17
 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 Carlomag no  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 Salvat ore  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 ra- tionale for assessing the efcacy of RTK blockade to treat patients who are resistant to or not candidates for treatment with radioactive iodine. The rst results of clinical trials based on the use of RTK inhibitors in thyroid cancer patients have recently been published. Here we discuss targeting of specic RTKs as a potential therapeutic strategy for the treatment of thyroid cancer. Key words:  kinase; thyroid; monoclonal antibody; small-molecule inhibitor. * Corresponding author. Tel.:  þ39-081-7463056; Fax:  þ39-081-7463037. E-mail address:  [email protected]  (M. Santoro). 1521-690X/$ - see front matter  ª 2008 Elsevier Ltd. All rights reserved. Best Pract ice & Resear ch Clinical Endocrino logy & Metabolism Vol. 22, No. 6, pp. 1023–1038, 2008 doi:10.1016/j.beem.2008.09.012 available online at  http://www .sciencedirect.com

Inhibidores de Receptor Tirosina Kinasa en CA Tiroideo

Embed Size (px)

DESCRIPTION

inhibidores de TK para tratamiento de CA tiroides

Citation preview

  • 9Francesca Carlomagno MD, PhD

    targeting of specific RTKs as a potential therapeutic strategy for the treatment of thyroid cancer.

    1521-690X/$ - see front matter 2008 Elsevier Ltd. All rights reserved.

    Best Practice & Research Clinical Endocrinology & MetabolismVol. 22, No. 6, pp. 10231038, 2008

    doi:10.1016/j.beem.2008.09.012available online at http://www.sciencedirect.com* Corresponding author. Tel.: 39-081-7463056; Fax: 39-081-7463037.E-mail address: [email protected] (M. Santoro).Key words: kinase; thyroid; monoclonal antibody; small-molecule inhibitor.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, PhDAssociate Professor

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

    Massimo Santoro* MD, PhDProfessor 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 tyrosinekinases (RTKs) or their downstream signalling molecules. Hence, there is a strong biological ra-tionale for assessing the efficacy of RTK blockade to treat patients who are resistant to or notcandidates for treatment with radioactive iodine. The first results of clinical trials based on theuse of RTK inhibitors in thyroid cancer patients have recently been published. Here we discussReceptor tyrosine kinase inhibitors in thyroid

    cancer

    Maria Domenica Castellone MD, PhDResearch 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

  • CANCER GENES CODING FOR RECEPTOR TYROSINE KINASES

    The protein kinase complement of the human genome (the kinome) contains 518protein kinase genes, including 58 receptor tyrosine kinases (RTKs) and 32 non-RTKs (www.kinase.com/human/kinome). In high-throughput genome-sequencing pro-jects, approximately 120 genes encoding protein kinases were found to be mutatedand 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 juxtamembranesegment (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 pointmutations, cancers may feature illicit expression of the RTK or its cognate growthfactor and RTK gene rearrangements.35 Thyroid cancer, as discussed below, is oftenassociated to the oncogenic conversion of genes coding for RTKs or their signallingeffectors.6,7

    MECHANISMS OF RTK ACTIVATION

    Upon growth factor binding, RTKs undergo a dimerization process that activatesthe enzymatic function.3 All kinase domains are divided into an amino-terminal

    1024 M. D. Castellone et alFigure 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 do-

    main (JMR), and tyrosine kinase domain (TK), is illustrated. Blue boxes summarize major biological outcomes

    of the activation of specific signalling components.

  • (N-terminal) and a carboxy-terminal (C-terminal) lobe. The active site is located in thecleft between the N- and C-terminal lobes. Mg-ATP binds to the base of the cleft, the

    RTK inhibitors in thyroid cancer 1025non-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 sitecan also bind small-molecule inhibitors (see below).4,5 In the inactive kinase, the activesite is closed by the activation segment of the C-terminal lobe. Growth-factor-bindingcauses RTK dimerization, followed by extensive movements of the activation segmentand the JMR domain; this process allows ATP binding and catalysis.

    Oncogenic mutations disrupt normal regulatory mechanisms and lead to constitu-tive activation of the kinase. Point mutations or rearrangements in the extracellulardomain mimic ligand-binding, thereby causing constitutive dimerization. Mutations inthe 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 recruitsintracellular proteins endowed with phosphotyrosine binding domains.35 Proximaltargets of the RTKs invoke the intracellular signalling cascades RAS-RAF-MAPK (theERK pathway) and the phosphatidylinositol 3-kinase (PI3K) AKT that ultimatelylead to diverse biological responses: i.e. mitogenesis, survival, differentiation and mo-tility (Figure 1).35

    Genes coding for proteins working in the RTK-initiated signal transduction path-ways are frequently mutated in cancer. For instance, systematic DNA sequencing ofthe best-annotated protein-coding genes in breast and colorectal cancer revealedfrequent mutations in the PI3K and nuclear factor kB (NFkB) pathways, both of whichare involved in RTK signalling (Figure 1).8 In the case of thyroid cancer, paradigmaticexamples of this concept are mutations in BRAF, a RAF family serine/threonine kinase,in papillary thyroid carcinoma (PTC) (2969%) and PTC-derived anaplastic thyroid car-cinoma (ATC) (1035%), and in the RAS small GTPase that is mutated in the follicularvariant 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 PI3Kcatalytic subunit, is associated with ATC and FTC, and loss of expression of PTEN, themajor phosphatase antagonist of PI3K function, is frequent in ATC.9 Finally, functionalactivation of NFkB has been described in ATC.10 NFkB is crucial for RET and BRAFoncogene signalling in thyroid cancer cells11,12, and the proteasome inhibitor bortezo-mib (PS-341, Velcade, Millennium Pharmaceuticals), whose complex mechanism ofaction 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 target-ing a feasible therapeutic strategy for cancer.4,5 Monoclonal antibodies (mAbs) canblock the growth factor or the RTK itself. Some humanized mAbs such as trastuzu-mab (herceptin; Genentech/Roche) against the HER2/ErbB2 receptor, cetuximab (er-bitux; ImClone) against the epidermal growth factor receptor (EGFR/HER1/ErbB1),and bevacizumab (avastin; Genentech/Roche) against vascular endothelial growth fac-tor (VEGF) are now part of the treatment regimen for specific tumours.4,5 The anti-tumour activity of RTK-directed mAbs has been ascribed to different mechanisms: (1)

  • blockade of ligand-binding; (2) blockade of RTK homo- or hetero-dimerization; (3) in-terference with the active-like RTK conformation; (4) down-regulation of the receptor

    1026 M. D. Castellone et alfrom the cell surface (receptor internalization); (5) shedding of the extracellulardomain 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 ob-struct kinase activity by binding to the ATP pocket of the kinase in competition withcellular ATP. The effectiveness of imatinib (imatinib mesylate/gleevec/glivec, STI571; No-vartis), an inhibitor of ABL, KITand platelet-derived growth factor receptor (PDGFR), inBCR-ABL-positive chronicmyelogenous leukaemia (CML) andKITor PDGFR-amutant gas-trointestinal stromal tumours (GISTs) has illustrated the power of this approach.4 TheATP binding site is highly conserved across the kinome. Thus, at best, TKIs may be se-lective but not specific and affect more than one RTK. As most cancers are the resultof a number of mutations, it is reasonable to suppose that TKIs able to target multiplekinases or a rational combination of TKIs will be clinically more effective than agentsblocking a single kinase. As discussed in the next paragraph, multi-targeting TKIs orcombination 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 sec-ondary mutations of the target kinase. These mutations either prevent the kinase fromadopting the conformation to which the compound binds or alter the compound con-tact 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 tobe highly dependent on specific genes and related pathways. This dependence canbe exploited therapeutically by appropriate targeting. Thus, the use of TKIs shouldbe restricted to those tumours that are addicted to the kinase that is targeted bythat specific agent. Finally, compensatory cross-talk between RTKs may attenuateTKI efficacy. Tumour cells become resistant to an EGFR TKI by an adaptation mecha-nism involving activation of alternative RTKs, such as METor PDGFR.14 Another studyshowed that tumour cells are resistant to EGFR TKI up front, because they simulta-neously activate the MET, EGFR and PDGFR RTKs.15 Cocktails of drugs or multi-targeting 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 stud-ies based on the use of TKIs for thyroid cancer patients (www.clinicaltrials.gov). Theseare summarized in Table 1, and include imatinib mesylate1618, gefitinib (ZD1839/Ire-ssa; 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 TheRTKs targeted by these agents are illustrated in Figure 2, and their involvement inthyroid 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)

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

    RTK inhibitors in thyroid cancer 1027Name Other names Tyrosine kinase targets

    (only those IC50< 1 mM)

    Clinical development

    Imatinib mesylate

    (Novartis)

    Gleevec, glivec,

    STI571

    ABL, KIT, PDGFR Approved for

    CML, GIST

    Gefitinib

    (AstraZeneca)

    ZD1839, iressa EGFR Approved in some

    countries for NSCLC

    Axitinib

    (Pfizer)

    AG-013736 VEGFR-1, -2, -3, PDGFR, KIT Investigational

    Vandetanib

    (AstraZeneca)

    Zactima,

    ZD6474

    EGFR, RET, VEGFR-2, -3 Investigational

    Sunitinib (Pfizer) Sutent,

    SU11248

    VEGFR-2, PDGFR, KIT, FLT3,

    RET, FGFR-1, CSF-1R

    Approved for metastatic

    renal carcinoma and

    imatinib-resistant GIST

    Sorafenib

    (Bayer)

    Nexavar,

    BAY 43-9006

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

    PDGFR, FLT3, RET, KIT, FGFR-1

    Approved for metastatic

    renal carcinoma

    Motesanib

    diphosphate

    (Amgen)

    AMG 706 VEGFR-1, -2, -3, KIT, RET,

    PDGFR, FLT3

    Investigational

    XL184 (Exelixis) e RET, VEGFR-2, MET Investigational

    Pazopanib GW-786034 VEGFR-1, -2, -3, PDGFR, Investigationalligands. In PTC, chromosomal inversions or translocations cause the fusion of the RETkinase-encoding domain with the 50-end of heterologous genes. The resulting chimericsequences are called RET/PTC.6,7 Germline point mutations in RET cause MEN-2(multiple endocrine neoplasia type 2) syndromes that predisposes to MTC. Somaticmutations of RET are also found in sporadic MTC.29 Transgenic mouse models dem-onstrated that REToncogenes are able to drive PTC and MTC formation.6 Moreover,RET knock-down by dominant-negative mutants or RNAi impairs proliferation ofMTC and PTC cell lines harbouring RET-derived oncogenes.30,31 In this scenarioRET appears to be a promising target for the molecular therapy of PTC and MTC.However, the heterogeneous distribution of RET mutations in some tumour sampleshas challenged the notion that RET alteration is the driving lesion in those individualpatients.32,33

    Some of the TKIs in clinical experimentation in thyroid cancer patients have anti-RET activity (Table 1 and Figure 2). Most of them e.g. vandetanib34, sunitinib35, sor-afenib36, XL-18428, and motesanib37, inhibit both RETand VEGFRs. Thus, potentiallythese drugs can simultaneously attack both neoplastic and endothelial cells. X-raystructural analysis has demonstrated that vandetanib binds the ATP pocket of thekinase and is therefore an ATP-competitive RET TK inhibitor.38

    (GlaxoSmithKline) KIT, FGFR-1

    ABL, Abelson murine leukaemia viral (v-abl) oncogene homologue; PDGFR, platelet-derived growth fac-

    tor 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 transfec-

    tion; FGFR, fibroblast growth factor receptor; MET, hepatocyte growth factor receptor; CSF-1R, colony-

    stimulating factor receptor.

    **CML chronic myelogenous leukaemia, GIST gastro intestinal stromal tumours; NSCLC nonsmall cell lung carcinoma.

  • 1028 M. D. Castellone et alNTRK1 (NERVE GROWTH FACTOR RECEPTOR)

    NTRK1 (also known as TRKA) belongs to the RTK neurotrophin receptor family thatincludes NTRK2 (TRKB) and NTRK3 (TRKC) (Figure 2).39 NTRK1 is the high-affinityreceptor for nerve growth factor. In 513% of PTC cases, the NTRK1 TK-encodingdomain is illegitimately recombined with heterologous sequences, thereby generatingTRK-T oncogenes.7,39 As yet there is no evidence that other members of the NTRKfamily are mutated in thyroid carcinoma.40 However, changes in their expressionwere involved in thyroid C-cell transformation, with NTRK2 being reduced andNTRK3 being up-regulated in MTC.41 The CEP-751 TKI, which is active againstboth 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 scat-ter factor) (Figure 2).43 The MET homologue, RON, is the receptor for macrophage-stimulating protein (MSP). Amplification of the MET gene has been reported in severalhuman tumours; more rarely, MET carries activating point mutations. Most often, MET

    Figure 2. Schematic representation of some receptor tyrosine kinases (RTKs). Specific domains of the ex-

    tracellular portion are indicated: immunoglobulin-like, plexin and transcription factor domain (IPT); sema-

    phorin 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).

  • 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

    RTK inhibitors in thyroid cancer 1029Once activated, MET stimulates cell scattering, invasion, protection from apoptosisand anoikis, and angiogenesis.43

    Approximately 70% of PTCs over-express MET, whereas normal thyrocytes do notexpress MET. HGF is locally produced by PTC stromal fibroblasts.44,45 METwas iden-tified as one of the most consistently up-regulated markers of thyroid cancer, partic-ularly 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 copygains have been identified in approximately 10% of ATCs and less frequently inFTCs,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, HGFwas 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 cor-related with advanced clinical stage.54

    Monoclonal antibodies against HGF or the extracellular MET domain and METATP-competitive TKIs have been described.43 The TKI PF2341066 (Pfizer) is undergoingphase-I/II clinical trials, and the related compound, PHA665752, was effective againstPTC cells in vitro.55 Another two TKIs, XL880 and XL184 (Exelixis), are undergoingclinical experimentation. It is noteworthy that XL184, which is also a RET inhibitor, isbeing 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) be-longs 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 twocysteine-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 devoidof intrinsic kinase activity, are non-autonomous receptors and require heterodimeri-zation with other ErbB family members for activation and signalling. ErbB2 is themost potent oncoprotein in the ErbB family and it is the preferred heterodimeric part-ner of the other ErbBs. ErbB3 is a potent PI3K activator.56 The EGF family includesvarious 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, EGF-related growth factors are produced either by the tumour cells or by stromal cells.56

    In gliomas, EGFR amplification is often accompanied by structural rearrangements. So-matic 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 inthe 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 EGFRmutation (exons 1821) was found in 31 ATCs58, and only two mutations were de-tected in 62 thyroid cancers and 11 cell lines.59 In another study, no EGFR mutationwas found in 51 ATC and 64 FTC samples; however, EGFR gene copy gains were fre-quent 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

  • 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,

    1030 M. D. Castellone et alEGF is mitogenic for thyrocytes, and long-term treatment with EGF induced gene ex-pression 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 forma complex that mediates EGFR-dependent RET phosphorylation.66

    Four different anti-EGFR agents are approved in several countries for NSCLC andfor colorectal, pancreatic and head and neck carcinoma (Figure 2). These include twomAbs, cetuximab (a mouse/human chimeric mAb) and panitumumab (vectibix; Abge-nix) (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 HER2-over-expressing breast cancers.4,5,57

    In preclinical studies, micromolar doses of gefitinib60,67 or NVP-AEE788 (Novartis),an EGFR and VEGFRTKI with additional activity against RET59,68,69, were effective in cul-tured ATC cells. NVP-AEE788 and cetuximab reduced the secretion of VEGF from thy-roid cancer cells, but only NVP-AEE788 dose-dependently inhibited proliferation.70 Incells expressing activated REToncogenes, NVP-AEE788 and PKI166 (Novartis), anotherEGFRTKI, were active at submicromolar concentrations, secondary to interaction be-tween EGFR and RET.66 No objective response was obtained in a phase-II study of gefi-tinib in patients with locally advanced or metastatic thyroid cancer, although a fraction ofpatients achieved prolonged stable disease and there was a reduction in tumour size andplasma thyroglobulin concentration. Therefore, EGFR inhibition is not sufficient, at leastby 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 membersthat are important regulators of angiogenesis and tumourigenesis. FGFs signal throughfour RTKs (FGFR-1, -2, -3 and -4).7 Each receptor has two or three immunoglobulin-like extracellular domains, a transmembrane domain, and an intracellular TK(Figure 2).

    Thus far, no mutations or rearrangements involving FGFRs have been identified inthyroid cancer.7 Expression of FGF1 and FGF2 (also known as basic FGF, a potent an-giogenic factor) is increased in thyroid cancer.7173 Increased expression of FGFR-1, -3and -4 has been observed in benign and malignant thyroid tumours.71,74 FGFR-2 ex-pression, instead, was down-regulated in thyroid cancer, and its re-expression in thy-roid carcinoma cells interrupted signalling upstream of BRAF and MAPK and reducedcell growth.75 FGFR-4 is expressed predominantly in aggressive thyroid tumour typesand MTC cells. Molecular targeting of FGFR-4 with the ATP-competitive inhibitorPD173074 (Pfizer) inhibited growth and reduced tumourigenesis of MTC cells.76 Itis noteworthy that sorafenib, sunitinib and pazopanib, which are undergoing clinical ex-perimentation in thyroid cancer patients, exert anti-FGFR activity (Table 1 andFigure 2).

    IGF-1R (INSULIN-LIKE GROWTH FACTOR RECEPTOR 1)

    IGF-1R (insulin-like growth factor receptor 1) is a ubiquitous transmembrane tyrosinekinase structurally similar to the insulin receptor (IR). IGF-1R is composed of two

  • extracellular a-subunits and two intracellular b-subunits (Figure 2). The a-subunitsbind ligands (IGF-I, IGF-II, and insulin at supraphysiological doses), whereas b-subunits

    77

    well as in endothelial cells.

    RTK inhibitors in thyroid cancer 1031VEGF targeting is a promising anti-cancer therapeutic approach. VEGF-targetedtherapy acts through various mechanisms: inhibition of new vessel formation, apopto-sis of pre-existing vessels, blockade of endothelial cell progenitors, and vessel constric-tion (with reduced blood flow and ischaemia).81,82 There are currently more than 20different VEGF-targeted agents undergoing clinical evaluation (Figure 2). These includeneutralizing mAbs against VEGF or VEGFRs. Bevacizumab is a humanized mAb againstVEGF currently registered for colorectal cancer, breast cancer and NSCLC.4,5,81 Sev-eral multi-targeting TKIs that block VEGFRs have shown promising clinical activityagainst various solid tumours, and two of them (sorafenib and sunitinib) are registeredas anti-cancer agents.4,5,81 Unfortunately, in most cases the effects of anti-angiogenictreatment are only transitory. Various mechanisms have been evoked to explain thisphenomenon: production by cancer cells of angiogenic growth factors other thanVEGF (FGF2, PDGF, ephrins, angiopoietin, IL-8), recruitment of angiogeniccontain the TK domain. Most of the effects of IGF-I are mediated by IGF-1R. Theeffects of IGF-II may be mediated by both IGF-IR and an alternatively spliced variantof IR.

    IGF-II and IGF-1R are over-expressed in many cancer types. High circulating levelsof IGF-I have been indicated as risk factors for various tumour types. Moreover, IGF-1R up-regulation was found to mediate resistance to TKIs in different types of cancercells. IGF-I and IGF-1R are over-expressed in thyroid cancer, particularly in the mostaggressive variants.78 Importantly, IGF-I and insulin are essential for the mitogenic ac-tion of TSH and EGF in thyroid follicular cells.79 Both anti-IGF-R mAbs and TKIs arebeing developed (Figure 2). The TKI NVP-ADW742 (Novartis) is cytotoxic for FTC-and MTC-derived cancer cells.80 IMC-A12 (ImClone) is a fully human mAb that targetsthe human IGF-1R. IMC-A12 was effective in treating an orthotopic mouse model ofATC.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 isVEGFA (commonly referred to as VEGF). VEGFA and VEGFB are angiogenic proteins,whereas VEGFC and VEGFD are primarily lymphangiogenic proteins.81,82 The VEGFreceptors 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 vasculatureand also in other cell types. VEGFR-3 is the receptor for VEGFC and VEGFD and isexpressed primarily on lymphatic endothelium.81,82 However, it has been recentlydemonstrated that VEGFR-3 is also expressed in tumour blood vessels where it con-tributes to angiogenic sprouting, and simultaneous blockade of VEGFR-3 and VEGFR-2resulted 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, tu-mour size and metastasis. Over-expression of VEGFC in PTC has been reported inthree 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

  • bone-marrow precursors, survival of blood vessels covered by pericytes, and migra-tion of cancer cells outside the primary tumour to co-opt pre-existing vessels.91

    1032 M. D. Castellone et alImportantly, clinical trials with VEGFR-blocking multi-target TKIs (sorafenib,axitinib, motesanib, sunitinib, vandetanib and XL-184) have yielded promising resultsin thyroid cancer patients in the last few months (Table 1).2028 In addition, pazopanib(GW-786034; GlaxoSmithKline), a multi-targeted pan-VEGFR inhibitor, is undergoingclinical experimentation in thyroid cancer patients (www.clinicaltrials.gov).

    Preclinical studies with NVP-AEE788, a dual VEGFR and EGFR inhibitor, in FTC andATC cells6870, and with PTK787/ZK222584 (Novartis and Schering), a pan-VEGFRTKI, in thyroid carcinoma mouse xenografts93 showed promising results. Cediranib(AZD2171; AstraZeneca), another pan-VEGFRTKI, inhibited tumour growth and pro-longed animal survival in an orthotopic nude mouse model of ATC.92 Finally, VEGFmAbs, including the murine version of bevacizumab, inhibited the growth of mousexenografts 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 immuno-globulin (Ig) loops and a split intracellular TK domain (Figure 2).95 PDGF-AA andPDGF-CC ligands act via PDGFR-a, while PDGF-BB and probably PDGF-DD act viaPDGFR-b. PDGF-B is expressed mainly in vascular endothelial cells, megakaryocytesand neurons. PDGF-A and PDGF-C are expressed in epithelial cells, muscle, and neu-ronal 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 ofthe PDGFR genes have been reported in myeloid disorders and leukaemias. Autocrineor paracrine loops involving PDGFs and their receptors are commonly observed insolid tumours.95,96 Autocrine PDGF loops are involved in tumours that originatefrom PDGFR-positive cells, such as tumours of glial origin and sarcomas. Autocrinesignalling may also play a role in carcinomas in conjunction with ectopic PDGFRexpression. Moreover, a paracrine PDGF loop is commonly observed in epithelialcancers. In fact, PDGF is expressed in the neoplastic component whereas PDGFR isexpressed in the stromal compartment. Tumour stroma contains a vascular part con-sisting of endothelial cells and associated mural cells (PDGFR-b-positive pericytes) anda fibrous part consisting of mesenchymal cells (PDGFR-a-positive tumour fibroblasts).Thus PDGF enhances pericyte recruitment to tumour vessels and recruits fibroblaststhat secrete angiogenic and tumour growth factors.95,96 PDGFR-b signalling could alsomediate 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 lesserextent FTC, featured frequent PDGFR-b and PDGFR-a gene copy gains.51 Normalthyroid cells lack PDGFR, whereas FTC, PTC and ATC cancer cells over-expressPDGFR-b.58,97

    Several PDGFRs TKIs have been developed. Imatinib, which inhibits both PDGFR-b and PDGFR-a, has been approved for the treatment of GIST carrying activatingmutations in PDGFR-a.4,5 Imatinib was poorly effective in cultured ATC and MTCcells,98,99 and had practically no effect in MTC patients, suggesting that at least inthis thyroid cancer type PDGFR inhibition is not sufficient to achieve a clinical

  • 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

    renchyma, have given promising results in thyroid cancer patients. It may be envis-aged that a more detailed understanding of the mechanism of action of thoseprotein kinases, such as RET, that already have a strong track record of involvementin thyroid oncogenesis, together with the systematic analysis of other genes of theRTK family by way of such post-genomic techniques as high-throughput sequencingand copy number analysis, as well as functional screens such as silencing by kinomeRNAi (RNA interference) libraries, will foster more research on RTK targeting forthe treatment of thyroid cancer. Meanwhile, clinical trials are of paramount impor-tance to address the efficacy of the RTK inhibitors and possible side-effects. Whencoupled with the genotyping of patients (identifying a mutation in the targeted ki-nase, such as RET for example) and measurement of surrogate markers of targetinhibition (for instance, RET phosphorylation in pre- and post-treatment tumour bi-opsies), clinical trials will also help to validate the RTK as a target for therapeuticintervention.

    Research agenda

    preclinical studies in appropriate cell system models are needed to character-

    RTK inhibitors in thyroid cancer 1033ACKNOWLEDGEMENTS

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

    ize the activity of RTK inhibitors and to validate RTK targets preclinical studies in appropriate animal models (tissue-specific transgenic miceor nude mice xenografts) are needed to validate RTK targets and the activity ofthe 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 enzyme-linked immunosorbent assay; shedding of the receptor ectodomain) for targetinhibition

    combinations of agents against different targets should be investigatedare undergoing clinical experimentation in patients affected by thyroid cancer, maymount 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 pa-

  • CONFLICT OF INTEREST STATEMENT

    1034 M. D. Castellone et alM. Santoro has received research funding support from AstraZeneca, Bayer andAmgen.

    REFERENCES

    *1. Bardelli A, Parsons DW, Silliman N et al. Mutational analysis of the tyrosine kinome in colorectal can-

    cers. 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 Metab-

    olism 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.

  • *24. Wells SA, Gosnell JE, Gagel RF et al. Vandetanib in metastatic hereditary medullary thyroid cancer: fol-

    low-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.

    RTK inhibitors in thyroid cancer 1035*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 suni-

    tinib 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 subpop-

    ulations 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 car-

    cinomas: effects of the detection methods and genetic heterogeneity. The Journal of Clinical Endocrinology

    and Metabolism 2006; 91: 36033610.

    34. Carlomagno F, Vitagliano D, Guida Tet 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.

  • 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.

    1036 M. D. Castellone et al47. 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 Recep-

    tor (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 Endocri-

    nology and Metabolism 2008 (Epub ahead of print).

    52. Wasenius VM, Hemmer S, Karjalainen-Lindsberg ML et al. METreceptor tyrosine kinase sequence alter-

    ations 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 StaverenWC, Delys L et al. Long-term EGF/serum-treated human thyrocytes mimic papil-

    lary 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 Yet al. Efficacy of epidermal growth factor receptor-targeted molec-

    ular 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.

  • 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 epider-

    RTK inhibitors in thyroid cancer 1037mal 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 factor-

    I 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 neo-

    plasms. World Journal of Surgery 2001; 25: 302306.

    90. YuXM, LoCY,ChanWFet al. Increased expressionof vascular endothelial growth factorC in papillary thyroid

    carcinoma correlates with cervical lymph node metastases. Clinical Cancer Research 2005; 11: 80638069.

  • 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, in-

    hibits 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 platelet-

    derived 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 hu-

    man 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.

    1038 M. D. Castellone et al

    Receptor tyrosine kinase inhibitors in thyroid cancerCancer genes coding for receptor tyrosine kinasesMechanisms of RTK activationMechanisms of RTK signallingRTKs as molecular targets for cancer treatmentRTK inhibitors in thyroid cancerRET (glial-derived growth factor receptor)NTRK1 (nerve growth factor receptor)MET (hepatocyte growth factor receptor)EGFR (epidermal growth factor receptor)FGFR (fibroblast growth factor receptor)IGF-1R (insulin-like growth factor receptor 1)VEGFR (vascular endothelial growth factor receptor)PDGFR (Plateled-derived growth factor receptor)SummaryAcknowledgementsConflict of interest statementReferences