ArticlePDF AvailableLiterature Review

BRAF Mutation in Papillary Thyroid Carcinoma: Pathogenic Role and Clinical Implications

Authors:

Abstract and Figures

Papillary thyroid cancer (PTC) is the most common endocrine malignancy, accounting for 85-90% of all thyroid cancers. Genetic alternations involving the mitogen-activated protein kinase (MAPK) pathway are frequently demonstrated in PTC, such as RET/PTC, RAS, and B-type Raf kinase (BRAF) mutations. Over 90% of BRAF mutations are T1799A, resulting in a BRAF(V600E) mutation. BRAF(V600E) is present in approximately 50% of PTC and also found in aggressive histologic variants and PTC-derived anaplastic thyroid cancer, but is rare in follicular variants, and not found in follicular thyroid cancer. The tumorigenic role of BRAF(V600E) in the development of PTC was documented in thyroid-targeted BRAF(V600E) transgenic mice, and rat thyroid cells overexpressed with BRAF(V600E) suggested that BRAF(V600E) is an initiator of tumorigenesis and is required for tumor progression in PTC. Most clinical studies have demonstrated an association of BRAF(V600E) mutation with aggressive clinicopathologic characteristics and high tumor recurrence, although the results are controversial. The association is also observed in patients with papillary thyroid microcarcinomas and low-risk PTC. As a highly specific and unique mutation in PTC, testing for BRAF(V600E) in fine-needle aspiration specimens has been shown to refine the diagnostic accuracy of PTC in indeterminate cytology. Preoperative BRAF(V600E) analysis in low-risk patients may provide important value for prognostication, and these patients might benefit from receiving more intensive management and frequent follow-up. BRAF-targeted therapies have been developed to treat various human cancers including advanced thyroid cancers. Preclinical results are encouraging, but the anticancer effects of clinical trials are disappointing. Studies of multi-kinase inhibitors and/or combination with other regimens are underway in the treatment of advanced thyroid cancers. In this article, we review the pathogenesis of PTC, and the clinical implications of BRAF(V600E) mutation in the diagnosis, prognosis and potential targeted therapeutic strategies for thyroid cancers.
Content may be subject to copyright.
J Chin Med Assoc March 2010 Vol 73 No 3 113
Introduction
Thyroid cancer is the most common endocrine ma-
lignancy and accounts for 1% of all cancers. In the
United States, it was estimated that 37,200 men and
women (10,000 men, 27,200 women) would be diag-
nosed with thyroid cancer in 2009.1The incidence of
thyroid cancer has increased 50% since 1973, and it
is the most rapidly increasing cancer among women
and the 2nd most among men. Follicular cell-derived
thyroid cancers are classified into papillary thyroid
cancer (PTC), follicular thyroid cancer (FTC), and
anaplastic thyroid cancer (ATC). PTC is the most
common type and accounts for 85–90% of all thyroid
malignancies.2,3 Differentiated thyroid cancers (DTC)
including PTC and FTC exhibit evidence of follicular
epithelial cell differentiation such as iodine uptake and
organification, and are usually treated successfully by
primary surgical excision, radioiodine therapy, and levo-
thyroxine suppression. The overall 10-year relative
survival rate of DTC is over 90%.1,4 However, up to
35% of patients suffered from disease recurrence dur-
ing a 40-year follow-up, and over 1,600 people in the
United States and 35,000 worldwide die of thyroid
cancer each year. The death rate for thyroid cancer in
the United States is 0.5 per 100,000 each year.1,5,6
Thyroid cancer with undifferentiated/aggressive his-
tologic variants, or loss of iodine uptake due to sub-
sequent dedifferentiation, are often inoperable and
exhibit poor response to radioiodine therapy, leading
REVIEW ARTICLE
BRAF Mutation in Papillary Thyroid Carcinoma:
Pathogenic Role and Clinical Implications
Kam-Tsun Tang1,3, Chen-Hsen Lee2,3*
Departments of 1Medical Education and Research, and 2Surgery, Taipei Veterans General Hospital, and
3National Yang-Ming University School of Medicine, Taipei, Taiwan, R.O.C.
Papillary thyroid cancer (PTC) is the most common endocrine malignancy, accounting for 85–90% of all thyroid cancers.
Genetic alternations involving the mitogen-activated protein kinase (MAPK) pathway are frequently demonstrated in PTC,
such as RET/PTC, RAS, and B-type Raf kinase (BRAF) mutations. Over 90% of BRAF mutations are T1799A, resulting in a
BRAFV600E mutation. BRAFV600E is present in 50% of PTC and also found in aggressive histologic variants and PTC-
derived anaplastic thyroid cancer, but is rare in follicular variants, and not found in follicular thyroid cancer. The tumori-
genic role of BRAFV600E in the development of PTC was documented in thyroid-targeted BRAFV600E transgenic mice, and
rat thyroid cells overexpressed with BRAFV600E suggested that BRAFV600E is an initiator of tumorigenesis and is required
for tumor progression in PTC. Most clinical studies have demonstrated an association of BRAFV600E mutation with aggres-
sive clinicopathologic characteristics and high tumor recurrence, although the results are controversial. The association
is also observed in patients with papillary thyroid microcarcinomas and low-risk PTC. As a highly specific and unique
mutation in PTC, testing for BRAFV600E in fine-needle aspiration specimens has been shown to refine the diagnostic accu-
racy of PTC in indeterminate cytology. Preoperative BRAFV600E analysis in low-risk patients may provide important value
for prognostication, and these patients might benefit from receiving more intensive management and frequent follow-up.
BRAF-targeted therapies have been developed to treat various human cancers including advanced thyroid cancers.
Preclinical results are encouraging, but the anticancer effects of clinical trials are disappointing. Studies of multi-kinase
inhibitors and/or combination with other regimens are underway in the treatment of advanced thyroid cancers. In this
article, we review the pathogenesis of PTC, and the clinical implications of BRAFV600E mutation in the diagnosis, prognosis
and potential targeted therapeutic strategies for thyroid cancers. [J Chin Med Assoc 2010;73(3):113–128]
Key Words: BRAF mutation, fine-needle aspiration cytology, papillary thyroid cancer
© 2010 Elsevier Taiwan LLC and the Chinese Medical Association. All rights reserved.
*Correspondence to: Dr Chen-Hsen Lee, Department of Surgery, Taipei Veterans General Hospital,
201, Section 2, Shih-Pai Road, Taipei 112, Taiwan, R.O.C.
E-mail: chlee@vghtpe.gov.tw Received: September 1, 2009 Accepted: December 11, 2009
to a high recurrence rate and unfavorable prognosis.
Traditional chemotherapy has low response rates, and
long-term efficacy is unsatisfactory.7,8 There is currently
no effective treatment for these patients.
Conventional treatment strategies for DTC are
based on various staging systems (such as the TNM
system), which are designed according to a patient’s
clinicopathological characteristics, allowing patients at
low risk to undergo less intensive therapy and less fre-
quent follow-up than patients at high risk.9,10 Unfor-
tunately, 15% of tumor recurrence after a median of
11 years’ follow-up, and over 10% of cancer deaths were
initially classified as low risk.11,12 Therefore, more
accurate risk stratification and effective treatment for
advanced thyroid cancer are important to reduce tumor
recurrence and mortality.
Recently, significant progress has been made in
the understanding of the B-type Raf kinase (BRAF)
mutation and the mitogen-activated protein kinase
(MAPK) pathway in the tumorigenesis of human
cancers.13,14 Thyroid cancers, particularly PTCs, are
frequently found to have genetic alterations. A thymi-
dine-to-adenosine transversion at exon 15 nucleotide
1799 (T1799A) of the BRAF gene, resulting in the
replacement of valine with glutamic acid at position
600 (BRAFV600E), is the most prevalent mutation in
PTC.14 As a result, BRAF mutation has recently been
the subject of intensive study to investigate its tumori-
genic role and its clinical implications.15–17 In this arti-
cle, we review the mechanism of BRAFV600E mutation
and MAPK signal transduction pathway in the patho-
genesis of PTC, the clinical implications of BRAFV600E
mutation in preoperative diagnosis and prognostic
stratification, and recent advances in the use of the
BRAFV600E mutation as a potential target of thera-
peutic strategies for thyroid cancers.
MAPK Signal Transduction Pathway
The MAPK pathway is an intracellular signal transduc-
tion pathway that is required for maintaining cellular
activities such as cell growth, proliferation, differenti-
ation, and apoptosis responsive to cell surface receptor
tyrosine kinase (RTK) stimulation.18,19 This pathway
relays the extracellular signals from various growth fac-
tors, hormones and cytokines to the nucleus through
the activation of signal cascades. As shown in Figure 1,
the binding of the ligands to their surface RTKs lead
to the dimerization of receptors and tyrosine residue
autophosphorylation. The activated receptors, through
adaptor proteins, activate RAS kinase. Then, RAS ki-
nase activates the phosphorylation of Raf kinases, which
in turn activate the dual-specificity protein kinases:
MAP kinase kinases (MAPKK; also known as MAP/
extracellular signal-regulated kinase, MEK) 1 and 2.
MEK1/2 phosphorylate and activate extracellular
signal-regulated kinases (ERK) 1 and 2. ERK1/2
regulate various transcription factors leading to gene
expression.
RAS kinase belongs to a family of small G-proteins
(KRAS, HRAS, NRAS) located on the inner surface
of cell membranes and function as a GTPase, switching
between active GTP-bound form and inactive GDP-
bound form. Cycling between GDP/GTP is regulated
by adaptors (e.g. growth-factor-receptor bound-2;
GRB2) and guanine nucleotide exchange factors (e.g.
son of sevenless; SOS). These proteins facilitate the RAS
active GTP-bound form formation, and RAS GTPase
catalyzes GTP hydrolysis, resulting in return to its
inactive GDP-bound form.20–22 Raf kinase was the first
identified and most characterized downstream cyto-
solic effector of RAS.23,24 It belongs to a family of
serine/threonine kinases (A-Raf, B-Raf, and C-Raf or
Raf-1), and all isoforms share 3 common conserved
regions—CR1 (RAS-binding domain and cysteine-rich
domain), CR2 (N-terminal regulatory domain), and
CR3 (C-terminal kinase catalytic domain)—as well as
several regulatory phosphorylation sites (Figure 2).25,26
The activation of Raf is a complex process taking
place at the membrane, where Raf undergoes multi-
site phosphorylation and protein interactions before
being rendered active.17,27–29 The binding of RAS to
RAS-binding domain (RBD) of Raf is regulated by di-
meric adaptors such as 14-3-3 proteins that are bound
to the phosphorylated proteins. BRAF contains con-
served phosphorylation sites at S365 and S729 that
are phosphorylated in the inactive state. Dimeric pro-
teins 14-3-3 bind to these phosphorylated sites, creat-
ing a conformation that interferes with the binding of
RAS to RBD.29 The activation of BRAF is initiated with
the recruitment of the inactive BRAF to the inner mem-
brane, where the N-terminal 14-3-3 binding site is
dephosphorylated to dissociate 14-3-3 proteins, and
followed by the phosphorylation of T599 and S602
at the activation segment (Figures 1 and 2).26 Inactive
BRAF exhibits a characteristic bilobar structure by
forming a hydrophobic interaction between residues
G596-V600 of the activation loop and residues G464-
V471 of the P loop (ATP binding sites), resulting in a
conformation that the catalytic residues cannot bind
to ATP. It is postulated that phosphorylation of T599
disrupts the hydrophobic interaction between these 2
loops, resulting in the binding of ATP and the activation
of BRAF.17 The basic mechanisms in the activation of
the 3 Raf isoforms are similar, except that A-Raf and
J Chin Med Assoc March 2010 Vol 73 No 3
114
K.T. Tang, C.H. Lee
J Chin Med Assoc March 2010 Vol 73 No 3 115
BRAF mutation in papillary thyroid cancer
Figure 1. MAPK signal transduction pathway. In normal cells, ligands’ (L) binding of the extracellular domain of their membrane tyrosine
kinase receptors (RTK) triggers dimerization of the receptor autophosphorylation of tyrosine residues in the intracellular TK domain
activation of adaptor proteins/guanine nucleotide exchange factors (e.g. Grb2 and SOS) inactive GDP-bound form of RAS active
GTP-bound form BRAF recruitment to the membrane BRAF phosphorylation MEK phosphorylation ERK phosphorylation
nuclear translocation ERK-induced phosphorylation of nuclear transcription factors gene expression proliferation, survival,
senescence and differentiation. Raf–MEK–ERK kinase cascade is scaffolded by kinase suppressor of Ras (KSR). Protein phosphatase
2A (PP2A) is involved in the dephosphorylation of inhibitory sites of Raf kinases during their activation process.
C-Raf require additional kinases (e.g. SRC) and more
phosphorylation steps at the N-terminal side of CR3
(N-region). Negative charge within the N-region is
essential for Raf kinase activation. BRAF is constitu-
tively phosphorylated at S446, and a regulatory tyro-
sine residue is occupied by an aspartic acid at D449,
such that its constant negative charge will act like the
phosphorylation at this site.30 As a result, BRAF is
activation-ready and only requires the RAS-mediated
membrane recruitment of BRAF.
MEK1/2 are the physiological downstream effec-
tors of BRAF. BRAF has the highest basal kinase activ-
ity and is the strongest Raf activator of downstream
MEK.31,32 Activated BRAF induces phosphorylation at
2 serine residues, S217 and S221, within the activation
segment of MEK. Downstream of MEK are ERK1/2,
which are activated by phosphorylation at the T202
and Y204 residues of ERK.32 Phosphorylation of ERK
activates substrates located in the nucleus and cyto-
plasm. The majority of ERK substrates are nuclear
proteins, and the nuclear translocation of ERK phos-
phorylates various transcription factors, which in turn
regulate gene expression.33–35 Meticulous regulation
of ERK is crucial to maintain biological homeostasis
responsive to various extracellular signals. For example,
hyperactivation of the ERK pathway can induce cell
cycle arrest and senecense.36,37 In contrast, aberrant
activation of the pathway may induce tumor transfor-
mation (Figure 3).13,17 The kinetics and amplitude of
ERK signaling induced by different ligands can regulate
biological programs differentially, such as proliferation,
differentiation or apoptosis. The regulation of cellular
responses is a complicated mechanism that may in-
volve various substrates at different levels of the cascade,
such as scaffold proteins and feedback inhibitors (Figure
4).38 Apparently, tumors prefer ERK activities’ program-
ming for proliferation and survival.
Genetic Alterations in PTC
Aberrant activation of the MAPK pathway is frequently
found in human cancers (Figure 3). The consistent
finding of RAS and BRAF mutations in similar cancer
J Chin Med Assoc March 2010 Vol 73 No 3
116
K.T. Tang, C.H. Lee
Figure 2. Structure of the Raf proteins. The Raf isoforms, A-Raf, B-Raf and C-Raf, share 3 conserved regions: CR1, CR2 and CR3. The
amino acids shown refer to known phosphorylation sites. CR1 contains the RAS-binding domain (RBD) and the cysteine-rich domain
(CRD), which are both required for membrane recruitment. CR2 and C-terminal contain the 14-3-3 binding sites. CR3 contains the cat-
alytic domain (including the activation segment). The negative-charge regulatory region (N-region) contains residue C-Raf (Y341), which
is conserved in A-Raf (Y302) but is replaced by aspartic acid at D449 in BRAF. S338 of C-Raf is conserved in all RAF proteins (S299 in
A-Raf and S446 in BRAF), but it is constitutively phosphorylated in BRAF (star shape). The catalytic domain contains the 2 activation-
segment phosphorylation sites C-Raf (T491 and S494), which are conserved in A-Raf (T452 and T455) and BRAF (T599 and S602).
Figure 3. Activation of MAPK signaling pathway by RAS, RET/PTC and BRAFV600E mutations. The mechanism is similar to the physiolog-
ical condition described in Figure 1, except that the signal is generated through RAS, RET/PTC and BRAFV600E mutations. The activation
of the MAPK pathway becomes constitutive to induce cell transformation.
types, and the mutation rarely involving more than 1
component of the pathway (mutually exclusive), sug-
gest that the constitutive activation of these mutants
in the pathway might be the pathogenesis of tumor
formation. They also imply that a single gene muta-
tion in the pathway is sufficient to induce cell trans-
formation.13,38–40 About 70% of patients with PTC are
found to have genetic alterations related to the MAPK
pathway, such as RET/PTC rearrangement, and RAS
and BRAF mutations, indicating that the MAPK sig-
naling pathway plays an important role in the patho-
genesis of PTC.40,41
RET protooncogene is a tyrosine kinase receptor
that is highly expressed in parafollicular C cells, but its
expression is low in thyroid follicular cells. The RET
gene can be activated in follicular cells by chromosomal
rearrangements, linking the promoter and N-terminal
domains of unrelated genes to the tyrosine kinase do-
main of the RET gene. The aberrant production of
different chimeric forms of the receptor is known as
RET/PTC. More than 11 types of RET/PTCs have
been reported, and RET/PTC1 and RET/PTC3 are
the most common rearrangements seen in PTCs, prob-
ably because RET/PTC1 and RET/PTC3 are intra-
chromosomal (chromosome 10q) paracentric inversions
by fusion of the 3’ portion of RET to the 5’ portion of
the H4 (D10S170) and NCOA4 (ELE1) genes, res-
pectively.42,43 The other RET/PTCs are rearranged by
interchromosomal translocations. As shown in Figure 3,
RET/PTCs constitutively activate the RAS/BRAF/
MAPK pathway,44–46 and the transformation of thyroid
cells could be induced by overexpression of either
RET/PTC1 or RET/PTC3 in cultured thyroid fol-
licular cells and transgenic mice. Silencing the BRAF in
RET/PTC-transformed follicular cells reversed the
tumorigenic effect of RET/PTC, confirming that sig-
naling along the BRAF-MAPK pathway is required for
its tumorigenesis.43,44,47–49 The prevalence of RET/
PTC rearrangements in PTC is varied in different geo-
graphic regions and account for 20% of PTC, which
is particularly common in young patients and individ-
uals with history of previous radiation exposure.50
RAS proteins are plasma membrane GTPases
switching between active GTP-bound form and inac-
tive GDP-bound form to activate downstream effector
pathways.20–22 Point mutations of RAS, either with
increased affinity for GTP (at codons 12 and 13) or
with decreased autocatalytic GTPase function (at codon
61), will withhold RAS at the active GTP-bound con-
dition leading to the constitutive activation of the
J Chin Med Assoc March 2010 Vol 73 No 3 117
BRAF mutation in papillary thyroid cancer
Figure 4. Proposed model of feedback inhibition in tumor cells with RET/PTC or RTKs and with BRAFV600E. RET/PTC or RTKs activate
the tumor cell feedback mechanism and inhibit the MAPK pathway at multiple levels. Feedback mechanism or mediators such as dual-
specificity phosphatases (DUSPs) downregulate both RAF/MEK activation and ERK phosphorylation in RTK cells. BRAFV600E is constitu-
tively active and insusceptible to negative feedback.
MAPK pathway (Figure 3). Mutations of all family
members of RAS genes have been reported in differ-
ent types of thyroid follicular cell-derived tumors,
particularly in follicular adenomas and carcinomas.
However, it rarely occurs in PTCs (10%) and is almost
solely found in follicular variants.51–53
BRAF is highly expressed in hematopoietic cells,
neurons, testis and thyroid follicular cells.54,55 Contrary
to the extremely rare mutation found in A-Raf and C-
Raf, BRAF is the most common Raf mutation and is the
second most common somatic mutation in all human
cancers. The BRAF mutation is frequently detected in
malignant melanomas, and in colon, ovarian and thy-
roid carcinomas.13,14,17 Although >45 BRAF mutations
have been identified in human cancers, about 90% of
BRAF mutations are T A transversion in exon 15 at
nucleotide 1799 (T1799A) leading to a valine glu-
tamic acid replacement at position 600 (BRAFV600E).
Except for the rare BRAF mutations (K601E, AKAP9-
BRAF, V600E +K601del, V599ins, V600D +
FGLAT601-605ins) reported in thyroid cancer, over
90% of BRAF mutation in PTCs are BRAFV600E.13,56–60
The prevalence of BRAFV600E in PTCs varies from
29% to 83%, and is 60% in classic PTC, 77% in tall-
cell variant, and 25% in PTC-derived ATC, but it is
rare (0–12%) in follicular-variant PTC and is not
found in FTC.15 An in vitro study showed that the
replacement of a negative-charge residue glutamate at
V600 adjacent to T599 induces an effect similar to
phosphorylation at T599 and S602, which disrupts
the hydrophobic interaction between the P loop and
the activation loop.17 The kinase activity of BRAFV600E
is 460-fold higher than the wild-type BRAF, and this
active conformation can constitutively activate its down-
stream effectors to transform normal cells or induce
cancer proliferation without the need of RAS for acti-
vation.13,17
BRAFV600E mutation is frequently detected in pap-
illary thyroid microcarcinomas (PTMC), suggesting
that the mutation is an early event during PTC devel-
opment.61–65 The tumorigenic role of BRAFV600E in
the development of PTC was documented in thyroid-
targeted BRAFV600E transgenic mice, and tumors devel-
oped from these transgenic mice were found to
progressively transform into poorly differentiated can-
cers with aggressive characteristics.66 In vitro,
BRAFV600E-overexpressed rat thyroid cells grown on
MatrigelTM showed an increase in migration of thyroid
cells. It has also been reported to be associated with
the upregulation of metalloproteinases (MMPs), par-
ticularly matrix MMP3, MMP9 and MMP13 genes,
which are related to tumor invasion.45,67 The prolifer-
ation of BRAFV600E-harbored or transfected cell lines
could be inhibited by MAPK pathway inhibitors or
siRNA specific BRAF knockdown.68,69 The above ob-
servations suggest that BRAFV600E is an initiator of
tumorigenesis through the MAPK pathway, and is
required for the progression of PTC.70 Both RET/
PTC and BRAFV600E mutations can constitutively acti-
vate MAPK pathways, resulting in follicular cell transfor-
mation. However, several studies have shown that RET/
PTC-expressed rat thyroid cells induce a weaker tumor-
igenic effect than BRAFV600E mutation.71 Conditional
expression of BRAFV600E in thyroid cells markedly
increased the MatrigelTM invasion of the transformed
thyroid cells, which is more invasive than RET/PTC
expressed cells.67 Furthermore, microarray studies have
also shown that human PTCs harbor BRAFV600E and
RAS mutation, and RET/PTCs exhibit different gene
expression profiles, suggesting that different mutants
may affect the pathway outcomes differently. Among
them, BRAFV600E mutation is the most potent activa-
tor in the stimulation of MAPK pathway output.72
Interestingly, the high kinase activity of BRAFV600E
mutation seems to be not effectively translated to ERK
activity; it only increased ERK activity 2- to 4.6-fold,
suggesting the existence of regulatory mechanisms in
the controlling of the signal output.17,27 Several pos-
sible feedback mechanisms have been reported to
inhibit the ERK pathway output.73–76 ERK stimulates
gene expression of feedback regulators (e.g. dual-
specificity phosphatases; Sprouty) to inhibit RAS ac-
tivating proteins (e.g. SOS), and also ERK’s own
activities (Figure 4).73,74 Phosphorylated-ERK has been
demonstrated to phosphorylate Raf directly to induce
hyperphosphorylation of Raf, leading to conforma-
tional changes that might interfere with the binding
of Raf to RAS, MEK or scaffold proteins.77 The phys-
iological role of these feedback loops is unclear, and it
may aim to prevent cell cycle arrest and senescence
due to hyperactivation of the ERK pathway. The con-
stitutive activation of the MAPK signal pathway by
RAS and BRAFV600E mutations was also found to
induce feedback downregulation similar to the physi-
ological condition.78 Conceivably, these oncoproteins
might take steps to minimize the effects of feedback
inhibition by either insensitivity of the mutant protein
to normal negative feedback or directly affecting the
mediators of the feedback mechanism. Recent investi-
gation demonstrated that feedback inhibition of
Raf/MEK signaling was found to downregulate ERK
output in RTK cells, but not in BRAFV600E cells
(Figure 4). The evasion of the feedback mechanism in
BRAFV600E cells was evidenced by the increase in
transcriptional output and MEK/ERK-dependent
transformation. This phenomenon may partly explain
J Chin Med Assoc March 2010 Vol 73 No 3
118
K.T. Tang, C.H. Lee
the stronger tumorigenic effect observed in the
BRAFV600E-expressed cells than in the RET/PTC-
expressed cells.78
Tumorigenesis is a complex process involving mul-
tiple signaling networks instead of a single linear uni-
directional cascade of the MAPK pathway. The high
prevalence of BRAFV600E mutation in tall-cell variant
and PTC-derived ATC suggests that BRAFV600E muta-
tions may play a role in the progression of PTC to
more aggressive thyroid carcinomas.15 Recent studies
have shown that induction of BRAFV600E expression
in rat thyroid cells facilitated the acquisition of sec-
ondary genetic events through induction of genomic
instability, but not in RET/PTC-expressed cells.79
Genetic alternations in the PI3K/Akt pathway and
PTEN have also been found in thyroid cancers such
as ATC and metastatic tumors from radioactive iodine-
refractory (RAIR) PTC, particularly in the later stages
of cancer progression.80 Aberrant activation of the
PI3K/Akt pathway is often coexistent with BRAFV600E
in ATC and RAIR tumors, suggesting that the genetic
instability induced by the primary BRAFV600E mutation
in PTC may facilitate the secondary genetic alterna-
tions involving the PI3K/Akt pathway. The secondary
mutation might lead to the progression of DTC to the
more aggressive thyroid cancer, and dedifferentiation
of the cancer cells.81 This hypothesis is supported by
observations that tumors in thyroid-targeted BRAFV600E
transgenic mice progressed to more aggressive phe-
notype, and BRAFV600E mutation is associated with
advanced patient age and not frequently detected in
childhood PTCs.15,66,82 Further investigations of the
feedback and alternation pathways are important in
understanding the mechanisms involved in the tu-
morigenesis of PTC, which might provide valuable
information regarding clinical implications.
Clinical Implications Associated With
BRAFV600E Mutation in PTC
Recently, BRAFV600E has taken center stage due to the
findings that it may be associated with tumorigenesis
and aggressiveness. Many studies have been done to
investigate the clinicopathologic characteristics, and
the potential utility of BRAFV600E mutation on the
diagnostic, prognostic and therapeutic aspects of PTC.
Unlike the highly consistent results obtained from
in vitro studies, the current clinical data show con-
troversial results regarding BRAFV600E mutation as a
genetic prognostic marker of PTC.
Environmental and various predisposing factors have
been reported to increase the risk of thyroid cancer,
such as radiation exposure, dietary iodine, genetics
and life style.83 Radiation-associated PTCs are usually
associated with RET/PTC and to a lesser extent with
NTRK1, but not with BRAFV600E mutation.84–86
Ciampi et al57 reported that AKAP9-BRAF fusion
was more commonly found in radiation-induced PTC
than sporadic PTC. AKAP9-BRAF is caused by para-
centric inversion of chromosome 7q, resulting in an in-
frame fusion between exons 1–8 of the AKAP9 gene
and exons 9–10 of BRAF.87 This implies that radiation-
induced PTCs are likely caused by the chromosome-
paracentric inversion linked to constitutive activators,
while sporadic PTCs are predominantly activated by
point mutations on the effector kinases of the MAPK
pathway. In Italy, a higher incidence of thyroid cancer
and BRAFV600E-PTC were found in people residing
in Eastern Sicily, including the volcanic area of Etna
(45.9%), than people in Western Sicily (22.7%). Iodine
deficiency was not found to be the cause of the differ-
ence, and the unidentified carcinogens were suspected
to be the volcanic soil, water or atmosphere.88 A large
Chinese epidemiological study of 1,032 conventional
PTCs demonstrated that cities with high iodine con-
tent had significantly higher incidence of BRAFV600E
mutation (69%) than cities with normal iodine content
(53%). The results suggest that high iodine intake is a
risk factor for BRAFV600E mutation and may therefore
be a risk factor for PTC development.89
Over 30 studies on the relationship between
BRAFV600E and the clinicopathological characteristics
in PTC have been reported worldwide. The majority
of them suggested that BRAFV600E mutation was
associated with advanced disease stages and aggressive
phenotype, while others did not find this association;
the results remain controversial to date. Several stud-
ies reported a significant association of BRAFV600E
mutation with high-risk clinicopathological character-
istics such as older age,65,90–96 male sex,97,98 tumor
size,88,98–101 and aggressive subtype.63,65,92,94,102–104
Many studies found that extrathyroidal invasion, lymph
node metastasis, and advanced stages III/VI are the 3
most common risk factors consistently associated with
BRAFV600E mutation.63,65,87,88,90–96,98–100,102,105,106
Oler et al60 and Vasko et al107 observed that BRAFV600E
mutation in lymph node metastasis was occasionally
not found in their primary lesion, suggesting that tu-
mor cells that acquire the mutation de novo are proba-
bly prompted to metastasis. Rodolico et al95 further
demonstrated that metastatic lymph nodes harboring
the BRAFV600E mutation were larger in size and had a
higher prevalence of extracapsular invasion than those
without the mutation. However, other studies on
paired primary and lymph node metastatic lesions did
J Chin Med Assoc March 2010 Vol 73 No 3 119
BRAF mutation in papillary thyroid cancer
not find discordant mutation in most of the lesion
pairs, indicating that the acquisition of BRAFV600E
mutations are not a requirement in the progression
from localized to metastatic PTC.92,105,108 In a large
Italian cohort study, Lupi et al63 found that BRAFV600E
mutation was associated with the absence of tumor
capsule, particularly in follicular- and micro-PTC vari-
ants, but not in conventional variant. Two meta-
analyses also reported an association of BRAFV600E
mutation with extrathyroidal invasion, aggressive his-
totype and advanced disease stages, but not with age,
sex, or tumor size, and the association of BRAFV600E
mutation with lymph node metastasis is not a uniform
finding.15,109 In fact, a recent large Chinese cohort
study investigating the association of iodine intake
with BRAFV600E mutation in different cities demon-
strated that overall results of extrathyroidal invasion,
lymph node metastasis and advanced disease stages were
significantly associated with BRAFV600E mutation, but
the association with extrathyroidal invasion and lymph
node metastasis were not seen in all cities when ana-
lyzed city by city.89
Clinicopathological characteristics and staging sys-
tems are designed to predict tumor recurrence and
disease prognosis.9Three studies, including a multi-
center study of 219 patients, an American study of
245 conventional PTC cases, and an Italian study of
102 patients, demonstrated that BRAFV600E mutation
was associated with aggressive clinicopathological fea-
tures, and was also an independent predictor of tumor
recurrence after a median of 15 months, 6 years, and
15 years of follow-up, respectively.92,100,106 Kim et al98
reported that BRAFV600E mutation was associated with
tumor recurrence in 203 conventional PTC follow-ups
for a median of 7.3 years, but was not an independent
predictor after adjustment for clinicopathological prog-
nostic factors. More importantly, the association was
also observed in patients with low disease stages I/II,
and conventional PTC.92,98,106 Another finding from
a study of 54 recurrent PTCs showed that 77.8% of
tumors were found to harbor BRAFV600E mutation and
9.3% had both BRAF and RET/PTC mutations.110
The results give further support to the premise that
secondary mutations may cause tumors to progress to
a more aggressive status. In contrast to the studies
above, in a recent large Japanese cohort study of 631
patients with PTC who were followed-up for a median
of 83 months, neither clinicopathological character-
istics nor tumor recurrence was associated with
BRAFV600E mutation.111 A number of studies from
various ethnic groups and geographic regions also did
not find any association of BRAFV600E mutation with
the aggressive clinicopathological features.62,64,112–119
The conflicting results of these studies might be due
to variations in the study populations in terms of size,
age distribution, histological variants, genetic factors,
environmental factors, disease stages at the time of
initial diagnosis, and the methods or criteria used.
Some studies demonstrated that BRAFV600E-
mutated PTC is associated with high recurrence rate,
and a decrease in radioiodine uptake in the recurrent
tumor.102,106 These observations were supported by a
recent study of patients with RAIR-differentiated PTC,
in which 62% of patients were found to be BRAFV600E-
positive, and 54% were 18F-fluorodeoxyglucose pos-
itron emission tomography (18-FDG-PET)-positive.
Interestingly, all of the 18-FDG-PET-positive patients
were found to be BRAFV600E-positive.80 In the process
of thyroid hormone synthesis, inorganic iodine is
actively transported into the thyroid cells via a basal
membrane protein—sodium iodide symporter (NIS).
Iodide is, in turn, transported into the follicle via an
apical protein—pendrin, where iodide is oxidized by
thyroid peroxidase (TPO) and incorporated into thy-
roglobulin in the synthesis of thyroid hormone. This
process is regulated by thyroid-stimulating hormone
(TSH) through the binding of the membranous TSH
receptor (TSHR). Recent immunohistochemistry study
showed that tumor tissues with BRAFV600E mutation
had lower NIS expression, and failure of NIS targeted
to the membrane when compared with PTC without
the mutation.102 BRAFV600E mutation was also found
to be associated with a decrease in gene expression of
TSHR, TPO, NIS, thyroglobulin and pendrin in pri-
mary or recurrent tumors.72,99,102,113,120,121 Instead
of iodide-metabolizing gene silencing, glucose trans-
porter-1 (GLUT-1) expression was found to be in-
creased in PTC, which was significantly higher in
tumors with BRAFV600E mutation than wild-type.120
These data support the biological basis for the clinical
use of 18-FDG-PET to detect recurrent/metastatic
lesions in patients with RAIR PTC. Recently, Romei
et al119 reported that there was neither association of
BRAFV600E mutation with clinicopathological charac-
teristics nor with GLUT-1/3 expression in PTC, but
there was consistently lower expressions of NIS and
TPO in BRAFV600E-mutated PTC; the lower expres-
sions of NIS and TPO were not seen in PTC with
RET/PTC rearrangement. A recent Brazilian study
also reported that decreased NIS gene expression was
found in conventional PTC and PTMC harboring
BRAFV600E mutation.99 In vitro, conditional BRAFV600E
expression in rat thyroid cell lines suppressed iodide-
metabolizing genes. Inhibition of MAPK pathway or
silencing BRAF using inhibitors or siRNA restored the
expression of the iodide-metabolizing genes.79,102,122
J Chin Med Assoc March 2010 Vol 73 No 3
120
K.T. Tang, C.H. Lee
The results indicate that the MAPK pathway plays an
important role in the regulation of iodide-metabolizing
genes, and several studies further demonstrated that
the MAPK pathway promotes expression of DNA
methyltransferase, which silences these genes through
promoter methylation.122–124
The current treatment strategies for PTC allow
patients at low risk to undergo less intensive postop-
erative adjunctive therapy and less frequent follow-up
than patients at high risk, whereas patients in advanced
stages, with aggressive histotype, or at high risk are
managed by more aggressive therapy such as total/
near total thyroidectomy, lymph node dissection, adju-
vant radioiodine ablation and thyroid hormone sup-
pression therapy.10,125,126 For patients at high risk, the
preoperative diagnosis of BRAFV600E mutation seems
to have little additional value in the current treatment
and follow-up protocols. However, the majority of
patients with DTC are classified as low risk, and many
of them suffer from tumor recurrence in later years.
Testing for BRAFV600E mutation might be helpful in
tailoring the therapeutic strategies for these patients,
in case BRAFV600E mutation is proven to be a marker
of poor prognosis.
The incidence of PTMC (tumor size <1 cm in
diameter) has dramatically increased since the intro-
duction of high-resolution ultrasound-guided fine-
needle aspiration (FNA) biopsy for patients with
nodular thyroid disease. The prevalence of PTMC has
increased approximately 2- to 4-fold in various coun-
tries during the last 2 decades, and the rapid rise of
PTC in recent years is mainly due to the increased rate
of diagnosis of PTMC.127–130 PTMC accounts for ap-
proximately a quarter of thyroid cancers.131 It is gen-
erally considered to be a low-risk cancer, and most cases
are classified as stages I/II. Recommended treatment
for these small low-risk tumors, in the absence of known
risk factors or lymph node metastasis, is lobectomy
with or without isthmectomy.10,125 However, multi-
focality, extrathyroidal extension, and lymph node me-
tastasis are often reported in PTMC, with incidences
of 7.1–56.8%, 2–62.1%, and 0–64%, respectively.131
Several studies have compared the clinical and histo-
logic characteristics between PTC and incidental or
nonincidental PTMC, and found that the prevalence
of multifocality, extrathyroidal extension and lymph
node metastasis are similar in PTMC and PTC. The
aggressive phenotypes are more frequently found in
nonincidental PTMC and PTMC with size larger than
5 mm and 8 mm, respectively.132–135 BRAFV600E muta-
tion is also the most common genetic alteration in
PTMC, accounting for 17–65.6%, with incidence lower
than that of PTC in general.61–65,88,136 Recently, several
studies have investigated the relationship between clin-
icopathological characteristics and BRAFV600E-mutated
PTMC. Park et al136 reported a surprisingly high preva-
lence of extrathyroidal invasion (52.2%) and lymph
node metastasis (32.9%) in PTMC; the frequency of
BRAFV600E mutation and the recurrence rate of PTMC
were similar to those of PTC. Lupi et al63 and Frasca
et al88 observed that BRAFV600E-PTMC was associ-
ated with extrathyroidal extension and advanced dis-
ease stages. Their findings are consistent with the recent
report from Lee et al137 that more BRAFV600E mutation
was detected in PTMC with advanced disease stages,
extrathyroidal extension, and nodal metastasis than in
those without these aggressive clinicopathological char-
acteristics. Ugolini et al61 reported an association of
BRAFV600E mutation with the lack of tumor capsule in
PTMC. Rodolico et al95 further found that BRAFV600E
mutation was associated with lymph node metastases,
a wider diameter of the largest metastatic area, a higher
number of involved lymph nodes, and a higher percent-
age of metastatic lesions with extracapsular extension
in PTMC. Together, the clinicopathological charac-
teristics of BRAFV600E-PTMC seem to be no different
from those of its larger counterpart, and BRAFV600E-
PTMCs exhibit signs of greater aggressiveness and
higher recurrence rate than wild-type. Most patients
with PTMC are classified as stages I/II. According to
current treatment strategies, these patients might re-
ceive “inadequate” treatment and less frequent follow-
up. Thus, some investigators have suggested evaluating
BRAFV600E mutation in these patients preoperatively,
and treating patients with positive BRAFV600E muta-
tion more aggressively.
Use of BRAF Mutation for
Preoperative Diagnosis
Preoperative evaluation of nodular goiter is based on
FNA cytology to select patients for surgical treatment
or medical follow-up.138 As BRAFV600E mutation and
RET/PTC rearrangements are exclusively found in
PTC, examining these markers in DNA specimens
obtained from FNA can make a diagnosis in most
PTCs. Salvatore et al139 detected 38% and 18% of
BRAFV600E and RET/PTC in FNA samples, respec-
tively. The identification of BRAFV600E mutation and
RET/PTC refined the diagnosis of PTC in 5 of 15
samples that were considered either indeterminate or
insufficient at cytology. Using FNA BRAFV600E analy-
sis, Cohen et al140 confirmed the BRAFV600E mutation
in 72% of carcinomas within the malignant group,
and established the diagnosis of PTC in 16% of the
J Chin Med Assoc March 2010 Vol 73 No 3 121
BRAF mutation in papillary thyroid cancer
indeterminate group. No BRAFV600E mutation was de-
tected in the benign group. Two recent studies from
Marchetti et al141 and Zatelli et al142 demonstrated
that combining traditional cytology and molecular
analysis of BRAFV600E mutation on FNA specimens
improved the diagnostic accuracy of PTCs from 62.3%
to 82.2% and from 77.3% to 86.7%, respectively. How-
ever, BRAFV600E mutation is only positive in 50% of
PTCs, and negative results cannot exclude malig-
nancy. Therefore, the sensitivity of BRAFV600E muta-
tion analysis for PTC diagnosis is limited, although the
specificity is high. Most of the indeterminate speci-
mens are follicular neoplasm and follicular variant of
PTC, which are rarely found to harbor BRAFV600E
mutation. Inadequate FNA may lead to insufficient
tumor DNA recovery from the nucleic acid prepara-
tions, which might lead to false-negative results. In
fact, traditional FNA cytology by expert pathologists
can provide reliable information on PTCs with an
overall accuracy >90%.138 Therefore, the value of rou-
tine BRAFV600E mutation analysis for PTC diagnosis in
FNA is marginal. It is more reasonable to reserve the
test for patients with indeterminate/inadequate FNA
cytology, which may improve the diagnostic yield in
these patients.
A recent study reported that RAS and PAX8/PPAR
gene analysis in addition to BRAFV600E and RET/PTC
analysis in FNA specimens enhanced the diagnostic
accuracy of FNA cytology, particularly in indetermi-
nate cytology. In the study, 97% of nodules with posi-
tive mutations were ultimately found to be malignant,
suggesting that additional RAS and PAX8/PPAR analy-
sis improved the diagnostic accuracy of indeterminate
cytology which are predominantly follicular neoplasm
and follicular variant of PTC.143 Xing et al144 investi-
gated the utility of BRAFV600E mutation analysis of
FNA specimens for preoperative risk stratification in
PTC. Their results showed a significant association of
BRAFV600E mutation with poor clinicopathological
outcomes, and BRAFV600E mutation predicted extra-
thyroidal extension, thyroid capsular invasion, and
lymph node metastasis. More importantly, 36% of
PTCs with BRAFV600E mutation were found to have
tumor persistence/recurrence, compared with 12% of
PTCs without BRAFV600E mutation, after a median of
3 years’ follow-up (odds ratio, 4.16). The positive and
negative predictive values for the test to predict tumor
persistence/recurrence were 36% and 88% for all PTCs,
and 34% and 92% for conventional PTCs, respec-
tively.144 Preoperative FNA for BRAFV600E diagnosis
seems to be helpful for tailoring the treatment strate-
gies for PTC with low grade and for PTMC. However,
further investigations in large randomized, controlled,
prospective trials are necessary to confirm the role of
BRAFV600E mutation as a clinically useful prognostic
marker.
Use of BRAF Mutation for Therapeutic
Decisions
As mentioned above, the constitutive activation of
BRAFV600E mutation in the MAPK pathway seems to
be the cause of tumorigenesis and progression in
PTC.13,14,17 Using inhibitors that target BRAF kinase
or its downstream effectors is the logical therapeutic
approach to inhibit tumor growth and progression of
PTCs. Different trials have evaluated the anticancer
effects of BRAF inhibitors, and the preclinical results
are encouraging. Nonselective BRAF inhibitors AAL-
881 and LBT-613 are isoquinolone compounds that
have been shown to inhibit cell cycle progression from
S-phase to G2-M phase and G0-G1 arrest, resulting
in growth reduction and apoptosis in several thyroid
cancer cell lines and in xenograft tumors.68 Specific
knockdown of BRAFV600E by siRNA inhibited the
growth of ATC cell lines, the growth and transfor-
mation of BRAFV600E-mutated PTC cells, and prolif-
eration and tumorigenesis in xenograft tumors.69,70
PLX4032, a small-molecule-specific BRAF inhibitor,
arrested the cell growth of ATC cells and NPA human
thyroid cancer cell lines harboring BRAFV600E mu-
tation.145 Prolonged treatment of ATC cells with
PLX4032 induced the re-expression of NIS. In thy-
roid cancer cell lines bearing the RET/PTC1 and
wild-type BRAF, PLX4032 showed an approximately
50-fold higher IC50 value than BRAFV600E cell lines,
indicating that PLX4032 has selective growth in-
hibitory effect on BRAFV600E-mutated thyroid cancer
cells.145 U0126 is a MEK inhibitor that has been
reported to inhibit the growth of the thyroid cell-
expressed BRAFV600E and restore the expression of
iodide-metabolizing genes.122 AZD6244 is a potent
MEK1/2 inhibitor that has been demonstrated to
inhibit ERK phosphorylation in thyroid cancer cell
lines regardless of the status of BRAFV600E mutation,
and the dose required to inhibit the cell growth in 4
BRAFV600E mutant cell lines is lower than that for the
2 wild-type cell lines. AZD6244 has also been shown to
inhibit the growth of xenograft tumors derived from
BRAFV600E mutant ATC cell lines.146 Another small-
molecule potent MEK1/2-selective inhibitor, CI-1040
(PD-184352), inhibited cancer cell proliferation and
tumor xenografts derived from various cancer cells
harboring BRAFV600E or RAS mutations.147 CI-1040
inhibited the growth and induced re-expression of
J Chin Med Assoc March 2010 Vol 73 No 3
122
K.T. Tang, C.H. Lee
some thyroid genes in thyroid cancer cell lines with
BRAFV600E mutation, but not in cells with RET/PTC
or wild-type alleles.148 The results suggest that the
inhibition effects of CI-1040 on tumor cell prolifera-
tion are BRAF or RAS mutation-selective. CI-1040 is
the first MEK inhibitor to enter clinical trials to evalu-
ate its performance in the treatment of lung, colon,
breast and pancreatic cancers. However, no significant
clinical anticancer effect was observed.149,150
BAY43-9006 (sorafenib), is the most studied
multi-kinase inhibitor for targeting BRAF and angio-
genesis-related RTK. It has been shown to inhibit the
proliferation of ATC lines and tumor xenografts. How-
ever, the effect seems to be caused by blocking angio-
genesis via VEGFR signaling rather than by inhibiting
BRAF selectively.151 Several clinical trials that studied
sorafenib monotherapy for the treatment of various
malignancies including iodine-resistant thyroid can-
cer have been completed recently. Although phase I
trials showed encouraging results that sorafenib was a
well tolerated agent, phase II trials showed little or no
antitumor effects in advanced melanoma patients when
sorafenib was used as a single-agent therapy.152 Re-
cently, a longer than 16-week phase II trial of sora-
fenib in 30 patients with metastatic iodine-refractory
thyroid carcinoma showed an overall clinical benefit
of 77%, 70% with thyroglobulin reduction, and a me-
dian 79-week progression-free survival.153 Another
phase II trial of sorafenib in patients with metastatic
thyroid cancer also showed a similar antitumor activity,
with a median progression-free survival of 64 weeks,
and a reduction in the levels of VEGFR phosphoryla-
tion, ERK phosphorylation, and VEGF expression in
tumor biopsies.154 To date, there is no evidence to show
that the antitumor effects of sorafenib are through
the inhibition of BRAF. Sorafenib is a multi-kinase
inhibitor that may also target other kinase pathways
such as VEGFR to inhibit tumorigenesis. Indeed, anti-
tumor effects were also observed with other kinase
inhibitors such as axitinib and motesanib that inhibit
VEGFRs and PDGFRs.155,156 Factors other than BRAF
mutation may affect tumor response to sorafenib, and
the combination of different kinase inhibitors and/or
chemotherapy may be a potential therapeutic strategy
in the future.
Conclusions and Perspectives
The discovery that BRAFV600E is the most common
mutation in PTC, and molecular studies demonstrating
its tumorigenic role in PTC, suggest that BRAF muta-
tion is the initiator of PTC. The notion of the inherited
strong kinase potency of BRAFV600E with the ability to
induce genetic instability, silence iodide-metabolizing
genes, and evade the feedback mechanisms which
may promote the progression and aggressiveness of
PTCs is comprehensible. Indeed, the majority of clin-
ical studies that support the association of BRAFV600E
with aggressive clinicopathological characteristics and
higher tumor recurrence make BRAFV600E mutation a
potential diagnostic and prognostic marker, although
quite a number of studies did not find any association.
Using BRAFV600E mutation in FNA specimens as a
diagnostic marker to improve the diagnostic accuracy
of PTC in indeterminate and inadequate cytology is
feasible. However, the high frequency of follicular neo-
plasm and follicular-variant PTCs in indeterminate
samples rarely being BRAFV600E-positive, and the low
tumor DNA yield in inadequate specimens, limit the
clinical use of the test. Multiple-genotype analysis, such
as for RAS and PAX8-PPAR, improves the diagnostic
accuracy in indeterminate cytology but may cause false-
positive results because RAS mutation is also positive
in follicular adenomas. It seems that future discovery
of more specific markers is the ultimate solution for
this issue.
PTC is a relatively benign cancer when compared
with other malignancies, and the majority of patients
can be cured after the initial treatment with an opti-
mistic outcome. Preoperative BRAFV600E analysis might
have value for predicting which patients with low-grade
disease classified by the current staging systems will
eventually have an aggressive clinical course. It would
be beneficial for these patients (including BRAFV600E-
mutated PTMC) to receive more intensive primary
treatment, higher dose of radio-iodide ablation, and
frequent follow-up to reduce the risk of tumor metas-
tasis and recurrence later in the course of tumor pro-
gression. It might also be clinically indicated to use
18-FDG-PET to detect early recurrence of RAIR PTC.
It is noteworthy that there is currently no effective
therapy for tumors that are inoperable or lose iodine
avidity. However, current available data regarding the
negative clinical implications of BRAFV600E mutation in
low-risk PTC are inconclusive. Further investigations in
large randomized, controlled, prospective trials are nec-
essary to confirm the prognostic role of BRAFV600E
mutation before it is applied in routine clinical practice.
To date, the understanding of in vitro molecular
pathways involved in thyroid carcinogenesis supports
the rationale to develop BRAF-targeted therapies for
PTC. Although the preclinical results are encouraging,
the anticancer effects of clinical trials for specific BRAF-
targeted therapies are unsatisfactory. Tumorigenesis is
a complex process that involves other signal pathways
J Chin Med Assoc March 2010 Vol 73 No 3 123
BRAF mutation in papillary thyroid cancer
to effect tumor aggressiveness and progression. As new
targets are disclosed, future research might use selective
target inhibitors or multi-kinase inhibitors alone or in
combination with other regimens (e.g. cytotoxic drugs,
radioiodine therapy after re-expression of thyroid-
specific genes) for the treatment of advanced thyroid
cancers, with the hope that these approaches can pro-
vide satisfactory results.
Acknowledgments
This work was supported by a grant (V99C1-171) from
Taipei Veterans General Hospital.
References
1. Horner MJ, Ries LAG, Krapcho M, Neyman N, Aminou R,
Howlader N, Altekruse SF, et al, eds. SEER Cancer Statistics
Review, 1975–2006. Bethesda, MD: National Cancer Institute.
Available at http://seer.cancer.gov/csr/1975_2006/, based
on November 2008 SEER data submission, posted to the
SEER website, 2009. [Date accessed: December 15, 2009]
2. Hundahl SA, Fleming ID, Fremgen AM, Menck HR.
A National Cancer Data Base report on 53,856 cases of thyroid
carcinoma treated in the U.S., 1985–1995. Cancer 1998;83:
2638–48.
3. Dohan O, Baloch Z, Banrevi Z, Livolsi V, Carrasco N.
Rapid communication: predominant intracellular overexpres-
sion of the Na(+)/I(–) symporter (NIS) in a large sampling
of thyroid cancer cases. J Clin Endocrinol Metab 2001;86:
2697–700.
4. Mazzaferri EL, Jhiang SM. Long-term impact of initial surgical
and medical therapy on papillary and follicular thyroid cancer.
Am J Med 1994;97:418–28.
5. Mazzaferri EL, Kloos RT. Current approaches to primary ther-
apy for papillary and follicular thyroid cancer. J Clin Endocrinol
Metab 2001;86:1447–63.
6. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics,
2002. CA Cancer J Clin 2005;55:74–108.
7. Gottlieb JA, Hill CS. Chemotherapy of thyroid cancer with
adriamycin: experience with 30 patients. N Engl J Med 1974;
290:193–7.
8. Shimaoka K, Schoenfeld DA, DeWys WD, Creech RH,
DeConti R. A randomized trial of doxorubicin versus dox-
orubicin plus cisplatin in patients with advanced thyroid carci-
noma. Cancer 1985;56:2155–60.
9. Döbert N, Menzel C, Oeschger S, Grünwald F. Differentiated
thyroid carcinoma: the new UICC 6th edition TNM classifica-
tion system in a retrospective analysis of 169 patients. Thyroid
2004;14:65–70.
10. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL,
Mandel SJ, Mazzaferri EL, et al. Management guidelines for
patients with thyroid nodules and differentiated thyroid cancer.
Thyroid 2006;16:109–42.
11. Loh KC, Greenspan FS, Gee L, Miller TR, Yeo PP. Pathologi-
cal tumor-node-metastasis (pTNM) staging for papillary and
follicular thyroid carcinomas: a retrospective analysis of 700
patients. J Clin Endocrinol Metab 1997;82:3553–62.
12. Kitamura Y, Shimizu K, Nagahama M, Sugino K, Ozaki O,
Mimura T, Ito K, et al. Immediate causes of death in thyroid
carcinoma: clinicopathological analysis of 161 fatal cases.
J Clin Endocrinol Metab 1999;84:4043–9.
13. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S,
Teague J, et al. Mutations of the BRAF gene in human cancer.
2002;417:949–54.
14. Garnett MJ, Marais R. Guilty as charged: B-RAF is a human
oncogene. Cancer Cell 2004;6:313–9.
15. Xing M. BRAF mutation in papillary thyroid cancer: pathogenic
role, molecular bases, and clinical implications. Endoc Rev
2007;28:742–62.
16. Puxeddu E, Durante C, Avenia N, Filetti S, Russo D. Clinical
implications of BRAF mutation in thyroid carcinoma. Trends
Endocrinol Metab 2008;19:138–45.
17. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D,
Good VM, Jones CM, et al. Mechanism of activation of the
RAF-ERK signaling pathway by oncogenic mutations of B-RAF.
Cell 2004;116:855–67.
18. Robinson MJ, Cobb MH. Mitogen-activated protein kinase
pathways. Curr Opin Cell Biol 1997;9:180–6.
19. MacCorkle RA, Tan TH. Mitogen-activated protein kinases
in cell-cycle control. Cell Biochem Biophys 2005;43:451–61.
20. Wittinghofer A, Nassar N. How Ras-related proteins talk to
their effectors. Trends Biochem Sci 1996;21:488–91.
21. Hancock JF. Ras proteins: different signals from different
locations. Nat Rev Mol Cell Biol 2003;4:373–84.
22. Weber CK, Slupsky JR, Herrmann C, Schuler M, Rapp UR,
Block C. Mitogenic signaling of Ras is regulated by differential
interaction with Raf isozymes. Oncogene 2000;19:169–76.
23. Avruch J, Khokhlatchev A, Kyriakis JM, Luo Z, Tzivion G,
Vavvas D, Zhang XF. Ras activation of the Raf kinase: tyrosine
kinase recruitment of the MAP kinase cascade. Recent Prog
Horm Res 2001;56:127–55.
24. Malumbres M, Barbacid M. RAS oncogenes: the first 30 years.
Nat Rev Cancer 2003;3:459–65.
25. Chong H, Lee J, Guan KL. Positive and negative regulation
of Raf kinase activity and function by phosphorylation.
EMBO J 2001;20:3716–27.
26. Zhang BH, Guan KL. Activation of B-Raf kinase requires
phosphorylation of the conserved residues Thr598 and
Ser601. EMBO J 2001;19:5429–39.
27. Kolch W. Meaningful relationships: the regulation of the
Ras/Raf/MEK/ERK pathway by protein interactions.
Biochem J 2000;351:289–305.
28. Morrison DK, Cutler RE. The complexity of Raf-1 regulation.
Curr Opin Cell Biol 1997;9:174–9.
29. Leicht DT, Balan V, Kaplun A, Singh-Gupta V, Kaplun L,
Dobson M, Tzivion G. Raf kinases: function, regulation and
role in human cancer. Biochim Biophys Acta 2007;1773:
1196–212.
30. Mason CS, Springer C, Cooper RG, Superti-Furga G,
Marshall CJ, Marais R. Serine and tyrosine phosphorylations
cooperate in Raf-1, but not B-Raf activation. EMBO J 1999;
18:2137–48.
31. Mercer KE, Pritchard CA. Raf proteins and cancer: B-Raf is
identified as a mutational target. Biochim Biophys Acta 2003;
1653:25–40.
32. Marais R, Light Y, Paterson HF, Mason CS, Marshall CJ. Dif-
ferential regulation of Raf-1, A-Raf, and B-Raf by oncogenic
ras and tyrosine kinases. J Biol Chem 1997;272:4378–83.
33. Peyssonnaux C, Eychène A. The Raf/MEK/ERK pathway:
new concepts of activation. Biol Cell 2001;93:53–62.
34. Chang F, Steelman LS, Lee JT, Shelton JG, Navolanic PM,
Blalock WL, Franklin RA, et al. Signal transduction mediated
by the Ras/Raf/MEK/ERK pathway from cytokine recep-
tors to transcription factors: potential targeting for therapeu-
tic intervention. Leukemia 2003;17:1263–93.
J Chin Med Assoc March 2010 Vol 73 No 3
124
K.T. Tang, C.H. Lee
35. Hagemann C, Rapp UR. Isotype-specific functions of Raf
kinases. Exp Cell Res 1999;253:34–46.
36. Ravi RK, Weber E, McMahon M, Williams JR, Baylin S, Mal A,
Harter ML, et al. Activated Raf-1 causes growth arrest in
human small cell lung cancer cells. J Clin Invest 1998;101:
153–9.
37. Zhu J, Woods D, McMahon M, Bishop JM. Senescence of
human fibroblasts induced by oncogenic Raf. Genes Dev 1998;
12:2997–3007.
38. Wellbrock C, Karasarides M, Marais R. The raf proteins take
centre stage. Nat Rev Mol Cell Biol 2004;5:875–85.
39. Hoshino R, Chatani Y, Yamori T, Tsuruo T, Oka H, Yoshida O,
Shimada Y, et al. Constitutive activation of the 41-/43-kDa
mitogen-activated protein kinase signaling pathway in human
tumors. Oncogene 1999;18:813–22.
40. Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE,
Fagin JA. High prevalence of BRAF mutations in thyroid cancer:
genetic evidence for constitutive activation of the RET/PTC-
RAS-BRAF signaling pathway in papillary thyroid carcinoma.
Cancer Res 2003;63:1454–7.
41. Kondo T, Ezzat S, Asa SL. Pathogenetic mechanisms in thyroid
follicular-cell neoplasia. Nat Rev 2006;6:292–306.
42. Ciampi R, Nikiforov YE. RET/PTC rearrangements and BRAF
mutations in thyroid tumorigenesis. Endocrinology 2007;148:
936–41.
43. Santoro M, Melillo RM, Fusco A. RET/PTC activation
in papillary thyroid carcinoma: European Journal of
Endocrinology Prize Lecture. Eur J Endocrinol 2006;155:
645–53.
44. Knauf JA, Kuroda H, Basu S, Fagin JA. RET/PTC-induced
dedifferentiation of thyroid cells is mediated through Y1062
signaling through SHC-RAS-MAP kinase. Oncogene 2003;
22:4406–12.
45. Melillo RM, Castellone MD, Guarino V, De Falco V,
Cirafici AM, Salvatore G, Caiazzo F, et al. The RET/PTC-
RAS-BRAF linear signaling cascade mediates the motile and
mitogenic phenotype of thyroid cancer cells. J Clin Invest
2005;115:1068–81.
46. Mitsutake N, Miyagishi M, Mitsutake S, Akeno N, Mesa C Jr,
Knauf JA, Zhang L, et al. BRAF mediates RET/PTC-
induced mitogen-activated protein kinase activation in thyroid
cells: functional support for requirement of the RET/
PTC-RAS-BRAF pathway in papillary thyroid carcinogenesis.
Endocrinology 2006;147:1014–9.
47. Jhiang SM, Sagartz JE, Tong Q, Parker-Thornburg J,
Capen CC, Cho JY, Xing S, et al. Targeted expression of the
ret/PTC1 oncogene induces papillary thyroid carcinomas.
Endocrinology 1996;137:375–8.
48. Santoro M, Chiappetta G, Cerrato A, Salvatore D, Zhang L,
Manzo G, Picone A, et al. Development of thyroid papillary
carcinomas secondary to tissue-specific expression of the
RET/PTC1 oncogene in transgenic mice. Oncogene 1996;
12:1821–6.
49. Powell DJ Jr, Russell J, Nibu K, Li G, Rhee E, Liao M,
Goldstein M, et al. The RET/PTC3 oncogene: metastatic
solid-type papillary carcinomas in murine thyroids. Cancer Res
1998;58:5523–28.
50. Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H,
Fagin JA. Distinct pattern of ret oncogene rearrangements in
morphological variants of radiation-induced and sporadic thy-
roid papillary carcinomas in children. Cancer Res 1997;57:
1690–4.
51. Suarez HG, du Villard JA, Caillou B, Schlumberger M,
Tubiana M, Parmentier C, Monier R. Detection of activated ras
oncogenes in human thyroid carcinomas. Oncogene 1988;2:
403–6.
52. Namba H, Rubin SA, Fagin JA. Point mutations of ras on-
cogenes are an early event in thyroid tumorigenesis. Mol
Endocrinol 1990;4:1474–9.
53. Zhu Z, Gandhi M, Nikiforova MN, Fischer AH, Nikiforov YE.
Molecular profile and clinical-pathologic features of the follic-
ular variant of papillary thyroid carcinoma: an unusually high
prevalence of ras mutations. Am J Clin Pathol 2003;120:
71–7.
54. Daum G, Eisenmann-Tappe I, Fries HW, Troppmair J,
Rapp UR. The ins and outs of Raf kinases. Trends Biochem Sci
1994;19:474–80.
55. Kondo T, Nakazawa T, Murata S, Kurebayashi J, Ezzat S,
Asa SL, Katoh R. Enhanced B-Raf protein expression is inde-
pendent of V600E mutant status in thyroid carcinomas. Hum
Pathol 2007;38:1810–8.
56. Trovisco V, Vieira de Castro I, Soares P, Máximo V, Silva P,
Magalhães J, Abrosimov A, et al. BRAF mutations are associ-
ated with some histological types of papillary thyroid carci-
noma. J Pathol 2004;202:247–51.
57. Ciampi R, Knauf JA, Kerler R, Gandhi M, Zhu Z, Nikiforova
MN, Rabes HM, et al. Oncogenic AKAP9-BRAF fusion is
a novel mechanism of MAPK pathway activation in thyroid
cancer. J Clin Invest 2005;115:94–101.
58. Moretti S, Macchiarulo A, De Falco V, Avenia N, Barbi F,
Carta C, Cavaliere A, et al. Biochemical and molecular charac-
terization of the novel BRAF (V599Ins) mutation detected
in a classic papillary thyroid carcinoma. Oncogene 2006;25:
4235–40.
59. Hou P, Liu D, Xing M. Functional characterization of the
T1799–1801del and A1799–1816ins BRAF mutations in
papillary thyroid cancer. Cell Cycle 2007;6:377–9.
60. Oler G, Ebina KN, Michaluart P Jr, Kimura ET, Cerutti J.
Investigation of BRAF mutation in a series of papillary thy-
roid carcinoma and matched-lymph node metastasis reveals
a new mutation in metastasis. Clin Endocrinol (Oxf) 2005;62:
509–11.
61. Ugolini C, Giannini R, Lupi C, Salvatore G, Miccoli P,
Proietti A, Elisei R, et al. Presence of BRAF V600E in very
early stages of papillary thyroid carcinoma. Thyroid 2007;17:
381–8.
62. Kim TY, Kim WB, Song JY, Rhee YS, Gong G, Cho YM,
Kim SY, et al. The BRAF mutation is not associated with poor
prognostic factors in Korean patients with conventional papil-
lary thyroid microcarcinoma. Clin Endocrinol (Oxf) 2005;
63:588–93.
63. Lupi C, Giannini R, Ugolini C, Proietti A, Berti P, Minuto M,
Materazzi G, et al. Association of BRAF V600E mutation
with poor clinicopathological outcomes in 500 consecutive
cases of papillary thyroid carcinoma. J Clin Endocrinol Metab
2007;92:4085–90.
64. Sedliarou I, Saenko V, Lantsov D, Rogounovitch T, Namba H,
Abrosimov A, Lushnikov E, et al. The BRAFT1796A trans-
version is a prevalent mutational event in human thyroid
microcarcinoma. Int J Oncol 2004;25:1729–35.
65. Trovisco V, Soares P, Preto A, de Castro IV, Lima J, Castro P,
Máximo V, et al. Type and prevalence of BRAF mutations are
closely associated with papillary thyroid carcinoma histotype
and patients’ age but not with tumour aggressiveness. Virchows
Arch 2005;446:589–95.
66. Knauf JA, Ma X, Smith EP, Zhang L, Mitsutake N, Liao XH,
Refetoff S, et al. Targeted expression of BRAFV600E in thyroid
cells of transgenic mice results in papillary thyroid cancers that
undergo dedifferentiation. Cancer Res 2005;65:4238–45.
67. Mesa Jr C, Mirza M, Mitsutake N, Sartor M, Medvedovic M,
Tomlinson C, Knauf JA, et al. Conditional activation of
RET/PTC3 and BRAFV600E in thyroid cells is associated
J Chin Med Assoc March 2010 Vol 73 No 3 125
BRAF mutation in papillary thyroid cancer
with gene expression profiles that predict a preferential role of
BRAF in extracellular matrix remodeling. Cancer Res 2006;
66:6521–9.
68. Ouyang B, Knauf JA, Smith EP, Zhang L, Ramsey T, Yusuff N,
Batt D, et al. Inhibitors of Raf kinase activity block growth
of thyroid cancer cells with RET/PTC or BRAF mutations
in vitro and in vivo. Clin Cancer Res 2006;12:1785–93.
69. Salvatore G, Falco V, Salerno P, Nappi T, Pepe S, Troncone G,
Carlomagno F, et al. BRAF is a therapeutic target in aggres-
sive thyroid carcinoma. Clin Cancer Res 2006;12:1623–9.
70. Liu D, Liu Z, Condouris S, Xing M. BRAF V600E maintains
proliferation, transformation, and tumorigenicity of BRAF-
mutant papillary thyroid cancer cells. J Clin Endocrinol Metab
2007;92:2264–71.
71. Wang J, Knauf JA, Basu S, Puxeddu E, Kuroda H, Santoro M,
Fusco A, et al. Conditional expression of RET/PTC induces
a weak oncogenic drive in thyroid PCCL3 cells and inhibits
thyrotropin action at multiple levels. Mol Endocrinol 2003;
17:1425–36.
72. Giordano TJ, Kuick R, Thomas DG, Misek DE, Vinco M,
Sanders D, Zhu Z, et al. Molecular classification of papillary
thyroid carcinoma: distinct BRAF, RAS, and RET/PTC
mutation-specific gene expression profiles discovered by
DNA microarray analysis. Oncogene 2005;24:6646–56.
73. Owens DM, Keyse SM. Differential regulation of MAP kinase
signaling by dual specificity protein phosphatases. Oncogene
2007;26:3203–13.
74. Kim HJ, Bar-Sagi D. Modulation of signaling by Sprouty:
a developing story. Nat Rev Mol Cell Biol 2004;5:441–50.
75. Traverse S, Gomez N, Paterson H, Marshall C, Cohen P.
Sustained activation of the mitogen-activated protein (MAP)
kinase cascade may be required for differentiation of PC12
cells: comparison of the effects of nerve growth factor and
epidermal growth factor. Biochem J 1992;288:351–5.
76. Amit I, Citri A, Shay T, Lu Y, Katz M, Zhang F, Tarcic G,
et al. A module of negative feedback regulators defines growth
factor signaling. Nat Genet 2007;39:503–12.
77. Dougherty MK, Müller J, Ritt DA, Zhou M, Zhou XZ,
Copeland TD, Conrads TP, et al. Regulation of Raf-1 by
direct feedback phosphorylation. Mol Cell 2005;17:215–24.
78. Pratilas CA, Taylor BS, Ye Q, Viale A, Sander C, Solit DB,
Rosen N. V600EBRAF is associated with disabled feedback
inhibition of RAF–MEK signaling and elevated transcriptional
output of the pathway. Proc Natl Acad Sci USA 2009;106:
4519–24.
79. Mitsutake N, Knauf JA, Mitsutake S, Mesa C Jr, Zhang L,
Fagin JA. Conditional BRAFV600E expression induces
DNA synthesis, apoptosis, dedifferentiation, and chromoso-
mal instability in thyroid PCCL3 cells. Cancer Res 2005;65:
2465–73.
80. Ricarte-Filho JC, Ryder M, Chitale DA, Rivera M, Heguy A,
Ladanyi M, Janakiraman M, et al. Mutational profile of
advanced primary and metastatic radioactive iodine-refractory
thyroid cancers reveals distinct pathogenetic roles for BRAF,
PIK3CA, and AKT1. Cancer Res 2009;69:4885–93.
81. Paes JE, Ringel MD. Dysregulation of the phosphatidylinosi-
tol 3-kinase pathway in thyroid neoplasia. Endocrinol Metab
Clin North Am 2008;37:375–87.
82. Yamashita S, Saenko V. Mechanisms of disease: molecular
genetics of childhood thyroid cancers. Nat Clin Pract
Endocrinol Metab 2007;3:422–9.
83. Dal Maso L, Bosetti C, La Vecchia C, Franceschi S. Risk fac-
tors for thyroid cancer: an epidemiological review focused on
nutritional factors. Cancer Causes Control 2009;20:75–86.
84. Ron E, Lubin JH, Shore RE, Mabuchi K, Modan B, Pottern
LM, Schneider AB, et al. Thyroid cancer after exposure to
external radiation: a pooled analysis of seven studies. Radiat
Res 1995;141:259–77.
85. Moysich KB, Menezes RJ, Michalek AM. Chernobyl-related
ionising radiation exposure and cancer risk: an epidemiologi-
cal review. Lancet Oncol 2002;3:269–79.
86. Nikiforova MN, Ciampi R, Salvatore G, Santoro M, Gandhi
M, Knauf JA, Thomas GA, et al. Low prevalence of BRAF
mutations in radiation-induced thyroid tumors in contrast to
sporadic papillary carcinomas. Cancer Lett 2004;209:1–6.
87. Lee JH, Lee ES, Kim YS, Won NH, Chae YS. BRAF mutation
and AKAP9 expression in sporadic papillary thyroid carcino-
mas. Pathology 2006;38:201–4.
88. Frasca F, Nucera C, Pellegriti G, Gangemi P, Attard M, Stella
M, Loda M, et al. BRAF(V600E) mutation and the biology
of papillary thyroid cancer. Endocr Relat Cancer 2008;15:
191–205.
89. Guan H, Ji M, Bao R, Yu H, Wang Y, Hou P, Zhang Y, et al.
Association of high iodine intake with the T1799A BRAF
mutation in papillary thyroid cancer. J Clin Endocrinol Metab
2009;94:1612–7.
90. Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP,
Giordano TJ, Biddinger PW, et al. Correlation between
genetic alterations and microscopic features, clinical manifes-
tations, and prognostic characteristics of thyroid papillary car-
cinomas. Am J Surg Pathol 2006;30:216–22.
91. Fugazzola L, Puxeddu E, Avenia N, Romei C, Cirello V,
Cavaliere A, Faviana P, et al. Correlation between B-RAFV600E
mutation and clinico-pathologic parameters in papillary thyroid
carcinoma: data from a multicentric Italian study and review
of the literature. Endocr Relat Cancer 2006;13:455–64.
92. Kebebew E, Weng J, Bauer J, Ranvier G, Clark OH, Duh QY,
Shibru D, et al. The prevalence and prognostic value of BRAF
mutation in thyroid cancer. Ann Surg 2007;246:466–71.
93. Kim KH, Suh KS, Kang DW, Kang DY. Mutations of the
BRAF gene in papillary thyroid carcinoma and in Hashimoto’s
thyroiditis. Pathol Int 2005;55:540–5.
94. Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW,
Knauf JA, Basolo F, Zhu Z, et al. BRAF mutations in thyroid
tumors are restricted to papillary carcinomas and anaplastic or
poorly differentiated carcinomas arising from papillary carci-
nomas. J Clin Endocrinol Metab 2003;88:5399–404.
95. Rodolico V, Cabibi D, Pizzolanti G, Richiusa P, Gebbia N,
Martorana A, Russo A, et al. BRAF(V600E) mutation and
p27(kip1) expression in papillary carcinomas of the thyroid
1 cm and their paired lymph node metastases. Cancer 2007;
110:1218–26.
96. Powell N, Jeremiah S, Morishita M, Dudley E, Bethel J,
Bogdanova T, Tronko M, et al. Frequency of BRAF T1796A
mutation in papillary thyroid carcinoma relates to age of patient
at diagnosis and not to radiation exposure. J Pathol 2005;
205:558–64.
97. Xu X, Quiros RM, Gattuso P, Ain KB, Prinz RA. High preva-
lence of BRAF gene mutation in papillary thyroid carcinomas
and thyroid tumor cell lines. Cancer Res 2003;63:4561–7.
98. Kim TY, Kim WB, Rhee YS, Song JY, Kim JM, Gong G,
Lee S, et al. The BRAF mutation is useful for prediction
of clinical recurrence in low-risk patients with conventional
papillary thyroid carcinoma. Clin Endocrinol (Oxf) 2006;65:
364–8.
99. Oler G, Cerutti JM. High prevalence of BRAF mutation in
a Brazilian cohort of patients with sporadic papillary thyroid
carcinomas: correlation with more aggressive phenotype and
decreased expression of iodide-metabolizing genes. Cancer
2009;115:972–80.
100. Elisei R, Ugolini C, Viola D, Lupi C, Biagini A, Giannini R,
Romei C, et al. BRAF(V600E) mutation and outcome of
J Chin Med Assoc March 2010 Vol 73 No 3
126
K.T. Tang, C.H. Lee
patients with papillary thyroid carcinoma: a 15-year median
follow-up study. J Clin Endocrinol Metab 2008;93:3943–9.
101. Jo YS, Li S, Song JH, Kwon KH, Lee JC, Rha SY, Lee HJ, et al.
Influence of the BRAF V600E mutation on expression of vas-
cular endothelial growth factor in papillary thyroid cancer.
J Clin Endocrinol Metab 2006;91:3667–70.
102. Riesco-Eizaguirre G, Gutierrez-Martinez P, Garcia-Cabezas
MA, Nistal M, Santisteban P. The oncogene BRAF V600E is
associated with a high risk of recurrence and less differenti-
ated papillary thyroid carcinoma due to the impairment of
Na +/I– targeting to the membrane. Endocr Relat Cancer
2006;13:257–69.
103. Giannini R, Ugolini C, Lupi C, Proietti A, Elisei R, Salvatore G,
Berti P, et al. The heterogeneous distribution of BRAF muta-
tion supports the independent clonal origin of distinct tumor
foci in multifocal papillary thyroid carcinoma. J Clin Endocrinol
Metab 2007;92:3511–6.
104. Frattini M, Ferrario C, Bressan P, Balestra D, De Cecco L,
Mondellini P, Bongarzone I, et al. Alternative mutations of
BRAF, RET and NTRK1 are associated with similar but dis-
tinct gene expression patterns in papillary thyroid cancer.
Oncogene 2007;23:7436–40.
105. Namba H, Nakashima M, Hayashi T, Hayashida N, Maeda S,
Rogounovitch TI, Ohtsuru A, et al. Clinical implication of
hot spot BRAF mutation, V599E, in papillary thyroid cancers.
J Clin Endocrinol Metab 2003;88:4393–7.
106. Xing M, Westra WH, Tufano RP, Cohen Y, Rosenbaum E,
Rhoden KJ, Carson KA, et al. BRAF mutation predicts a poorer
clinical prognosis for papillary thyroid cancer. J Clin Endocrinol
Metab 2005;90:6373–9.
107. Vasko V, Hu S, Wu G, Xing JC, Larin A, Savchenko V, Trink
B, et al. High prevalence and possible de novo formation of
BRAF mutation in metastasized papillary thyroid cancer in
lymph nodes. J Clin Endocrinol Metab 2005;90:5265–9.
108. Trovisco V, Couto JP, Cameselle-Teijeiro J, de Castro IV,
Fonseca E, Soares P, Sobrinho-Simões M. Acquisition of BRAF
gene mutations is not a requirement for nodal metastasis of
papillary thyroid carcinoma. Clin Endocrinol (Oxf) 2008;69:
683–5.
109. Lee J, Lee ES, Kim YS. Clinicopathologic significance of
BRAF V600E mutation in papillary carcinomas of the thyroid:
a meta-analysis. Cancer 2007;110:38–46.
110. Henderson YC, Shellenberger TD, Williams MD, El-Naggar
AK, Fredrick MJ, Cieply KM, Clayman GL. High rate of
BRAF and RET/PTC dual mutations associated with recurrent
papillary thyroid carcinoma. Clin Cancer Res 2009;15:485–91.
111. Ito Y, Yoshida H, Maruo R, Morita S, Takano T, Hirokawa
M, Yabuta T, et al. BRAF mutation in papillary thyroid carci-
noma in a Japanese population: its lack of correlation with
high-risk clinicopathological features and disease-free survival
of patients. Endocr J 2009;56:89–97.
112. Abrosimov A, Saenko V, Rogounovitch T, Namba H,
Lushnikov E, Mitsutake N, Yamashita S. Different structural
components of conventional papillary thyroid carcinoma dis-
play mostly identical BRAF status. Int J Cancer 2007;120:
196–200.
113. Durante C, Puxeddu E, Ferretti E, Morisi R, Moretti S,
Bruno R, Barbi F, et al. BRAF mutations in papillary thyroid
carcinomas inhibit genes involved in iodine metabolism.
J Clin Endocrinol Metab 2007;92:2840–3.
114. Liu RT, Chen YJ, Chou FF, Li CL, Wu WL, Tsai PC, Huang
CC, et al. No correlation between BRAFV600E mutation
and clinicopathological features of papillary thyroid carcino-
mas in Taiwan. Clin Endocrinol (Oxf) 2005;63:461–6.
115. Mitsiades CS, Negri J, McMullan C, McMillin DW,
Sozopoulos E, Fanourakis G, Voutsinas G, et al. Targeting
BRAFV600E in thyroid carcinoma: therapeutic implications.
Mol Cancer Ther 2007;6:1070–8.
116. Park SY, Park YJ, Lee YJ, Lee HS, Choi SH, Choe G, Jang HC,
et al. Analysis of differential BRAF(V600E) mutational status
in multifocal papillary thyroid carcinoma: evidence of inde-
pendent clonal origin in distinct tumor foci. Cancer 2006;
107:1831–8.
117. Puxeddu E, Moretti S, Elisei R, Romei C, Pascucci R,
Martinelli M, Marino C, et al. BRAF(V599E) mutation is the
leading genetic event in adult sporadic papillary thyroid carci-
nomas. J Clin Endocrinol Metab 2004;89:2414–20.
118. Sapio MR, Posca D, Troncone G, Pettinato G, Palombini L,
Rossi G, Fenzi G, et al. Detection of BRAF mutation in thy-
roid papillary carcinomas by mutant allele-specific PCR
amplification (MASA). Eur J Endocrinol 2006;154:341–8.
119. Romei C, Ciampi R, Faviana P, Agate L, Molinaro E, Bottici
V, Basolo F, et al. BRAFV600E mutation, but not RET/PTC
rearrangements, is correlated with a lower expression of both
thyroperoxidase and sodium iodide symporter genes in papil-
lary thyroid cancer. Endocr Relat Cancer 2008;15:511–20.
120. Mian C, Barollo S, Pennelli G, Pavan N, Rugge M, Pelizzo MR,
Mazzarotto R, et al. Molecular characteristics in papillary thy-
roid cancers (PTCs) with no (131)I uptake. Clin Endocrinol
(Oxf) 2008;68:108–16.
121. Di Cristofaro J, Silvy M, Lanteaume A, Marcy M, Carayon P,
De Micco C. Expression of tpo mRNA in thyroid tumors:
quantitative PCR analysis and correlation with alterations of
ret, Braf, ras and pax8 genes. Endocr Relat Cancer 2006;13:
485–95.
122. Liu D, Hu S, Hou P, Jiang D, Condouris S, Xing M.
Suppression of BRAF/MEK/MAP kinase pathway restores
expression of iodide-metabolizing genes in thyroid cells express-
ing the V600E BRAF mutant. Clin Cancer Res 2007;13:
1341–9.
123. Ikuyama S, Niller HH, Shimura H, Akamizu T, Kohn LD.
Characterization of the 5-flanking region of the rat thyrotropin
receptor gene. Mol Endocrinol 1992;6:793–804.
124. Yokomori N, Tawata M, Saito T, Shimura H, Onaya T. Regula-
tion of the rat thyrotropin receptor gene by the methylation-
sensitive transcription factor GA-binding protein. Mol Endocrinol
1998;12:1241–9.
125. American Association of Clinical Endocrinologists and
Associazione MediciEndocrinologi medical guidelines for
clinical practice for the diagnosis and management of thyroid
nodules. Endocr Pract 2006;12:63–102.
126. Pacini F, Schlumberger M, Dralle H, Elisei R, Smit JW,
Wiersinga W. European consensus for the management of
patients with differentiated thyroid carcinoma of the follicular
epithelium. Eur Endocrinol 2006;154:787–803.
127. Leenhardt L, Grosclaude P, Chérié-Challine L, Thyroid
Cancer Committee. Increased incidence of thyroid carcinoma
in France: a true epidemic or thyroid nodule management
effects? Report from the French thyroid cancer committee.
Thyroid 2004;14:1056–60.
128. Davies L, Welch HG. Increasing incidence of thyroid cancer
in the United States, 1973–2002. JAMA 2006;295:2164–7.
129. Burgess JR, Dwyer T, McArdle K, Tucker P, Shugg D. The
changing incidence and spectrum of thyroid carcinoma in
Tasmania (1978–1998) during a transition from iodine suffi-
ciency to iodine deficiency. J Clin Endocrinol Metab 2000;85:
1513–7.
130. Chow SM, Law SC, Au SK, Mang O, Yau S, Yuen KT, Lau WH.
Changes in clinical presentation, management and outcome
in 1348 patients with differentiated thyroid carcinoma: expe-
rience in a single institute in Hong Kong, 1960–2000. Clin
Oncol 2003;15:329–36.
J Chin Med Assoc March 2010 Vol 73 No 3 127
BRAF mutation in papillary thyroid cancer
131. Roti E, degli Uberti EC, Bondanelli M, Braverman LE.
Thyroid papillary microcarcinoma: a descriptive and meta-
analysis study. Eur J Endocrinol 2008;159:659–73.
132. Cappelli C, Castellano M, Braga M, Gandossi E, Pirola I,
De Martino E, Agosti B, et al. Aggressiveness and outcome
of papillary thyroid carcinoma (PTC) versus microcarcinoma
(PMC): a mono-institutional experience. J Surg Oncol 2007;
95:555–60.
133. Baudin E, Travagli JP, Ropers J, Mancusi F, Bruno-Bossio G,
Caillou B, Cailleux AF, et al. Microcarcinoma of the thyroid
gland: the Gustave Roussy Institute experience. Cancer 1998;
83:553–9.
134. Roti E, Rossi R, Trasforini G, Bertelli F, Ambrosio MR,
Busutti L, Pearce EN, et al. Clinical and histological charac-
teristics of papillary thyroid microcarcinoma: results of a ret-
rospective study in 243 patients. J Clin Endocrinol Metab
2006;91:2171–8.
135. Chan BK, Desser TS, McDougall IR, Weigel RJ, Jeffrey RB
Jr. Common and uncommon sonographic features of papil-
lary thyroid carcinoma. J Ultrasound Med 2003;22:1083–90.
136. Park YJ, Kim YA, Lee YJ, Kim SH, Park SY, Kim KW, Chung
JK, et al. Papillary microcarcinoma in comparison with larger
papillary thyroid carcinoma in BRAFV600E mutation, clini-
copathological features, and immunohistochemical findings.
Head Neck 2010;32:38–45.
137. Lee X, Gao M, Ji Y, Yu Y, Feng Y, Li Y, Zhang Y, et al.
Analysis of differential BRAF(V600E) mutational status in high
aggressive papillary thyroid microcarcinoma. Ann Surg Oncol
2009;16:240–5.
138. Lee TI, Yang HJ, Lin SY, Lee MT, Lin HD, Braverman LE,
Tang KT. The accuracy of fine-needle aspiration biopsy and
frozen section in patients with thyroid cancer. Thyroid 2002;
12:619–26.
139. Salvatore G, Giannini R, Faviana P, Caleo A, Migliaccio I,
Fagin JA, Nikiforov YE, et al. Analysis of BRAF point mutation
and RET/PTC rearrangement refines the fine-needle aspiration
diagnosis of papillary thyroid carcinoma. J Clin Endocrinol
Metab 2004;89:5175–80.
140. Cohen Y, Rosenbaum E, Clark DP, Zeiger MA, Umbricht CB,
Tufano RP, Sidransky D, et al. Mutational analysis of BRAF
in fine-needle aspiration biopsies of the thyroid: a potential
application for the preoperative assessment of thyroid nodules.
Clin Cancer Res 2004;10:2761–5.
141. Marchetti I, Lessi F, Mazzanti CM, Bertacca G, Elisei R, Di
Coscio G, Pinchera A, et al. A morpho-molecular diagnosis of
papillary thyroid carcinoma: BRAF V600E detection as an
important tool in preoperative evaluation of fine-needle aspi-
rates. Thyroid 2009;19:837–42.
142. Zatelli MC, Trasforini G, Leoni S, Frigato G, Buratto M,
Tagliati F, Rossi R, et al. BRAF V600E mutation analysis
increases diagnostic accuracy for papillary thyroid carcinoma
in fine-needle aspiration biopsies. Eur J Endocrinol 2009;161:
467–73.
143. Nikiforov YE, Steward DL, Robinson-Smith TM, Haugen BR,
Klopper JP, Zhu Z, Fagin JA, et al. Molecular testing for
mutations in improving the fine-needle aspiration diagnosis of
thyroid nodules. J Clin Endocrinol Metab 2009;94:2092–8.
144. Xing M, Clark D, Guan H, Ji M, Dackiw A, Carson KA, Kim M,
et al. BRAF mutation testing of thyroid fine-needle aspiration
biopsy specimens for preoperative risk stratification in papillary
thyroid cancer. J Clin Oncol 2009;27:2977–82.
145. Sala E, Mologni L, Truffa S, Gaetano C, Bollag GE,
Gambacorti-Passerini C. BRAF silencing by short hairpin
RNA or chemical blockade by PLX4032 leads to different
responses in melanoma and thyroid carcinoma cells. Mol
Cancer Res 2008;6:751–9.
146. Friday BB, Yu C, Dy GK, Smith PD, Wang L, Thibodeau SN,
Adjei AA. BRAF V600E disrupts AZD6244-induced abro-
gation of negative feedback pathways between extracellular
signal-regulated kinase and Raf proteins. Cancer Res 2008;
68:6145–53.
147. Solit DB, Garraway LA, Pratilas CA, Sawai A, Getz G,
Basso A, Ye Q, et al. BRAF mutation predicts sensitivity to
MEK inhibition. Nature 2006;439:358–62.
148. Liu D, Liu Z, Jiang D, Dackiw AP, Xing M. Inhibitory effects
of the MEK inhibitor CI-1040 on the proliferation and tumor
growth of thyroid cancer cells with BRAF or RAS mutations.
J Clin Endocrinol Metab 2007;92:4686–95.
149. Lorusso PM, Adjei AA, Varterasian M, Gadgeel S, Reid J,
Mitchell DY, Hanson L, et al. Phase I and pharmacodynamic
study of the oral MEK inhibitor CI-1040 in patients with
advanced malignancies. J Clin Oncol 2005;23:5281–93.
150. Rinehart J, Adjei AA, Lorusso PM, Waterhouse D, Hecht JR,
Natale RB, Hamid O, et al. Multicenter phase II study of the
oral MEK inhibitor, CI-1040, in patients with advanced non-
small-cell lung, breast, colon, and pancreatic cancer. J Clin
Oncol 2004;22:4456–62.
151. Kim S, Yazici YD, Calzada G, Wang ZY, Younes MN, Jasser
SA, El-Naggar AK, et al. Sorafenib inhibits the angiogenesis
and growth of orthotopic anaplastic thyroid carcinoma
xenografts in nude mice. Mol Cancer Ther 2007;6:1785–92.
152. Eisen T, Ahmad T, Flaherty KT, Gore M, Kaye S, Marais R,
Gibbens I, et al. Sorafenib in advanced melanoma: a phase II
randomised discontinuation trial analysis. Br J Cancer 2006;
95:581–6.
153. Gupta-Abramson V, Troxel AB, Nellore A, Puttaswamy K,
Redlinger M, Ransone K, Mandel SJ, et al. Phase II trial of
sorafenib in advanced thyroid cancer. J Clin Oncol 2008;26:
4714–9.
154. Kloos RT, Ringel MD, Knopp MV, Hall NC, King M,
Stevens R, Liang J, et al. Phase II trial of sorafenib in metastatic
thyroid cancer. J Clin Oncol 2009;27:1675–84.
155. Sherman SI, Wirth LJ, Droz JP, Hofmann M, Bastholt L,
Martins RG, Licitra L, et al. Motesanib diphosphate in pro-
gressive differentiated thyroid cancer. N Engl J Med 2008;
359:31–42.
156. Cohen EE, Rosen LS, Vokes EE, Kies MS, Forastiere AA,
Worden FP, Kane MA, et al. Axitinib is an active treatment for
all histologic subtypes of advanced thyroid cancer: results
from a phase II study. J Clin Oncol 2008;26:4708–13.
J Chin Med Assoc March 2010 Vol 73 No 3
128
K.T. Tang, C.H. Lee
... As the two main types of thyroid cancer, PTC and FTC demonstrate good prognoses with an overall survival rate of 76% in a 30-year follow-up period. Thyroid cancer is generally considered a malignancy with benign behavior compared to cancers derived from other organs [1][2][3]. However, despite the low mortality rate, PTC and FTC display a high risk of recurrence of approximately 30% [3], and patients should undergo lifelong follow-up [2]. ...
... Thyroid cancer is generally considered a malignancy with benign behavior compared to cancers derived from other organs [1][2][3]. However, despite the low mortality rate, PTC and FTC display a high risk of recurrence of approximately 30% [3], and patients should undergo lifelong follow-up [2]. Current management modalities for PTC and FTC are surgical resection of the thyroid gland, lymph node (LN) dissection, subsequent postoperative radioiodine ablation to eradicate residual cancer and normal thyroid cells, and levothyroxine replacement and suppression to prevent thyroid cancer recurrence. ...
Article
Full-text available
Fatty acid-binding protein 4 (FABP4), a fatty acid transporter that coordinates lipid metabolism, is reported to exert a tumorigenic role in certain cancers. We investigated the effects of FABP4 in the carcinogenesis of thyroid cancer. Bioinformatics data about FABP4 in thyroid cancer were collected from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). Sixteen paired papillary thyroid cancer (PTC) tissues from Taipei Medical University (TMU) were gathered, and commercial thyroid cancer complementary (c)DNA and tissue arrays were purchased to measure FABP4 messenger (m)RNA and protein levels. By analyzing data from the GEO and TCGA, we showed that FABP4 mRNA was reduced in PTC and follicular thyroid carcinoma (FTC). In addition, a lower FABP4 mRNA level in PTC was associated with poor clinical parameters and outcomes in the TCGA database. Moreover, FABP4 transcripts and proteins were downregulated in PTC and FTC, and its mRNA expression was associated with PTC staging in clinical specimens. In the TCGA database and TMU cohort, FABP4 mRNA levels were associated with thyroglobulin (r = 0.511 and r = 0.656, respectively), thyroid peroxidase (r = 0.612 and r = 0.909, respectively), and sodium iodide symporter (r = 0.485 and r = 0.637, respectively) transcripts. In conclusion, FABP4 mRNA and protein levels were reduced in PTC and FTC, and may be used as a potential indicator for thyroid cancer evolution in clinical settings. Further, well-designed research to dissect the molecular mechanism of FABP4 in modulating thyroid carcinogenesis is needed.
... Afterward, cells were washed three times with an incubation medium containing 2 mM MgCl 2 , 120 mM NaCl, 5 mM KCl, 10 mM glucose, and 20 mM HEPES -pH 7.4 (Sigma-Aldrich, St. Louis, U.S.A.). The reaction was initiated by adding 200 μL of incubation medium containing 2 mM of AMP, followed by incubation at 37 °C for different durations (15,30, and 60 minutes). After the specified incubation times, 150 μL of the incubation medium with nucleotides was transferred into microtubes containing 150 μL of 10% w/v trichloroacetic acid. ...
... BCPAP harbors BRAF V600E mutation, which is associated with more aggressive clinicopathologic characteristics, and exhibits a heightened expression of genes associated with cell cycle and DNA replication [27]. Whereas TPC is characterized by the RET/PTC1 rearrangement, which is associated with a relatively benign clinical course [28][29][30][31][32]. ...
Article
Full-text available
Background ZEB1, a core transcription factor involved in epithelial-mesenchymal transition (EMT), is associated with aggressive cancer cell behavior, treatment resistance, and poor prognosis across various tumor types. Similarly, the expression and activity of CD73, an ectonucleotidase implicated in adenosine generation, is an important marker of tumor malignancy. Growing evidence suggests that EMT and the adenosinergic pathway are intricately linked and play a pivotal role in cancer development. Therefore, this study focuses on exploring the correlations between CD73 and ZEB1, considering their impact on tumor progression. Methods We employed CRISPR/Cas9 technology to silence CD73 expression in cell lines derived from papillary thyroid carcinoma. These same cells underwent lentiviral transduction of a reporter of ZEB1 non-coding RNA regulation. We conducted studies on cell migration using scratch assays and analyses of cellular speed and polarity. Additionally, we examined ZEB1 reporter expression through flow cytometry and immunocytochemistry, complemented by Western blot analysis for protein quantification. For further insights, we applied gene signatures representing different EMT states in an RNA-seq expression analysis of papillary thyroid carcinoma samples from The Cancer Genome Atlas. Results Silencing CD73 expression led to a reduction in ZEB1 non-coding RNA regulation reporter expression in a papillary thyroid carcinoma-derived cell line. Additionally, it also mitigated ZEB1 protein expression. Moreover, the expression of CD73 and ZEB1 was correlated with alterations in cell morphology characteristics crucial for cell migration, promoting an increase in cell polarity index and cell migration speed. RNA-seq analysis revealed higher expression of NT5E (CD73) in samples with BRAF mutations, accompanied by a prevalence of partial-EMT/hybrid state signature expression. Conclusions Collectively, our findings suggest an association between CD73 expression and/or activity and the post-transcriptional regulation of ZEB1 by non-coding RNA, indicating a reduction in its absence. Further investigations are warranted to elucidate the relationship between CD73 and ZEB1, with the potential for targeting them as therapeutic alternatives for cancer treatment in the near future.
... BRAFV600E mutation is typically present in the classic and tall-cell variants, commonly related to a higher level of aggression. 5 Patients with BRAF mutation are twice as likely to experience a relapse of their illness and possess greater mortality rates than those without the mutation. 6,7 The human RAS gene is divided into Kirsten RAT sarcoma (KRAS), neuroblastoma (NRAS), and Harvey (HRAS), as opposed to BRAFV600E mutation, are more prevalent in the follicular variant of PTC. 5 Better prognosis and more indolent disease behavior have been associated with RAS mutation. ...
... 6,7 The human RAS gene is divided into Kirsten RAT sarcoma (KRAS), neuroblastoma (NRAS), and Harvey (HRAS), as opposed to BRAFV600E mutation, are more prevalent in the follicular variant of PTC. 5 Better prognosis and more indolent disease behavior have been associated with RAS mutation. 8,9 The prevalence of BRAFV600E and RAS mutations varied between Western and Asian countries, which was believed to be caused by geographic heterogeneity, race, and other risk factors. ...
Article
Full-text available
Introduction: BRAFV600E and RAS mutations are the most common gene mutations in papillary thyroid carcinoma (PTC) that may be correlated with its biological behavior. There are still limited data about BRAFV600E and RAS mutations in Indonesia. This study aims to determine the prevalence of BRAFV600E and RAS mutations, and their association with clinicopathologic characteristics. Methods: Patients who had total thyroidectomy from 2019 to 2021 and those who met our study criteria underwent PCR and DNA sequencing analysis for BRAFV600E, BRAFK601E, exon 2 and 3 of NRAS, HRAS, and KRAS. Analyses were performed to determine the associations of BRAFV600E and RAS mutations with clinicopathologic characteristics. Results: Of 172 PTC patients, BRAFV600E mutation was observed in 37.8% of the patients and RAS mutations were found in 21.5%. One patient harbored BRAFK601E mutation. There was a significant association of BRAFV600E with a high-stage (p = 0.033, OR: 3.279; 95% CI: 1.048-10.259), tall-cell variants (p ≤0.001, OR: 41.143; 95% CI: 11.979-141.308), non-encapsulated (p = 0.001, OR: 4.176; 95% CI: 2.008-8.685), lymphovascular invasion (p = 0.043, OR: 1.912; 95% CI: 1.018-3.592), extrathyroidal extension (p = <0.001, OR: 3.983; 95% CI: 1.970-8.054), and lymph node metastasis (p = 0.009, OR: 2.301; 95% CI: 1.224-4.326). Follicular variant (p = 0.001, OR: 7.011; 95% CI: 2.690-18.268), encapsulated (p = 0.017, OR: 2.433; 95% CI: 1.161-5.100), and absent of extrathyroidal extension (p = 0.033, OR: 2.890; 95% CI: 1.052-7.940) were associated with RAS mutations. Conclusion: A significant association between BRAFV600E mutation and high clinical stage, tall-cell variants, non-encapsulated morphology, lymphovascular invasion, extrathyroidal extension, and lymph node metastasis in PTC was observed. RAS mutations were associated with the follicular variant, encapsulated tumor, and no extrathyroidal extension. HRAS-mutated PTC frequently exhibited tumor multifocality.
... Based on these results, the PI3K-Akt pathway seemed to be suppressed by PTEN activation. Next, the mitogen-activated protein kinase (MAPK) pathway, which is another signaling pathway that regulates a wide variety of essential cellular processes, including proliferation, differentiation, apoptosis, and stress responses, is considered a main driving force for the proliferation of papillary thyroid carcinoma if mutations result in the activation of this pathway [9,10]. We focused on the activated p38 MAPK pathway, which is frequently a treatment target [11][12][13]. ...
Article
Full-text available
Background and aims: Clinicians sometimes encounter papillary thyroid microcarcinoma (PMC) that is less than 10 mm, associated with lymph node metastasis. In this study, we assessed PMC clinicopathologically to clarify risk factors for poor prognosis. Patients and methods: Fifty-one patients who underwent thyroid surgery at Aichi Medical University from September 2009 to October 2016 were included. Patients were divided into two groups, pEX-positive (23 patients) and pEX-negative (28 patients), based on the pathological finding of thyroid capsule invasion. The former indicates that the tumor infiltrated the thyroid capsule and spread to the neighboring tissue, and the latter indicates no capsule invasion. We analyzed factors such as patient characteristics, pathological findings, and serum levels of thyroid hormones in the two groups. Results: No statistical differences were observed between the two groups in gender distribution or age at surgery. Preoperative cancer diagnoses were established for more patients in the pEX-positive group than in the pEX-negative group (n = 21 and 14, respectively; P = 0.004). The mean (±SD) pathological tumor diameter was 5.42 ± 2.77 in the pEX-negative group and 8.32 ± 1.61 in the pEX-positive group (P < 0.001). No significant differences in preoperative serum levels of free T3, free T4, thyroid-stimulating hormone, or thyroglobulin were observed between the two groups. The odds ratio for node positivity in tumors invading thyroid capsules (pEX-positive) compared to those with no capsule invasion (pEX-negative) was 13.20 (95% confidence interval, 3.45-50.42). Immunohistological staining for phosphatase and tensin homolog deleted from chromosome 10 (PTEN) and Akt (protein kinase B) revealed the facilitation of PTEN and suppression of Akt, which might indicate downregulation of the phosphoinositide 3-kinase-Akt (PI3K-Akt) cascade. Discussion: In general, the prognosis of PMC is favorable. However, the prognosis is less favorable in patients with nodal metastasis or extrathyroidal invasion. It is controversial whether resection is required for proven PMCs. For PMCs associated with extrathyroidal invasion, regional lymph node resection with lobectomy should be performed due to the high risk for lymphatic spread. There might be a possibility that the natural progression of PMC seems to be controlled by the facilitation of PTEN. However, a tumor in the lateral peripheral region of the thyroid parenchyma might be associated with capsule invasion followed by lymphatic spread.
... Mutation of the BRAF gene is the most common genetic event in PTC, and it has been found in more than 40% of these tumors [4][5][6]. BRAF V600E point mutation is accounting for 98% of all BRAF point mutations and plays an important role in the pathogenesis and progression of PTC. BRAF V600E point mutation is reported to be present in only PTC and its congeners, but not in benign thyroid lesions [7]. ...
Article
Full-text available
Background The incidental finding of thyroid inclusions in lymph nodes of neck dissections of non-thyroid cancer patients is an unusual event. It is still controversial for pathologists about whether this represents benign inclusions or metastatic papillary thyroid carcinoma (PTC). This study is to analyze clinicopathological features of such cases in an attempt to explore their clinical implications. Methods Pathological data were searched for incidentally detected PTC of cervical lymph nodes in non-thyroid cancer cases. Clinicopathological characteristics were reevaluated and recorded. BRAF V600E protein expression and sequencing analysis was then performed in cases with sufficient tissues. Results 31 patients had an incidental finding of PTC in lymph nodes of patients with non-thyroid cancer. BRAF immunohistochemical staining were performed in 17 metastatic lymph nodes with sufficient tumor tissues, and 6 were positive. BRAF V600E point mutation was detected in 5 of 6 BRAF V600E positive cases. Subsequent imaging examinations of the thyroid showed no nodules or calcifications/benign nodules in 20 patients, and suspected malignant nodules in 5 patients. 12 patients underwent total thyroidectomy or ipsilateral lobectomy, and 6 showed PTC in postoperative pathological examinations. The remaining 19 patients without surgery were kept under active surveillance, and no one had recurrence of PTC. Conclusion Incidentally discovered PTC in lymph nodes has usually interpreted as metastasis from a clinical occult thyroid primary cancer, but primary PTC was not always detected. This suggests it could be double occult lesions. With regards to concurrence with highly malignant tumor, most patients could keep regular surveillance.
... MAPK cascades involve RAF, MEK and ERK kinases. Approximately 50% of patients with metastatic melanoma harbor a BRAF mutation (with >90% being the BRAF V600E mutation), which mediates overactivation of the MAPK signaling pathway and the survival, differentiation and proliferation of melanocytes (24,25). This oncogenic signaling may be blocked by BRAF (vemurafenib, dabrafenib and encorafenib) or MEK (cobimetinib, trametinib and binimetinib) inhibitors (Fig. 1). ...
Article
Melanoma is the most aggressive and deadly type of skin cancer and is known for its poor prognosis as soon as metastasis occurs. Since 2011, new and effective therapies for metastatic melanoma have emerged, with US Food and Drug Administration approval of multiple targeted agents, such as V-Raf murine sarcoma viral oncogene homolog B1/mitogen-activated protein kinase kinase inhibitors and multiple immunotherapy agents, such as cytotoxic T lymphocyte-associated protein 4 and anti-programmed cell death protein 1/ligand 1 blockade. Based on insight into the respective advantages of the above two strategies, the present article provided a review of clinical trials of the application of targeted therapy and immunotherapy, as well as novel approaches of their combinations for the treatment of metastatic melanoma in recent years, with a focus on upcoming initiatives to improve the efficacy of these treatment approaches for metastatic melanoma.
... The most frequent genetic mutations involve BRAF, RAS, and TSHR instabilities. To be more specific, BRAF mutations are associated with papillary thyroid carcinoma [19] and TSH-R with the gain-offunction role is associated with hyperfunctioning thyroid nodules in 20-82% of cases [20][21][22]. Nonetheless, current literature regarding these TSH-R mutations and hot nodules is still unclear [20]. ...
Article
Background: Malignant hyperfunctioning thyroid nodules are rare and more likely to occur in follicular cancer types rather than papillary variants. The authors present a case of a papillary thyroid carcinoma associated with a hyperfunctioning nodule. Methods: A single adult patient submitted to total thyroidectomy with the presence of thyroid carcinoma within hyperfunctioning nodules was selected. Additionally, brief literature was conducted. Results: An asymptomatic 58-year-old male was subjected to routine blood analysis and a TSH level of <0.003 mIU/L. Ultrasonography revealed a 21mm solid, hypoechoic, and heterogenous nodule with microcalcifications in the right lobe. A fine needle aspiration guided by ultrasound resulted in a follicular lesion of undetermined significance. A 99mTc thyroid scintigram was followed and identified a right-sided hyperfunctioning nodule. Another cytology was performed and a papillary thyroid carcinoma was derived as a result. The patient underwent a total thyroidectomy. Postoperative histology confirmed the diagnosis and a tumor-free margin with no vascular or capsular invasions. Conclusion: Hyperfunctioning malignant nodules are a rare association, although a careful approach should be led since major clinical implications arise. Selective fine needle aspiration in all suspicious ≥ 1 cm nodules should be considered.
Article
The follicular variant (FV) of papillary thyroid carcinoma is characterized by a follicular growth pattern and cytologic features of papillary carcinoma. ret/PTC rearrangements are common in classic papillary thyroid carcinoma (PTC) and PAX8-PPARγ and ras mutations in follicular thyroid carcinoma. Their prevalence in FV has not been established. We studied these genetic alterations and clinical-pathologic features in 30 FV cases and compared those with 46 non-FV papillary carcinomas. FV cases revealed 1 ret/PTC rearrangement (3%) and 13 ras mutations (43%). Non-FV cases harbored 13 ret/PTC (28%) (P = .006) and no ras mutations (P = .0002). No PAX8-PPARγ was found in either group. FV cases demonstrated a significantly higher prevalence of tumor encapsulation, angiovascular invasion, and poorly differentiated areas and a lower rate of lymph node metastases. These data indicate that the FV of papillary carcinoma has a distinct set of molecular alterations and is characterized by a high frequency of ras point mutations.
Article
Genomic clones containing 1.7 kilobases of the 5'-flanking region of the rat TSH receptor (TSHR) plus coding sequence from the ATG initiation codon [1 basepair (bp)] to the start of the first intron (170 bp) have been isolated and characterized. RNAase protection, primer extension, and cDNA sequences cloned by the anchored polymerase chain reaction identified multiple transcriptional start sites, the major ones clustered between -89 to -68 bp. This portion of the 5'-flanking region has neither a TATA nor a CCAAT box, is GC rich but has no GC box motif, and has features of promoters seen in "housekeeping" genes. Chimeras containing 1.7 kilobases (-1707 to -2 bp) of the 5'-flanking region, or deletions thereof, and the bacterial chloramphenicol acetyltransferase (CAT) gene expressed significant CAT activity when transfected into rat thyroid cell lines, FRTL-5 and FRT, but not BRL rat liver or HeLa cells. TSH decreased CAT activity in the FRTL-5 thyroid cells that had been stably transfected with the TSHR-CAT chimeric constructs. Negative regulation of promoter activity by TSH was duplicated by 10 microM forskolin in FRT thyroid cells, which express no TSHR mRNA. Deletion analyses indicated that a "minimal" region, exhibiting promoter activity, tissue specificity, and negative regulation by TSH, is located between -195 and -39 bp; this region is highly conserved in rat and human TSHR genes. Differential digestion of genomic DNA by MspI and HpaII revealed that the TSHR promoter is methylated in FRT, but not FRTL-5, cells; methylation of the promoter may be associated with loss of endogenous TSHR gene expression in FRT cells.
Article
The ret/PTC oncogene, a rearranged form of the ret proto-oncogene, has been found to be restricted to human papillary thyroid carcinomas. This report shows that transgenic mice with thyroid-targeted expression of the ret/PTC1 oncogene developed thyroid carcinomas with considerable similarities to human papillary thyroid carcinomas, particularly in the nuclear cytologic features and the presence of local invasion. Our findings indicate that ret/PTC2 is not only a biomarker associated with papillary thyroid carcinomas, but is also the only proven specific genetic event leading to the development of papillary thyroid carcinoma.
Article
Thyroid nodules are common and are frequently benign. Current data suggest that the prevalence of palpable thyroid nodules is 3% to 7% in North America; the prevalence is as high as 50% based on ultrasonography (US) or autopsy data. The introduction of sensitive thyrotropin (thyroid-stimulating hormone or TSH) assays, the widespread application of fine-needle aspiration (FNA) biopsy, and the availability of high-resolution US have substantially improved the management of thyroid nodules. This document was prepared as a collaborative effort between the American Association of Clinical Endocrinologists (AACE) and the Associazione Medici Endocrinologi (AME). Most Task Force members are members of AACE. We have used the AACE protocol for clinical practice guidelines, with rating of available evidence, linking the guidelines to the strength of recommendations. Key observations include the following. Although most patients with thyroid nodules are asymptomatic, occasionally patients complain of dysphagia, dysphonia, pressure, pain, or symptoms of hyperthyroidism or hypothyroidism. Absence of symptoms does not rule out a malignant lesion; thus, it is important to review risk factors for malignant disease. Thyroid US should not be performed as a screening test. All patients with a palpable thyroid nodule, however, should undergo US examination. US-guided FNA (US-FNA) is recommended for nodules ≥10 mm; US-FNA is suggested for nodules <10 mm only if clinical information or US features are suspicious. Thyroid FNA is reliable and safe, and smears should be interpreted by an experienced pathologist. Patients with benign thyroid nodules should undergo follow-up, and malignant or suspicious nodules should be treated surgically. A radioisotope scan of the thyroid is useful if the TSH level is low or suppressed. Measurement of serum TSH is the best initial laboratory test of thyroid function and should be followed by measurement of free thyroxine if the TSH value is low and of thyroid peroxidase antibody if the TSH value is high. Percutaneous ethanol injection is useful in the treatment of cystic thyroid lesions; large, symptomatic goiters may be treated surgically or with radioiodine. Routine measurement of serum calcitonin is not recommended. Suggestions for thyroid nodule management during pregnancy are presented. We believe that these guidelines will be useful to clinical endocrinologists, endocrine surgeons, pediatricians, and internists whose practices include management of patients with thyroid disorders. These guidelines are thorough and practical, and they offer reasoned and balanced recommendations based on the best available evidence.
Article
Here we report the analysis of the Na(+)/I(-) symporter (NIS) protein expression in 57 thyroid cancer samples by immunohistochemistry with high-affinity anti-NIS Abs. As many as 70% of these samples exhibited increased NIS expression with respect to the normal surrounding thyroid tissue. Most significantly, NIS was located in these samples either in both the plasma membrane and intracellular compartments simultaneously, or exclusively in intracellular compartments. This suggests that NIS is clearly expressed or even overexpressed in most thyroid cancer cells, but malignant transformation in some of these cells interferes either with the proper targeting of NIS to the plasma membrane, or with the mechanisms that retain NIS in the plasma membrane after it has been targeted. The results further indicate that, in addition to indicating NIS expression in cases where it is absent (approximately 30%), improvements in (131)I radioablation therapy might result from promoting targeting of NIS to the plasma membrane in the majority (approximately 70%) of thyroid cancers.