Sei sulla pagina 1di 9

European Journal of Endocrinology (2006) 155 645653 ISSN 0804-4643

INVITED REVIEW

RET/PTC activation in papillary thyroid carcinoma: European


Journal of Endocrinology Prize Lecture
Massimo Santoro1, Rosa Marina Melillo1 and Alfredo Fusco1,2
1
Istituto di Endocrinologia ed Oncologia Sperimentale del CNR G. Salvatore, c/o Dipartimento di Biologia e Patologia Cellulare e Molecolare, University
Federico II, Via S. Pansini, 5, 80131 Naples, Italy and 2NOGEC (Naples Oncogenomic Center)CEINGE, Biotecnologie Avanzate & SEMM, European
School of Molecular Medicine, Naples, Italy
(Correspondence should be addressed to M Santoro; Email: masantor@unina.it)

Abstract
Papillary thyroid carcinoma (PTC) is frequently associated with RET gene rearrangements that
generate the so-called RET/PTC oncogenes. In this review, we examine the data about the mechanisms
of thyroid cell transformation, activation of downstream signal transduction pathways and
modulation of gene expression induced by RET/PTC. These findings have advanced our understanding
of the processes underlying PTC formation and provide the basis for novel therapeutic approaches to
this disease.

European Journal of Endocrinology 155 645653

RET/PTC rearrangements in papillary growth factor, have been described in a fraction of PTC
thyroid carcinoma patients (7). As illustrated in figure 1, many different
genes have been found to be rearranged with RET in
The rearranged during tansfection (RET) proto-onco- individual PTC patients. RET/PTC1 and 3 account for
gene, located on chromosome 10q11.2, was isolated in more than 90% of all rearrangements and are hence, by
1985 and shown to be activated by a DNA rearrange- far, the most frequent variants (811). They result from
ment (rearranged during transfection) (1). As the fusion of RET to the coiled-coil domain containing
illustrated in Fig. 1, it encodes a single-pass trans- gene 6 (CCDC6, formerly called H4/D10S170) or to the
membrane tyrosine kinase that functions as the nuclear receptor co-activator gene 4 (NcoA4, formerly
receptor (receptor tyrosine kinase, RTK) for the growth called RET fused gene (RFG)/ELE1/androgen receptor
factors of the glial cell line-derived neurotropic factor activator 70(ARA70)) (811).
family (2). RET is essential for the development of the
sympathetic, parasympathetic, and enteric nervous
systems, the kidney and the testis (2). Germline point Prevalence of RET/PTC rearrangements
mutations in RET lead to multiple endocrine neoplasia
type 2 syndromes thereby predisposing to thyroid C-cell- For a detailed discussion of the prevalence of RET/PTC
derived medullary thyroid carcinoma (3). rearrangements and their correlation with clinico-
With an incidence ranging between 0.5 and 10 cases pathological features of thyroid carcinomas, we refer
per 100 000 population, thyroid cancer is the most the reader to three excellent reviews (810). The
frequent endocrine malignancy (4). Papillary thyroid prevalence of RET/PTC rearrangements in thyroid
carcinoma (PTC), which accounts for approximately cancer varies widely among studies. In an adult
population from the United States, the prevalence of
80% of cases, is the most frequent of all thyroid
rearrangements was approximately 35% (9). Preva-
malignancies (4). In PTC, genomic rearrangements
lences between 3 and 85% have been reported for other
juxtapose the RET kinase and COOH-terminus encoding
regions (810). This wide range of values probably
domains (exons 1121) to unrelated genes, thereby reflects not only the geographic variation but also the
creating dominantly transforming oncogenes called different procedures used to identify RET/PTC
RET/PTC (5, 6). Similar rearrangements of another rearrangements, i.e. various reverse transcriptase
neurotropic RTK, namely the neurotropic tyrosine (RT)-PCR methods, Southern blot, and fluorescence
receptor kinase type 1 (NTRK1), the receptor for nerve in situ hybridization. An exhaustive study by Zhu and
co-workers (12) demonstrated that the method used has
Presented at the 7th European Congress of Endocrinology, a striking effect on the efficacy of RET/PTC detection,
Gothenburg, Sweden, 2005. and hence on the prevalence reported. Tumor

q 2006 Society of the European Journal of Endocrinology DOI: 10.1530/eje.1.02289


Online version via www.eje-online.org
646 M Santoro and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155

Figure 1 Schematic drawing of the RET


protein with the four extracellular cadherin-like
domains, the cysteine-rich box adjacent to the
plasma membrane, the juxtamembrane
domain and the split tyrosine kinase domain
(TK). In PTCs, RET is rearranged with diverse
genes, encoding protein dimerization motifs
(highlighted) that mediate ligand-independent
RET dimerization. The arrowheads indicate
RET breakpoints.

heterogeneity is another factor that can affect the the presence of multiple RET/PTC variants in individ-
evaluation of RET/PTC prevalence. In addition to PTC ual PTC samples (14).
samples with clonal RET/PTC rearrangements (those RET/PTC rearrangements have so far been identified
affecting the majority of tumor cells), there are samples only in thyroid lesions, and in particular in PTC (810).
with non-clonal rearrangements (those affecting a Most studies concur that RET/PTC rearrangements are
small portion of tumor cells) (12, 13). Whereas RET/ rare or absent in benign follicular adenomas, and absent
PTC is likely to be important for tumor formation in in follicular and medullary carcinomas (810). Various
samples with clonal rearrangements, RET/PTC could PTC histological variants have been identified, namely
not be a driver mutation in PTC samples with non- classic, follicular, diffuse-sclerosing, columnar-cell,
clonal rearrangements (13). This distinction has Hurthle-cell, cribriform, solid and tall-cell variants (4).
important implications for the stratification of patients Classic (w45%) and follicular (w18%) variants are the
who could potentially benefit from novel therapeutic most prevalent (4). RET/PTCs are more frequent in
approaches based on the use of RET kinase inhibitors tumors that have a classic architecture (15) and in
(see below). It is still controversial the presence of RET/ microcarcinomas (!1 cm) (16). They are rare in the
PTC rearrangements in non-neoplastic cells in Hashi- follicular variant of PTC (15). RET/PTCs have been
motos thyroiditis. It is possible that a heterogeneous reported in the cribriform variant, which is typically
presence of the rearrangement may account, at least in associated with familial adenomatous polyposis (17), in
part, for such a controversy (8, 9, 13). Tumor the Hurthle-cell variant (18, 19), and in hyalinizing
heterogeneity and multiclonality is also indicated by trabecular adenoma, a rare tumor that can be

www.eje-online.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155 RET/PTC in thyroid cancer 647

morphologically similar to PTC (20, 21). The solid of childhood PTC was reported in contaminated areas.
variant, which is an aggressive PTC subtype, is closely Post-Chernobyl PTC featured a high prevalence (over
associated with the RET/PTC3 oncogene, whereas the 60%) of RET/PTC rearrangements (22, 3639).
classic variant is associated with RET/PTC1 (22). RET/PTC3 was more prevalent among short latency
Clinical, epidemiologic, and pathologic evidence solid PTCs, whereas RET/PTC1 was more frequently
supports the possibility of a stepwise progression from found after a long latency period (9, 22, 3639). Rapid
well-differentiated carcinoma, including PTC, to poorly thyroid cell proliferation may account for the particu-
differentiated and anaplastic carcinoma (23). Accor- larly high sensitivity to radiation-induced RET/PTC
dingly, some anaplastic carcinomas are associated with rearrangements among children (40). However, also in
genetic lesions (rat sarcoma viral oncogene homolog cases of PTC in non-exposed populations, the prevalence
(RAS) and BRAF mutations) that overlap those present of RET/PTC was higher in children than in adults,
in PTC and in other well-differentiated carcinomas (23). which suggests that in general RET/PTC recombina-
Conflicting results have been reported about the tions occur more frequently in young thyroids (41, 42).
presence of RET/PTC in poorly-differentiated and RET/PTC rearrangements are not the only intrachro-
anaplastic carcinomas and, therefore about whether mosomal aberration found in radiation-associated PTC.
or not RET/PTC predisposes to thyroid tumor pro- Also NTRK1 rearrangements, caused by paracentric
gression (2328). Again, methodological differences inversions of chromosome 1q, have been reported in
and tumor heterogeneity may account for these post-Chernobyl PTC (7, 38). Moreover, in 11% of the post-
controversies. A large multicenter study in which all Chernobyl PTCs there was a paracentric inversion of
centers use the same methodology should resolve this chromosome 7q that resulted in the A-kinase anchor
issue. It is also important to study the different RET/PTC protein (AKAP)9BRAF fusion oncogene (43). Differently,
variants individually, because, as mentioned above, they point mutations in BRAF, which are very frequent in
could confer different degrees of aggressiveness on PTC. sporadic PTC (see below) are rare in Chernobyl cancers
It should be noted that intercross of RET/PTC transgenic affecting children (44). This suggests that young age and
mice with p53null mice induced the formation of radiation exposure are specifically linked to gene
anaplastic carcinomas, suggesting that at least in a rearrangements in PTC.
specific genetic background RET/PTC is able to induce The reason for the high frequency of chromosomal
thyroid tumor progression (29). rearrangements, and particularly paracentric inver-
sions, in thyroid cancer after radiation exposure is not
clear. Nikiforova and co-workers (45) shed new light on
this issue when they demonstrated that, although the
Chromosomal basis for RET/PTC two loci participating in the formation of the RET/PTC1
rearrangements fusion are about 30 megabases apart, they frequently
CCDC6 (the RET fusion partner in RET/PTC1) and juxtapose within the interfase nuclei of thyroid cells.
NcoA4 (the RET fusion partner in RET/PTC3 and 4) This spatial contiguity can provide a structural basis for
map together with RET on chromosome 10, whereas radiation-induced illegitimate recombination of the two
the other RET fusion partners are located on different genes. More recently, the same group showed that
chromosomes. Therefore, RET/PTC recombination NcoA4 is also in close proximity to RET in thyroid cell
events are caused either by a paracentric inversion of chromatin (46). A similar mechanism was proposed for
chromosome 10 (in the case of RET/PTC1 and NTRK1 fusions in PTC (47). Taken together, these
RET/PTC3) or balanced chromosomal translocations findings suggest that tissue-specific chromatin folding is
involving chromosome 10 and other chromosomes the genomic basis underlying the high prevalence of
(in the case of the other RET/PTC subtypes). It is well gene rearrangements in PTC.
known that ionizing radiations promote DNA double-
strand breaks and that PTC is the most common form of
solid neoplasm associated with radiation exposure. Molecular mechanisms of activation of
These findings prompted the notion that there is a RET/PTC oncoproteins
direct link between radiations, RET/PTC rearrange-
ments, and PTC formation. At least two observations Adoptive expression of RET/PTC transforms murine
support this concept. First, X-ray irradiation induces the fibroblasts (5, 6). Chronic RET/PTC expression causes
formation of RET/PTC1 (3032). Second, RET/PTC thyrotropin-independent proliferation and impairs the
rearrangements are very frequent in PTC samples from expression of the thyroid differentiation markers of rat
patients, who had received external radiations (3335) thyroid follicular PC Cl 3 cells (4850). On the other
and in post-Chernobyl PTCs (22, 3639). The Cherno- hand, acute expression of RET/PTC oncogenes induces
byl nuclear power plant accident in April 1986 resulted apoptosis of PC Cl 3 cells (49, 51), a phenomenon that is
in the release of large amounts of iodine isotopes, often seen when dominant oncogenes are acutely
mainly 131I, and, therefore, there was widespread introduced in normal cells. RET/PTC expression in
radiation exposure to the thyroid (36). A high incidence primary cultures of human thyrocytes results in changes

www.eje-online.org
648 M Santoro and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155

of nuclear morphology (irregular nuclear contours and other receptors, the juxtamembrane domain negatively
euchromasia) resembling those characterizing human affects RET mitogenic signaling although the molecular
PTC (52). Finally, thyroid targeting of RET/PTC1 (53, 54) mechanisms governing this process are not clear (60).
or RET/PTC3 (55) in transgenic mice leads to the On the other hand, all the translocated amino termini
development of tumors that histologically and cytologi- fused to RET are predicted to fold into coiled-coils (Figs 1
cally resemble human PTC. Interestingly, mice carrying and 2 mechanism no. 3). Coiled-coils are protein
RET/PTC3 develop thyroid tumors, whose solid pheno- protein interaction domains able to mediate dimeri-
type resembles those associated with activation of this zation and activation of the kinase. Accordingly, RET/
RET/PTC variant in humans (22, 55). RET/PTC3 is more PTC proteins form dimers that are essential for
potently transforming than RET/PTC1 also when tested oncogenic activation (58, 61). Whereas wild-type RET
in cultured thyrocytes in vitro (56). is a transmembrane protein, as a further consequence of
RET/PTC can exert oncogenic activity via various the deletion (mechanism no. 4 in Fig. 2), RET/PTC
mechanisms (schematically illustrated in Fig. 2); altered oncoproteins are re-localized to the cytosolic compart-
expression, ligand-independent kinase activation, sub- ment. This could prevent RET/PTC from interacting
cellular relocalization and functional alteration of the with negative regulators located at plasma membrane
RET fusion partner. Whereas wild-type RET expression level, for instance, the tyrosine phosphatase protein
is tightly regulated (2), its fusion partners are tyrosine phosphatase receptor type-J (PTPRJ) (62)
ubiquitously expressed (57, 58). Therefore, RET/PTC (Fig. 2). However, it should be noted that by interacting
rearrangements may lead to the unscheduled with proteins resident at the plasma membrane, e.g.
expression of RET in the thyroid cell (mechanism no. Enigma (63) or FRS2 (64), RET/PTC proteins may be
1 in Fig. 2). However, also unrearranged RET is re-directed to the membrane compartment. Finally
expressed in thyroid follicular cells and in tumors that (mechanism no. 5 in Fig. 2), RET/PTC rearrangements,
derive from them (59), and therefore the rearrangement besides activating RET, can also alter the function of
might be not essential for RET expression in the thyroid. RET-fused genes. This could explain why the RET/PTC
The mechanisms driving the ligand-independent kinase variants constituted by different fusion partners might
activity of RET/PTC oncoproteins have been partially have, at least in part, non-overlapping biological effects.
elucidated. The rearrangement deletes the signal Perhaps, the best example of a RET fusion partner,
sequence, the extracellular ligand-binding domain and whose altered function may contribute to neoplastic
the intracellular juxtamembrane domains of the transformation is the regulatory subunit type I-a of
receptor (mechanism no. 2 in Fig. 2). As in the case of protein kinase A (PRKAR1A) that is involved in RET/

Figure 2 Mechanisms for RET/PTC activation. The wild-type RET protein, the PTPRJ tyrosine phosphatase, and the rearranged
RET/PTC1 oncoproteins are depicted.

www.eje-online.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155 RET/PTC in thyroid cancer 649

PTC2. PRKAR1A is, indeed, a bona fide tumor


suppressor (65) and its ablation in the mouse induced
the formation of thyroid tumors (66). The knowledge
about the function of the other RET fusion partners is
limited. Some of them are involved in transcriptional
regulation, such as Ncoa4, tripartite motif-containing
(TRIM)24 (formerly called HTIF1, human transcrip-
tional intermediary factor I), methyl-CpG binding
domain protein 1 (MBD1) (formerly called PCM1,
pericentriolar material 1), and RET finger protein
(RFP) (formerly called TRIM27) (www.ncbi.nlm.nih.
gov/entrez/), or in cell fate determination, such as
TRIM33 (formerly called RFG7/Ectodermin/HTIFg)
(67). Ncoa4 functions as a co-activator of several
nuclear receptors. It interacts with and promotes
androgen receptor activity via the consensus FXXLF
motif (located at amino acids 328332) (68), and
interacts with and promotes peroxisome-proliferator-
activated receptor-g via the LXXLL motif located at
amino acids 9296 (69). Therefore, its rearrangement
may affect the function of these steroid hormone
receptors. Surprisingly, we still know very little about
CCDC6, the first isolated RET fusion partner (6, 57).
According to the conserved domain database (www.
ncbi.nlm.nih.gov/Structure/cdd/), this protein, like
golgi autoantigen glogin subfamily A (GOLGA)5
(formerly called RFG5) and KTN1 (kinectin; the fusion
partners in RET/PTC5 and 8 respectively) contains the
signature sequence of the ATP-binding cassette family of
ATPases, which are proteins involved in DNA mainten-
ance and repair. It is still unknown whether such a
function is altered consequent to the RET/PTC1
rearrangement. Of note, CCDC6 overexpression induces
apoptosis, suggesting that abrogation of its function
may promote PTC cell survival (57). Intriguingly, some
of the RET fusion partners are found rearranged in
tumors other than PTC; CCDC6 rearranges with the
kinase domain of the platelet-derived growth factor Figure 3 RET/PTC binds adaptor proteins such as growth factor
receptor-b in myeloproliferative disorders (70), TRIM24 receptor-bound protein 2 (GRB2) that, in cooperation with the
guanine nucleotide exchange factor SOS, activates RAS. In its
with BRAF in mouse hepatocellular carcinomas (71), GTP-bound form, RAS activates RAF kinases (such as BRAF), and
and PCM1 with JAK2 in chronic myeloid leukemia (72). its downstream signaling cascade (MEK and ERK). There are
several negative regulators of this pathway but the possible role of
these mediators in RET/PTC signaling has not been studied yet.
These proteins include the sprouty family of proteins (SPRY), which
RET/PTC and the MAPK signaling can inhibit the pathway at various levels; RAF kinase inhibitor
protein (RKIP); RAS and RAB interactor 1 (RIN1); IMP (impedes
cascade mitogenic signal propagation); and the MKP (dual specificity
phosphatase) family of MAPK phosphatases.
The most common genetic alteration found in PTC (up to
45% of cases) is the activating point mutation (most often
V600E) in BRAF (73). Together with the other onco- adaptors, IRS1/2, FRS2, and DOK1/4/5 (2, 11, 75).
proteins implicated in PTC (RET, NTRK1 and RAS), BRAF Binding to Shc and FRS2 mediates recruitment of growth
functions in the mitogen-activated protein kinase (MAPK) factor receptor-bound protein 2 (GRB2)/son of sevenless
pathway. By promoting GTP loading on RAS, RET and (SOS) complexes, thereby leading to RAS and, in turn,
NTRK1 stimulate RAF family kinases, which leads to the BRAF/MAPK stimulation (48, 50).
phosphorylation of MEK (MAP kinase or ERK) and ERK A wealth of experimental evidence supports the role
(extracellular signal-regulated kinases or MAPK) (74) of this signaling cascade in thyroid cell transformation:
(Fig. 3). One tyrosine residue (tyrosine 1062) in RET (i) RET/PTC-mediated transformation requires BRAF
mediates the firing of this cascade. This tyrosine is the (48, 50, 76); (ii) PTC formation in transgenic mice
binding site for several proteins including Shc family requires the integrity of the RET/PTC tyrosine

www.eje-online.org
650 M Santoro and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155

corresponding to Y1062 in wild-type RET (77); and (iii) reaction might be part of the oncogenic effect of RET/
transcriptional responses to RET/PTC and BRAF PTC on thyroid cancer and may explain the chronic
partially overlap (see also below) (48, 76, 78). Such inflammatory reaction that is characteristic of this
an assembly of oncogenic proteins in one signaling tumor type.
cascade has numerous implications; one is that other
genes implicated in PTC formations may encode
proteins functioning in the same cascade. Many
proteins (schematically represented in Fig. 3) are Concluding remarks
potentially able to modulate the RET/PTC-BRAF- Since the RET/PTC oncogene was isolated almost three
MAPK axis (79); it would be well worth investigating decades ago (5), much progress has been made in our
their activity in thyroid cell transformation. understanding of the effect exerted by this oncogene. Its
Not all the biological activities of the RET/PTC and relationship with radiation exposure has been estab-
BRAF oncoproteins overlap. Indeed, activated BRAF but lished, the chromosomal basis for its activation has been
not RET/PTC induces genomic instability in cultured clarified, its transforming and signaling properties have
thyroid cells (80); BRAF (but not RET/PTC) transgenic been characterized, and finally we now know that RET/
mice develop PTCs that progress to undifferentiated PTC triggers a pro-inflammatory response. Over the
carcinomas (81); and RET/PTC and BRAF regulate the last few years, it has become clear that cancers
expression of specific sets of genes as well as the characterized by activating mutations in dominant
expression of common genes (48, 78, 82, 83). Thus, oncogenes can be treated by directly targeting the
although functioning on the same pathway, RET/PTC disease-causing oncoprotein. In this framework, RET
and BRAF are endowed with differential signaling and the downstream signaling cascade are promising
abilities. This could explain the different clinico- therapeutic targets. Several inhibitors of RET, BRAF, and
pathologic features of PTCs associated with the two MEK kinases have been isolated (11, 91) and hopefully,
oncogenes. In fact, whereas most RET/PTC-positive their clinical use will lead to novel therapeutic options
PTCs belong to the classic variant and are associated for patients afflicted by thyroid cancer.
with a young age and an early stage at presentation,
BRAF-positive PTCs belong to the classic and tall cell
variants and are associated with old age and an
advanced tumor stage (15, 73). Acknowledgements
We thank members of our laboratory for scientific
advice and Jean A Gilder for text editing. We apologize
The pro-inflammatory transcriptional to many of our colleagues for failure to cite their work
program regulated by RET/PTC owing to space limitations.
The transcriptional response to RET/PTC has been
evaluated both in rat thyroid PC Cl 3 cells, either stably
(48, 84) or conditionally (76, 78, 85) expressing RET/ References
PTC3, and in human thyrocytes expressing RET/PTC1 1 Takahashi M, Ritz J & Cooper GM. Activation of a novel human
(86). Although some differences were noted (perhaps transforming gene, ret, by DNA rearrangement. Cell 1985 42
linked to the different model systems used), these studies 581588.
reached two conclusions. First, as anticipated above, 2 Airaksinen MS & Saarma M. The GDNF family: signalling,
tyrosine 1062 and the downstream signaling cascade is biological functions and therapeutic value. Nature Reviews
Neuroscience 2002 3 383394.
particularly important for RET/PTC-mediated effects 3 Marx SJ. Molecular genetics of multiple endocrine neoplasia types
(48, 76, 84, 86). Second, RET/PTC induced the 1 and 2. Nature Reviews. Cancer 2005 5 367375.
expression of a pro-inflammatory transcriptional 4 DeLellis RA & Williams ED. Thyroid and parathyroid tumors. In
program. Such a program included the up-regulation World Health Organization Classification of Tumours. Pathology and
Genetics. Tumours of Endocrine organs, pp 5156. Ed. RA DeLellis,
of various cytokines (OPN/SPP1, GM-CSF, M-CSF, RV Lloyd, PU Heitz & C Eng, Geneva: WHO Press, 2003.
G-CSF, IL1A, IL1B, IL6, and IL24) (8487), chemokines 5 Fusco A, Grieco M, Santoro M, Berlingieri MT, Pilotti S,
(CCL2, CCL20, CXCL8, CXCL1, CXCL10, and CXCL12) Pierotti MA, Della Porta G & Vecchio G. A new oncogene in
(48, 8486), chemokine receptors (CXCR4) (48, 86, human thyroid papillary carcinomas and their lymph-nodal
88), pro-inflammatory enzymes (cyclooxygenase-2, and metastases. Nature 1987 328 170172.
6 Grieco M, Santoro M, Berlingieri MT, Melillo RM, Donghi R,
microsomal prostaglandin E2 synthase) (84, 85, 89), Bongarzone I, Pierotti MA, Della Porta G, Fusco A & Vecchio G.
and interferon-dependent genes (85). Many of these PTC is a novel rearranged form of the ret proto-oncogene and is
mediators were also upregulated in the thyroids of RET/ frequently detected in vivo in human thyroid papillary carcinomas.
PTC3 transgenics (90). Moreover, injection of RET/PTC- Cell 1990 60 557563.
7 Pierotti MA & Greco A. Oncogenic rearrangements of the
transformed PC Cl 3 cells induced angiogenesis and NTRK1/NGF receptor. Cancer Letters 2006 232 9098.
inflammatory responses in SCID mice (84). These 8 Tallini G & Asa SL. RET oncogene activation in papillary thyroid
findings suggest that induction of an inflammatory-type carcinoma. Advances in Anatomic Pathology 2001 8 345354.

www.eje-online.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155 RET/PTC in thyroid cancer 651

9 Nikiforov YE. RET/PTC rearrangement in thyroid tumors. 26 Soares P, Fonseca E, Wynford-Thomas D & Sobrinho-Simoes M.
Endocrine Pathology 2002 13 316. Sporadic ret-rearranged papillary carcinoma of the thyroid: a
10 Kondo T, Ezzat S & Asa SL. Pathogenetic mechanisms in thyroid subset of slow growing, less aggressive thyroid neoplasms?
follicular-cell neoplasia. Nature Reviews Cancer 2006 6 292306. Journal of Pathology 1998 185 7178.
11 Santoro M & Carlomagno F. Drug insight: small-molecule inhibitors 27 Tallini G, Santoro M, Helie M, Carlomagno F, Salvatore G,
of protein kinases in the treatment of thyroid cancer. Nature Clinical Chiappetta G, Carcangiu ML & Fusco A. RET/PTC oncogene
Practice. Endocrinology and Metabolism 2006 2 4252. activation defines a subset of papillary thyroid carcinomas lacking
12 Zhu Z, Ciampi R, Nikiforova MN, Gandhi M & Nikiforov YE. evidence of progression to poorly differentiated or undifferentiated
Prevalence of RET/PTC rearrangements in thyroid papillary tumor phenotypes. Clinical Cancer Research 1998 4 287294.
carcinomas: effects of the detection methods and genetic 28 Santoro M, Papotti M, Chiappetta G, Garcia-Rostan G, Volante M,
heterogeneity. Journal of Clinical Endocrinology and Metabolism Johnson C, Camp RL, Pentimalli F, Monaco C, Herrero A,
2006 91 36033610. Carcangiu ML, Fusco A & Tallini G. RET activation and
13 Rhoden KJ, Unger K, Salvatore G, Yilmaz Y, Vovk V, Chiappetta G, clinicopathologic features in poorly differentiated thyroid tumors.
Qumsiyeh MB, Rothstein JL, Fusco A, Santoro M, Zitzelsberger H & Journal of Clinical Endocrinology and Metabolism 2002 87
Tallini G. RET/papillary thyroid cancer rearrangement in non- 370379.
neoplastic thyrocytes: follicular cells of Hashimotos thyroiditis share 29 La Perle KM, Jhiang SM & Capen CC. Loss of p53 promotes
low-level recombination events with a subset of papillary carcinoma. anaplasia and local invasion in ret/PTC1-induced thyroid
Journal of Clinical Endocrinology and Metabolism 2006 91 24142423. carcinomas. American Journal of Pathology 2000 157 671677.
14 Sugg SL, Ezzat S, Rosen IB, Freeman JL & Asa SL. Distinct multiple 30 Ito T, Seyama T, Iwamoto KS, Hayashi T, Mizuno T, Tsuyama N,
RET/PTC gene rearrangements in multifocal papillary thyroid Dohi K, Nakamura N & Akiyama M. In vitro irradiation is able to
neoplasia. Journal of Clinical Endocrinology and Metabolism 1998 83 cause RET oncogene rearrangement. Cancer Research 1993 53
41164122. 29402943.
15 Adeniran AJ, Zhu Z, Gandhi M, Steward DL, Fidler JP, Giordano TJ, 31 Mizuno T, Kyoizumi S, Suzuki T, Iwamoto KS & Seyama T.
Biddinger PW & Nikiforov YE. Correlation between genetic Continued expression of a tissue specific activated oncogene in the
alterations and microscopic features, clinical manifestations, and early steps of radiation-induced human thyroid carcinogenesis.
prognostic characteristics of thyroid papillary carcinomas. Oncogene 1997 15 14551460.
American Journal of Surgical Pathology 2006 30 216222. 32 Mizuno T, Iwamoto KS, Kyoizumi S, Nagamura H, Shinohara T,
16 Fusco A, Chiappetta G, Hui P, Garcia-Rostan G, Golden L, Koyama K, Seyama T & Hamatani K. Preferential induction of
Kinder BK, Dillon DA, Giuliano A, Cirafici AM, Santoro M, RET/PTC1 rearrangement by X-ray irradiation. Oncogene 2000
Rosai J & Tallini G. Assessment of RET/PTC oncogene activation 19 438443.
and clonality in thyroid nodules with incomplete morphological 33 Bounacer A, Wicker R, Caillou B, Cailleux AF, Sarasin A,
evidence of papillary carcinoma: a search for the early precursors
Schlumberger M & Suarez HG. High prevalence of activating ret
of papillary cancer. American Journal of Pathology 2002 60
proto-oncogene rearrangements, in thyroid tumors from patients
21572167.
who had received external radiation. Oncogene 1997 15
17 Cetta F, Chiappetta G, Melillo RM, Petracci M, Montalto G,
12631273.
Santoro M & Fusco A. The RET/PTC1 oncogene is activated in
34 Collins BJ, Chiappetta G, Schneider AB, Santoro M, Pentimalli F,
familial adenomatous polyposis-associated thyroid papillary
Fogelfeld L, Gierlowski T, Shore-Freedman E, Jaffe G & Fusco A.
carcinomas. Journal of Clinical Endocrinology and Metabolism
RET expression in papillary thyroid cancer from patients irradiated
1998 83 10031006.
in childhood for benign conditions. Journal of Clinical Endocrinology
18 Cheung CC, Ezzat S, Ramyar L, Freeman JL & Asa SL. Molecular
basis of Hurthle cell papillary thyroid carcinoma. Journal of Clinical and Metabolism 2002 87 39413946.
Endocrinology and Metabolism 2000 85 878882. 35 Alipov G, Ito M, Prouglo Y, Takamura N & Yamashita S. Ret proto-
19 Chiappetta G, Toti P, Cetta F, Giuliano A, Pentimalli F, Amendola I, oncogene rearrangement in thyroid cancer around Semipalatinsk
Lazzi S, Monaco M, Mazzuchelli L, Tosi P, Santoro M & Fusco A. nuclear testing site. Lancet 1999 354 15281529.
The RET/PTC oncogene is frequently activated in oncocytic 36 Williams D. Cancer after nuclear fallout: lessons from the
thyroid tumors (Hurthle cell adenomas and carcinomas), but Chernobyl accident. Nature Reviews Cancer 2002 2 543549.
not in oncocytic hyperplastic lesions. Journal of Clinical 37 Fugazzola L, Pilotti S, Pinchera A, Vorontsova TV, Mondellini P,
Endocrinology and Metabolism 2002 87 364369. Bongarzone I, Greco A, Astakhova L, Butti MG, Demidchik EP &
20 Cheung CC, Boerner SL, MacMillan CM, Ramyar L & Asa SL. Pierotti M. Oncogenic rearrangements of the RET proto-oncogene
Hyalinizing trabecular tumor of the thyroid: a variant of papillary in papillary thyroid carcinomas from children exposed to the
carcinoma proved by molecular genetics. American Journal of Chernobyl nuclear accident. Cancer Research 1995 55 56175620.
Surgical Pathology 2000 24 16221626. 38 Rabes HM, Demidchik EP, Sidorow JD, Lengfelder E, Beimfohr C,
21 Papotti M, Volante M, Giuliano A, Fassina A, Fusco A, Bussolati G, Hoelzel D & Klugbauer S. Pattern of radiation-induced RET
Santoro M & Chiappetta G. RET/PTC activation in hyalinizing and NTRK1 rearrangements in 191 post-chernobyl papillary
trabecular tumors of the thyroid. American Journal of Surgical thyroid carcinomas: biological, phenotypic, and clinical impli-
Pathology 2000 24 16151621. cations. Clinical Cancer Research 2000 6 10931103.
22 Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H & 39 Thomas GA, Bunnell H, Cook HA, Williams ED, Nerovnya A,
Fagin JA. Distinct pattern of ret oncogene rearrangements in Cherstvoy ED, Tronko ND, Bogdanova TI, Chiappetta G,
morphological variants of radiation-induced and sporadic thyroid Viglietto G, Pentimalli F, Salvatore G, Fusco A, Santoro M &
papillary carcinomas in children. Cancer Research 1997 57 Vecchio G. High prevalence of RET/PTC rearrangements in
16901694. Ukrainian and Belarussian post-Chernobyl thyroid papillary
23 Nikiforov YE. Genetic alterations involved in the transition from carcinomas: a strong correlation between RET/PTC3 and the
well-differentiated to poorly differentiated and anaplastic thyroid solid-follicular variant. Journal of Clinical Endocrinology and
carcinomas. Endocrine Pathology 2004 15 319327. Metabolism 1999 84 42324238.
24 Bongarzone I, Vigneri P, Mariani L, Collini P, Pilotti S & 40 Saad AG, Kumar S, Ron E, Lubin JH, Stanek J, Bove KE &
Pierotti MA. RET/NTRK1 rearrangements in thyroid gland tumors Nikiforov YE. Proliferative activity of human thyroid cells in
of the papillary carcinoma family: correlation with clinicopatho- various age groups and its correlation with the risk of thyroid
logical features. Clinical Cancer Research 1998 4 223228. cancer after radiation exposure. Journal of Clinical Endocrinology
25 Jhiang SM, Caruso DR, Gilmore E, Ishizaka Y, Tahira T, Nagao M, and Metabolism 2006 91 26722677.
Chiu IM & Mazzaferri EL. Detection of the PTC/retTPC oncogene in 41 Bongarzone I, Fugazzola L, Vigneri P, Mariani L, Mondellini P,
human thyroid cancers. Oncogene 1992 7 13311337. Pacini F, Basolo F, Pinchera A, Pilotti S & Pierotti MA. Age-related

www.eje-online.org
652 M Santoro and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155

activation of the tyrosine kinase receptor protooncogenes RET and RET/PTC3 oncogene correlates with its prevalence in tall-cell
NTRK1 in papillary thyroid carcinoma. Journal of Clinical variant of papillary thyroid carcinoma. American Journal of
Endocrinology and Metabolism 1996 81 20062009. Pathology 2002 160 247254.
42 Powell N, Jeremiah S, Morishita M, Dudley E, Bethel J, 57 Celetti A, Cerrato A, Merolla F, Vitagliano D, Vecchio G & Grieco M.
Bogdanova T, Tronko M & Thomas G. Frequency of BRAF H4(D10S170), a gene frequently rearranged with RET in papillary
T1796A mutation in papillary thyroid carcinoma relates to age thyroid carcinomas: functional characterization. Oncogene 2004
of patient at diagnosis and not to radiation exposure. Journal of 23 109121.
Pathology 2005 205 558564. 58 Monaco C, Visconti R, Barone MV, Pierantoni GM, Berlingieri MT,
43 Ciampi R, Knauf JA, Kerler R, Gandhi M, Zhu Z, De Lorenzo C, Mineo A, Vecchio G, Fusco A & Santoro M. The RFG
Nikiforova MN, Rabes HM, Fagin JA & Nikiforov YE. oligomerization domain mediates kinase activation and re-locali-
Oncogenic AKAP9-BRAF fusion is a novel mechanism of zation of the RET/PTC3 oncoprotein to the plasma membrane.
MAPK pathway activation in thyroid cancer. Journal of Clinical Oncogene 2001 20 599608.
Investigation 2005 115 94101. 59 Bunone G, Uggeri M, Mondellini P, Pierotti MA & Bongarzone I.
44 Lima J, Trovisco V, Soares P, Maximo V, Magalhaes J, Salvatore G, RET receptor expression in thyroid follicular epithelial cell-derived
Santoro M, Bogdanova T, Tronko M, Abrosimov A, Jeremiah S, tumors. Cancer Research 2000 60 28452849.
Thomas G, Williams D & Sobrinho-Simoes M. BRAF mutations are 60 Melillo RM, Cirafici AM, De Falco V, Bellantoni M, Chiappetta G,
not a major event in post-Chernobyl childhood thyroid carci- Fusco A, Carlomagno F, Picascia A, Tramontano D, Tallini G &
nomas. Journal of Clinical Endocrinology and Metabolism 2004 89 Santoro M. The oncogenic activity of RET point mutants
42674271. for follicular thyroid cells may account for the occurrence of
45 Nikiforova MN, Stringer JR, Blough R, Medvedovic M, Fagin JA & papillary thyroid carcinoma in patients affected by familial
Nikiforov YE. Proximity of chromosomal loci that participate in medullary thyroid carcinoma. American Journal of Pathology
radiation-induced rearrangements in human cells. Science 2000 2004 165 511521.
290 138141. 61 Tong Q, Xing S & Jhiang SM. Leucine zipper-mediated dimerization
46 Gandhi M, Medvedovic M, Stringer JR & Nikiforov YE. Interphase is essential for the PTC1 oncogenic activity. Journal of Biological
chromosome folding determines spatial proximity of genes Chemistry 1993 272 90439047.
participating in carcinogenic RET/PTC rearrangements. Oncogene 62 Iervolino A, Iuliano R, Trapasso F, Viglietto G, Melillo RM,
2006 25 23602366. Carlomagno F, Santoro M & Fusco A. The receptor-type protein
47 Roccato E, Bressan P, Sabatella G, Rumio C, Vizzotto L, Pierotti MA tyrosine phosphatase J antagonizes the biochemical and biological
& Greco A. Proximity of TPR and NTRK1 rearranging loci in effects of RET-derived oncoproteins. Cancer Research 2006 66
human thyrocytes. Cancer Research 2005 65 25722576. 62806287.
63 Durick K, Gill GN & Taylor SS. Shc and Enigma are both required
48 Melillo RM, Castellone MD, Guarino V, De Falco V, Cirafici AM,
for mitogenic signaling by Ret/ptc2. Molecular and Cellular Biology
Salvatore G, Caiazzo F, Basolo F, Giannini R, Kruhoffer M,
1998 18 22982308.
Orntoft T, Fusco A & Santoro M. The RET/PTC-RAS-BRAF linear
64 Melillo RM, Santoro M, Ong SH, Billaud M, Fusco A, Hadari YR,
signaling cascade mediates the motile and mitogenic phenotype of
Schlessinger J & Lax I. Docking protein FRS2 links the protein
thyroid cancer cells. Journal of Clinical Investigation 2005 115
tyrosine kinase RET and its oncogenic forms with the mitogen-
10681081.
activated protein kinase signaling cascade. Molecular and Cellular
49 Wang J, Knauf JA, Basu S, Puxeddu E, Kuroda H, Santoro M,
Biology 2001 21 41774187.
Fusco A & Fagin JA. Conditional expression of RET/PTC induces a
65 Bossis I & Stratakis CA. Minireview: PRKAR1A: normal and
weak oncogenic drive in thyroid PCCL3 cells and inhibits abnormal functions. Endocrinology 2004 145 54525458.
thyrotropin action at multiple levels. Molecular Endocrinology 66 Griffin KJ, Kirschner LS, Matyakhina L, Stergiopoulos S, Robinson-
2003 17 14251436. White A, Lenherr S, Weinberg FD, Claflin E, Meoli E, Cho-
50 Knauf JA, Kuroda H, Basu S & Fagin JA. RET/PTC-induced Chung YS & Stratakis CA. Down-regulation of regulatory subunit
dedifferentiation of thyroid cells is mediated through Y1062 type 1A of protein kinase A leads to endocrine and other tumors.
signaling through SHC-RAS-MAP kinase. Oncogene 2003 22 Cancer Research 2004 64 88118815.
44064412. 67 Dupont S, Zacchigna L, Cordenonsi M, Soligo S, Adorno M,
51 Castellone MD, Cirafici AM, De Vita G, De Falco V, Malorni L, Rugge M & Piccolo S. Germ-layer specification and control of cell
Tallini G, Fagin JA, Fusco A, Melillo RM & Santoro M. Ras- growth by Ectodermin, a Smad4 ubiquitin ligase. Cell 2005 121
mediated apoptosis of PC CL 3 rat thyroid cells induced by 8799.
RET/PTC oncogenes. Oncogene 2003 22 246255. 68 Yeh S & Chang C. Cloning and characterization of a specific
52 Fischer AH, Bond JA, Taysavang P, Battles OE & Wynford- coactivator, ARA70, for the androgen receptor in human prostate
Thomas D. Papillary thyroid carcinoma oncogene (RET/PTC) cells. PNAS 1996 93 55175521.
alters the nuclear envelope and chromatin structure. American 69 Heinlein CA, Ting HJ, Yeh S & Chang C. Identification of ARA70 as
Journal of Pathology 1998 153 14431450. a ligand-enhanced coactivator for the peroxisome proliferator-
53 Santoro M, Chiappetta G, Cerrato A, Salvatore D, Zhang L, activated receptor gamma. Journal of Biological Chemistry 1999
Manzo G, Picone A, Portella G, Santelli G, Vecchio G & Fusco A. 274 1614716152.
Development of thyroid papillary carcinomas secondary to tissue- 70 Kulkarni S, Heath C, Parker S, Chase A, Iqbal S, Pocock CF,
specific expression of the RET/PTC1 oncogene in transgenic mice. Kaeda J, Cwynarski K, Goldman JM & Cross NC. Fusion of
Oncogene 1996 12 18211826. H4/D10S170 to the platelet-derived growth factor receptor beta in
54 Jhiang SM, Sagartz JE, Tong Q, Parker-Thornburg J, Capen CC, BCR-ABL-negative myeloproliferative disorders with a
Cho JY, Xing S & Ledent C. Targeted expression of the ret/PTC1 t(5;10)(q33;q21). Cancer Research 2000 60 35923598.
oncogene induces papillary thyroid carcinomas. Endocrinology 71 Zhong S, Delva L, Rachez C, Cenciarelli C, Gandini D, Zhang H,
1996 137 375378. Kalantry S, Freedman LP & Pandolfi PP. A RA-dependent, tumour-
55 Powell DJ Jr, Russell J, Nibu K, Li G, Rhee E, Liao M, Goldstein M, growth suppressive transcription complex is the target of the
Keane WM, Santoro M, Fusco A & Rothstein JL. The RET/PTC3 PML-RARalpha and T18 oncoproteins. Nature Genetics 1999 23
oncogene: metastatic solid-type papillary carcinomas in murine 287295.
thyroids. Cancer Research 1998 58 55235528. 72 Bousquet M, Quelen C, De Mas V, Duchayne E, Roquefeuil B,
56 Basolo F, Giannini R, Monaco C, Melillo RM, Carlomagno F, Delsol G, Laurent G, Dastugue N & Brousset P. The t(8;9)(p22;p24)
Pancrazi M, Salvatore G, Chiappetta G, Pacini F, Elisei R, Miccoli P, translocation in atypical chronic myeloid leukaemia yields a new
Pinchera A, Fusco A & Santoro M. Potent mitogenicity of the PCM1-JAK2 fusion gene. Oncogene 2005 24 72487252.

www.eje-online.org
EUROPEAN JOURNAL OF ENDOCRINOLOGY (2006) 155 RET/PTC in thyroid cancer 653

73 Xing M. BRAF mutation in thyroid cancer. Endocrine-Related with similar but distinct gene expression patterns in papillary
Cancer 2005 12 245262. thyroid cancer. Oncogene 2004 23 74367440.
74 Fagin JA. Challenging dogma in thyroid cancer molecular genetics- 84 Russell JP, Shinohara S, Melillo RM, Castellone MD, Santoro M &
role of RET/PTC and BRAF in tumor initiation. Journal of Clinical Rothstein JL. Tyrosine kinase oncoprotein, RET/PTC3, induces the
Endocrinology and Metabolism 2004 89 42644266. secretion of myeloid growth and chemotactic factors. Oncogene
75 Kodama Y, Asai N, Kawai K, Jijiwa M, Murakumo Y, Ichihara M & 2003 22 45694577.
Takahashi M. The RET proto-oncogene: a molecular therapeutic 85 Puxeddu E, Knauf JA, Sartor MA, Mitsutake N, Smith EP,
target in thyroid cancer. Cancer Science 2005 96 143148. Medvedovic M, Tomlinson CR, Moretti S & Fagin JA. RET/PTC-
76 Mitsutake N, Miyagishi M, Mitsutake S, Akeno N, Mesa C, Jr, induced gene expression in thyroid PCCL3 cells reveals early
Knauf JA, Zhang L, Taira K & Fagin JA. BRAF mediates RET/PTC- activation of genes involved in regulation of the immune response.
induced mitogen-activated protein kinase activation in thyroid Endocrine-Related Cancer 2005 12 319334.
cells: functional support for requirement of the RET/PTC-RAS- 86 Borrello MG, Alberti L, Fischer A, Deglinnocenti D, Ferrario C,
BRAF pathway in papillary thyroid carcinogenesis. Endocrinology Gariboldi M, Marchesi F, Allavena P, Greco A, Collini P, Pilotti S,
2006 147 10141019. Cassinelli G, Bressan P, Fugazzola L, Mantovani A & Pierotti MA.
77 Buckwalter TL, Venkateswaran A, Lavender M, La Perle KM, Induction of a proinflammatory program in normal human
Cho JY, Robinson ML & Jhiang SM. The roles of phosphotyrosines- thyrocytes by the RET/PTC1 oncogene. PNAS 2005 102
294, -404, and -451 in RET/PTC1-induced thyroid tumor 1482514830.
formation. Oncogene 2002 21 81668172. 87 Castellone MD, Celetti A, Guarino V, Cirafici AM, Basolo F,
78 Mesa C Jr, Mirza M, Mitsutake N, Sartor M, Medvedovic M, Giannini R, Medico E, Kruhoffer M, Orntoft TF, Curcio F, Fusco A,
Tomlinson C, Knauf JA, Weber GF & Fagin JA. Conditional Melillo RM & Santoro M. Autocrine stimulation by osteopontin
activation of RET/PTC3 and BRAFV600E in thyroid cells is plays a pivotal role in the expression of the mitogenic and invasive
associated with gene expression profiles that predict a preferential phenotype of RET/PTC-transformed thyroid cells. Oncogene 2004
role of BRAF in extracellular matrix remodeling. Cancer Research 23 21882196.
2006 66 65216529. 88 Castellone MD, Guarino V, De Falco V, Carlomagno F, Basolo F,
79 Sebolt-Leopold JS & Herrera R. Targeting the mitogen-activated Faviana P, Kruhoffer M, Orntoft T, Russell JP, Rothstein JL, Fusco A,
protein kinase cascade to treat cancer. Nature Reviews Cancer 2004 Santoro M & Melillo RM. Functional expression of the CXCR4
4 937947. chemokine receptor is induced by RET/PTC oncogenes and is a
80 Knauf JA, Ouyang B, Knudsen ES, Fukasawa K, Babcock G & common event in human papillary thyroid carcinomas. Oncogene
Fagin JA. Oncogenic RAS induces accelerated transition through 2004 23 59585967.
G2/M and promotes defects in the G2 DNA damage and mitotic 89 Puxeddu E, Mitsutake N, Knauf JA, Moretti S, Kim HW, Seta KA,
spindle checkpoints. Journal of Biological Chemistry 2006 281 Brockman D, Myatt L, Millhorn DE & Fagin JA. Microsomal
38003809. prostaglandin E2 synthase-1 is induced by conditional expression
81 Knauf JA, Ma X, Smith EP, Zhang L, Mitsutake N, Liao XH, of RET/PTC in thyroid PCCL3 cells through the activation of the
Refetoff S, Nikiforov YE & Fagin JA. Targeted expression of MEK-ERK pathway. Journal of Biological Chemistry 2003 278
BRAFV600E in thyroid cells of transgenic mice results in papillary 5213152138.
thyroid cancers that undergo dedifferentiation. Cancer Research 90 Russell JP, Engiles JB & Rothstein JL. Proinflammatory mediators
2005 65 42384245. and genetic background in oncogene mediated tumor progression.
82 Giordano TJ, Kuick R, Thomas DG, Misek DE, Vinco M, Sanders D, Journal of Immunology 2004 172 40594067.
Zhu Z, Ciampi R, Roh M, Shedden K, Gauger P, Doherty G, 91 Ouyang B, Knauf JA, Smith EP, Zhang L, Ramsey T, Yusuff N,
Thompson NW, Hanash S, Koenig RJ & Nikiforov YE. Molecular Batt D & Fagin JA. Inhibitors of Raf kinase activity block growth of
classification of papillary thyroid carcinoma: distinct BRAF, RAS, thyroid cancer cells with RET/PTC or BRAF mutations in vitro and
and RET/PTC mutation-specific gene expression profiles discov- in vivo. Clinical Cancer Research 2006 12 17851793.
ered by DNA microarray analysis. Oncogene 2005 24 66466656.
83 Frattini M, Ferrario C, Bressan P, Balestra D, De Cecco L, Mondellini P,
Bongarzone I, Collini P, Gariboldi M, Pilotti S, Pierotti MA & Greco A. Received 4 August 2006
Alternative mutations of BRAF, RET and NTRK1 are associated Accepted 4 September 2006

www.eje-online.org

Potrebbero piacerti anche