Вы находитесь на странице: 1из 16

Best Practice & Research Clinical Endocrinology & Metabolism

Vol. 22, No. 6, pp. 10231038, 2008


doi:10.1016/j.beem.2008.09.012
available online at http://www.sciencedirect.com

9
Receptor tyrosine kinase inhibitors in thyroid
cancer
Maria Domenica Castellone

MD, PhD

Research Associate
Istituto di Endocrinologia ed Oncologia Sperimentale CNR, 80131 Naples, Italy c/o Dipartimento di Biologia
e Patologia Cellulare e Molecolare, Universita Federico II, 80131 Naples, Italy

Francesca Carlomagno

MD, PhD

Research Associate
Istituto di Endocrinologia ed Oncologia Sperimentale CNR, 80131 Naples, Italy c/o Dipartimento di Biologia
e Patologia Cellulare e Molecolare, Universita Federico II, 80131 Naples, Italy

Giuliana Salvatore

MD, PhD

Associate Professor
Dipartimento di Studi delle Istituzioni e dei Sistemi Territoriali, Universita Parthenope, 80133 Naples, Italy

Massimo Santoro *

MD, PhD

Professor of Pathology
Dipartimento di Biologia e Patologia Cellulare e Molecolare, L. Califano, Universita` Federico II, via S. Pansini 5,
80131 Naples, Italy

Thyroid cancer is frequently associated with the oncogenic conversion of receptor tyrosine
kinases (RTKs) or their downstream signalling molecules. Hence, there is a strong biological rationale for assessing the efficacy of RTK blockade to treat patients who are resistant to or not
candidates for treatment with radioactive iodine. The first results of clinical trials based on the
use of RTK inhibitors in thyroid cancer patients have recently been published. Here we discuss
targeting of specific RTKs as a potential therapeutic strategy for the treatment of thyroid cancer.
Key words: kinase; thyroid; monoclonal antibody; small-molecule inhibitor.

* Corresponding author. Tel.: 39-081-7463056; Fax: 39-081-7463037.


E-mail address: masantor@unina.it (M. Santoro).
1521-690X/$ - see front matter 2008 Elsevier Ltd. All rights reserved.

1024 M. D. Castellone et al

CANCER GENES CODING FOR RECEPTOR TYROSINE KINASES


The protein kinase complement of the human genome (the kinome) contains 518
protein kinase genes, including 58 receptor tyrosine kinases (RTKs) and 32 nonRTKs (www.kinase.com/human/kinome). In high-throughput genome-sequencing projects, approximately 120 genes encoding protein kinases were found to be mutated
and causally implicated in cancer development.1,2
RTKs are transmembrane proteins that have an extracellular ligand-binding domain,
a transmembrane segment, and an intracellular domain containing the juxtamembrane
segment (JMR), the tyrosine kinase catalytic domain (TK) and a carboxy-terminal tail
(Figure 1). RTKs are often involved in cancer development.1,2 Besides RTK point
mutations, cancers may feature illicit expression of the RTK or its cognate growth
factor and RTK gene rearrangements.35 Thyroid cancer, as discussed below, is often
associated to the oncogenic conversion of genes coding for RTKs or their signalling
effectors.6,7
MECHANISMS OF RTK ACTIVATION
Upon growth factor binding, RTKs undergo a dimerization process that activates
the enzymatic function.3 All kinase domains are divided into an amino-terminal

Figure 1. Overview of the signalling mechanism of receptor tyrosine kinases (RTKs). The general structure
of a prototypic RTK, with the extracellular domain (EC), transmembrane domain (TM), juxtamembrane domain (JMR), and tyrosine kinase domain (TK), is illustrated. Blue boxes summarize major biological outcomes
of the activation of specific signalling components.

RTK inhibitors in thyroid cancer 1025

(N-terminal) and a carboxy-terminal (C-terminal) lobe. The active site is located in the
cleft between the N- and C-terminal lobes. Mg-ATP binds to the base of the cleft, the
non-transferable b-phosphate of Mg-ATP binds to the P-loop of the N-terminal lobe,
and the peptide substrate binds along the surface of the cleft.3,4 The ATP-binding site
can also bind small-molecule inhibitors (see below).4,5 In the inactive kinase, the active
site is closed by the activation segment of the C-terminal lobe. Growth-factor-binding
causes RTK dimerization, followed by extensive movements of the activation segment
and the JMR domain; this process allows ATP binding and catalysis.
Oncogenic mutations disrupt normal regulatory mechanisms and lead to constitutive activation of the kinase. Point mutations or rearrangements in the extracellular
domain mimic ligand-binding, thereby causing constitutive dimerization. Mutations in
the intracellular domain target regulatory domains of the kinase, such as the P-loop,
the activation segment, or the JMR, thereby disrupting auto-inhibitory mechanisms.3
MECHANISMS OF RTK SIGNALLING
Mutual trans-phosphorylation of tyrosine residues within active RTK dimers recruits
intracellular proteins endowed with phosphotyrosine binding domains.35 Proximal
targets of the RTKs invoke the intracellular signalling cascades RAS-RAF-MAPK (the
ERK pathway) and the phosphatidylinositol 3-kinase (PI3K) AKT that ultimately
lead to diverse biological responses: i.e. mitogenesis, survival, differentiation and motility (Figure 1).35
Genes coding for proteins working in the RTK-initiated signal transduction pathways are frequently mutated in cancer. For instance, systematic DNA sequencing of
the best-annotated protein-coding genes in breast and colorectal cancer revealed
frequent mutations in the PI3K and nuclear factor kB (NFkB) pathways, both of which
are involved in RTK signalling (Figure 1).8 In the case of thyroid cancer, paradigmatic
examples of this concept are mutations in BRAF, a RAF family serine/threonine kinase,
in papillary thyroid carcinoma (PTC) (2969%) and PTC-derived anaplastic thyroid carcinoma (ATC) (1035%), and in the RAS small GTPase that is mutated in the follicular
variant of PTC (up to 40%), in follicular thyroid carcinoma (FTC) (4053%) and in ATC
(2060%).7 Moreover, mutations or amplification of PIK3CA, which codes for the PI3K
catalytic subunit, is associated with ATC and FTC, and loss of expression of PTEN, the
major phosphatase antagonist of PI3K function, is frequent in ATC.9 Finally, functional
activation of NFkB has been described in ATC.10 NFkB is crucial for RET and BRAF
oncogene signalling in thyroid cancer cells11,12, and the proteasome inhibitor bortezomib (PS-341, Velcade, Millennium Pharmaceuticals), whose complex mechanism of
action includes NFkB blockade, was effective in thyroid carcinoma cell lines.13
RTKs AS MOLECULAR TARGETS FOR CANCER TREATMENT
The advent of small-molecule drugs and monoclonal antibodies have made RTK targeting a feasible therapeutic strategy for cancer.4,5 Monoclonal antibodies (mAbs) can
block the growth factor or the RTK itself. Some humanized mAbs such as trastuzumab (herceptin; Genentech/Roche) against the HER2/ErbB2 receptor, cetuximab (erbitux; ImClone) against the epidermal growth factor receptor (EGFR/HER1/ErbB1),
and bevacizumab (avastin; Genentech/Roche) against vascular endothelial growth factor (VEGF) are now part of the treatment regimen for specific tumours.4,5 The antitumour activity of RTK-directed mAbs has been ascribed to different mechanisms: (1)

1026 M. D. Castellone et al

blockade of ligand-binding; (2) blockade of RTK homo- or hetero-dimerization; (3) interference with the active-like RTK conformation; (4) down-regulation of the receptor
from the cell surface (receptor internalization); (5) shedding of the extracellular
domain of the receptor; or (6) antibody-dependent cell-mediated cytotoxicity
(ADCC).4,5
Most RTK-directed small-molecule drugs are tyrosine kinase inhibitors (TKIs) and obstruct kinase activity by binding to the ATP pocket of the kinase in competition with
cellular ATP. The effectiveness of imatinib (imatinib mesylate/gleevec/glivec, STI571; Novartis), an inhibitor of ABL, KITand platelet-derived growth factor receptor (PDGFR), in
BCR-ABL-positive chronic myelogenous leukaemia (CML) and KITor PDGFR-a mutant gastrointestinal stromal tumours (GISTs) has illustrated the power of this approach.4 The
ATP binding site is highly conserved across the kinome. Thus, at best, TKIs may be selective but not specific and affect more than one RTK. As most cancers are the result
of a number of mutations, it is reasonable to suppose that TKIs able to target multiple
kinases or a rational combination of TKIs will be clinically more effective than agents
blocking a single kinase. As discussed in the next paragraph, multi-targeting TKIs or
combination therapies may also attenuate resistance formation.
Resistance development is a critical issue in the use of TKIs in cancer treatment.
Resistance is primarily mediated by the clonal expansion of cancer cells carrying secondary mutations of the target kinase. These mutations either prevent the kinase from
adopting the conformation to which the compound binds or alter the compound contact point.4,5 Another important point in the clinical use of TKIs is target selection.
Although cancer cells frequently contain mutations in multiple genes, they appear to
be highly dependent on specific genes and related pathways. This dependence can
be exploited therapeutically by appropriate targeting. Thus, the use of TKIs should
be restricted to those tumours that are addicted to the kinase that is targeted by
that specific agent. Finally, compensatory cross-talk between RTKs may attenuate
TKI efficacy. Tumour cells become resistant to an EGFR TKI by an adaptation mechanism involving activation of alternative RTKs, such as MET or PDGFR.14 Another study
showed that tumour cells are resistant to EGFR TKI up front, because they simultaneously activate the MET, EGFR and PDGFR RTKs.15 Cocktails of drugs or multitargeting TKIs may be used to overcome these mechanisms.
RTK INHIBITORS IN THYROID CANCER
The last few months have witnessed the publication of the first results of clinical studies based on the use of TKIs for thyroid cancer patients (www.clinicaltrials.gov). These
are summarized in Table 1, and include imatinib mesylate1618, gefitinib (ZD1839/Iressa; Astra Zeneca)19, axitinib (AG-013736; Pfizer)20, sorafenib (nexavar/BAY 43-9006;
Bayer)21,22, and motesanib (motesanib diphosphate/AMG706; Amgen).23 Moreover,
promising early results have emerged from studies with vandetanib (ZD6474/zactima;
AstraZeneca)24, sunitinib (sutent/SU11248; Pfizer)2527, and XL184 (Exelixis).28 The
RTKs targeted by these agents are illustrated in Figure 2, and their involvement in
thyroid cancer is discussed hereafter.
RET (GLIAL-DERIVED GROWTH FACTOR RECEPTOR)
RET (REarranged during Transfection) has been extensively reviewed elsewhere
(Figure 2).6,7,29 RET is the receptor of glial-derived neurotrophic factor (GDNF)

RTK inhibitors in thyroid cancer 1027

Table 1. Receptor tyrosine kinase inhibitors in clinical trials in thyroid cancer.


Name
Imatinib mesylate
(Novartis)
Gefitinib
(AstraZeneca)
Axitinib
(Pfizer)
Vandetanib
(AstraZeneca)
Sunitinib (Pfizer)

Sorafenib
(Bayer)
Motesanib
diphosphate
(Amgen)
XL184 (Exelixis)
Pazopanib
(GlaxoSmithKline)

Other names

Tyrosine kinase targets


(only those IC50 < 1 mM)
ABL, KIT, PDGFR

Clinical development

Gleevec, glivec,
STI571
ZD1839, iressa

EGFR

AG-013736

VEGFR-1, -2, -3, PDGFR, KIT

Approved for
CML, GIST
Approved in some
countries for NSCLC
Investigational

Zactima,
ZD6474
Sutent,
SU11248

EGFR, RET, VEGFR-2, -3

Investigational

VEGFR-2, PDGFR, KIT, FLT3,


RET, FGFR-1, CSF-1R

Nexavar,
BAY 43-9006
AMG 706

RAF, BRAF, P38, VEGFR-1, -2, -3,


PDGFR, FLT3, RET, KIT, FGFR-1
VEGFR-1, -2, -3, KIT, RET,
PDGFR, FLT3

Approved for metastatic


renal carcinoma and
imatinib-resistant GIST
Approved for metastatic
renal carcinoma
Investigational

e
GW-786034

RET, VEGFR-2, MET


VEGFR-1, -2, -3, PDGFR,
KIT, FGFR-1

Investigational
Investigational

ABL, Abelson murine leukaemia viral (v-abl) oncogene homologue; PDGFR, platelet-derived growth factor receptor; KIT, stem-cell factor receptor; EGFR, epidermal growth factor receptor; VEGFR, vascular
endothelial growth factor receptor; FLT3, fms-related tyrosine kinase; RET, rearranged during transfection; FGFR, fibroblast growth factor receptor; MET, hepatocyte growth factor receptor; CSF-1R, colonystimulating factor receptor.
**CML chronic myelogenous leukaemia, GIST gastro intestinal stromal tumours; NSCLC non
small cell lung carcinoma.

ligands. In PTC, chromosomal inversions or translocations cause the fusion of the RET
kinase-encoding domain with the 50 -end of heterologous genes. The resulting chimeric
sequences are called RET/PTC.6,7 Germline point mutations in RET cause MEN-2
(multiple endocrine neoplasia type 2) syndromes that predisposes to MTC. Somatic
mutations of RET are also found in sporadic MTC.29 Transgenic mouse models demonstrated that RET oncogenes are able to drive PTC and MTC formation.6 Moreover,
RET knock-down by dominant-negative mutants or RNAi impairs proliferation of
MTC and PTC cell lines harbouring RET-derived oncogenes.30,31 In this scenario
RET appears to be a promising target for the molecular therapy of PTC and MTC.
However, the heterogeneous distribution of RET mutations in some tumour samples
has challenged the notion that RET alteration is the driving lesion in those individual
patients.32,33
Some of the TKIs in clinical experimentation in thyroid cancer patients have antiRET activity (Table 1 and Figure 2). Most of them e.g. vandetanib34, sunitinib35, sorafenib36, XL-18428, and motesanib37, inhibit both RET and VEGFRs. Thus, potentially
these drugs can simultaneously attack both neoplastic and endothelial cells. X-ray
structural analysis has demonstrated that vandetanib binds the ATP pocket of the
kinase and is therefore an ATP-competitive RET TK inhibitor.38

1028 M. D. Castellone et al

Figure 2. Schematic representation of some receptor tyrosine kinases (RTKs). Specific domains of the extracellular portion are indicated: immunoglobulin-like, plexin and transcription factor domain (IPT); semaphorin domain (Sema); plexins, semaphorins, and integrins domain (Psi). Some RTKs have a tyrosine
kinase domain (TK) domain split by a peptide insert. Examples of agents monoclonal antibodies (mAbs)
or tyrosine kinase inhibitors (TKIs) able to obstruct RTK activity are indicated at the bottom (see text
for details).

NTRK1 (NERVE GROWTH FACTOR RECEPTOR)


NTRK1 (also known as TRKA) belongs to the RTK neurotrophin receptor family that
includes NTRK2 (TRKB) and NTRK3 (TRKC) (Figure 2).39 NTRK1 is the high-affinity
receptor for nerve growth factor. In 513% of PTC cases, the NTRK1 TK-encoding
domain is illegitimately recombined with heterologous sequences, thereby generating
TRK-T oncogenes.7,39 As yet there is no evidence that other members of the NTRK
family are mutated in thyroid carcinoma.40 However, changes in their expression
were involved in thyroid C-cell transformation, with NTRK2 being reduced and
NTRK3 being up-regulated in MTC.41 The CEP-751 TKI, which is active against
both RET and NTRK, had a cytostatic effect in MTC cells.42
MET (HEPATOCYTE GROWTH FACTOR RECEPTOR)
MET is a cell-surface receptor for hepatocyte growth factor (HGF, also known as scatter factor) (Figure 2).43 The MET homologue, RON, is the receptor for macrophagestimulating protein (MSP). Amplification of the MET gene has been reported in several
human tumours; more rarely, MET carries activating point mutations. Most often, MET

RTK inhibitors in thyroid cancer 1029

is transcriptionally up-regulated in carcinomas. Since HGF is ubiquitously expressed,


particularly in tumour stroma, this is believed to lead to constitutive MET activation.43
Once activated, MET stimulates cell scattering, invasion, protection from apoptosis
and anoikis, and angiogenesis.43
Approximately 70% of PTCs over-express MET, whereas normal thyrocytes do not
express MET. HGF is locally produced by PTC stromal fibroblasts.44,45 MET was identified as one of the most consistently up-regulated markers of thyroid cancer, particularly in BRAF mutant PTC cases and in PTCs of the aggressive tall-cell variant.4649
Moreover, MET and HGF are co-expressed in a subset of MTCs.50 MET gene copy
gains have been identified in approximately 10% of ATCs and less frequently in
FTCs,51 MET mutations are rare in thyroid cancer; a missense mutation, T1010I,
was found in 6% of thyroid carcinomas and in germline DNA.52 Importantly, HGF
was identified as one of the most potent growth factors for cultured thyrocytes.53
The RON RTK was reported to be over-expressed in PTC and FTC, and its levels correlated with advanced clinical stage.54
Monoclonal antibodies against HGF or the extracellular MET domain and MET ATPcompetitive TKIs have been described.43 The TKI PF2341066 (Pfizer) is undergoing
phase-I/II clinical trials, and the related compound, PHA665752, was effective against
PTC cells in vitro.55 Another two TKIs, XL880 and XL184 (Exelixis), are undergoing
clinical experimentation. It is noteworthy that XL184, which is also a RET inhibitor, is
being studied also in thyroid cancer patients (Table 1, Figure 2).28
EGFR (EPIDERMAL GROWTH FACTOR RECEPTOR)
The epidermal growth factor (EGF) receptor (EGFR, also named ErbB1 or HER1) belongs to the ErbB/HER family of RTKs, which in addition includes ErbB2/Neu/HER2,
ErbB3/HER3 and ErbB4/HER4.56,57 The four ErbBs share an overall structure of two
cysteine-rich regions in their extracellular region and an intracellular TK domain
(Figure 2). ErbB2, which does not have a direct ligand, and ErbB3, which is devoid
of intrinsic kinase activity, are non-autonomous receptors and require heterodimerization with other ErbB family members for activation and signalling. ErbB2 is the
most potent oncoprotein in the ErbB family and it is the preferred heterodimeric partner of the other ErbBs. ErbB3 is a potent PI3K activator.56 The EGF family includes
various ligands: EGF, transforming growth factor-a (TGF-a) and amphiregulin
(AREG), which bind specifically to EGFR; b-cellulin (BTC), heparin-binding EGF
(HB-EGF) and epiregulin (EREG), which bind EGFR and ErbB4; and neuregulins
(NRGs), which bind either ErbB3/ErbB4 heterodimers or ErbB4 homodimers.56,57
Gene amplification of EGFR is often found in human cancers. In many tumours, EGFrelated growth factors are produced either by the tumour cells or by stromal cells.56
In gliomas, EGFR amplification is often accompanied by structural rearrangements. Somatic mutations in the TK domain of EGFR are present in non-small-cell lung cancers
(NSCLCs). Amplification of ErbB2 occurs in breast and other carcinomas. Mutations in
the kinase domain of ErbB2 occur in a small number of NSCLCs.4,5,56,57
Structural alterations in the EGFR gene are uncommon in thyroid cancer. No EGFR
mutation (exons 1821) was found in 31 ATCs58, and only two mutations were detected in 62 thyroid cancers and 11 cell lines.59 In another study, no EGFR mutation
was found in 51 ATC and 64 FTC samples; however, EGFR gene copy gains were frequent in these tissues.51 Over-expression of EGFR in thyroid cancer is controversial:
some studies report up-regulation in thyroid carcinomas, particularly in ATC58,60,61,
whereas others report expression levels similar to those observed in normal

1030 M. D. Castellone et al

tissue.59,62 Growth factors of the EGF family (TGF-a, AREG, EREG)63 as well as ErbB2,
ErbB3 and ErbB4 were found to be up-regulated in PTC61,64 and ATC.58 Importantly,
EGF is mitogenic for thyrocytes, and long-term treatment with EGF induced gene expression profiles similar to those of PTC samples.65 It was recently reported that RET/
PTC oncogenes induce EGFR expression, and that EGFR and RET/PTC proteins form
a complex that mediates EGFR-dependent RET phosphorylation.66
Four different anti-EGFR agents are approved in several countries for NSCLC and
for colorectal, pancreatic and head and neck carcinoma (Figure 2). These include two
mAbs, cetuximab (a mouse/human chimeric mAb) and panitumumab (vectibix; Abgenix) (a fully human mAb), and two TKIs, gefitinib and erlotinib (tarceva; Genentech).57
Trastuzumab (the humanized mAb targeting HER2) and lapatinib (GW572016/Tykerb;
GlaxoSmithKline), a pan-ErbB TKI, have been approved for the treatment of HER2over-expressing breast cancers.4,5,57
In preclinical studies, micromolar doses of gefitinib60,67 or NVP-AEE788 (Novartis),
an EGFR and VEGFR TKI with additional activity against RET59,68,69, were effective in cultured ATC cells. NVP-AEE788 and cetuximab reduced the secretion of VEGF from thyroid cancer cells, but only NVP-AEE788 dose-dependently inhibited proliferation.70 In
cells expressing activated REToncogenes, NVP-AEE788 and PKI166 (Novartis), another
EGFR TKI, were active at submicromolar concentrations, secondary to interaction between EGFR and RET.66 No objective response was obtained in a phase-II study of gefitinib in patients with locally advanced or metastatic thyroid cancer, although a fraction of
patients achieved prolonged stable disease and there was a reduction in tumour size and
plasma thyroglobulin concentration. Therefore, EGFR inhibition is not sufficient, at least
by itself, to induce a major clinical response in thyroid cancer patients.19
FGFR (FIBROBLAST GROWTH FACTOR RECEPTOR)
The fibroblast growth factor (FGF) family currently includes more than 20 members
that are important regulators of angiogenesis and tumourigenesis. FGFs signal through
four RTKs (FGFR-1, -2, -3 and -4).7 Each receptor has two or three immunoglobulinlike extracellular domains, a transmembrane domain, and an intracellular TK
(Figure 2).
Thus far, no mutations or rearrangements involving FGFRs have been identified in
thyroid cancer.7 Expression of FGF1 and FGF2 (also known as basic FGF, a potent angiogenic factor) is increased in thyroid cancer.7173 Increased expression of FGFR-1, -3
and -4 has been observed in benign and malignant thyroid tumours.71,74 FGFR-2 expression, instead, was down-regulated in thyroid cancer, and its re-expression in thyroid carcinoma cells interrupted signalling upstream of BRAF and MAPK and reduced
cell growth.75 FGFR-4 is expressed predominantly in aggressive thyroid tumour types
and MTC cells. Molecular targeting of FGFR-4 with the ATP-competitive inhibitor
PD173074 (Pfizer) inhibited growth and reduced tumourigenesis of MTC cells.76 It
is noteworthy that sorafenib, sunitinib and pazopanib, which are undergoing clinical experimentation in thyroid cancer patients, exert anti-FGFR activity (Table 1 and
Figure 2).
IGF-1R (INSULIN-LIKE GROWTH FACTOR RECEPTOR 1)
IGF-1R (insulin-like growth factor receptor 1) is a ubiquitous transmembrane tyrosine
kinase structurally similar to the insulin receptor (IR). IGF-1R is composed of two

RTK inhibitors in thyroid cancer 1031

extracellular a-subunits and two intracellular b-subunits (Figure 2). The a-subunits
bind ligands (IGF-I, IGF-II, and insulin at supraphysiological doses), whereas b-subunits
contain the TK domain.77 Most of the effects of IGF-I are mediated by IGF-1R. The
effects of IGF-II may be mediated by both IGF-IR and an alternatively spliced variant
of IR.
IGF-II and IGF-1R are over-expressed in many cancer types. High circulating levels
of IGF-I have been indicated as risk factors for various tumour types. Moreover, IGF1R up-regulation was found to mediate resistance to TKIs in different types of cancer
cells. IGF-I and IGF-1R are over-expressed in thyroid cancer, particularly in the most
aggressive variants.78 Importantly, IGF-I and insulin are essential for the mitogenic action of TSH and EGF in thyroid follicular cells.79 Both anti-IGF-R mAbs and TKIs are
being developed (Figure 2). The TKI NVP-ADW742 (Novartis) is cytotoxic for FTCand MTC-derived cancer cells.80 IMC-A12 (ImClone) is a fully human mAb that targets
the human IGF-1R. IMC-A12 was effective in treating an orthotopic mouse model of
ATC.78
VEGFR (VASCULAR ENDOTHELIAL GROWTH FACTOR RECEPTOR)
The vascular endothelial growth factor (VEGF) family consists of five ligands: VEGFA,
VEGFB, VEGFC, VEGFD and PGF (placenta growth factor).81 The best characterized is
VEGFA (commonly referred to as VEGF). VEGFA and VEGFB are angiogenic proteins,
whereas VEGFC and VEGFD are primarily lymphangiogenic proteins.81,82 The VEGF
receptors are VEGFR-1 (FLT-1), VEGFR-2 (KDR) and VEGFR-3 (FLT-4) (Figure 2).
VEGFR-2 binds VEGFA and is expressed primarily on blood vessel endothelium.
VEGFR-1 binds VEGFA, VEGFB and PGF and is expressed mainly in the vasculature
and also in other cell types. VEGFR-3 is the receptor for VEGFC and VEGFD and is
expressed primarily on lymphatic endothelium.81,82 However, it has been recently
demonstrated that VEGFR-3 is also expressed in tumour blood vessels where it contributes to angiogenic sprouting, and simultaneous blockade of VEGFR-3 and VEGFR-2
resulted in potent inhibition of tumour angiogenesis.83
Over-expression of VEGFA has been reported in thyroid carcinoma tissue73,8486
and plasma.87 In most85,87,88 but not all73,89 reports this was correlated with stage, tumour size and metastasis. Over-expression of VEGFC in PTC has been reported in
three studies85,90,91, whereas over-expression of VEGFC in MTC is controversial.85,91
Finally, expression of VEGFR-192 and VEGFR-285,93 has been detected in thyrocytes as
well as in endothelial cells.
VEGF targeting is a promising anti-cancer therapeutic approach. VEGF-targeted
therapy acts through various mechanisms: inhibition of new vessel formation, apoptosis of pre-existing vessels, blockade of endothelial cell progenitors, and vessel constriction (with reduced blood flow and ischaemia).81,82 There are currently more than 20
different VEGF-targeted agents undergoing clinical evaluation (Figure 2). These include
neutralizing mAbs against VEGF or VEGFRs. Bevacizumab is a humanized mAb against
VEGF currently registered for colorectal cancer, breast cancer and NSCLC.4,5,81 Several multi-targeting TKIs that block VEGFRs have shown promising clinical activity
against various solid tumours, and two of them (sorafenib and sunitinib) are registered
as anti-cancer agents.4,5,81 Unfortunately, in most cases the effects of anti-angiogenic
treatment are only transitory. Various mechanisms have been evoked to explain this
phenomenon: production by cancer cells of angiogenic growth factors other than
VEGF (FGF2, PDGF, ephrins, angiopoietin, IL-8), recruitment of angiogenic

1032 M. D. Castellone et al

bone-marrow precursors, survival of blood vessels covered by pericytes, and migration of cancer cells outside the primary tumour to co-opt pre-existing vessels.91
Importantly, clinical trials with VEGFR-blocking multi-target TKIs (sorafenib,
axitinib, motesanib, sunitinib, vandetanib and XL-184) have yielded promising results
in thyroid cancer patients in the last few months (Table 1).2028 In addition, pazopanib
(GW-786034; GlaxoSmithKline), a multi-targeted pan-VEGFR inhibitor, is undergoing
clinical experimentation in thyroid cancer patients (www.clinicaltrials.gov).
Preclinical studies with NVP-AEE788, a dual VEGFR and EGFR inhibitor, in FTC and
ATC cells6870, and with PTK787/ZK222584 (Novartis and Schering), a pan-VEGFR
TKI, in thyroid carcinoma mouse xenografts93 showed promising results. Cediranib
(AZD2171; AstraZeneca), another pan-VEGFR TKI, inhibited tumour growth and prolonged animal survival in an orthotopic nude mouse model of ATC.92 Finally, VEGF
mAbs, including the murine version of bevacizumab, inhibited the growth of mouse
xenografts of thyroid cancer cells.94
PDGFR (PLATELED-DERIVED GROWTH FACTOR RECEPTOR)
The plateled-derived growth factor (PDGF) family consists of polypeptides PDGF-A, -B,
-C and D that form homo- or more rarely hetero-dimers. PDGFs act via two RTKs
(PDGFR-a and PDGFR-b) with common structures, including extracellular immunoglobulin (Ig) loops and a split intracellular TK domain (Figure 2).95 PDGF-AA and
PDGF-CC ligands act via PDGFR-a, while PDGF-BB and probably PDGF-DD act via
PDGFR-b. PDGF-B is expressed mainly in vascular endothelial cells, megakaryocytes
and neurons. PDGF-A and PDGF-C are expressed in epithelial cells, muscle, and neuronal progenitors. PDGFR-b is expressed in vascular smooth cells and pericytes,
whereas PDGFR-a is expressed in mesenchymal cells.95,96
Amplification of PDGFs or PDGFRs genes is a frequent finding in glial tumours.
A subset of GIST carries activating point mutations in PDGFR-a. Translocations of
the PDGFR genes have been reported in myeloid disorders and leukaemias. Autocrine
or paracrine loops involving PDGFs and their receptors are commonly observed in
solid tumours.95,96 Autocrine PDGF loops are involved in tumours that originate
from PDGFR-positive cells, such as tumours of glial origin and sarcomas. Autocrine
signalling may also play a role in carcinomas in conjunction with ectopic PDGFR
expression. Moreover, a paracrine PDGF loop is commonly observed in epithelial
cancers. In fact, PDGF is expressed in the neoplastic component whereas PDGFR is
expressed in the stromal compartment. Tumour stroma contains a vascular part consisting of endothelial cells and associated mural cells (PDGFR-b-positive pericytes) and
a fibrous part consisting of mesenchymal cells (PDGFR-a-positive tumour fibroblasts).
Thus PDGF enhances pericyte recruitment to tumour vessels and recruits fibroblasts
that secrete angiogenic and tumour growth factors.95,96 PDGFR-b signalling could also
mediate the increase in interstitial fluid pressure that reduces drug uptake.96
No mutation was found in PDGFR-a gene in ATC; however, ATC, and to a lesser
extent FTC, featured frequent PDGFR-b and PDGFR-a gene copy gains.51 Normal
thyroid cells lack PDGFR, whereas FTC, PTC and ATC cancer cells over-express
PDGFR-b.58,97
Several PDGFRs TKIs have been developed. Imatinib, which inhibits both PDGFRb and PDGFR-a, has been approved for the treatment of GIST carrying activating
mutations in PDGFR-a.4,5 Imatinib was poorly effective in cultured ATC and MTC
cells,98,99 and had practically no effect in MTC patients, suggesting that at least in
this thyroid cancer type PDGFR inhibition is not sufficient to achieve a clinical

RTK inhibitors in thyroid cancer 1033

response.1618 Notably, some VEGFR-targeted TKIs inhibit the activity of PDGFRs


(Table 1 and Figure 2), raising the possibility that these compounds, some of which
are undergoing clinical experimentation in patients affected by thyroid cancer, may
mount a dual attack (VEGFR against endothelial cells and PDGFR against pericytes)
on the tumour vasculature.
SUMMARY
Clinical trials with several agents that target RTKs, in either tumour stroma or parenchyma, have given promising results in thyroid cancer patients. It may be envisaged that a more detailed understanding of the mechanism of action of those
protein kinases, such as RET, that already have a strong track record of involvement
in thyroid oncogenesis, together with the systematic analysis of other genes of the
RTK family by way of such post-genomic techniques as high-throughput sequencing
and copy number analysis, as well as functional screens such as silencing by kinome
RNAi (RNA interference) libraries, will foster more research on RTK targeting for
the treatment of thyroid cancer. Meanwhile, clinical trials are of paramount importance to address the efficacy of the RTK inhibitors and possible side-effects. When
coupled with the genotyping of patients (identifying a mutation in the targeted kinase, such as RET for example) and measurement of surrogate markers of target
inhibition (for instance, RET phosphorylation in pre- and post-treatment tumour biopsies), clinical trials will also help to validate the RTK as a target for therapeutic
intervention.

Research agenda
 preclinical studies in appropriate cell system models are needed to characterize the activity of RTK inhibitors and to validate RTK targets
 preclinical studies in appropriate animal models (tissue-specific transgenic mice
or nude mice xenografts) are needed to validate RTK targets and the activity of
the compounds of interest
 patients enrolled in clinical studies need to be genotyped for the compound(s)
target(s) (example: RET mutation in patients treated with anti-RET agents)
 studies, both preclinical and clinical, are needed to identify surrogate markers
(for example, phosphorylation of the receptor by immunoblot or enzymelinked immunosorbent assay; shedding of the receptor ectodomain) for target
inhibition
 combinations of agents against different targets should be investigated

ACKNOWLEDGEMENTS
We gratefully acknowledge members of our laboratory for continuous support. We
are grateful to Jean Ann Gilder for text editing and StudioCiotola for art-work. We
thank ISO (Istituto Superiore di Oncologia) and Nogec (Naples Oncogenomic Center)
for support.

1034 M. D. Castellone et al

CONFLICT OF INTEREST STATEMENT


M. Santoro has received research funding support from AstraZeneca, Bayer and
Amgen.
REFERENCES
*1. Bardelli A, Parsons DW, Silliman N et al. Mutational analysis of the tyrosine kinome in colorectal cancers. Science 2003; 300: 949.
2. Greenman C, Stephens P, Smith R et al. Patterns of somatic mutation in human cancer genomes. Nature
2007; 446: 153158.
3. Dibb NJ, Dilworth SM & Mol CD. Switching on kinases: oncogenic activation of BRAF and the PDGFR
family. Nature Reviews. Cancer 2004; 4: 718727.
4. Baselga J. Targeting tyrosine kinases in cancer: the second wave. Science 2006; 312: 11751178.
5. Sebolt-Leopold JS & English JM. Mechanisms of drug inhibition of signalling molecules. Nature 2006; 441:
457462.
6. Santoro M & Carlomagno F. Drug insight: small-molecule inhibitors of protein kinases in the treatment
of thyroid cancer. Nature Clinical Practice. Endocrinology & Metabolism 2006; 2: 4252.
7. Kondo T, Ezzat S & Asa SL. Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nature Reviews.
Cancer 2006; 6: 292306.
*8. Wood LD, Parsons DW, Jones S et al. The genomic landscapes of human breast and colorectal cancers.
Science 2007; 318: 11081113.
9. Paes JE & Ringel MD. Dysregulation of the phosphatidylinositol 3-kinase pathway in thyroid neoplasia.
Endocrinology and Metabolism Clinics of North America 2008; 37: 375387.
10. Pacifico F, Mauro C, Barone C et al. Oncogenic and anti-apoptotic activity of NF-kappa B in human
thyroid carcinomas. The Journal of Biological Chemistry 2004; 279: 5461054619.
11. Ludwig L, Kessler H, Wagner M et al. Nuclear factor-kappaB is constitutively active in C-cell carcinoma
and required for RET-induced transformation. Cancer Research 2001; 61: 45264535.
12. Palona I, Namba H, Mitsutake N et al. BRAFV600E promotes invasiveness of thyroid cancer cells
through nuclear factor kappaB activation. Endocrinology 2006; 147: 56995707.
13. Mitsiades CS, McMillin D, Kotoula V et al. Antitumor effects of the proteasome inhibitor bortezomib in
medullary and anaplastic thyroid carcinoma cells in vitro. The Journal of Clinical Endocrinology and Metabolism 2006; 91: 40134021.
14. Engelman JA, Zejnullahu K, Mitsudomi T et al. MET amplification leads to gefitinib resistance in lung
cancer by activating ERBB3 signaling. Science 2007; 316: 10391043.
15. Stommel JM, Kimmelman AC, Ying H et al. Coactivation of receptor tyrosine kinases affects the
response of tumor cells to targeted therapies. Science 2007; 318: 287290.
16. Frank-Raue K, Fabel M, Delorme S et al. Efficacy of imatinib mesylate in advanced medullary thyroid
carcinoma. European Journal of Endocrinology 2007; 157: 215220.
17. de Groot JW, Zonnenberg BA, van Ufford-Mannesse PQ et al. A phase II trial of imatinib therapy for
metastatic medullary thyroid carcinoma. The Journal of Clinical Endocrinology and Metabolism 2007; 92:
34663469.
18. Gross DJ, Munter G, Bitan M et al. The role of imatinib mesylate (Glivec) for treatment of patients with
malignant endocrine tumors positive for c-kit or PDGF-R. Endocrine-Related Cancer 2006; 13: 535540.
19. Pennell NA, Daniels GH, Haddad RI et al. A phase II study of gefitinib in patients with advanced thyroid
cancer. Thyroid 2008; 18: 317323.
*20. Cohen EE, Rosen LS, Vokes EE et al. Axitinib is an active treatment for all histologic subtypes of advanced
thyroid cancer: results from a phase II study. Journal of Clinical Oncology 2008 (Epub ahead of print).
*21. Gupta-Abramson V, Troxel AB, Nellore A et al. Phase II trial of Sorafenib in advanced thyroid cancer.
Journal of Clinical Oncology 2008 (Epub ahead of print).
*22. Hong D, Ye L, Gagel R et al. Medullary thyroid cancer: targeting the RET kinase pathway with sorafenib/
tipifarnib. Molecular Cancer Therapeutics 2008; 7: 10011006.
*23. Sherman SI, Wirth LJ, Droz JP et al. Motesanib diphosphate in progressive differentiated thyroid cancer.
The New England Journal of Medicine 2008; 359: 3142.

RTK inhibitors in thyroid cancer 1035


*24. Wells SA, Gosnell JE, Gagel RF et al. Vandetanib in metastatic hereditary medullary thyroid cancer: follow-up results of an open-label phase II trial. Journal of Clinical Oncology 2007. ASCO Annual Meeting
Proceedings. Vol. 25 (June 20 Supplement): Abstract No: 6018.
*25. Cohen EE, Needles BM, Cullen KJ et al. Phase 2 study of sunitinib in refractory thyroid cancer. Journal
of Clinical Oncology 2008. ASCO Annual Meeting Proceedings Vol. 26 (May 20 Supplement): Abstract
No: 6025.
26. Kelleher FC & McDermott R. Response to sunitinib in medullary thyroid cancer. Annals of Internal
Medicine 2008; 148: 567.
27. Dawson SJ, Conus NM, Toner GC et al. Sustained clinical responses to tyrosine kinase inhibitor sunitinib in thyroid carcinoma. Anti-cancer Drugs 2008; 19: 547552.
*28. Salgia R, Sherman S, Hong DS et al. A phase I study of XL184, a RET, VEGFR2, and MET kinase inhibitor, in
patients (pts) with advanced malignancies, including pts with medullary thyroid cancer (MTC). Journal of
Clinical Oncology 2008. ASCO Annual Meeting Proceedings. Vol. 26 (May 20 supplement): Abstract No:
3522.
*29. Schlumberger M, Carlomagno F, Baudin E et al. New therapeutic approaches to treat medullary thyroid
carcinoma. Nature Clinical Practice. Endocrinology & Metabolism 2008; 4: 2232.
30. Drosten M, Hilken G, Bockmann M et al. Role of MEN2A-derived RET in maintenance and proliferation
of medullary thyroid carcinoma. Journal of the National Cancer Institute 2004; 96: 12311239.
31. de Martimprey H, Bertrand JR, Fusco A et al. siRNA nanoformulation against the ret/PTC1 junction
oncogene is efficient in an in vivo model of papillary thyroid carcinoma. Nucleic Acids Research 2008;
36: e2.
32. Eng C, Mulligan LM, Healey CS et al. Heterogeneous mutation of the RET proto-oncogene in subpopulations of medullary thyroid carcinoma. Cancer Research 1996; 56: 21672170.
33. Zhu Z, Ciampi R, Nikiforova MN et al. Prevalence of RET/PTC rearrangements in thyroid papillary carcinomas: effects of the detection methods and genetic heterogeneity. The Journal of Clinical Endocrinology
and Metabolism 2006; 91: 36033610.
34. Carlomagno F, Vitagliano D, Guida T et al. ZD6474, an orally available inhibitor of KDR tyrosine kinase
activity, efficiently blocks oncogenic RET kinases. Cancer Research 2002; 62: 72847290.
35. Kim DW, Jo YS, Jung HS et al. An orally administered multitarget tyrosine kinase inhibitor, SU11248, is
a novel potent inhibitor of thyroid oncogenic RET/papillary thyroid cancer kinases. The Journal of Clinical
Endocrinology and Metabolism 2006; 91: 40704076.
36. Carlomagno F, Anaganti S, Guida T et al. BAY 43-9006 inhibition of oncogenic RET mutants. Jouranl of
National Cancer Institute 2006; 98: 326334.
37. Polverino A, Coxon A, Starnes C et al. AMG 706, an oral, multikinase inhibitor that selectively targets
vascular endothelial growth factor, platelet-derived growth factor, and kit receptors, potently inhibits
angiogenesis and induces regression in tumor xenografts. Cancer Research 2006; 66: 87158721.
38. Knowles PP, Murray-Rust J, Kjaer S et al. Structure and chemical inhibition of the RET tyrosine kinase
domain. The Journal of Biological Chemistry 2006; 281: 3357733587.
39. Pierotti MA & Greco A. Oncogenic rearrangements of the NTRK1/NGF receptor. Cancer Letters 2006;
232: 9098.
40. Gimm O, Dziema H, Brown J et al. Mutation analysis of NTRK2 and NTRK3, encoding 2 tyrosine kinase
receptors, in sporadic human medullary thyroid carcinoma reveals novel sequence variants. International
Journal of Cancer 2001; 92: 7074.
41. McGregor LM, McCune BK, Graff JR et al. Roles of trk family neurotrophin receptors in medullary
thyroid carcinoma development and progression. Proceedings of the National Academy of Sciences of
the United States of America 1999; 96: 45404545.
42. Strock CJ, Park JI, Rosen DM et al. Activity of irinotecan and the tyrosine kinase inhibitor CEP-751 in
medullary thyroid cancer. The Journal of Clinical Endocrinology and Metabolism 2006; 91: 7984.
43. Comoglio PM, Giordano S & Trusolino L. Drug development of MET inhibitors: targeting oncogene
addiction and expedience. Nature Reviews. Drug Discovery 2008; 7: 504516.
44. Di Renzo MF, Olivero M, Ferro S et al. Overexpression of the c-MET/HGF receptor gene in human
thyroid carcinomas. Oncogene 1992; 7: 25492553.
45. Mineo R, Costantino A, Frasca F et al. Activation of the hepatocyte growth factor (HGF)-Met system in
papillary thyroid cancer: biological effects of HGF in thyroid cancer cells depend on Met expression
levels. Endocrinology 2004; 145: 43554365.

1036 M. D. Castellone et al
46. Fujarewicz K, Jarzab M, Eszlinger M et al. A multi-gene approach to differentiate papillary thyroid
carcinoma from benign lesions: gene selection using support vector machines with bootstrapping.
Endocrine-Related Cancer 2007; 14: 809826.
47. Griffith OL, Melck A, Jones SJ et al. Meta-analysis and meta-review of thyroid cancer gene expression
profiling studies identifies important diagnostic biomarkers. Journal of Clinical Oncology 2006; 24: 5043
5051.
48. Giordano TJ, Kuick R, Thomas DG 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: 66466656.
49. Nardone HC, Ziober AF, LiVolsi VA et al. c-Met expression in tall cell variant papillary carcinoma of the
thyroid. Cancer 2003; 98: 13861393.
50. Papotti M, Olivero M, Volante M et al. Expression of Hepatocyte Growth Factor (HGF) and its Receptor (MET) in Medullary Carcinoma of the Thyroid. Endocrine Pathology 2000; 11: 1930.
51. Liu Z, Hou P, Ji M et al. Highly Prevalent Genetic Alterations in Receptor Tyrosine Kinases and PI3K/Akt
and MAP Kinase Pathways in Anaplastic and Follicular Thyroid Cancers. The Journal of Clinical Endocrinology and Metabolism 2008 (Epub ahead of print).
52. Wasenius VM, Hemmer S, Karjalainen-Lindsberg ML et al. METreceptor tyrosine kinase sequence alterations in differentiated thyroid carcinoma. The American Journal of Surgical Pathology 2005; 29: 544549.
53. Dremier S, Taton M, Coulonval K et al. Mitogenic, dedifferentiating, and scattering effects of hepatocyte
growth factor on dog thyroid cells. Endocrinology 1994; 135: 135140.
54. Wang MH, Lee W, Luo YL et al. Altered expression of the RON receptor tyrosine kinase in various
epithelial cancers and its contribution to tumourigenic phenotypes in thyroid cancer cells. The Journal
of Pathology 2007; 213: 402411.
55. Chattopadhyay C, El-Naggar AK, Williams MD et al. Small molecule c-MET inhibitor PHA665752: effect
on cell growth and motility in papillary thyroid carcinoma. Head & Neck 2008; 30: 9911000.
56. Yarden Y & Sliwkowski MX. Untangling the ErbB signalling network. Nature Reviews. Molecular Cell
Biology 2001; 2: 127137.
57. Ciardiello F & Tortora G. EGFR antagonists in cancer treatment. The New England Journal of Medicine
2008; 358: 11601174.
58. Elliott DD, Sherman SI, Busaidy NL et al. Growth factor receptors expression in anaplastic thyroid
carcinoma: potential markers for therapeutic stratification. Human Pathology 2008; 39: 1520.
59. Mitsiades CS, Kotoula V, Poulaki V et al. Epidermal growth factor receptor as a therapeutic target
in human thyroid carcinoma: mutational and functional analysis. The Journal of Clinical Endocrinology
and Metabolism 2006; 91: 36623666.
60. Schiff BA, McMurphy AB, Jasser SA et al. Epidermal growth factor receptor (EGFR) is overexpressed in
anaplastic thyroid cancer, and the EGFR inhibitor gefitinib inhibits the growth of anaplastic thyroid
cancer. Clinical Cancer Research 2004; 10: 85948602.
61. Haugen DR, Akslen LA, Varhaug JE et al. Expression of c-erbB-3 and c-erbB-4 proteins in papillary
thyroid carcinomas. Cancer Research 1996; 56: 11841188.
62. Wiseman SM, Melck A, Masoudi H et al. Molecular Phenotyping of Thyroid Tumors Identifies a Marker
Panel for Differentiated Thyroid Cancer Diagnosis. Annals of Surgical Oncology 2008 (Epub ahead of print).
63. Delys L, Detours V, Franc B et al. Gene expression and the biological phenotype of papillary thyroid
carcinomas. Oncogene 2007; 26: 78947903.
64. Haugen DR, Akslen LA, Varhaug JE et al. Expression of c-erbB-2 protein in papillary thyroid carcinomas.
British Journal of Cancer 1992; 65: 832837.
65. Hebrant A, van Staveren WC, Delys L et al. Long-term EGF/serum-treated human thyrocytes mimic papillary thyroid carcinomas with regard to gene expression. Experimental Cell Research 2007; 313: 32763284.
66. Croyle M, Akeno N, Knauf JA et al. RET/PTC-induced cell growth is mediated in part by epidermal
growth factor receptor (EGFR) activation: evidence for molecular and functional interactions between
RET and EGFR. Cancer Research 2008; 68: 41834191.
67. Nobuhara Y, Onoda N, Yamashita Y et al. Efficacy of epidermal growth factor receptor-targeted molecular therapy in anaplastic thyroid cancer cell lines. British Journal of Cancer 2005; 92: 11101116.
68. Younes MN, Yigitbasi OG, Park YW et al. Antivascular therapy of human follicular thyroid cancer
experimental bone metastasis by blockade of epidermal growth factor receptor and vascular growth
factor receptor phosphorylation. Cancer Research 2005; 65: 47164727.

RTK inhibitors in thyroid cancer 1037


69. Kim S, Schiff BA, Yigitbasi OG et al. Targeted molecular therapy of anaplastic thyroid carcinoma with
AEE788. Molecular Cancer Therapeutics 2005; 4: 632640.
70. Hoffmann S, Burchert A, Wunderlich A et al. Differential effects of cetuximab and AEE 788 on epidermal growth factor receptor (EGF-R) and vascular endothelial growth factor receptor (VEGF-R) in
thyroid cancer cell lines. Endocrine 2007; 31: 105113.
71. Boelaert K, McCabe CJ, Tannahill LA et al. Pituitary tumor transforming gene and fibroblast growth
factor-2 expression: potential prognostic indicators in differentiated thyroid cancer. The Journal of
Clinical Endocrinology and Metabolism 2003; 88: 23412347.
72. Eggo MC, Hopkins JM, Franklyn JA et al. Expression of fibroblast growth factors in thyroid cancer. The
Journal of Clinical Endocrinology and Metabolism 1995; 80: 10061011.
73. de la Torre NG, Buley I, Wass JA et al. Angiogenesis and lymphangiogenesis in thyroid proliferative
lesions: relationship to type and tumour behaviour. Endocrine-Related Cancer 2006; 13: 931944.
74. St Bernard R, Zheng L, Liu W et al. Fibroblast growth factor receptors as molecular targets in thyroid
carcinoma. Endocrinology 2005; 146: 11451153.
75. Kondo T, Zheng L, Liu W et al. Epigenetically controlled fibroblast growth factor receptor 2 signaling
imposes on the RAS/BRAF/mitogen-activated protein kinase pathway to modulate thyroid cancer
progression. Cancer Research 2007; 67: 54615470.
76. Ezzat S, Huang P, Dackiw A et al. Dual inhibition of RET and FGFR4 restrains medullary thyroid cancer
cell growth. Clinical Cancer Research 2005; 11: 13361341.
77. Surmacz E. Growth factor receptors as therapeutic targets: strategies to inhibit the insulin-like growth
factor I receptor. Oncogene 2003; 22: 65896597.
78. Wang Z, Chakravarty G, Kim S et al. Growth-inhibitory effects of human anti-insulin-like growth factorI receptor antibody (A12) in an orthotopic nude mouse model of anaplastic thyroid carcinoma. Clinical
Cancer Research 2006; 12: 47554765.
79. Coulonval K, Vandeput F, Stein RC et al. Phosphatidylinositol 3-kinase, protein kinase B and ribosomal
S6 kinases in the stimulation of thyroid epithelial cell proliferation by cAMP and growth factors in the
presence of insulin. The Biochemical Journal 2000; 348: 351358.
80. Mitsiades CS, Mitsiades NS, McMullan CJ et al. Inhibition of the insulin-like growth factor receptor-1
tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematologic malignancies,
and solid tumors. Cancer Cell 2004; 5: 221230.
81. Ellis LM & Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nature Reviews.
Cancer 2008; 8: 579591.
82. Bergers G & Hanahan D. Modes of resistance to anti-angiogenic therapy. Nature Reviews. Cancer 2008; 8:
592603.
83. Tammela T, Zarkada G, Wallgard E et al. Blocking VEGFR-3 suppresses angiogenic sprouting and
vascular network formation. Nature 2008 Jun 25 (Epub ahead of print).
84. Viglietto G, Maglione D, Rambaldi M et al. Upregulation of vascular endothelial growth factor (VEGF)
and downregulation of placenta growth factor (PlGF) associated with malignancy in human thyroid
tumors and cell lines. Oncogene 1995; 11: 15691579.
85. Bunone G, Vigneri P, Mariani L et al. Expression of angiogenesis stimulators and inhibitors in human
thyroid tumors and correlation with clinical pathological features. The American Journal of Pathology
1999; 155: 19671976.
86. Soh EY, Duh QY, Sobhi SA et al. Vascular endothelial growth factor expression is higher in differentiated
thyroid cancer than in normal or benign thyroid. The Journal of Clinical Endocrinology and Metabolism
1997; 82: 37413747.
87. Tuttle RM, Fleisher M, Francis GL et al. Serum vascular endothelial growth factor levels are elevated in
metastatic differentiated thyroid cancer but not increased by short-term TSH stimulation. The Journal of
Clinical Endocrinology and Metabolism 2002; 87: 17371742.
88. Klein M, Vignaud JM, Hennequin V et al. Increased expression of the vascular endothelial growth factor
is a pejorative prognosis marker in papillary thyroid carcinoma. The Journal of Clinical Endocrinology and
Metabolism 2001; 86: 656658.
89. Huang SM, Lee JC, Wu TJ et al. Clinical relevance of vascular endothelial growth factor for thyroid neoplasms. World Journal of Surgery 2001; 25: 302306.
90. Yu XM, Lo CY, Chan WF et al. Increased expression of vascular endothelial growth factor C in papillary thyroid
carcinoma correlates with cervical lymph node metastases. Clinical Cancer Research 2005; 11: 80638069.

1038 M. D. Castellone et al
91. Hung CJ, Ginzinger DG, Zarnegar R et al. Expression of vascular endothelial growth factor-C in benign
and malignant thyroid tumors. The Journal of Clinical Endocrinology and Metabolism 2003; 88: 36943699.
92. Gomez-Rivera F, Santillan-Gomez AA, Younes MN et al. The tyrosine kinase inhibitor, AZD2171, inhibits vascular endothelial growth factor receptor signaling and growth of anaplastic thyroid cancer
in an orthotopic nude mouse model. Clinical Cancer Research 2007; 13: 45194527.
93. Schoenberger J, Grimm D, Kossmehl P et al. Effects of PTK787/ZK222584, a tyrosine kinase inhibitor,
on the growth of a poorly differentiated thyroid carcinoma: an animal study. Endocrinology 2004; 145:
10311038.
94. Soh EY, Eigelberger MS, Kim KJ et al. Neutralizing vascular endothelial growth factor activity inhibits
thyroid cancer growth in vivo. Surgery 2000; 128: 10591065.
95. Andrae J, Gallini R & Betsholtz C. Role of platelet-derived growth factors in physiology and medicine.
Genes & Development 2008; 22: 12761312.
96. Heldin CH, Rubin K, Pietras K et al. High interstitial fluid pressure - an obstacle in cancer therapy.
Nature Reviews. Cancer 2004; 4: 806813.
97. Heldin NE, Gustavsson B, Claesson-Welsh L et al. Aberrant expression of receptors for plateletderived growth factor in an anaplastic thyroid carcinoma cell line. Proceeding of the National Academy
of Sciences of the United States of America 1988; 85: 93029306.
98. Dziba JM & Ain KB. Imatinib mesylate (gleevec; STI571) monotherapy is ineffective in suppressing human anaplastic thyroid carcinoma cell growth in vitro. The Journal of Clinical Endocrinology and Metabolism
2004; 89: 21272135.
99. de Groot JW, Plaza Menacho I, Schepers H et al. Cellular effects of imatinib on medullary thyroid cancer
cells harboring multiple endocrine neoplasia Type 2A and 2B associated RET mutations. Surgery 2006;
139: 806814.

Вам также может понравиться