943 (2011).
Hoang‑Vu, C. & Dammann, R. CpG island methylation tumour microenvironment in thyroid tumorigenesis. 114. Guigon, C. J., Zhao, L., Willingham, M. C. &
of tumor-related promoters occurs preferentially in 94. Knauf, J. A. et al. Progression of BRAF-induced thyroid Cheng, S. Y. PTEN deficiency accelerates tumour
undifferentiated carcinoma. Thyroid 16, 633–642 cancer is associated with epithelial-mesenchymal progression in a mouse model of thyroid cancer.
(2006). transition requiring concomitant MAP kinase and Oncogene 28, 509–517 (2009).
75. Frisk, T. et al. Silencing of the PTEN tumor-suppressor TGFβ signaling. Oncogene 30, 3153–3162 (2011). 115. Karin, M. NF‑κB as a critical link between
gene in anaplastic thyroid cancer. Genes 95. Nucera, C., Lawler, J. & Parangi, S. BRAF(V600E) and inflammation and cancer. Cold Spring Harb. Perspect.
Chromosomes Cancer 35, 74–80 (2002). microenvironment in thyroid cancer: a functional link Biol. 1, a000141 (2009).
76. Bruni, P. et al. PTEN expression is reduced in a subset to drive cancer progression. Cancer Res. 71, 116. Pacifico, F. et al. Oncogenic and anti-apoptotic activity
of sporadic thyroid carcinomas: evidence that PTEN- 2417–2422 (2011). of NF‑κB in human thyroid carcinomas. J. Biol. Chem.
growth suppressing activity in thyroid cancer cells 96. Xing, M. Oxidative stress: a new risk factor for thyroid 279, 54610–54619 (2004).
mediated by p27kip1. Oncogene 19, 3146–3155 cancer. Endocr. Relat. Cancer 19, C7–C11 (2012). 117. Visconti, R. et al. Expression of the neoplastic
(2000). 97. Guo, M., Liu, W., Serra, S., Asa, S. L. & Ezzat, S. phenotype by human thyroid carcinoma cell lines
77. Kondo, T., Ezzat, S. & Asa, S. L. Pathogenetic FGFR2 isoforms support epithelial-stromal requires NFκB p65 protein expression. Oncogene 15,
mechanisms in thyroid follicular-cell neoplasia. Nature interactions in thyroid cancer progression. Cancer Res. 1987–1994 (1997).
Rev. Cancer 6, 292–306 (2006). 72, 2017–2027 (2012). 118. Mitsiades, C. S. et al. Antitumor effects of the
78. Xing, M. Recent advances in molecular biology of 98. Saji, M. & Ringel, M. D. The PI3K‑Akt-mTOR pathway proteasome inhibitor bortezomib in medullary and
thyroid cancer and their clinical implications. in initiation and progression of thyroid tumors. Mol. anaplastic thyroid carcinoma cells in vitro. J. Clin.
Otolaryngol. Clin. North Am. 41, 1135–1146 Cell Endocrinol. 321, 20–28 (2010). Endocrinol. Metab. 91, 4013–4021 (2006).
(2008). 99. Liaw, D. et al. Germline mutations of the PTEN gene in 119. Pacifico, F. & Leonardi, A. Role of NF‑κB in thyroid
79. Melillo, R. M. et al. The RET/PTC-RAS-BRAF linear Cowden disease, an inherited breast and thyroid cancer. Mol. Cell Endocrinol. 321, 29–35 (2010).
signaling cascade mediates the motile and mitogenic cancer syndrome. Nature Genet. 16, 64–67 (1997). 120. Li, X., Abdel-Mageed, A. B., Mondal, D. & Kandil, E.
phenotype of thyroid cancer cells. J. Clin. Invest. 115, 100. Miyakawa, M. et al. Increased expression of The nuclear factor κ‑B signaling pathway as a
1068–1081 (2005). phosphorylated p70S6 kinase and Akt in papillary therapeutic target against thyroid cancers. Thyroid
80. Oler, G. et al. Gene expression profiling of papillary thyroid cancer tissues. Endocr. J. 50, 77–83 (2003). 28 Dec 2012 (doi:10.1089/thy.2012.0237).
thyroid carcinoma identifies transcripts correlated 101. Ringel, M. D. et al. Overexpression and overactivation 121. Ikenoue, T. et al. Functional analysis of mutations
with BRAF mutational status and lymph node of Akt in thyroid carcinoma. Cancer Res. 61, within the kinase activation segment of B‑Raf in
metastasis. Clin. Cancer Res. 14, 4735–4742 6105–6111 (2001). human colorectal tumors. Cancer Res. 63,
(2008). This study provides the first functional evidence for 8132–8137 (2003).
81. Jo, Y. S. et al. Influence of the BRAF V600E mutation the involvement of the PI3K–AKT pathway in 122. Ikenoue, T. et al. Different effects of point mutations
on expression of vascular endothelial growth factor in thyroid tumorigenesis and thyroid cancer invasion. within the B‑Raf glycine-rich loop in colorectal tumors
papillary thyroid cancer. J. Clin. Endocrinol. Metab. 102. Vasko, V. et al. Akt activation and localisation on mitogen-activated protein/extracellular signal-
91, 3667–3670 (2006). correlate with tumour invasion and oncogene regulated kinase kinase/extracellular signal-regulated
82. Giordano, T. J. et al. Molecular classification of expression in thyroid cancer. J. Med. Genet. 41, kinase and nuclear factor κB pathway and cellular
papillary thyroid carcinoma: distinct BRAF, RAS, and 161–170 (2004). transformation. Cancer Res. 64, 3428–3435
RET/PTC mutation-specific gene expression profiles 103. Suzuki, H., Willingham, M. C. & Cheng, S. Y. Mice with (2004).
discovered by DNA microarray analysis. Oncogene. a mutation in the thyroid hormone receptor βgene 123. Vale, T., Ngo, T. T., White, M. A. & Lipsky, P. E. Raf-
24, 6646–6656 (2005). spontaneously develop thyroid carcinoma: a mouse induced transformation requires an interleukin 1
83. Kumagai, A. et al. Childhood thyroid carcinoma with model of thyroid carcinogenesis. Thyroid 12, autocrine loop. Cancer Res. 61, 602–607 (2001).
BRAFT1799A mutation shows unique pathological 963–969 (2002). 124. Liu, D. & Xing, M. Potent inhibition of thyroid cancer
features of poor differentiation. Oncol. Rep. 16, 104. Furuya, F., Hanover, J. A. & Cheng, S. Y. Activation of cells by the MEK inhibitor PD0325901 and its
123–126 (2006). phosphatidylinositol 3‑kinase signaling by a mutant potentiation by suppression of the PI3K and NF‑κB
84. Palona, I. et al. BRAFV600E promotes invasiveness of thyroid hormone β receptor. Proc. Natl Acad. Sci. USA pathways. Thyroid 18, 853–864 (2008).
thyroid cancer cells through nuclear factor κB 103, 1780–1785 (2006). 125. Oh, H. J. et al. Role of the tumor suppressor RASSF1A
activation. Endocrinology 147, 5699–5707 (2006). This study demonstrates robust activation of the in Mst1‑mediated apoptosis. Cancer Res. 66,
This study demonstrates a direct coupling of PI3K–AKT pathway by a mutant thyroid hormone 2562–2569 (2006).
BRAF‑V600E to the NF‑κB pathway that is β-receptor that causes FTC in mice, thus providing 126. Lehtinen, M. K. et al. A conserved MST-FOXO
independent of MEK–ERK signalling in thyroid the first animal data to support an important role of signaling pathway mediates oxidative-stress responses
cancer. the PI3K–AKT pathway in the tumorigenesis of FTC. and extends life span. Cell 125, 987–1001 (2006).
85. Mesa, C. Jr et al. Conditional activation of RET/PTC3 105. Kim, C. S. et al. AKT activation promotes metastasis in 127. Schagdarsurengin, U. et al. Frequent epigenetic
and BRAFV600E in thyroid cells is associated with a mouse model of follicular thyroid carcinoma. silencing of the CpG island promoter of RASSF1A in
gene expression profiles that predict a preferential Endocrinology 146, 4456–4463 (2005). thyroid carcinoma. Cancer Res. 62, 3698–3701
role of BRAF in extracellular matrix remodeling. 106. Saji, M. et al. Akt1 deficiency delays tumor (2002).
Cancer Res. 66, 6521–6529 (2006). progression, vascular invasion, and distant metastasis 128. Xing, M. et al. Early occurrence of RASSF1A
86. Franzoni, A. et al. Prohibitin is overexpressed in in a murine model of thyroid cancer. Oncogene 30, hypermethylation and its mutual exclusion with BRAF
papillary thyroid carcinomas bearing the 4307–4315 (2011). mutation in thyroid tumorigenesis. Cancer Res. 64,
BRAF(V600E) mutation. Thyroid 19, 247–255 107. Xu, P. Z., Chen, M. L., Jeon, S. M., Peng, X. D. & 1664–1668 (2004).
(2009). Hay, N. The effect Akt2 deletion on tumor development This is the first study to link BRAF‑V600E to the
87. Watanabe, R. et al. Possible involvement of in Pten+/− mice. Oncogene 31, 518–526 (2012). RASSF1 pathway in thyroid cancer.
BRAFV600E in altered gene expression in papillary 108. Liu, D., Hou, P., Liu, Z., Wu, G. & Xing, M. Genetic 129. Van Der Heide, L. P., Hoekman, M. F. & Smidt, M. P.
thyroid cancer. Endocr. J. 56, 407–414 (2009). alterations in the phosphoinositide 3‑kinase/Akt The ins and outs of FoxO shuttling: mechanisms of
88. Zerilli, M. et al. BRAF(V600E) mutation influences signaling pathway confer sensitivity of thyroid cancer FoxO translocation and transcriptional regulation.
hypoxia-inducible factor‑1α expression levels in cells to therapeutic targeting of Akt and mammalian Biochem. J. 380, 297–309 (2004).
papillary thyroid cancer. Mod. Pathol. 23, target of rapamycin. Cancer Res. 69, 7311–7319 130. Clevers, H. & Nusse, R. Wnt/β-catenin signaling and
1052–1060 (2010). (2009). disease. Cell 149, 1192–1205 (2012).

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 197

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

131. Wiseman, S. M., Griffith, O. L., Gown, A., Walker, B. & 151. Riesco-Eizaguirre, G. & Santisteban, P. New insights in 169. Kogai, T., Sajid-Crockett, S., Newmarch, L. S., Liu, Y. Y.
Jones, S. J. Immunophenotyping of thyroid tumors thyroid follicular cell biology and its impact in thyroid & Brent, G. A. Phosphoinositide-3‑kinase inhibition
identifies molecular markers altered during cancer therapy. Endocr. Relat. Cancer 14, 957–977 induces sodium/iodide symporter expression in rat
transformation of differentiated into anaplastic (2007). thyroid cells and human papillary thyroid cancer cells.
carcinoma. Am. J. Surg. 201, 580–586 (2011). 152. Jhiang, S. M. Regulation of sodium/iodide symporter. J. Endocrinol. 199, 243–252 (2008).
132. Sastre-Perona, A. & Santisteban, P. Role of the wnt Rev. Endocr. Metab. Disord. 1, 205–215 (2000). 170. Xing, M., Haugen, B. R. & Schlumberger, M. Promises
pathway in thyroid cancer. Front. Endocrinol. 3, 31 153. Riesco-Eizaguirre, G., Gutierrez-Martinez, P., Garcia- in molecular-based management of differentiated
(2012). Cabezas, M. A., Nistal, M. & Santisteban, P. The thyroid cancer. Lancet (in the press).
133. Lu, C., Zhu, X., Willingham, M. C. & Cheng, S. Y. oncogene BRAF V600E is associated with a high risk This is a review on the clinically promising
Activation of tumor cell proliferation by thyroid of recurrence and less differentiated papillary thyroid diagnostic and prognostic molecular markers and
hormone in a mouse model of follicular thyroid carcinoma due to the impairment of Na+/I− targeting molecular therapeutic targets in thyroid cancer. It
carcinoma. Oncogene 31, 2007–2016 (2012). to the membrane. Endocr. Relat. Cancer 13, summarizes the recent developments in clinical
134. Cross, D. A., Alessi, D. R., Cohen, P., Andjelkovich, M. 257–269 (2006). translational research of thyroid cancer medicine.
& Hemmings, B. A. Inhibition of glycogen synthase 154. Mian, C. et al. Molecular characteristics in papillary 171. Cohen, Y. et al. Mutational analysis of BRAF in fine
kinase‑3 by insulin mediated by protein kinase B. thyroid cancers (PTCs) with no 131I uptake. Clin. needle aspiration biopsies of the thyroid: a potential
Nature 378, 785–789 (1995). Endocrinol. 68, 108–116 (2008). application for the preoperative assessment of thyroid
135. Choi, K. S., Bae, M. K., Jeong, J. W., Moon, H. E. & 155. Barollo, S. et al. BRAF in primary and recurrent nodules. Clin. Cancer Res. 10, 2761–2765 (2004).
Kim, K. W. Hypoxia-induced angiogenesis during papillary thyroid cancers: the relationship with 131I and 172. Xing, M. et al. Detection of BRAF mutation on fine
carcinogenesis. J. Biochem. Mol. Biol. 36, 120–127 2-[18F]fluoro-2‑deoxy‑D‑glucose uptake ability. Eur. needle aspiration biopsy specimens: a new diagnostic
(2003). J. Endocrinol. 163, 659–663 (2010). tool for papillary thyroid cancer. J. Clin. Endocrinol.
136. Scarpino, S. et al. Increased expression of Met 156. Durante, C. et al. BRAF mutations in papillary thyroid Metab. 89, 2867–2872 (2004).
protein is associated with up‑regulation of hypoxia carcinomas inhibit genes involved in iodine References 171 and 172 are the first studies to
inducible factor‑1 (HIF‑1) in tumour cells in papillary metabolism. J. Clin. Endocrinol. Metab. 92, 2840– demonstrate the feasibility of BRAF mutation
carcinoma of the thyroid. J. Pathol. 202, 352–358 2843 (2007). testing on thyroid FNAB samples for the diagnosis
(2004). 157. Romei, C. et al. BRAFV600E mutation, but not RET/ of thyroid cancer.
137. Kimura, T. et al. Regulation of thyroid cell proliferation PTC rearrangements, is correlated with a lower 173. Xing, M. Prognostic utility of BRAF mutation in
by TSH and other factors: a critical evaluation of in expression of both thyroperoxidase and sodium iodide papillary thyroid cancer. Mol. Cell Endocrinol. 321,
vitro models. Endocr. Rev. 22, 631–656 (2001). symporter genes in papillary thyroid cancer. Endocr. 86–93 (2010).
138. Lu, C., Zhao, L., Ying, H., Willingham, M. C. & Relat. Cancer 15, 511–520 (2008). 174. Filicori, F. et al. Risk stratification of indeterminate
Cheng, S. Y. Growth activation alone is not sufficient to 158. Oler, G. & Cerutti, J. M. High prevalence of BRAF thyroid fine-needle aspiration biopsy specimens based
cause metastatic thyroid cancer in a mouse model of mutation in a Brazilian cohort of patients with on mutation analysis. Surgery 150, 1085–1091
follicular thyroid carcinoma. Endocrinology 151, sporadic papillary thyroid carcinomas: correlation with (2011).
1929–1939 (2010). more aggressive phenotype and decreased expression 175. Nikiforov, Y. E. et al. Impact of mutational testing on
This study demonstrated for the first time that a of iodide-metabolizing genes. Cancer 115, 972–980 the diagnosis and management of patients with
transgenic mouse model of thyroid tumorigenesis (2009). cytologically indeterminate thyroid nodules: a
promoted by a mutation (in this case, mutant 159. Espadinha, C., Santos, J. R., Sobrinho, L. G. & prospective analysis of 1056 FNA samples. J. Clin.
THRβ) requires an intact TSHR pathway. Bugalho, M. J. Expression of iodine metabolism genes Endocrinol. Metab. 96, 3390–3397 (2011).
139. Franco, A. T. et al. Thyrotrophin receptor signaling in human thyroid tissues: evidence for age and This is a large study of the mutational analyses of
dependence of Braf-induced thyroid tumor initiation in BRAFV600E mutation dependency. Clin. Endocrinol. thyroid nodules for their diagnostic utility. The
mice. Proc. Natl Acad. Sci. USA 108, 1615–1620 70, 629–635 (2009). analyses show high diagnostic specificity and
(2011). 160. Morari, E. C. et al. Use of sodium iodide symporter positive predictive value.
Similarly to reference 138, this study also expression in differentiated thyroid carcinomas. Clin. 176. Alexander, E. K. et al. Preoperative diagnosis of
demonstrated a requirement for an intact TSHR Endocrinol. 75, 247–254 (2011). benign thyroid nodules with indeterminate cytology.
pathway, this time for BRAF‑V600E‑induced 161. Liu, D. et al. Suppression of BRAF/MEK/MAP kinase N. Engl. J. Med. 367, 705–715 (2012).
thyroid tumorigenesis in transgenic mice. pathway restores expression of iodide-metabolizing This study demonstrates a high sensitivity and
140. Boelaert, K. et al. Serum thyrotropin concentration as genes in thyroid cells expressing the V600E BRAF negative predictive power of a gene expression
a novel predictor of malignancy in thyroid nodules mutant. Clin. Cancer Res. 13, 1341–1349 (2007). classifier for diagnosing benign thyroid tumours
investigated by fine-needle aspiration. J. Clin. This study for the first time directly demonstrated with indeterminate cytology.
Endocrinol. Metab. 91, 4295–4301 (2006). in an in vitro thyroid cell system that BRAF‑V600E 177. Xing, M. et al. The BRAF T1799A mutation and poor
141. Fiore, E. et al. Lower levels of TSH are associated with silences thyroid iodide-handling genes and that outcomes of papillary thyroid cancer-report from the
a lower risk of papillary thyroid cancer in patients with suppression of BRAF‑V600E or MEK can restore international collaborative BRAF study group. Thyroid
thyroid nodular disease: thyroid autonomy may play a the expression of these genes, which was later 21 (S1), Abstract 112 (2011).
protective role. Endocr. Relat. Cancer 16, 1251–1260 reproduced in transgenic mouse studies in 178. Kebebew, E. et al. The prevalence and prognostic
(2009). reference 163. value of BRAF mutation in thyroid cancer. Ann. Surg.
142. Haymart, M. R. et al. Higher serum thyroid stimulating 162. Vadysirisack, D. D., Venkateswaran, A., Zhang, Z. & 246, 466–470 (2007).
hormone level in thyroid nodule patients is associated Jhiang, S. M. MEK signaling modulates sodium 179. Basolo, F. et al. Correlation between the BRAF V600E
with greater risks of differentiated thyroid cancer and iodide symporter at multiple levels and in a mutation and tumor invasiveness in papillary thyroid
advanced tumor stage. J. Clin. Endocrinol. Metab. 93, paradoxical manner. Endocr. Relat. Cancer 14, carcinomas smaller than 20 millimeters: analysis of
809–814 (2008). 421–432 (2007). 1060 cases. J. Clin. Endocrinol. Metab. 95,
143. Lyons, J. et al. Two G protein oncogenes in human 163. Chakravarty, D. et al. Small-molecule MAPK inhibitors 4197–4205 (2010).
endocrine tumors. Science 249, 655–659 (1990). restore radioiodine incorporation in mouse thyroid 180. Lin, K. L. et al. The BRAF mutation is predictive of
144. Parma, J. et al. Somatic mutations in the thyrotropin cancers with conditional BRAF activation. J. Clin. aggressive clinicopathological characteristics in
receptor gene cause hyperfunctioning thyroid Invest. 121, 4700–4711 (2011). papillary thyroid microcarcinoma. Ann. Surg. Oncol.
adenomas. Nature 365, 649–651 (1993). 164. Hou, P., Bojdani, E. & Xing, M. Induction of thyroid 17, 3294–3300 (2010).
145. Gudmundsson, J. et al. Common variants on 9q22.33 gene expression and radioiodine uptake in thyroid 181. Elisei, R. et al. The BRAFV600E mutation is an
and 14q13.3 predispose to thyroid cancer in cancer cells by targeting major signaling pathways. independent, poor prognostic factor for the outcome
European populations. Nature Genet. 41, 460–464 J. Clin. Endocrinol. Metab. 95, 820–828 (2010). of patients with low-risk intrathyroid papillary thyroid
(2009). This study demonstrated robust expression of carcinoma: single-institution results from a large
This is the first large genome-wide association thyroid iodide-handling genes and radioiodine cohort study. J. Clin. Endocrinol. Metab. 97,
study to reveal several novel single-nucleotide uptake in thyroid cancer cells by simultaneously 4390–4398 (2012).
polymorphisms that are associated with an targeting MAPK, PI3K–AKT and histone 182. Xing, M. et al. BRAF mutation testing of thyroid fine-
increased risk of thyroid cancer and low levels of deacetylase systems, which could be enhanced by needle aspiration biopsy specimens for preoperative
serum TSH. TSH, thus providing strong clinical implications for risk stratification in papillary thyroid cancer. J. Clin.
146. Gudmundsson, J. et al. Discovery of common variants the treatment of thyroid cancer. Oncol. 27, 2977–2982 (2009).
associated with low TSH levels and thyroid cancer risk. 165. Liu, Z. & Xing, M. Induction of sodium/iodide 183. Joo, J. Y. et al. Prediction of occult central lymph node
Nature Genet. 44, 319–322 (2012). symporter (NIS) expression and radioiodine uptake in metastasis in papillary thyroid carcinoma by
147. Wu, G. et al. Uncommon mutation, but common non-thyroid cancer cells. PLoS ONE 7, e31729 preoperative BRAF analysis using fine-needle
amplifications, of the PIK3CA gene in thyroid (2012). aspiration biopsy: a prospective study. J. Clin.
tumors. J. Clin. Endocrinol. Metab. 90, 4688–4693 166. Costamagna, E., Garcia, B. & Santisteban, P. The Endocrinol. Metab. 97, 3996–4003 (2012).
(2005). functional interaction between the paired domain 184. Garcia-Rostan, G. et al. ras mutations are associated
148. Costa, A. M. et al. BRAF mutation associated with transcription factor Pax8 and Smad3 is involved in with aggressive tumor phenotypes and poor prognosis
other genetic events identifies a subset of aggressive transforming growth factor-β repression of the sodium/ in thyroid cancer. J. Clin. Oncol. 21, 3226–3235
papillary thyroid carcinoma. Clin. Endocrinol. 68, iodide symporter gene. J. Biol. Chem. 279, (2003).
618–634 (2008). 3439–3446 (2004). 185. Basolo, F. et al. N‑ras mutation in poorly differentiated
149. Henderson, Y. C. et al. High rate of BRAF and RET/PTC 167. de Souza, E. C. et al. MTOR downregulates iodide thyroid carcinomas: correlation with bone metastases
dual mutations associated with recurrent papillary uptake in thyrocytes. J. Endocrinol. 206, 113–120 and inverse correlation to thyroglobulin expression.
thyroid carcinoma. Clin. Cancer Res. 15, 485–491 (2010). Thyroid 10, 19–23 (2000).
(2009). 168. Garcia, B. & Santisteban, P. PI3K is involved in the 186. Volante, M. et al. RAS mutations are the predominant
150. Dankort, D. et al. BrafV600E cooperates with Pten loss IGF‑I inhibition of TSH-induced sodium/iodide molecular alteration in poorly differentiated thyroid
to induce metastatic melanoma. Nature Genet. 41, symporter gene expression. Mol. Endocrinol. 16, carcinomas and bear prognostic impact. J. Clin.
544–552 (2009). 342–352 (2002). Endocrinol. Metab. 94, 4735–4741 (2009).

198 | MARCH 2013 | VOLUME 13 www.nature.com/reviews/cancer

© 2013 Macmillan Publishers Limited. All rights reserved


REVIEWS

187. Fukahori, M. et al. The association between RAS 198. Schweppe, R. E. et al. Distinct genetic alterations in 209. Manenti, G., Pilotti, S., Re, F. C., Della, P. G. &
gene mutations and clinical characteristics in the mitogen-activated protein kinase pathway dictate Pierotti, M. A. Selective activation of ras oncogenes in
follicular thyroid tumors: new insights from a single sensitivity of thyroid cancer cells to mitogen-activated follicular and undifferentiated thyroid carcinomas. Eur.
center and a large patient cohort. Thyroid 22, protein kinase kinase 1/2 inhibition. Thyroid 19, J. Cancer 30A, 987–993 (1994).
683–689 (2012). 825–835 (2009). 210. Zhu, Z., Gandhi, M., Nikiforova, M. N., Fischer, A. H.
188. Cohen, E. E. et al. Axitinib is an active treatment for 199. Liu, D., Xing, J., Trink, B. & Xing, M. BRAF mutation- & Nikiforov, Y. E. Molecular profile and clinical-
all histologic subtypes of advanced thyroid cancer: selective inhibition of thyroid cancer cells by the novel pathologic features of the follicular variant of
results from a phase II study. J. Clin. Oncol. 26, MEK inhibitor RDEA119 and genetic-potentiated papillary thyroid carcinoma. An unusually high
4708–4713 (2008). synergism with the mTOR inhibitor temsirolimus. Int. prevalence of ras mutations. Am. J. Clin. Pathol.
189. Gupta-Abramson, V. et al. Phase II trial of sorafenib in J. Cancer 127, 2965–2973 (2010). 120, 71–77 (2003).
advanced thyroid cancer. J. Clin. Oncol. 26, 200. Salerno, P. et al. Cytostatic activity of adenosine 211. Santarpia, L. et al. Genetic alterations in the RAS/
4714–4719 (2008). triphosphate-competitive kinase inhibitors in BRAF RAF/mitogen-activated protein kinase and
190. Kloos, R. T. et al. Phase II trial of sorafenib in mutant thyroid carcinoma cells. J. Clin. Endocrinol. phosphatidylinositol 3‑kinase/Akt signaling pathways
metastatic thyroid cancer. J. Clin. Oncol. 27, Metab. 95, 450–455 (2010). in the follicular variant of papillary thyroid carcinoma.
1675–1684 (2009). 201. Xing, J., Liu, R., Xing, M. & Trink, B. The BRAFT1799A Cancer 116, 2974–2983 (2010).
191. Ahmed, M. et al. Analysis of the efficacy and toxicity of mutation confers sensitivity of thyroid cancer cells to the 212. Dahia, P. L. et al. Somatic deletions and mutations in
sorafenib in thyroid cancer: a phase II study in a UK BRAFV600E inhibitor PLX4032 (RG7204). Biochem. the Cowden disease gene, PTEN, in sporadic thyroid
based population. Eur. J. Endocrinol. 165, 315–322 Biophys. Res. Commun. 404, 958–962 (2011). tumors. Cancer Res. 57, 4710–4713 (1997).
(2011). 202. Flaherty, K. T. et al. Inhibition of mutated, activated
192. Sherman, S. I. et al. Motesanib diphosphate in BRAF in metastatic melanoma. N. Engl. J. Med. 363, Acknowledgements
progressive differentiated thyroid cancer. N. Engl. 809–819 (2010). This work is supported by the US National Institutes of Health
J. Med. 359, 31–42 (2008). 203. Janku, F. et al. PI3K/AKT/mTOR inhibitors in patients (NIH) R01 grants CA113507 and CA134225 to the author.
This study represents an outstanding example of with breast and gynecologic malignancies harboring The author thanks A. K. Murugan, Y. Trink and D. Liu for their
the success of recent clinical trials of novel PIK3CA mutations. J. Clin. Oncol. 30, 777–782 (2012). help in preparing the figures and references. The author also
small-molecule inhibitors targeting aberrantly 204. Jin, N., Jiang, T., Rosen, D. M., Nelkin, B. D. & thanks the numerous colleagues and investigators in this field
activated signalling pathways in thyroid cancer. Ball, D. W. Synergistic action of a RAF inhibitor and a whose outstanding work made it an extremely enjoyable
193. Bible, K. C. et al. Efficacy of pazopanib in dual PI3K/mTOR inhibitor in thyroid cancer. Clin. experience to write this Review. The author wishes to apolo-
progressive, radioiodine-refractory, metastatic Cancer Res. 17, 6482–6489 (2011). gize to those whose work is not cited owing to space
differentiated thyroid cancers: results of a phase 2 205. Liu, R., Liu, D. & Xing, M. The Akt inhibitor MK2206 limitations.
consortium study. Lancet Oncol. 11, 962–972 synergizes, but perifosine antagonizes, the
(2010). BRAF(V600E) inhibitor PLX4032 and the MEK1/2 Competing interests statement
194. Xing, M. Genetic-targeted therapy of thyroid cancer: a inhibitor AZD6244 in the inhibition of thyroid cancer The author declares competing financial interests: see Web
real promise. Thyroid 19, 805–809 (2009). cells. J. Clin. Endocrinol. Metab. 97, e173–e182 version for details.
195. Ball, D. W. et al. Selective growth inhibition in BRAF (2012).
mutant thyroid cancer by the MEK 1/2 inhibitor 206. Vasko, V. et al. Specific pattern of RAS oncogene DATABASES
AZD6244 (ARRY‑142886). J. Clin. Endocrinol. Metab. mutations in follicular thyroid tumors. J. Clin. National Cancer Institute Drug Dictionary:
92, 4712–4718 (2007). Endocrinol. Metab. 88, 2745–2752 (2003). http://www.cancer.gov/drugdictionary
196. Leboeuf, R. et al. BRAFV600E mutation is associated 207. Nikiforova, M. N. et al. RAS point mutations and axitinib | motesanib | PLX4032 | pazopanib | sorafenib |
with preferential sensitivity to mitogen-activated PAX8‑PPARγ rearrangement in thyroid tumors: vandetanib
protein kinase kinase inhibition in thyroid cancer cell evidence for distinct molecular pathways in thyroid
lines. J. Clin. Endocrinol. Metab. 93, 2194–2201 follicular carcinoma. J. Clin. Endocrinol. Metab. 88, FURTHER INFORMATION
(2008). 2318–2326 (2003). Mingzhao Xing’s homepages: http://www.hopkinsmedicine.
197. Liu, D., Liu, Z., Jiang, D., Dackiw, A. P. & Xing, M. 208. Rivera, M. et al. Molecular genotyping of papillary org/endocrinology/faculty/faculty/xing.html;
Inhibitory effects of the MEK inhibitor CI‑1040 on the thyroid carcinoma follicular variant according to its http://www.hopkinsmedicine.org/endocrinology/research/
proliferation and tumor growth of thyroid cancer cells histological subtypes (encapsulated versus infiltrative) faculty/xing.html
with BRAF or RAS mutations. J. Clin. Endocrinol. reveals distinct BRAF and RAS mutation patterns. ALL LINKS ARE ACTIVE IN THE ONLINE PDF
Metab. 92, 4686–4695 (2007). Mod. Pathol. 23, 1191–1200 (2010).

NATURE REVIEWS | CANCER VOLUME 13 | MARCH 2013 | 199

View publication stats © 2013 Macmillan Publishers Limited. All rights reserved

You might also like