Abstract
Thyroid cancer is rare, but it is the most frequent endocrine malignancy. Its prognosis is generally favorable, especially in cases of well-differentiated thyroid cancers (DTCs), such as papillary and follicular cancers, which have survival rates of approximately 95% at 40 years. However, 15–20% of cases became radioiodine refractory (RAI-R), and until now, no other treatments have been effective. The same problems are found in cases of poorly differentiated (PDTC) and anaplastic (ATC) thyroid cancers and in at least 30% of medullary thyroid cancer (MTC) cases, which are very aggressive and not sensitive to radioiodine. Tyrosine kinase inhibitors (TKIs) represent a new approach to the treatment of advanced cases of RAI-R DTC, MTC, PDTC, and, possibly, ATC. In the past 10 years, several TKIs have been tested for the treatment of advanced, progressive, and RAI-R thyroid tumors, and some of them have been recently approved for use in clinical practice: sorafenib and lenvatinib for DTC and PDTC and vandetanib and cabozantinib for MTC. The objective of this review is to present the current status of the treatment of advanced thyroid cancer with the use of innovative targeted therapies by describing both the benefits and the limits of their use based on the experiences reported so far. A comprehensive analysis and description of the molecular basis of these therapies, as well as new therapeutic perspectives, are reported. Some practical suggestions are given for both the choice of patients to be treated and their management, with particular regard to the potential side effects.
Introduction
Thyroid cancer is the most common endocrine malignancy, accounting for approximately 4% of all human malignancies (Hayat et al. 2007). The majority of thyroid tumors (85–90%), namely, differentiated thyroid cancer (DTCs), arise from follicular cells and are further classified as either papillary (PTCs, 75–80%) or follicular thyroid cancers (FTCs, 5–10%). Both anaplastic thyroid cancers (ATCs) and poorly DTCs (PDTCs) are also derived from follicular cells, and they represent 2–3% and 3–5% of all thyroid tumors, respectively. The remaining 1–2% of thyroid tumors originate from parafollicular C-cells and are classified as medullary thyroid cancers (MTCs) (Veiga et al. 2013).
The survival rates of patients affected by thyroid cancer are highly variable and depend on the histotype and the degree of differentiation. Rates are 95 and 80% after 35–40 years for PTC and FTC, respectively; 65% for MTC after 10 years; less than 20% for PDTC at 5 years; and less than 10% for ATC at 6 months after the initial diagnosis (Elisei & Pinchera 2012).
The cellular origin of the tumor has important implications for planning the therapeutic and follow-up strategies. In fact, tumor cells of both PTC and FTC are able to take up and organify iodine and to secrete thyroglobulin (Tg) under stimulus from thyrotropin-stimulating hormone (TSH). Because of the preservation of these properties, the majority of DTCs are curable via surgery and radioactive iodine (131I) therapy. Recurrences can be identified early and then cured by measuring the basal and/or TSH-stimulated serum Tg levels and via neck ultrasound (Pacini et al. 2001, Torlontano et al. 2006). However, in approximately 10% of cases, the patients have an advanced stage of the cancer at the time of diagnosis, with local invasion and/or distant metastases in the lungs (50%), bone (25%), lungs and bone (20%), and other sites (5%), and curing these cases with conventional therapeutic procedures is unlikely (Durante et al. 2006). In about one-third of advanced DTCs, the metastatic lesions have a very low avidity for iodine at the time of diagnosis, and 131I therapy has no effects. This is also what normally happens in cases of ATC and PDTC whose tumoral cells are so dedifferentiated compared to the follicular cells from which they originate that they are no longer able to take up iodine, secrete Tg, or respond to TSH stimulus. For these cases of ATC and PDTC, there is no rationale for the use of 131I, and, as an alternative, other conventional therapies, such as external beam radiotherapy (EBRT) and chemotherapy, have been unsuccessfully employed so far.
A similar approach has been used with advanced MTC tumors, which are not able to concentrate 131I because they are derived from parafollicular C-cells, which have a totally different embryological origin from follicular cells and are not involved in iodine metabolism, are not TSH dependent, and do not produce Tg. However, they do produce several other peptides, among which the most important and specific is calcitonin (Ct) (Pacini et al. 1991). Despite the different cellular origin that suggests that MTC should be unresponsive to RAI, a beneficial effect of RAI treatment has been described in vitro and in vivo in rat studies (Ott et al. 1987). The efficacy of this therapy for MTC was investigated several years ago also in humans, but the multiple studies performed showed contradictory results (Deftos & Stein 1980, Ott et al. 1987, Bayraktar et al. 1990). A recent controlled multicenter study concluded that RAI is not appropriate for the treatment of MTC and that the beneficial effect of RAI, if present, was due to the so-called bystander effect (Meijer et al. 2013).
Until recently, no effective therapeutic options have been available for patients with any type of advanced thyroid cancer. In fact, EBRT has significant toxicity and mainly plays a palliative role, and classical cytotoxic chemotherapies have shown disappointing efficacy. In fact, despite the relevant toxic effects of chemotherapy, studies have shown that they have only a transient and poor response rate (10–20%), with no prolongation of survival in response to the use of either a single therapeutic agent or in combination (De Besi et al. 1991, Orlandi et al. 1994).
Fortunately, in the last decade, an increase in our understanding of the molecular mechanisms underlying thyroid carcinogenesis and the first description of compounds able to inhibit the catalytic activity of a tyrosine kinase receptor involved in the process, resulting in antiproliferative effects, has opened up an era of targeted cancer therapies that represent new and important therapeutic options (Yaish et al. 1988).
The objective of this review is to present the current status of the treatment of advanced thyroid cancers using these innovative targeted therapies by describing both the benefits and the limits of their use based on the experiences reported so far.
Molecular alterations in thyroid cancer and the rationale for targeted therapies
In the past three decades, several molecular alterations have been described in tumors originating from follicular and parafollicular cells. In 1986, the first activated oncogene was found in the DNA extracted from an irradiated PTC tumor and transfected into a cell line (Fusco et al. 1995). After this report, several studies demonstrated the presence of the same activated oncogene in other instances of PTC and particularly in those related to radiation exposure (Nikiforov et al. 1997, Elisei et al. 2001). The oncogene of interest is RET, which lies on chromosome 10 and codes for a tyrosine kinase membrane receptor that is normally involved in cell proliferation and tumoral transformation via the mitogen-activated protein kinase (MAPK) pathway, also known as the Ras–Raf–MEK–ERK pathway (Fig. 1) (Arighi et al. 2005). The activation of the RET oncogene in PTC is due to a rearrangement (i.e., RET/PTC) of its intracellular tyrosine kinase region with a ubiquitous gene partner characterized by the presence of a coiled–coiled domain that determines the activation of RET in the follicular cells in which it is not usually expressed or is only expressed at a very low level (Tallini & Asa 2001). Several RET/PTC rearrangements (Table 1) have been described, almost exclusively in PTC cells. The exceptions are a few cases of leukemia and lung adenocarcinomas that show peculiar and exclusive RET/PTC rearrangements that have not yet been reported in PTC (Ballerini et al. 2012, Kohno et al. 2012, Lira et al. 2014, Nakaoku et al. 2014). The prevalence of RET/PTC-positive cases is approximately 20%, and it has apparently been decreasing in recent decades (Romei et al. 2012).
Different types of RET/PTC rearrangements in human tumors.
Oncogene | Donor gene | Chromosomal location | Tumor type |
---|---|---|---|
RET/PTC1 | CCDC6 | 10q21 | PTC, NSCLC, CRC |
RET/PTC2 | PRKAR1A | 17q23 | PTC |
RET/PTC3RET/PTC4 | NCOA4 | 10q11.2 | PTC, NSCLC, CRC |
RET/PTC5 | GOLGA5 | 14q | PTC |
RET/PTC6 | TRIM24 | 7q32-34 | PTC |
RET/PTC7 | TRIM33 | 1p13 | PTC, NSCLC |
ELKS-RET | ELKS | 12p13.3 | PTC |
RET/PTC8 | KTN1 | 14q22.1 | PTC |
RET/PTC9 | RFG9 | 18q21-22 | PTC |
PCM1-RET | PCM1 | 8p21-22 | PTC |
ΔRFP-RET | TRIM27 | 6p21 | PTC |
HOOK3-RET | HOOK3 | 8p11.21 | PTC |
ERC1-RET | ERC1 | 12p | PTC |
AKAP13-RET | AKAP13 | 15q24-25 | PTC |
TBL1XR1-RET | TBL1XR1 | 3q26.32 | PTC |
FKBP-RET | FKBP | 20p13 | PTC |
SPECC1L-RET | SPECC1L | 22q11.23 | PTC |
RET-ANK3 | ANK3 | 10q21 | PTC |
ACBD5/RET | ACBD5 | 10p12.1 | PTC |
MYH13-RET | MYH13 | 17p13.1 | MTC |
KIF5B-RET | KIF5B | 10p11.22 | NSCLC |
CUX1-RET | CUX1 | 7q22.1 | NSCLC |
KIAA1468-RET | KIAA1468 | 18q21.33 | NSCLC |
BCR-RET | BCR | 22q11.23 | CMML |
FGFR1OP-RET | FGFR1OP | 6q27 | CMML |
PTC, papillary thyroid cancer; MTC, medullary thyroid cancer; CRC, colorectal cancer; NSCLC, nonsmall cell lung cancer; CMML, chronic myelomonocytic leukemia.
Currently, the most frequent mutation found in PTC is BRAFV600E mutation, which is present in approximately 40% of cases and is being found at an increasing rate worldwide (Smyth et al. 2005, Mathur et al. 2011, Romei et al. 2012). The BRAFV600E mutation, which causes the constitutive activation of a serine/threonine kinase, was shown to be an initiating event for the disease and also to promote proliferation, tumorigenicity, and dedifferentiation processes through the activation of the MAPK pathway (Knauf et al. 2005, Liu et al. 2007). Moreover, BRAFV600E mutation seems to increase the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 alpha (HIF1α), and thus, targeting the product of the BRAFV600E mutation may have the dual effects of blocking the tumor’s progression and reducing tumor angiogenesis (Jo et al. 2006, Zerilli et al. 2010). Although still controversial, there is a general agreement that the BRAFV600E mutation is associated with more aggressive clinical–pathological features, loss of 131I avidity, and increased recurrence and mortality rates (Nikiforova et al. 2003, Elisei et al. 2008, Riesco-Eizaguirre et al. 2006, Xing et al. 2013, 2015).
H-, N-, and K-RAS-activating point mutations in specific hot spots at codons 12, 13, and 61 are also present in PTC cells, particularly in the follicular variant of PTC (Bhaijee & Nikiforov 2011). These mutations cause the loss of GTPase activity so that the kinase becomes constitutively activated. RAS mutations can activate two different pathways, namely, the MAPK and the phosphoinositide-3-(PI3K/AKT) pathways (Fig. 1), the latter being the preferential way for proliferation as demonstrated by a higher prevalence of AKT phosphorylation in RAS-mutated tumors (Xing 2013). Several other oncogenes have been found to be activated in PTC but with a much lower prevalence than those mentioned previously (Giordano et al. 2014). It is worth noting that these oncogene mutations are usually mutually exclusive, and only rarely are two or three present in the same tumoral tissue (Nikiforov 2011, Giordano et al. 2014).
Although H-, N-, and K-RAS mutations are found in a relatively small percentage of PTCs, they represent the most frequent genetic alterations in FTCs, and they are also significantly present in PDTCs and ATCs (Xing 2013). Other common genetic alterations in FTCs are PTEN deletion/mutation, paired box 8-peroxisome proliferator-activated receptor-gamma (PAX8/PPARg) rearrangement, and PIK3CA and IDH1 mutations. In PDTCs, beta-catenin (CTNNB1), p53, and BRAFV600E mutations can be also present. Although the most common oncogene alteration in ATCs are p53 point mutations, BRAFV600E, PIK3CA, PTEN, IDH1, and ALK mutations have all been reported in these aggressive thyroid tumors, in which, unlike in other hystotypes, it is not uncommon to have multiple genetic alterations in the same tumoral tissue (Eng et al. 1996, Smallridge et al. 2009, Soares et al. 2011).
The most common genetic alterations found in MTC cells are RET-activating point mutations. Unlike in follicular cells, parafollicular C-cells normally express the RET oncogene and a simple heterozygous nucleotide substitution can determine its constitutive activation in this cell lineage. Germline RET mutations are present in approximately 95% of hereditary forms of MTC, which represent approximately 25% of all MTCs, whereas somatic RET mutations (mainly M918T) are present in approximately 45% of sporadic cases, which represent the other 75% of the cases. Recently, RAS mutations, mainly H- and K-mutations, have been reported in approximately 17% of RET-negative sporadic MTCs (Ciampi et al. 2013). In addition, RAS and RET are mutually exclusive in MTC cases. A few anecdotal MTC cases harboring a RET or ALK rearrangement have been very recently described (Grubbs et al. 2015, Ji et al. 2015).
In addition to the above-mentioned molecular alterations, HGF, MET, and VEGF, as well as their receptors, are overexpressed in both MTCs and DTCs, and seem to play an important role in the pathogenesis, progression, and recurrence of these diseases (Papotti et al. 2000, Capp et al. 2010, Karaca et al. 2011, Koo et al. 2014).
Additional factors promoting thyroid cancer tumorigenesis are gene amplifications and copy number gains. These genetic abnormalities involve genes encoding tyrosine kinase receptors (TKRs), such as VEGFR, MET, EGFR, PDGFR, KIT, and PI3K/AKT pathway kinases, including PIK3CA, PIK3CB, 3-phosphoinositide-dependent protein kinase 1 (PDPK1), and AKT (Abubaker et al. 2008, Liu et al. 2008, Santarpia et al. 2008). The higher prevalence of these alterations in ATCs than in FTCs or PTCs and the consequent overexpression of the corresponding proteins are likely responsible of a more aggressive phenotype.
In addition to these genetic alterations, epigenetic abnormalities have been described to play an important role in human cancer and in thyroid cancer tumorigenesis, cell differentiation, and proliferation (Xing 2007). Oncogenes and the abnormal activation of the pathways involved in thyroid cancer tumorigenesis cause the aberrant methylation of thyroid-specific genes involved in iodine metabolism and/or of tumor suppressor genes, leading to the loss of radioiodine avidity and increased tumor growth, invasion, and metastasization. The activation of PI3K/AKT signaling through the aberrant methylation of PTEN leads to a self-amplifying loop that maintains the constitutive activation of PI3K/AKT signaling (Xing 2013).
Another important pathway that is linked to the tumorigenesis of thyroid cancer is the nuclear factor-kappa B (NF-κB) pathway. This pathway, which is physiologically involved in the inflammatory response, has recently been shown to control proliferative and antiapoptotic signaling in thyroid cancer cells (Xing 2013). Interestingly, the activation of this pathway seems to upregulate the same proteins that are overexpressed by the MAPK pathway, and the oncogenic mutations that act through the MAPK pathway (i.e., BRAFV600E, RET/PTC rearrangements, and RAS) cause the activation of NF-κB (Xing 2013). Targeting both pathways could have a synergistic effect on the proliferation of BRAFV600E-mutant cells (Xing 2013).
Despite the many genetic alterations that have been described for thyroid cancer and the most recent efforts to find other activated oncogenes, approximately 5–10% of PTCs, 50–60% of MTCs, and 10% of ATCs and PDTCs are still negative for all known genetic abnormalities (Soares et al. 2011, Giordano et al. 2014, Ji et al. 2015).
Targeted therapies: what are they and how do they act?
The increasing knowledge about the molecular alterations underlying thyroid cancer that has been obtained in the last decade has greatly increased the interest in developing new drugs for targeted treatments. The families of drugs that have primarily been investigated for the treatment of thyroid cancer are small molecules, namely, tyrosine kinase inhibitors (TKIs) and monoclonal antibodies (mAbs). Both families of drugs are able to bind to one or multiple RTKs, thus inhibiting their tyrosine kinase activity (Yaish et al. 1988). One of the main differences between TKIs and mAbs is that the latter are unable to penetrate the tumoral cell plasma membrane. For this reason, it is likely that mAbs would be more effective against circulating cancer cells than against solid tumors.
Of these two families, TKIs are the most extensively studied. Tyrosine kinases are enzymes responsible for the control of mitogenic signals via the phosphorylation/dephosphorylation of many intracellular proteins involved in the signal transduction cascade. The enhanced catalytic activity that is responsible for uncontrolled cell growth is the molecular rationale for the use of TKIs in thyroid cancer treatment. The first description of a drug able to inhibit the catalytic activity of RTKs and its potential use as an antiproliferative agent was demonstrated in 1988 (Yaish et al. 1988).
Since 2001, when the first TKI (imatinib) was approved for the treatment of chronic myelogenous leukemia, many of these drugs have been studied for the treatment of thyroid cancer (Druker et al. 2001). These drugs bind to different receptors with different affinities but share the same mechanism of action, namely, competitive ATP inhibition at the catalytic binding site of tyrosine kinase (Fig. 2). Almost all TKIs investigated in relation to thyroid cancer are multitarget drugs, with the exception of selumetinib, which binds to only one receptor (MEK) and, unlike the others, works by blocking tumor growth and acts via the reinduction of 131I uptake in dedifferentiated DTC cells (Table 2).
TKIs and their molecular targets.
Drug | VEGFR1 | VEGFR2 | VEGFR3 | c-KIT | RET | PDGFR | FGFR1-3 | EGFR | Others |
---|---|---|---|---|---|---|---|---|---|
Axitinib | + | − | + | + | − | + | − | − | |
BEZ235 | − | − | − | − | − | − | − | − | Dual PI3K/mTOR |
Cabozantinib | − | + | − | + | + | − | − | − | MET, KIF5B-RET rearrangement |
Imatinib | − | − | − | + | − | + | − | − | Bcr-Abl |
Lenvatinib | + | − | + | + | + | + | + | − | KIF5B-RET, CCDC6-RET, NcoA4-RET rearrangement |
Motesanib | + | − | + | + | + | + | − | − | |
Nintedanib | + | − | + | − | − | + | + | − | |
Pazopanib | + | − | + | + | − | + | − | − | |
Ponatinib | − | − | − | − | + | + | + | − | Bcr-Abl, FLT3, KIT |
Selumetinib | − | − | − | − | − | − | − | − | MEK |
Sorafenib | − | + | + | + | + | + | − | − | Raf, FLT3 |
Sunitinib | + | − | + | + | + | + | − | − | FLT3 |
Vandetanib | − | + | − | + | + | − | − | + | KIF5B-RET rearrangement |
Vemurafenib | − | − | − | − | − | − | − | − | BRAFV600E, CRAF |
Crizotinib | − | − | − | − | − | − | − | − | MET, ALK, ROS1 |
Bcr-Abl, Abelson and breakpoint cluster region fusion gene; EGFR, epidermal growth factor receptor; FGFR, fibroblast growth factor receptor; KIT, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene; Raf, v-raf murine sarcoma viral oncogene homolog; BRAFV600E, valine-to-glutamic acid substitution of BRAF gene; CRAF, v-raf murine sarcoma viral oncogene homolog 1; FLT3, Fms-like tyrosine kinase 3; MEK, mitogen-activated protein kinase; MET, hepatocyte growth factor receptor; PDGFR, platelet-derived growth factor receptor; RET, rearranged during transfection receptor; RET gene fusions, KIF5B-RET, CCDC6-RET, and NcoA4-RET; VEGFR1,2,3, vascular endothelial growth factor receptors 1, 2, and 3; ALK, anaplastic lymphoma kinase; ROS1, c-ros oncogene 1.
Due to the high prevalence of the BRAFV600E mutation in PTCs, drugs targeting the MAPK pathway, one component of which is a product of this gene, have been the most extensively studied. However, considering that the PI3K/AKT/mTOR pathway is another important pathway for the development of thyroid tumors, other drugs have also been investigated. The most studied group of drugs in relation to this pathway in thyroid cancer has been mammalian target of rapamycin (mTOR) inhibitors. More recently, due to the relative inefficacy of this group of compounds in treating solid tumors, a new drug (BEZ235) targeting both PI3K and mTOR has been developed (Lin et al. 2012). The possible synergistic effects of gene amplifications, copy number gains, PIK3CA alterations, and BRAF mutations suggest that a more effective result could be obtained by simultaneously targeting both the PI3K/AKT and MAPK pathways (Xing 2013).
Another important target that is often overactivated in DTC, MTC, and ATC is the NF-κB pathway. The inactivation of this pathway is now possible with a proteasome inhibitor, bortezomib, through a complex mechanism that prevents the degradation of a factor (i.e., inhibitory-kappa B) that normally inhibits this pathway. This drug is also able to induce programmed cell death by increasing the expression of tumor necrosis factor-related apoptosis-induced ligand that induces apoptosis through the activation of caspases (Gild et al. 2011, Xing 2013).
Angiogenesis is also a very important process in tumor progression and an attractive target for therapy (Xing 2013). Angiogenesis is promoted by VEGF, which is overexpressed in response to intratumoral hypoxia via the overactivation of HIF1α. This transcriptional factor is upregulated not only by hypoxia but also via growth factor signaling pathways, such as PI3K/AKT and MAPK, and is expressed in thyroid cancer cells, especially in ATC cells, but not in normal thyroid tissue (Burrows et al. 2010, Zerilli et al. 2010). An important target of HIF1α is the MET oncogene that is upregulated in many thyroid cancers and promotes angiogenesis as well as cellular motility, invasion, and metastasis (Rong et al. 1994, Ramirez et al. 2000, Scarpino et al. 2004). Almost all TKIs target VEGF receptor (VEGFR) with different affinities, but only one of them (i.e., cabozantinib) also blocks HIF1α signaling (Table 2).
Another promising drug that targets angiogenesis is combretastatin A-4 phosphate (CA4P), a microtubule-depolymerizing agent (Sosa et al. 2014). This drug is strictly related to colchicine, the classic tubulin-binding agent that was discovered to have a damaging effect on tumor vasculature in the 1930s. This vascular-targeting agent, also known as fosbretabulin, impairs tumoral vasculature function, reduces tumoral blood flow, and induces tumoral ischemic necrosis. CA4P exerts its activity via two mechanisms. First, it binds to tubulin dimers, interferes with microtubule polymerization, and induces mitotic arrest and apoptosis in endothelial cells. Second, it inhibits endothelial cell migration and capillary tube formation by disrupting the function of vascular endothelial-cadherin (VE-cadherin) that is an important determinant of microvascular integrity.
The role of mAbs in treating thyroid cancer is still uncertain. There are at least three ways of using this large, new, and varied family of drugs. The first is by using a mAb that is directed against tyrosine kinase receptors or their ligands, mainly VEGF (VEGF mAb) to block their function, thus exerting an antivascular effect (Bauer et al. 2002). The second is by using a mAb that is directed against an antigen expressed by the tumor (e.g. carcino embryonic antigen (CEA)) and is conjugated with a radioisotope (e.g. 131I, 90Y) as a way to reach the tumor cells and kill them with the radioactivity (Juweid et al. 2000). The third is by targeting and inhibiting specific receptors (e.g. CTLA-4, PD-1) with mAb (e.g. ipilimumab, pembrolizumab, etc.) to negatively regulate the immune response by promoting the immune system to attack tumor cells (Koguchi et al. 2015).
Clinical trials of targeted therapies in advanced, progressive thyroid cancer: the experiences of the last decade
After the discovery of in vitro evidence that some TKIs could interfere with thyroid cancer cell growth (Carlomagno et al. 2002, 2006, Salvatore et al. 2006), several clinical trials were designed (Table 3). The first international clinical trial started in 2005 and explored the efficacy of motesanib diphosphate (AMG706) on progressive, locally advanced or metastatic, radioiodine refractory (RAI-R) DTC (ClinicalTrials.gov identifier NCT00121628). Ninety-three patients with evidence of disease progression as assessed by the investigator, based on the Response Evaluation Criteria in Solid Tumors (RECIST), within the 6 months prior to the start of the drug study were treated with 125mg of AMG706, administered orally once daily (Sherman et al. 2008). At the end of the study, an objective response (OR) was achieved in 14% of the cases, a stable disease (SD) in 67% of the cases, and 8% of the cases had a progressive disease (PD) (Sherman et al. 2008). In 35% of the patients, a SD was obtained for 24 weeks or longer. The median progression-free survival (PFS) was estimated to be 40 weeks (Sherman et al. 2008). The same drug was investigated for the treatment of locally advanced or metastatic, progressive, or symptomatic MTC in a single-arm phase 2 study (ClinicalTrials.gov identifier, NCT00121628). A total of 91 patients were enrolled and treated with AMG706. Of these patients, only 2% achieved an OR, 81% had an SD (48% for 24 weeks or longer), and 8% had a PD; 9% of the patients were not able to be evaluated for a response. The median PFS was 48 weeks (Schlumberger et al. 2009). Although the rate of OR was rather low for both DTC and MTC, a significant proportion of patients achieved SD, and this can be considered a clinically beneficial outcome. Despite these promising results, the drug never reached the market. The main reasons for this are attributable to the relative inefficacy of the drug in treating other solid tumors and to the design of the studies. Both studies used a single-arm design were performed in a relatively small population of patients, and, particularly in case of the MTC trial, many patients were enrolled because they were symptomatic but did not have radiological evidence of disease progression. Moreover, the lack of a placebo arm makes the interpretations of the data from these studies regarding the drug’s efficacy and toxicity quite difficult.
Results of clinical trials with tyrosine kinase inhibitors in thyroid cancer patients.
Drug | Tumor | Phase | Patients (n) | PR (%) | SD>6 months (%) | Median PFS (months) | Median OS (months) | Most frequent AEs (%) (any grade) | References |
---|---|---|---|---|---|---|---|---|---|
Axitinib | MTCDTC | 2 | 52 | 35 | NE | 16.1 | 27.2 | Diarrhea (60) Hypertension (54) Fatigue (48) Anorexia (37) |
Locati et al. (2014) |
Motesanib | MTC | 2 | 91 | 2 | 48 | 12 | NE | Diarrhea (41) Fatigue (41) Hypothyroidism (29) Hypertension (27) Anorexia (27) |
Schlumberger et al. (2009) |
DTC | 2 | 93 | 14 | 35 | 10 | NE | Diarrhea (59) Hypertension (56) Fatigue (46) Weight loss (40) |
Sherman et al. (2008) | |
Vandetanib | MTC | 3 | 331 | 45 | 87 | NE | NE | Diarrhea (56) Skin rash (45) Nausea (33) Hypertension (32) |
Wells et al. (2012) |
DTC | 2 | 145 | 8 | 57 | 11.1 | NE | Diarrhea (74) Hypertension (34) Acneiform rash (27) Asthenia (26) Anorexia (26) |
Leboulleux et al. (2012) | |
Cabozantinib | MTC | 3 | 330 | 28 | NE | 11.2 | NE | Diarrhea (63) Hand-and-foot syndrome (50) Weight loss (47) Anorexia (45) Nausea (43) Fatigue (40) |
Elisei et al. (2013) |
Sorafenib | DTC | 3 | 417 | 12.2 | 42 | 10.8 | NE | Hand-and-foot syndrome (76) Diarrhea (68) Alopecia (67) Skin rash (50) Fatigue (49) Weight loss (46) Hypertension (40) Anorexia (31) |
Brose et al. (2013) |
ATC | 2 | 20 | 10 | 25 | 1.9 | 3.9 | Skin rash (65) Fatigue (60) Weight loss (60) Diarrhea (35) Hypertension (20) |
Savvides et al. (2013) | |
Lenvatinib | DTC | 3 | 392 | 64.8 | 29.8 | 18.3 | NE | Hypertension (67) Diarrhea (59) Fatigue or asthenia (59) Anorexia (50) Wight loss (46) Nausea (41) Stomatitis (35) |
Schlumberger et al. (2015) |
Fosbretabulin with Paclitaxel/Carboplatin | ATC | 2/3 | 80 | 20 | NE | 3.3 | 5.2 | Neutropenia (56) Anemia (34) Hypertension (33) Leukopenia (31) Fatigue (31) Alopecia (31) Diarrhea (23) |
Sosa et al. (2014) |
DTC, differentiated thyroid cancer; MTC, medullary thyroid cancer; ATC, anaplastic thyroid cancer; PR, partial response; SD, stable disease; PFS, progression-free survival; OS, overall survival; NE, not estimated.
Soon after the AMG706 study, an international, multicentric, phase 2 study examining the effect of axitinib (Table 3) on MTC and DTC was started (ClinicalTrials.gov identifier NCT00389441). Fifty-two cases of locally advanced, unresectable, or metastatic MTC or RAI-R DTC with disease progression demonstrated in the previous 12 months were treated with 5 mg axitinib, orally administered twice daily. At the end of the study, 35% had a PR and 35% had an SD for 16 weeks or longer. The median PFS was 16.1 months, and the median overall survival (OS) was 27.2 months (Locati et al. 2014). In this study, as in the previous studies, the single-arm design makes the interpretation of the results rather difficult and, although the data appeared encouraging, no further studies have been planned for this drug.
More recently, two new compounds, vandetanib and cabozantinib, have been investigated for the treatment of patients with advanced, unresectable, locally advanced or metastatic MTC in two phase 3 trials (ZETA and EXAM trials) (Table 3). In the first trial (ClinicalTrials.gov identifier NCT00410761), 331 patients were enrolled and randomly assigned to receive 300 mg of oral vandetanib once daily or a placebo (2:1). At the data cutoff point, after a median follow-up of 24 months, a significant PFS prolongation with vandetanib relative to the effect of the placebo (30.5 vs 19.3 months, respectively) was observed (hazard ratio (HR), 0.46; 95% CI, 0.31–0.69; P<0.001). Moreover, a statistically significant difference in the effect of vandetanib relative to that of the placebo was observed in the OR rates (P<0.001) and disease control rates (P=0.001), as well as for the biochemical response (P<0.001). Although there was apparently a better response to vandetanib in MTC patients with the somatic M918T RET mutation, MTC cases with no somatic RET mutation also showed a positive response (Wells et al. 2012). An OS analysis was not performed because at the end of the study, the data were still too preliminary to make any conclusions regarding the relative long-term survival of patients affected by MTC. These data allowed for the approval of vandetanib (Caprelsa, AstraZeneca) from the FDA (2011) and the EMA (2013) for use in the treatment of symptomatic or progressive, unresectable, locally advanced or metastatic MTC.
Currently, an international, multicentric phase 3 clinical trial (VERIFY trial) (ClinicalTrials.gov identifier NCT01876784) exploring the efficacy of vandetanib in treating RAI-R DTC is being conducted. The study was motivated by the positive results obtained in a phase 2 study performed in France (ClinicalTrials.gov identifier NCT00537095) in locally advanced or metastatic cases of RAI-R DTC (PTC, FTC, or PDTC). In this latter study, 72 patients were randomized to receive vandetanib (300 mg per day) or a placebo (1:1). At the end of the study, the patients in the vandetanib arm had a statistically significant increase in PFS compared to that of the placebo arm (11.1 vs 5.9 months for patients in the vandetanib and placebo arm, respectively; HR 0.63; 60% CI 0.54–0.74; one-sided P=0.008) (Leboulleux et al. 2012). As mentioned previously, vandetanib has been already approved for the treatment of advanced MTC, and this study might allow its use in treating RAI-R DTC as well if the data of the phase 2 study are confirmed. A limit of the VERIFY study is that to be enrolled, RAI-R DTC patients had to be ‘naïve’ of any other treatment; thus, the efficacy of vandetanib as second-line treatment cannot be evaluated.
The EXAM trial (ClinicalTrials.gov identifier NCT01908426) was also a double-blind, phase 3 trial comparing the safety and efficacy of cabozantinib to that of a placebo in treating advanced and progressive MTC patients (Table 3). Due to the positive results observed in the phase 1 study, the EXAM trial allowed for the inclusion of patients who had previously been treated with other TKIs (i.e., vandetanib, motesanib, sorafenib, etc.) (Kurzrock et al. 2011). A total of 330 MTC patients were randomly assigned to receive cabozantinib (140 mg orally) once daily or a placebo (2:1). In contrast with the ZETA trial, an important inclusion criterion was the documented, central radiographic progression of MTC per the RECIST. The OR rate was 28 and 0% for the cabozantinib and placebo treatments, respectively. The estimated median PFS was 11.2 and 4.0 months for the cabozantinib and placebo groups, respectively (HR, 0.28; 95% CI, 0.19–0.40; P<0.001). The longer median PFS durations observed in the ZETA trial with respect to that observed in the EXAM trial may be explained by the different levels of the severity of diseases in the two groups of patients, as demonstrated by the fact that the PFS durations of the two placebo groups were also significantly different (19.3 months in the ZETA trial and 4.0 months in the EXAM trial). In fact, although symptomatic patients without evidence of progressive disease were enrolled in the ZETA trial, the presence of disease progression was a fundamental inclusion criterion in the EXAM trial. In addition, in the EXAM trial, tumor response and PFS were both independent of RET mutation status, and more importantly, prolonged PFS with cabozantinib treatment was also observed in the subgroup of patients who had prior TKI treatment (Elisei et al. 2013). As for ZETA trial, OS was not improved in the patients treated with cabozantinib compared to those treated with the placebo. However, newly collected data show that OS was significantly increased in patients treated with cabozantinib when the analysis was restricted to the subgroup of M918T-RET-mutated MTC patients (Schlumberger et al. 2015b). The positive results in terms of the safety and efficacy of cabozantinib allowed for the approval of this drug (Cometriq, Exelixis, San Francisco, CA, USA) by the FDA in 2012 and the EMA in 2014 for the treatment of patients with progressive, metastatic MTC. At the time of this review, no other drugs are under clinical evaluation in phase 3 studies for the treatment of advanced MTC.
The inhibitory effect of sorafenib in the treatment of thyroid tumors has been recently explored in an international, multicentric, phase 3 study (DECISION trial) (ClinicalTrials.gov identifier NCT00984282) (Table 3), which included 417 patients affected by locally advanced/metastatic RAI-R DTC who were randomly assigned to receive sorafenib (400 mg orally) twice daily or a placebo (1:1) (Brose et al. 2014). Patients were allowed to enter the study only if they demonstrated disease progression per the RECIST within the previous 14 months and only if they had not been previously treated with other TKIs or chemotherapy. At the data cutoff point, patients treated with sorafenib had a longer and statistically significant difference in PFS relative to that of patients receiving the placebo (10.8 vs 5.8 months, respectively; HR, 0.587; 95% CI (0.45–0.76); P<0.0001). The median OS has not been reached yet and additional analyses of the OS are planned. However, the results will be affected by the large proportion of patients in the placebo arm (71%) who have crossed over to treatment. No complete response (CR) was observed, and PR was documented in 12.2% of the patients in the sorafenib arm and 0.5% in the placebo arm. The median duration of PR was 10.2 months. Valuable information has been obtained via the exploratory analysis of outcomes of patients receiving open-label sorafenib postprogression in the phase 3 DECISION trial (Schlumberger et al. 2014b). This analysis demonstrates that sorafenib may continue to suppress tumor growth rates following tumor progression because the median PFS of patients receiving this drug was still lower than that of patients treated with the placebo (6.7 vs 5.3 months). This suggests that, despite the evidence of tumor progression, in the absence of an alternative drug, it may be better to continue to treat the patients with sorafenib, especially if it is well tolerated. Moreover, this exploratory analysis also demonstrated that those patients in the placebo arm who started receiving sorafenib following tumor progression showed a comparable PFS to those who started receiving the drug from the beginning of the trial (9.6 vs 10.8 months). This indicates, although does not prove, that delaying the initiation of sorafenib treatment should not greatly affect the response to the drug (Schlumberger et al. 2014b). The positive results in terms of the safety and efficacy of sorafenib allowed for the approval of this drug (Nexavar, Bayer) by the FDA in 2013 and by the EMA in 2014 for the treatment of patients with RAI-R DTC. However, it is worth noting that sorafenib had already been approved by both the FDA and the EMA for the treatment of advanced hepatocellular carcinomas (Llovet et al. 2008) and advanced renal cell carcinomas (Escudier et al. 2007). For this reason, and based on the evidence of its clinical benefits obtained in previous phase 2 studies (Gupta-Abramson et al. 2008, Kloos et al. 2009), sorafenib can be used ‘off label’ in many countries, and several studies confirming the efficacy of the drug in treating RAI-R DTC have been already reported (Marotta et al. 2013, Pitoia 2014).
A phase 2 study exploring the efficacy of lenvatinib in treating RAI-R DTC shown to be progressing in the 12 months prior to the trial has been recently published (Cabanillas et al. 2015). Fifty-eight patients were enrolled and received 24 mg lenvatinib orally, once daily. A partial response (PR) was observed in 50% of the patients, and the median PFS was 12.6 months. It is worth noting that patients previously treated with a VEGFR-directed treatment had a higher rate of response in terms of PR than did naïve patients (59 vs 46%, respectively). The results were so promising that a phase 3, multicenter, randomized, placebo-controlled study with lenvatinib (SELECT trial) (ClinicalTrials.gov identifier NCT01321554) (Table 3) was immediately started. This trial was designed with the same therapeutic scheme, and 392 patients were randomized to receive either lenvatinib or the placebo (2:1). Patients were enrolled in the study based on cases of centrally assessed RAI-R disease and radiological evidence of disease progression within the 13 months prior to the study. In contrast to the DECISION trial, prior TKI treatment was not part of the exclusion criteria. At the end of the study, patients treated with lenvatinib had a longer and statistically significant difference in PFS relative to that of patients treated with a placebo (18.3 vs 3.6 months, respectively; HR 0.21; 99% CI (0.14–0.31); P<0.001) (Schlumberger et al. 2015c). A benefit in terms of PFS was present regardless of the BRAF or RAS mutation status of the patients or prior TKI treatment, and it was also observed in patients with PDTC. The overall response rates were more than 50% in all metastatic sites (brain, bone, liver, lungs, and lymph nodes), and a PFS benefit associated with lenvatinib was present in all cases independent of the sites of metastasis, with the exception of patients with brain metastases in which PFS fell to 8.8 months in patients treated with lenvatinib and to 3.7 months in those receiving the placebo (Habra et al. 2015). At the first data cutoff period, OS was not different in patients treated with lenvatinib than in those treated with the placebo, even if a higher rate of response was present in patients treated with lenvatinib, when the potential bias introduced by patient crossover was considered (Schlumberger et al. 2015c). More recently, a higher and statistically significant difference in the OS rate using a rank-preserving structural failure time model of patients treated with lenvatinib than in those treated with placebo in the SELECT study was reported at the European Cancer Congress (HR=0.53; 95% CI: 0.34–0.82, P=0.0051) (Guo et al. 2015). Moreover, when the OS analysis was performed on subgroups of patients, a statistically significant increase of OS was observed in patients >65 years (Brose et al. 2015) with respect to younger and in follicular with respect to papillary histotype (Elisei et al. 2015). Lenvatinib has also been tested in MTC patients in a phase 2 study from which very promising results have been recently published (Schlumberger et al. 2015a).
Another interesting drug that was investigated for the treatment of RAI-R, progressive, metastatic or unresectable PTC patients positive for the BRAFV600 mutation is vemurafenib (ClinicalTrials.gov identifier NCT01286753) (Table 3). In this open-label, exploratory, phase 2 study, 51 patients were assigned to two cohorts: patients naïve for TKI treatment (n=26) and patients previously treated with TKIs (n=25). Both cohorts were treated with oral vemurafenib (960 mg, twice daily). At the end of the study, no cases of CR were observed and PR was present in 35% of the TKI treatment-naïve cohort and in 26% of the TKI-treated cohort. The clinical benefit, namely, CR+PR+SD≥6 months, was present in 58% of the TKI treatment-naïve cohort and in 36% of the TKI-treated cohort. The median PFS was 15.6 months in the TKI treatment-naïve cohort and 6.8 months in the TKI-treated cohort (Brose et al. 2013). More recently, these results have been confirmed in an off-label study (Dadu et al. 2015).
A very interesting drug, selumetinib, has been recently evaluated for use in treating RAI-R thyroid cancer. This drug, in contrast to the other TKIs, acts as a highly selective uni-target therapy (Table 2) that does not work directly on tumor growth but has been demonstrated to induce or enhance 131I uptake and retention in a mouse model and in a subgroup of patients with thyroid cancer that was RAI-R (Chakravarty et al. 2011, Ho et al. 2013). The patients whose tumors were positive for RAS mutations showed a very good response, thus suggesting a major role for this drug in treating RAS-positive RAI-R DTCs. This study led to the design of a multicentric, international, phase 2 study (ASTRA trial) (ClinicalTrials.gov identifier NCT01843062) of selumetinib versus a placebo to improve the rate of thyroid remnant ablation in intermediate- to high-risk DTC patients. The ASTRA study is still enrolling DTC patients who have been recently submitted for total thyroidectomy and who have no gross residual disease but who have had a T3 or T4 thyroid tumor with at least five micro-lymph node metastases or one metastatic lymph node bigger than 1cm. The enrolment is expected to be completed in spring 2016. The use of selumetinib for the treatment of RAI-R metastatic disease is now being explored in two multicenter, phase 2 studies in North America. The first, which is investigating the possibility of treating patients with RAI-R PTC in terms of OR rate (CR and PR), is ongoing but no longer recruiting participants (ClinicalTrials.gov identifier NCT00559949). The second is a double-blind, phase 2 study of RAI comparing the use of selumetinib or a placebo for the treatment of RAI-avid recurrent or metastatic advanced (stage IV) DTC and PDTC (ClinicalTrials.gov identifier NCT02393690).
Another promising drug for the treatment of patients with advanced thyroid cancer is pazopanib. This drug, which targets VEGFR, PDGF, c-KIT, and other kinases, was investigated in 39 patients with RAI-R and rapidly progressing (in the 6-month period before enrolment) metastatic DTC in a phase 2 study (ClinicalTrials.gov identifier NCT00625846). Among the 37 patients who were able to be evaluated for a response, a PR was observed in 18 patients (49%), with a calculated response duration longer than 1 year in 66% (Bible et al. 2010).
The same authors investigated the efficacy of pazopanib in a cohort of patients with advanced and rapidly progressing (within 6 months) MTC. Among the 35 patients, a PR was obtained in 5 patients (14.3%) with a median PFS of 9.4 months and a median OS of 19.9 months (Bible et al. 2014). Despite the positive results of these phase 2 studies, no phase 3 trials are ongoing.
A phase 2 study of pazopanib as a monotherapy for ATC demonstrated only minimal clinical activity (Bible et al. 2012). More recently, an in vitro and in vivo study has reported a synergistic antitumor effect of combining pazopanib with a microtubule inhibitor, such as paclitaxel (Isham et al. 2013). Based on these findings, some efficacy may be demonstrated in a phase 2 study sponsored by the US National Cancer Institute (Bethesda, MD, USA) in which an intensity-modulated radiation therapy and the use of paclitaxel with or without pazopanib in ATC patients are being explored (ClinicalTrials.gov identifier NCT01236547).
Other than pazopanib, several other TKIs have been studied in the treatment of ATC but almost exclusively in phase 2 studies (Savvides et al. 2013). The major limit of these studies is that the rarity of ATC that does not allow trials to enroll large numbers of patients in a short period of time. A phase 2 trial in patients affected by ATC and treated with sorafenib was performed using the same scheme as the DECISION trial (Savvides et al. 2013). Among the 20 ATC patients enrolled, 10% experienced a PR and 25% SD according to RECIST. The duration of the response in the two patients with a PR was 10 and 27 months, whereas in the patients experiencing SD, the median duration of the response was 4 months, with a range of 3–11 months. The overall PFS was 1.9 months, with a median of 3.9 months and a 1-year survival rate of 20%. Interestingly, one of the patients who had a PR had previously shown disease progression while being treated with other antivascular agents (Savvides et al. 2013).
The biggest study on ATC was performed a few years ago using combretastatin (CA4P). In this open-label, multicentric, international study, 80 patients were randomly assigned in a 2:1 ratio to receive carboplatin/paclitaxel (CP) chemotherapy with or without CA4P. At the end of the study, the median OS was 5.2 months in the CP/CA4P arm (55) and 4.0 months in the CP arm (25). It was of interest that the 1-year survival rate was 26% for the CP/CA4P arm and 9% for the CP arm. Unfortunately, no statistically significant difference was observed in PFS time between the two arms. It is likely that if the population enrolled was larger, the primary objective of the study would have been reached, but unfortunately, ATC is a really rare disease and the number of patients enrolled was not enough to achieve the required statistical power (Sosa et al. 2014).
Interestingly, a remarkable response to crizotinib has been recently reported in a patient affected by ATC that presented an ALK rearrangement (Godbert et al. 2015). Despite this very interesting case, no clinical trials have yet been started.
The possibility of using mAbs to block ATC cells growth was investigated in a nude-mouse xenograft model using VEGF-mAbs. Despite the significant reduction of ATC growth, after more than 10 years of investigation, this treatment has not reached the clinical stage (Bauer et al. 2002). Since 1995, many studies have investigated the possibility of treating thyroid cancer patients with radioimmunotherapy (RIT). In particular, MTC patients were treated with mAbs directed against CEA and labeled with radioisotopes (Juweid et al. 1995, 1999). Moreover, the combination of RIT and doxorubicin has been shown to have a potential synergistic therapeutic effect, which may be due to a radiosensitizing effect of the chemotherapeutic agent (Behr et al. 1997). Nevertheless, there are no studies showing any actual clinical benefit of these types of therapies.
More recently, a phase 1 study has investigated the toxicity and therapeutic potential of an anti-CEA-mAb combined with autologous hematopoietic stem cell rescue in patients with rapidly progressing metastatic MTC and showed promising results (Juweid et al. 2000). A phase 1/2 study with a high-dose 90Y-labeled, humanized mAbs alone or in combination with doxorubicin and peripheral blood stem cell rescue has been recently completed, but the results are not yet available (ClinicalTrials.gov identifier NCT00004048). Despite these efforts, in the absence of a large, randomized, phase-3 study to compare the efficacy and toxicity of RIT with a placebo or TKI treatment, the benefits of this type treatment remain unclear.
The results of all these clinical trials are summarized in Table 3. Numerous additional clinical trials on the same and on different drugs that are still ongoing and/or actively recruiting participants are summarized in Table 4.
Clinical trials still ongoing and/or actively recruiting participants.
Drug | Tumor | Study type | Recruitment status | ClinicalTrials.gov identifier |
---|---|---|---|---|
Selumetinib | PTC | Phase 2 | Active, not recruiting | NCT00559949 |
DTC, PDTC | Phase 2 | Recruiting | NCT02393690 | |
DTC | Phase 3 | Recruiting | NCT01843062 | |
Everolimus | DTC | Phase 2 | Active, not recruiting | NCT00936858 |
Everolimus and Vatalanib | MTC | Phase 1 | Active, not recruiting | NCT00655655 |
Lenvatinib | DTC | Phase 1/2 | Recruiting | NCT02432274 |
DTC, PDTC, ATC, MTC | Observational | Recruiting | NCT02430714 | |
DTC | Phase 4 | Recruiting | NCT02211222 | |
Cabozantinib | MTC | Phase 4 | Recruiting | NCT01896479 |
Vandetanib | MTC | Phase 4 | Active, not recruiting | NCT01496313 |
PTC, papillary thyroid cancer; DTC, differentiated thyroid cancer; PDTC, poorly differentiated thyroid cancer; MTC, medullary thyroid cancer; ATC, anaplastic thyroid cancer.
Pitfalls of targeted therapies
The escape phenomenon
To date, the main limitation of targeted therapy is the fact that after a variable period of time from the beginning of the treatment, cancer cells start to grow again, likely due to the development of an escape mechanism. This phenomenon is almost always present, independent of the type of TKI used and the type of human tumor treated. The most likely explanation is that the tumoral cells develop a mechanism of resistance to the treatment (Wang et al. 2009, Arao et al. 2011, Finke et al. 2011).
There are two main types of resistance: a ‘primary’ resistance that is intrinsic to the tumor cells and is present before the treatment has been started and a ‘secondary’ resistance that develops after a certain period of exposure to the TKI. An example of primary resistance in MTCs is represented by the V804M-RET mutation that confers resistance to vandetanib treatment in vitro by preventing the binding of the drug to the receptor (Carlomagno et al. 2004). An example of secondary resistance in thyroid tumors treated with TKIs is still unknown, but it is possible that secondary site mutations could develop during therapy. These new mutations have been demonstrated in lung adenocarcinoma cells treated with TKIs and can be represented by MET amplification or by RAS, BRAF, and PIK3CA gene mutations (Ohashi et al. 2012). Usually, they are located downstream from the TKI target or in parallel pathways and result in a mechanism to bypass the action of the drug.
The challenge is to find the mutations or other alterations that determine the resistance to the drug because patients should not be further treated with the same drug once the efficacy of the TKI is lost and should be treated with other drugs that specifically target the new alterations. To accomplish this, tissue samples should be collected in clinical trials both before starting the TKI treatment and during the treatment when the escape phenomenon arises (Bible et al. 2015). Supporting this concept are the results obtained by studying the mechanisms of resistance to imatinib in patients affected by chronic myeloid leukemia. After the discovery of imatinib resistance mutations, new drugs, such as dasatinib, nilotinib, and bosutinib, have been developed for use as second- and third-line therapies for use in treating CML patients after the development of escape from imatinib (Stein & Smith 2010, Amsberg & Koschmieder 2013). This type of research and its findings are also important for RAI-R DTC patients who become resistant to sorafenib, vandetanib, or lenvatinib (Isham et al. 2014).
The cytostatic action
All TKIs act as cytostatic and not as cytotoxic agents, implying that tumoral cells are not killed but made quiescent and nonproliferative. This is also the reason why CR, with very few exceptions (Schlumberger et al. 2015c), has never been reported in clinical trials employing TKIs. It is also why the shrinkages of the tumoral masses observed in many cases and the findings of a significant PR are mainly due to the antiangiogenic actions and the partial ischemia of the tumoral core. The cytostatic action represents a limit because, although clinically satisfactory, once started, TKIs should be continued indefinitely until evidence of tumoral progression appears or until the appearance of severe side effects. Moreover, there is also some evidence that once the TKI treatment is stopped, the progression of the disease can become even more rapid. This is of particular relevance in patients, either women or men, who intend to have children. These patients should stop taking the drug for at least 12–14 and 3–6 months, respectively. There is not yet sufficient evidence to predict how much an interruption of TKI treatment of this length affects the disease progression or if it can make the progression even worse because of recovery by the tumor from the cytostatic action. For this specific problem (i.e., having children), the preservation of gametes before starting the TKI treatment should be taken into consideration. However, this would still not be enough to avoid drug interruption in women.
Drug toxicities and side effects
Despite the different receptors targeted, the vast majority of TKI-related adverse events (AEs) are common to different drugs. The most frequent AEs are diarrhea, anorexia, weight loss, fatigue, hypertension, hypothyroidism, hand-and-foot syndrome, and skin rash. The most frequent drug-related AEs for the different drugs investigated in the largest clinical trials are summarized in Table 5. The majority of these AEs are generally mild or moderate (G1–G2), and only in less than 5–10% of cases are they severe or life threatening (G3–G4), based on the common terminology criteria for adverse events (Shah et al. 2013). Death related to AEs (G5) is, fortunately, a very rare event.
Prevalence of TKI-related adverse events in different studies.
Drug | Tumor | Phase | Adverse events | References | |||||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Hypertension (%) | Diarrhea (%) | Skinrash (%) | Anorexia (%) | Nausea (%) | Weight loss (%) | Fatigue (%) | QTcprolangation (%) | ||||||||||||
G1–2 | G3–4 | G1–2 | G3–4 | G1–2 | G3–4 | G1–2 | G3–4 | G1–2 | G3–4 | G1–2 | G3–4 | G1–2 | G3–4 | G1–2 | G3–4 | ||||
Axitinib | MTC DTC |
II | 48 | 6 | 50 | 10 | NE | NE | 31 | 6 | 27 | 0 | 25 | 10 | 36 | 12 | NE | NE | Locati et al. (2014) |
Motesanib | DTC | II | 31 | 25 | 46 | 13 | NE | NE | 23 | 4 | 26 | 2 | 35 | 5 | 42 | 4 | NE | NE | Sherman et al. (2008) |
MTC | II | 17 | 10 | 28 | 13 | NE | NE | 24 | 3 | 24 | 2 | 20 | 2 | 33 | 8 | NE | NE | Schlumberger et al. (2009) | |
Vandetanib | MTC | III | 23 | 9 | 45 | 11 | 41 | 4 | 17 | 4 | 33 | 0 | 10 | 0 | 18 | 6 | 6 | 8 | Wells et al. (2012) |
DTC | II | 34 | 0 | 64 | 10 | 25 | 0 | 25 | 1 | 25 | 0 | NE | NE | 18 | 5 | 9 | 14 | Leboulleux et al. (2012) | |
Cabozantinib | MTC | III | 24 | 8 | 48 | 15 | 18 | 1 | 41 | 4 | 42 | 1 | 43 | 4 | 31 | 9 | NE | NE | Elisei et al. (2013) |
Sorafenib | ATC | II | 20 | 0 | 35 | 0 | 50 | 15 | 5 | 5 | 25 | 0 | 55 | 5 | 55 | 5 | NE | NE | Savvides et al. (2013) |
DTC | III | 31 | 9 | 63 | 5 | 45 | 5 | 30 | 2 | 20 | 0 | 41 | 6 | 44 | 6 | NE | NE | Brose et al. (2015) | |
Lenvatinib | DTC | III | 26 | 41 | 51 | 8 | 15 | <1 | 44 | 5 | 39 | 2 | 36 | 9 | 50 | 9 | 7 | 1 | Schlumberger et al. (2015c) |
Combretastatin | ATC | II/III | 25 | 4 | 19 | 2 | NE | NE | 11 | 0 | 23 | 0 | NE | NE | 27 | 2 | 7 | 4 | Sosa et al. (2014) |
MTC, medullary thyroid cancer; DTC, differentiated thyroid cancer; ATC, anaplastic thyroid cancer; NE, not estimated.
The majority of AEs are easily prevented or managed with drug treatment, but in a nonnegligible percentage of cases, dose reduction (up to 79% for cabozantinib), interruption (up to 66.2% for sorafenib), or withdrawal (up to 26% for lenvatinib) was needed in clinical trials (Brose 2013, Elisei et al. 2013, Cabanillas et al. 2015).
Because of this, after drug approval, a number of phase 4 studies were designed to evaluate the efficacy/tolerability of the drugs at lower doses, which can be better tolerated, and to determine if the same results on the disease progression are observed (vandetanib: NCT01496313; cabozantinib: NCT01896479; lenvatinib NCT02211222) (Table 4). From a practical point of view, side effects should be known and prevented by both patients and doctors to avoid the need for and the risk of unnecessarily stopping the drug treatment. Some practical suggestions are given in the ‘Management of thyroid cancer patients undergoing TKI treatment: practical suggestions’ section. In particular, patients should be instructed to report any types of side effects as soon as they appear to allow doctors to immediately start a therapeutic strategy.
According to some observations, TKI toxicities could also be used as a surrogate marker of the efficacy of the drug. TKI toxicities have recently been classified as being an ‘on-target’ toxicity (On-TT) and an ‘off-target’ toxicity (Off-TT) (Shah et al. 2013). An On-TT is a primary effect of the drug that occurs because of a common pathway/target among neoplastic and normal cells, whereas an Off-TT is a secondary effect of the drug that is due to the inhibition of the kinases that are not the intended target of the drug. A paradigmatic example is the association between TKI-induced hypertension and drug efficacy in terms of OR, PFS, and OS outcomes in patients treated with sunitinib and sorafenib for renal cell carcinoma and hepatocellular carcinoma (Rini et al. 2011, Estfan et al. 2013). In cases of thyroid cancer, this phenomenon has been recently reported for hypertension in patients treated with lenvatinib (Choi et al. 2015).
Future steps: targeting different pathways and combined therapies
A genotype-directed therapy is the goal of targeted therapy; however, due to the cross talk between signaling pathways, it is unlikely that treatment with a single TKI or targeted drug will be effective. In fact, blocking a single pathway has been shown to lead to the activation of other pathways that then overcome the drug’s effects (Bernards 2012). Moreover, it is likely that concurrent and/or subsequent genetic molecular alterations drive tumor growth, invasion, and metastases simultaneously or at different times through different pathways in aggressive tumors. This hypothesis is consistent with several studies, showing that more aggressive thyroid cancers such as PDTCs and ATCs often show more than one gene alteration (Sobrinho-Simoes et al. 2008, Smallridge et al. 2009). Nevertheless, dual inhibition of the MAPK and mTOR pathways or the MEK and mTOR pathways has shown a strong inhibitory synergism in thyroid cancer cell lines, including those from ATC (Jin et al. 2009, Liu et al. 2010). Similarly, a synergistic effect was demonstrated through the combination of a dual inhibitor of PI3K/mTOR (BEZ235) and an RAF inhibitor (RAF265) in MTC and DTC cell lines both in vitro and in a mouse xenograft model (Jin et al. 2011). Another way of overcoming drug resistance mechanism and/or enhancing drug efficacy is the combination of TKIs or other target agents with chemotherapy or radiotherapy. In fact, it has been demonstrated that treatment with BEZ235 and paclitaxel, imatinib and docetaxel, and efatutazone, a PPARγ agonist, and paclitaxel have synergistic effects in vitro compared to the effect of treatment with a single agent (Copland et al. 2006, Kim et al. 2012, Lin et al. 2012). These combinations were demonstrated to be safe and tolerable in ATC patients in a phase 1 trial (Smallridge et al. 2013). Similarly, it was demonstrated that an mTOR inhibitor (everolimus) enhances the response to cytotoxic chemotherapy (doxorubicin) and EBRT in PTC (Lin et al. 2010).
Which thyroid tumors are good candidates for targeted therapies?
In general, good candidates for targeted therapies are those thyroid tumors that are defined as RAI-R according to well-defined criteria (Schlumberger et al. 2014a). This group generally includes approximately 66% of DTCs that have distant metastases at the time of diagnosis, which cannot be cured with 131I because they were initially or became RAI-R over time. RAI-R diseases are more frequent in older patients because of the presence of macroscopic metastatic diseases and/or a PDTC. It is worth noting that RAI-R thyroid tumors generally show an 18FDG-PET-positive scan and that, independent of the ability of metastatic lesions to take up 131I, 18FDG-PET-positive metastatic patients have a very lower survival rate (Robbins et al. 2006) and they are very good candidates for targeted therapies.
The group of good candidates for targeted therapies also includes the vast majority of ATC and PDTC cases that have locally advanced disease and distant metastases at the time of presentation because, despite the fact that these tumors originate from follicular cells, the complete lack of cell differentiation makes the disease unresponsive or scarcely responsive to 131I treatment and also to other conventional treatments such as ERBT and chemotherapy.
Approximately 30% of patients with MTC, particularly those with a somatic RET mutation and those with distant metastases at diagnosis, have a survival time that is not longer than 5–10 years (Gharib et al. 1992). These MTC patients are good candidates for targeted therapies because no other therapies are available at the present time (Orlandi et al. 1994).
Although necessary, the RAI-R feature is not sufficient per se to determine if a thyroid tumor is a good candidate for targeted therapy. It is well known that, with the exception of ATC, thyroid cancer grows slowly, especially DTC and MTC. Patients with these cancers often survive for a long time, and in many cases, the quality of their life is very good. For this reason, only patients with an advanced and progressive disease should undergo a treatment with targeted therapies. According to expert opinion (Schlumberger et al. 2014a), good candidates are patients with a RAI-R metastatic thyroid tumor with lesions that are radiologically measurable and have been in progression over the previous 12–14 months, as defined by the RECIST (Eisenhauer et al. 2009). Due to the presence of two specific serum markers in thyroid cancer that could correlate with the tumor burden, namely, calcitonin (Ct) for MTC and Tg for DTC, many researchers have investigated their doubling time to assess the progression of the disease (Laure Giraudet et al. 2008, Miyauchi et al. 2011). Despite the reliability of these markers, progressive diseases must be assessed or confirmed with standardized imaging, which should be repeated every 6–12 months, and the rate of progression should be calculated using the RECIST (Eisenhauer et al. 2009). Only a few exceptions to this general rule are accepted and include cases with a large tumor burden, the presence of the disease in sites where its progression can be harmful (e.g. trachea, spinal cord, brain), or a high level of 18-FDG uptake (Deandreis et al. 2011, Cabanillas & Sherman 2012). Other exceptions can be made for symptomatic patients and patients with paraneoplastic diseases, such as Cushing’s syndrome in those with MTC. In fact, several reports have demonstrated the utility of TKIs for hypercortisolism control. The fact that several different TKIs can induce this positive effect demonstrates that this is a class effect (Baudry et al. 2013, Fox et al. 2013, Barroso-Sousa et al. 2014, Marques et al. 2015, Pitoia et al. 2015).
Another very important consideration is that, in any patient, before starting a targeted therapy, the possibility of controlling the disease via local treatments (e.g. surgical treatment, chemoembolization, radiofrequency) should be considered, especially if the number of metastatic lesions is relatively small (Schlumberger et al. 2012, 2014a).
Management of thyroid cancer patients undergoing TKI treatment: practical suggestions
Once the decision to start the TKI treatment has been made, the patient should be evaluated for any concomitant diseases and treated for these if necessary. Frequent clinical and biochemical control tests should be performed, especially during the first weeks of treatment. When using TKIs that can result in hand-and-foot syndrome, the use of urea-based cream, as well as comfortable shoes and gloves, should be suggested during the winter for the care of the patient’s hands and feet. Patients treated with vandetanib or cabozantinib should be advised to avoid exposing their skin to aggressive soaps and direct sunlight. Diarrhea should be controlled with the use of loperamid, and attention should be paid to identifying and avoiding those aliments that worsen the symptoms. A supportive multivitamin drug can also be suggested. Many patients become anorexic and lose weight, sometimes very significantly. The use of Megace and/or steroids can be suggested to improve the appetite and regain weight. However, in some cases, side effects such as fatigue and severe anorexia cannot be treated with any drug, and only the reduction of the daily dose of the TKI can control the symptoms. The reduction of the daily dose often allows for a good compromise between the possibility of continuing the treatment and the tolerability of the side effects. As a matter of fact, in almost all phase 3 studies, the median dose used by patients was lower than the starting dose, and phase 4 studies aimed at determining if lower doses are still effective are ongoing for several TKIs. From a practical point of view, it is important that patients are aware of the risk they run if they stop the drug (i.e., a rapid increase in the rate of cell growth) because sometimes they spontaneously decide to stop the drug to better manage the side effects. Patients should be instructed to report any side effects as earlier as possible to allow the doctors to immediately take care of them. Providing good information and good communication with doctors and/or dedicated nurses is the best way to help patients during this therapeutic program (Worden et al. 2015).
Conclusions and future perspectives
TKIs represent a new and promising approach to the treatment of advanced RAI-R DTC, MTC, and likely also PDTC and ATC. At the present time, sorafenib and lenvatinib have been approved by the FDA and the EMA for the treatment of progressive RAI-DTC, and vandetanib and cabozantinib have been approved for treating advanced and progressive MTC. After 10 years, patients with these diseases, who have been without any therapeutic options until now, finally have options for slowing tumor growth and better controlling some symptoms.
However, many areas of uncertainty in the treatment of thyroid cancer patients with targeted therapies remain to be elucidated. Despite the efficacy of TKIs demonstrated in many phase 3 studies in terms of PFS, only a limited amount of data regarding the actual prolongation of patient survival are available at the moment. In addition, these drugs have a nonnegligible toxicity that has an impact on the patients’ quality of life; for this reason, determining the right time to start these treatments represents a goal for future clinical research. We still do not know if these diseases should be treated at the very early stages or when the tumor burden becomes significant. While waiting for a better clarification of this issue, only advanced and progressive diseases should be treated with TKIs. Comparative and sequential clinical studies will tell us which treatment strategies are the best to begin with and which are the best for a sequential treatment regimen or if it is better to combine different targeted therapies, possibly at lower doses to decrease toxicities and at the same time increase efficacy. The use of new drugs when the escape phenomenon occurs is necessary, and molecular studies of metastatic and growing tumoral tissues during the treatment will be fundamental. We still do not know if the molecular signature of the primary tumor could help determine the choice of the most appropriate drug; indeed, the results of the phase 3 studies do not strongly support the hypothesis that patients with molecular alterations in genes considered as the main targets of these TKIs (e.g. M918T-RET for vandetanib or cabozantinib, and BRAFV600E for sorafenib) respond to treatment better than those without the mutations. Cross-resistance between drugs should be discovered to avoid unnecessary, useless, and harmful treatments. The associations between different TKIs, between these drugs and innovative compounds, and their association with classical cytotoxic chemotherapy and EBRT should be carefully evaluated in preclinical models as well as in clinical trials. Further understanding of the molecular basis of cancer growth, invasion, and metastasization, and the mechanisms through which resistance develops will hopefully lead to the development of new targeted drugs that could selectively and definitely kill cancer cells and spare normal cells or at least lower drug toxicity to ameliorate the patient’s quality of life.
Declaration of interest
Rossella Elisei is a consultant and/or keynote speaker for Exelixis/Sobi, AstraZeneca, Genzyme, Bayer, and Eisai. D Viola is a consultant and/or keynote speaker for Sobi and Genzyme. E Molinaro and V Bottici have been keynote speakers for Genzyme. The other coauthors have nothing to disclose. Nevertheless, this review is an unconditioned and independent work reporting published data and reflecting personal experience.
Funding
This research did not receive any specific grant from any funding agency in the public, commercial, or not-for-profit sector.
References
Abubaker J, Jehan Z, Bavi P, Sultana M, Al-Harbi S, Ibrahim M, Al-Nuaim A, Ahmed M, Amin T & Al-Fehaily M et al. 2008 Clinicopathological analysis of papillary thyroid cancer with PIK3CA alterations in a Middle Eastern population. Journal of Clinical Endocrinology and Metabolism 93 611–618.
Amsberg GK & Koschmieder S 2013 Profile of bosutinib and its clinical potential in the treatment of chronic myeloid leukemia. OncoTargets and Therapy 6 99–106 (doi:10.2147/OTT.S19901).
Arao T, Matsumoto K, Furuta K, Kudo K, Kaneda H, Nagai T, Sakai K, Fujita Y, Tamura D & Aomatsu K et al. 2011 Acquired drug resistance to vascular endothelial growth factor receptor 2 tyrosine kinase inhibitor in human vascular endothelial cells. Anticancer Research 31 2787–2796.
Arighi E, Borrello MG & Sariola H 2005 RET tyrosine kinase signaling in development and cancer. Cytokine and Growth Factor Reviews 16 441–467. (doi:10.1016/j.cytogfr.2005.05.010)
Ballerini P, Struski S, Cresson C, Prade N, Toujani S, Deswarte C, Dobbelstein S, Petit A, Lapillonne H & Gautier EF et al. 2012 RET fusion genes are associated with chronic myelomonocytic leukemia and enhance monocytic differentiation. Leukemia 26 2384–2389 (doi:10.1038/leu.2012.109).
Barroso-Sousa R, Lerario AM, Evangelista J, Papadia C, Lourenco DM Jr., Lin CS, Kulcsar MA, Fragoso MC & Hoff AO 2014 Complete resolution of hypercortisolism with sorafenib in a patient with advanced medullary thyroid carcinoma and ectopic ACTH (adrenocorticotropic hormone) syndrome. Thyroid 24 1062–1066 (doi:10.1089/thy.2013.0571).
Baudry C, Paepegaey AC & Groussin L 2013 Reversal of Cushing's syndrome by vandetanib in medullary thyroid carcinoma. New England Journal of Medicine 369 584–586. (doi:10.1056/NEJMc1301428)
Bauer AJ, Terrell R, Doniparthi NK, Patel A, Tuttle RM, Saji M, Ringel MD & Francis GL 2002 Vascular endothelial growth factor monoclonal antibody inhibits growth of anaplastic thyroid cancer xenografts in nude mice. Thyroid 12 953–961. (doi:10.1089/105072502320908286)
Bayraktar M, Gedik O, Akalin S, Usman A, Adalar N & Telatar F 1990 The effect of radioactive iodine treatment on thyroid C cells. Clinical Endocrinology 33 625–630.
Behr TM, Wulst E, Radetzky S, Blumenthal RD, Dunn RM, Gratz S, Rave-Frank M, Schmidberger H, Raue F & Becker W 1997 Improved treatment of medullary thyroid cancer in a nude mouse model by combined radioimmunochemotherapy: doxorubicin potentiates the therapeutic efficacy of radiolabeled antibodies in a radioresistant tumor type. Cancer Research 57 5309–5319.
Bernards R 2012 A missing link in genotype-directed cancer therapy. Cell 151 465–468 (doi:10.1016/j.cell.2012.10.014).
Bhaijee F & Nikiforov YE 2011 Molecular analysis of thyroid tumors. Endocrine Pathology 22 126–133. (doi:10.1007/s12022-011-9170-y)
Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, Rubin J, Sideras K, Morris JC 3rd & McIver B et al. 2010 Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study. Lancet Oncology 11 962–972 (doi:10.1016/S1470-2045(10)70203-5).
Bible KC, Suman VJ, Menefee ME, Smallridge RC, Molina JR, Maples WJ, Karlin NJ, Traynor AM, Kumar P & Goh BC et al. 2012 A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. Journal of Clinical Endocrinology and Metabolism 97 3179–3184 (doi:10.1210/jc.2012-1520).
Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, Rubin J, Karlin N, Sideras K & Morris JC 3rd et al. 2014 A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: Mbib57H. Journal of Clinical Endocrinology and Metabolism 99 1687–1693 (doi:10.1210/jc.2013-3713).
Bible KC, Cote GJ, Demeure MJ, Elisei R, Jhiang S & Ringel MD 2015 Correlative studies in clinical trials: a position statement from the International Thyroid Oncology Group. Journal of Clinical Endocrinology and Metabolism 100 4387–4395 (doi:10.1210/jc.2015-2818).
Brose MS 2013 Sorafenib in locally advanced or metastatic patients with radioactive iodine-refractory differentiated thyroid cancer: the phase III DECISION trial. Journal of Clinical Oncology (abstract 4).
Brose M, Cabanillas M, Cohen E, Wirth L, Sherman S, Riehl T, Yue H & Sherman E 2013 An open-label, multi-center phase 2 study of the BRAF inhibitor vemurafenib in patients with metastatic or unresectable papillary thyroid cancer (ptc) positive for the BRAF V600 mutation and resistant to radioactive iodine. Abstract LBA28 presented at the European Cancer Congress, September 27–October 1 2013, Amsterdam, The Netherlands. Philadelphia, PA, USA: Elsevier.
Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, de la Fouchardiere C, Pacini F, Paschke R & Shong YK et al. 2014 Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet 384 319–328. (doi:10.1016/S0140-6736(14)60421-9)
Brose MS, Teng A, Rietschel P & Habra MA 2015 Lenvatinib and the effect of age on overall survival for patients with radioiodine-refractory differentiated thyroid cancer. Abstract 731 presented at the 15th International Thyroid Congress, 18–23 October 2015, Orlando, Florida, USA. Thyroid 25 (supplement 1) A-290 abstract 731. (doi:10.1089/thy.2015.29004.abstracts)
Burrows N, Resch J, Cowen RL, von Wasielewski R, Hoang-Vu C, West CM, Williams KJ & Brabant G 2010 Expression of hypoxia-inducible factor 1 alpha in thyroid carcinomas. Endocrine-Related Cancer 17 61–72. (doi:10.1677/ERC-08-0251)
Cabanillas ME, Schlumberger M, Jarzab B, Martins RG, Pacini F, Robinson B, McCaffrey JC, Shah MH, Bodenner DL & Topliss D et al. 2015 A phase 2 trial of lenvatinib (E7080) in advanced, progressive, radioiodine-refractory, differentiated thyroid cancer: a clinical outcomes and biomarker assessment. Cancer 121 2749–2756. (doi:10.1002/cncr.29395)
Cabanillas ME & Sherman SI 2012 Applying new clinicopathological characteristics to prognostication in advanced thyroid carcinoma. Endocrine-Related Cancer 19 C19–C22. (doi:10.1530/ERC-11-0371)
Capp C, Wajner SM, Siqueira DR, Brasil BA, Meurer L & Maia AL 2010 Increased expression of vascular endothelial growth factor and its receptors, VEGFR-1 and VEGFR-2, in medullary thyroid carcinoma. Thyroid 20 863–871. (doi:10.1089/thy.2009.0417)
Carlomagno F, Vitagliano D, Guida T, Ciardiello F, Tortora G, Vecchio G, Ryan AJ, Fontanini G, Fusco A & Santoro M 2002 ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Research 62 7284–7290.
Carlomagno F, Guida T, Anaganti S, Vecchio G, Fusco A, Ryan AJ, Billaud M & Santoro M 2004 Disease associated mutations at valine 804 in the RET receptor tyrosine kinase confer resistance to selective kinase inhibitors. Oncogene 23 6056–6063. (doi:10.1038/sj.onc.1207810)
Carlomagno F, Anaganti S, Guida T, Salvatore G, Troncone G, Wilhelm SM & Santoro M 2006 BAY 43-9006 inhibition of oncogenic RET mutants. Journal of National Cancer Institute 98 326–334. (doi:10.1093/jnci/djj069)
Chakravarty D, Santos E, Ryder M, Knauf JA, Liao XH, West BL, Bollag G, Kolesnick R, Thin TH & Rosen N et al. 2011 Small-molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. Journal of Clinical Investigation 121 4700–4711. (doi:10.1172/JCI46382)
Choi J, Abouzaid X, Li X & Rietschel P 2015 Characteristics of patients on lenvatinib with treatment-emergent hypertension in the select trial. Thyroid 25 (Supplement 1) A-250 abstract 629. (doi:10.1089/thy.2015.29004.abstracts)
Ciampi R, Mian C, Fugazzola L, Cosci B, Romei C, Barollo S, Cirello V, Bottici V, Marconcini G & Rosa PM et al. 2013 Evidence of a low prevalence of RAS mutations in a large medullary thyroid cancer series. Thyroid 23 50–57. (doi:10.1089/thy.2012.0207)
Copland JA, Marlow LA, Kurakata S, Fujiwara K, Wong AK, Kreinest PA, Williams SF, Haugen BR, Klopper JP & Smallridge RC 2006 Novel high-affinity PPARgamma agonist alone and in combination with paclitaxel inhibits human anaplastic thyroid carcinoma tumor growth via p21WAF1/CIP1. Oncogene 25 2304–2317. (doi:10.1038/sj.onc.1209267)
Dadu R, Shah K, Busaidy NL, Waguespack SG, Habra MA, Ying AK, Hu MI, Bassett R, Jimenez C & Sherman SI et al. 2015 Efficacy and tolerability of vemurafenib in patients with BRAF(V600E) -positive papillary thyroid cancer: M.D. Anderson Cancer Center off label experience. Journal of Clinical Endocrinology and Metabolism 100 E77–E81. (doi:10.1210/jc.2014-2246)
Deandreis D, Al Ghuzlan A, Leboulleux S, Lacroix L, Garsi JP, Talbot M, Lumbroso J, Baudin E, Caillou B & Bidart JM et al. 2011 Do histological, immunohistochemical, and metabolic (radioiodine and fluorodeoxyglucose uptakes) patterns of metastatic thyroid cancer correlate with patient outcome? Endocrine-Related Cancer 18 159–169. (doi:10.1677/ERC-10-0233)
De Besi P, Busnardo B, Toso S, Girelli ME, Nacamulli D, Simioni N, Casara D, Zorat P & Fiorentino MV 1991 Combined chemotherapy with bleomycin, adriamycin, and platinum in advanced thyroid cancer. Journal of Endocrinological Investigation 14 475–480. (doi:10.1007/BF03346846)
Deftos LJ & Stein MF 1980 Radioiodine as an adjunct to the surgical treatment of medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 50 967–968. (doi:10.1210/jcem-50-5-967)
Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, Capdeville R & Talpaz M 2001 Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. New England Journal of Medicine 344 1038–1042. (doi:10.1056/NEJM200104053441402)
Durante C, Haddy N, Baudin E, Leboulleux S, Hartl D, Travagli JP, Caillou B, Ricard M, Lumbroso JD & De Vathaire F et al. 2006 Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy. Journal of Clinical Endocrinology and Metabolism 91 2892–2899.
Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S & Mooney M et al. 2009 New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). European Journal of Cancer 45 228–247. (doi:10.1016/j.ejca.2008.10.026)
Elisei R, Romei C, Vorontsova T, Cosci B, Veremeychik V, Kuchinskaya E, Basolo F, Demidchik EP, Miccoli P & Pinchera A et al. 2001 RET/PTC rearrangements in thyroid nodules: studies in irradiated and not irradiated, malignant and benign thyroid lesions in children and adults. Journal of Clinical Endocrinology and Metabolism 86 3211–3216.
Elisei R, Ugolini C, Viola D, Lupi C, Biagini A, Giannini R, Romei C, Miccoli P, Pinchera A & Basolo F 2008 BRAF(V600E) mutation and outcome of patients with papillary thyroid carcinoma: a 15-year median follow-up study. Journal of Clinical Endocrinology and Metabolism 93 3943–3949. (doi:10.1210/jc.2008-0607)
Elisei R & Pinchera A 2012 Advances in the follow-up of differentiated or medullary thyroid cancer. Nature Reviews Endocrinology 8 466–475. (doi:10.1038/nrendo.2012.38)
Elisei R, Schlumberger MJ, Muller SP, Schoffski P, Brose MS, Shah MH, Licitra L, Jarzab B, Medvedev V & Kreissl MC et al. 2013 Cabozantinib in progressive medullary thyroid cancer. Journal of Clinical Oncology 31 3639–3646. (doi:10.1200/JCO.2012.48.4659)
Elisei R, Schlumberger M, Tahara M, Robinson B, Brose M, Dutcus C, Zhu J, Newbold K, Kiyota N & Kim SB et al. 2015 Subgroup analysis according to differentiated thyroid cancer histology in phase 3 (SELECT) trial of lenvatinib. Oncology Research and Treatment 38 (Supplement 5) 1–270 (abstract 91).
Eng C, Clayton D, Schuffenecker I, Lenoir G, Cote G, Gagel RF, van Amstel HK, Lips CJ, Nishisho I & Takai SI et al. 1996 The relationship between specific RET proto-oncogene mutations and disease phenotype in multiple endocrine neoplasia type 2. International RET mutation consortium analysis. JAMA 276 1575–1579.
Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E & Desai AA et al. 2007 Sorafenib in advanced clear-cell renal-cell carcinoma. New England Journal of Medicine 356 125–134.
Estfan B, Byrne M & Kim R 2013 Sorafenib in advanced hepatocellular carcinoma: hypertension as a potential surrogate marker for efficacy. American Journal of Clinical Oncology 36 319–324. (doi:10.1097/COC.0b013e3182468039)
Finke J, Ko J, Rini B, Rayman P, Ireland J & Cohen P 2011 MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy. International Immunopharmacology 11 856–861. (doi:10.1016/j.intimp.2011.01.030)
Fox E, Widemann BC, Chuk MK, Marcus L, Aikin A, Whitcomb PO, Merino MJ, Lodish M, Dombi E & Steinberg SM et al. 2013 Vandetanib in children and adolescents with multiple endocrine neoplasia type 2B associated medullary thyroid carcinoma. Clinical Cancer Research 19 4239–4248. (doi:10.1158/1078-0432.CCR-13-0071)
Fusco A, Santoro M, Grieco M, Carlomagno F, Dathan N, Fabien N, Berlingieri MT, Li Z, De Franciscis V & Salvatore D et al. 1995 RET/PTC activation in human thyroid carcinomas. Journal of Endocrinological Investigation 18 127–129.
Gharib H, McConahey WM, Tiegs RD, Bergstralh EJ, Goellner JR, Grant CS, van Heerden JA, Sizemore GW & Hay ID 1992 Medullary thyroid carcinoma: clinicopathologic features and long-term follow-up of 65 patients treated during 1946 through 1970. Mayo Clinic Proceedings 67 934–940. (doi:10.1016/S0025-6196(12)60923-9)
Gild ML, Bullock M, Robinson BG & Clifton-Bligh R 2011 Multikinase inhibitors: a new option for the treatment of thyroid cancer. Nature Reviews Endocrinology 7 617–624. (doi:10.1038/nrendo.2011.141)
Giordano TJ, Beaudenon-Huibregtse S, Shinde R, Langfield L, Vinco M, Laosinchai-Wolf W & Labourier E 2014 Molecular testing for oncogenic gene mutations in thyroid lesions: a case-control validation study in 413 postsurgical specimens. Human Pathology 45 1339–1347. (doi:10.1016/j.humpath.2014.03.010)
Godbert Y, Henriques de Figueiredo B, Bonichon F, Chibon F, Hostein I, Perot G, Dupin C, Daubech A, Belleannee G & Gros A et al. 2015 Remarkable response to crizotinib in woman with anaplastic lymphoma kinase-rearranged anaplastic thyroid carcinoma. Journal of Clinical Oncology 33 e84–e87. (doi:10.1200/JCO.2013.49.6596)
Grubbs EG, Ng PK, Bui J, Busaidy NL, Chen K, Lee JE, Lu X, Lu H, Meric-Bernstam F & Mills GB et al. 2015 RET fusion as a novel driver of medullary thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 100 788–793. (doi:10.1210/jc.2014-4153)
Guo M, Sherman L, Wirth L, Schlumberger M, Dutcus C, Robinson B, Tahara M & Latimer N 2015 Overall survival gain with lenvatinib vs. placebo in radioactive iodine refractory differentiated thyroid cancer (RR-DTC): An updated analysis. Abstract 2805 presented at the European Cancer Congress, 25–29 September 2015, Vienna, Austria. Philadelphia, PA, USA: Elsevier.
Gupta-Abramson V, Troxel AB, Nellore A, Puttaswamy K, Redlinger M, Ransone K, Mandel SJ, Flaherty KT, Loevner LA & O'Dwyer PJ et al. 2008 Phase II trial of sorafenib in advanced thyroid cancer. Journal of Clinical Oncology 26 4714–4719. (doi:10.1200/JCO.2008.16.3279)
Habra MA, Song JH & Rietschel P 2015 Outcomes by site of metastasis for patients with radioiodine-refractory differentiated thyroid cancer treated with lenvatinib versus placebo: results from a phase 3, randomized trial. Abstract 55 presented at the 15th International Thyroid Congress, 18–23 October 2015, Orlando, Florida, USA. Thyroid 25 (supplement 1) A-23 abstract 55. (doi:10.1089/thy.2015.29004.abstracts)
Hayat MJ, Howlader N, Reichman ME & Edwards BK 2007 Cancer statistics, trends, and multiple primary cancer analyses from the Surveillance, Epidemiology, and End Results (SEER) Program. Oncologist 12 20–37. (doi:10.1634/theoncologist.12-1-20)
Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, Pentlow KS, Zanzonico PB, Haque S & Gavane S et al. 2013 Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. New England Journal of Medicine 368 623–632. (doi:10.1056/NEJMoa1209288)
Isham CR, Bossou AR, Negron V, Fisher KE, Kumar R, Marlow L, Lingle WL, Smallridge RC, Sherman EJ & Suman VJ et al. 2013 Pazopanib enhances paclitaxel-induced mitotic catastrophe in anaplastic thyroid cancer. Science Translational Medicine 5 166ra163. (doi:10.1126/scitranslmed.3004358)
Isham CR, Netzel BC, Bossou AR, Milosevic D, Cradic KW, Grebe SK & Bible KC 2014 Development and characterization of a differentiated thyroid cancer cell line resistant to VEGFR-targeted kinase inhibitors. Journal of Clinical Endocrinology and Metabolism 99 E936–E943. (doi:10.1210/jc.2013-2658)
Ji JH, Oh YL, Hong M, Yun JW, Lee HW, Kim D, Ji Y, Kim DH, Park WY & Shin HT et al. 2015 Identification of driving ALK fusion genes and genomic landscape of medullary thyroid cancer. PLoS Genetics 11 e1005467. (doi:10.1371/journal.pgen.1005467)
Jin N, Jiang T, Rosen DM, Nelkin BD & Ball DW 2009 Dual inhibition of mitogen-activated protein kinase kinase and mammalian target of rapamycin in differentiated and anaplastic thyroid cancer. Journal of Clinical Endocrinology and Metabolism 94 4107–4112. (doi:10.1210/jc.2009-0662)
Jin N, Jiang T, Rosen DM, Nelkin BD & Ball DW 2011 Synergistic action of a RAF inhibitor and a dual PI3K/mTOR inhibitor in thyroid cancer. Clinical Cancer Research 17 6482–6489. (doi:10.1158/1078-0432.CCR-11-0933)
Jo YS, Li S, Song JH, Kwon KH, Lee JC, Rha SY, Lee HJ, Sul JY, Kweon GR & Ro HK et al. 2006 Influence of the BRAFV600E mutation on expression of vascular endothelial growth factor in papillary thyroid cancer. Journal of Clinical Endocrinology and Metabolism 91 3667–3670.
Juweid M, Sharkey RM, Behr T, Swayne LC, Rubin AD, Hanley D, Herskovic T, Markowitz A, Siegel J & Goldenberg DM 1995 Targeting and initial radioimmunotherapy of medullary thyroid carcinoma with 131I-labeled monoclonal antibodies to carcinoembryonic antigen. Cancer Research 55 5946s–5951s.
Juweid ME, Hajjar G, Swayne LC, Sharkey RM, Suleiman S, Herskovic T, Pereira M, Rubin AD & Goldenberg DM 1999 Phase I/II trial of (131)I-MN-14F(ab)2 anti-carcinoembryonic antigen monoclonal antibody in the treatment of patients with metastatic medullary thyroid carcinoma. Cancer 85 1828–1842. (doi:10.1002/(sici)1097-0142(19990415)85:8<1828::aid-cncr25>3.0.co;2-h)
Juweid ME, Hajjar G, Stein R, Sharkey RM, Herskovic T, Swayne LC, Suleiman S, Pereira M, Rubin AD & Goldenberg DM 2000 Initial experience with high-dose radioimmunotherapy of metastatic medullary thyroid cancer using 131I-MN-14 F(ab)2 anti-carcinoembryonic antigen MAb and AHSCR. Journal of Nuclear Medicine 41 93–103.
Karaca Z, Tanriverdi F, Unluhizarci K, Ozturk F, Gokahmetoglu S, Elbuken G, Cakir I, Bayram F & Kelestimur F 2011 VEGFR1 expression is related to lymph node metastasis and serum VEGF may be a marker of progression in the follow-up of patients with differentiated thyroid carcinoma. European Journal of Endocrinology 164 277–284. (doi:10.1530/EJE-10-0967)
Kim E, Matsuse M, Saenko V, Suzuki K, Ohtsuru A, Mitsutake N & Yamashita S 2012 Imatinib enhances docetaxel-induced apoptosis through inhibition of nuclear factor-kappaB activation in anaplastic thyroid carcinoma cells. Thyroid 22 717–724. (doi:10.1089/thy.2011.0380)
Kloos RT, Ringel MD, Knopp MV, Hall NC, King M, Stevens R, Liang J, Wakely PE Jr., Vasko VV & Saji M et al. 2009 Phase II trial of sorafenib in metastatic thyroid cancer. Journal of Clinical Oncology 27 1675–1684.
Knauf JA, Ma X, Smith EP, Zhang L, Mitsutake N, Liao XH, Refetoff S, Nikiforov YE & Fagin JA 2005 Targeted expression of BRAFV600E in thyroid cells of transgenic mice results in papillary thyroid cancers that undergo dedifferentiation. Cancer Research 65 4238–4245. (doi:10.1158/0008-5472.CAN-05-0047)
Koguchi Y, Hoen HM, Bambina SA, Rynning MD, Fuerstenberg RK, Curti BD, Urba WJ, Milburn C, Bahjat FR & Korman AJ et al. 2015 Serum immunoregulatory proteins as predictors of overall survival of metastatic melanoma patients treated with ipilimumab. Cancer Research 75 5084–5092. (doi:10.1158/0008-5472.CAN-15-2303)
Kohno T, Ichikawa H, Totoki Y, Yasuda K, Hiramoto M, Nammo T, Sakamoto H, Tsuta K, Furuta K & Shimada Y et al. 2012 KIF5B-RET fusions in lung adenocarcinoma. Nature Medicine 18 375–377. (doi:10.1038/nm.2644)
Koo BS, Kim JM, Seo ST, Yoon YH, Kwon KR, Kim SH, Kwon HW, Bae WJ & Lim YC 2014 Upregulation of HGF and c-MET is Associated with subclinical central lymph node metastasis in papillary thyroid microcarcinoma. Annals of Surgical Oncology 21 2310–2317 (doi:10.1245/s10434-014-3553-5).
Kurzrock R, Sherman SI, Ball DW, Forastiere AA, Cohen RB, Mehra R, Pfister DG, Cohen EE, Janisch L & Nauling F et al. 2011 Activity of XL184 (Cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. Journal of Clinical Oncology 29 2660–2666. (doi:10.1200/JCO.2010.32.4145)
Laure Giraudet A, Al Ghulzan A, Auperin A, Leboulleux S, Chehboun A, Troalen F, Dromain C, Lumbroso J, Baudin E & Schlumberger M 2008 Progression of medullary thyroid carcinoma: assessment with calcitonin and carcinoembryonic antigen doubling times. European Journal of Endocrinology 158 239–246. (doi:10.1530/EJE-07-0667)
Leboulleux S, Bastholt L, Krause T, de la Fouchardiere C, Tennvall J, Awada A, Gomez JM, Bonichon F, Leenhardt L & Soufflet C et al. 2012 Vandetanib in locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 2 trial. Lancet Oncology 13 897–905. (doi:10.1016/S1470-2045(12)70335-2)
Lin CI, Whang EE, Donner DB, Du J, Lorch J, He F, Jiang X, Price BD, Moore FD Jr. & Ruan DT 2010 Autophagy induction with RAD001 enhances chemosensitivity and radiosensitivity through Met inhibition in papillary thyroid cancer. Molecular Cancer Research 8 1217–1226. (doi:10.1158/1541-7786.MCR-10-0162)
Lin SF, Huang YY, Lin JD, Chou TC, Hsueh C & Wong RJ 2012 Utility of a PI3K/mTOR inhibitor (NVP-BEZ235) for thyroid cancer therapy. PLoS One 7 e46726. (doi:10.1371/journal.pone.0046726)
Lira ME, Choi YL, Lim SM, Deng S, Huang D, Ozeck M, Han J, Jeong JY, Shim HS & Cho BC et al. 2014 A single-tube multiplexed assay for detecting ALK, ROS1, and RET fusions in lung cancer. Journal of Molecular Diagnostics 16 229–243. (doi:10.1016/j.jmoldx.2013.11.007)
Liu D, Liu Z, Jiang D, Dackiw AP & Xing M 2007 Inhibitory effects of the mitogen-activated protein kinase kinase inhibitor CI-1040 on the proliferation and tumor growth of thyroid cancer cells with BRAF or RAS mutations. Journal of Clinical Endocrinology and Metabolism 92 4686–4695. (doi:10.1210/jc.2007-0097)
Liu Z, Hou P, Ji M, Guan H, Studeman K, Jensen K, Vasko V, El-Naggar AK & Xing M 2008 Highly prevalent genetic alterations in receptor tyrosine kinases and phosphatidylinositol 3-kinase/akt and mitogen-activated protein kinase pathways in anaplastic and follicular thyroid cancers. Journal of Clinical Endocrinology and Metabolism 93 3106–3116. (doi:10.1210/jc.2008-0273)
Liu D, Xing J, Trink B & Xing M 2010 BRAF mutation-selective inhibition of thyroid cancer cells by the novel MEK inhibitor RDEA119 and genetic-potentiated synergism with the mTOR inhibitor temsirolimus. International Journal of Cancer 127 2965–2973. (doi:10.1002/ijc.v127:12)
Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL & Forner A et al. 2008 Sorafenib in advanced hepatocellular carcinoma. New England Journal of Medicine 359 378–390. (doi:10.1056/NEJMoa0708857)
Locati LD, Licitra L, Agate L, Ou SH, Boucher A, Jarzab B, Qin S, Kane MA, Wirth LJ & Chen C et al. 2014 Treatment of advanced thyroid cancer with axitinib: Phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments. Cancer 120 2694–2703. (doi:10.1002/cncr.28766)
Marotta V, Ramundo V, Camera L, Del Prete M, Fonti R, Esposito R, Palmieri G, Salvatore M, Vitale M & Colao A et al. 2013 Sorafenib in advanced iodine-refractory differentiated thyroid cancer: efficacy, safety and exploratory analysis of role of serum thyroglobulin and FDG-PET. Clinical Endocrinology 78 760–767. (doi:10.1111/cen.12057)
Marques P, Vieira Mda S & Bugalho MJ 2015 Ectopic cushing in a patient with medullary thyroid carcinoma: hypercortisolism control and tumor reduction with sunitinib. Endocrine 49 290–292. (doi:10.1007/s12020-014-0352-5)
Mathur A, Moses W, Rahbari R, Khanafshar E, Duh QY, Clark O & Kebebew E 2011 Higher rate of BRAF mutation in papillary thyroid cancer over time: a single-institution study. Cancer 117 4390–4395. (doi:10.1002/cncr.26072)
Meijer JA, Bakker LE, Valk GD, de Herder WW, de Wilt JH, Netea-Maier RT, Schaper N, Fliers E, Lips P & Plukker JT et al. 2013 Radioactive iodine in the treatment of medullary thyroid carcinoma: a controlled multicenter study. European Journal of Endocrinology 168 779–786. (doi:10.1530/EJE-12-0943)
Miyauchi A, Kudo T, Miya A, Kobayashi K, Ito Y, Takamura Y, Higashiyama T, Fukushima M, Kihara M & Inoue H et al. 2011 Prognostic impact of serum thyroglobulin doubling-time under thyrotropin suppression in patients with papillary thyroid carcinoma who underwent total thyroidectomy. Thyroid 21 707–716. (doi:10.1089/thy.2010.0355)
Nakaoku T, Tsuta K, Ichikawa H, Shiraishi K, Sakamoto H, Enari M, Furuta K, Shimada Y, Ogiwara H & Watanabe S et al. 2014 Druggable oncogene fusions in invasive mucinous lung adenocarcinoma. Clinical Cancer Research 20 3087–3093. (doi:10.1158/1078-0432.CCR-14-0107)
Nikiforov YE 2011 Molecular diagnostics of thyroid tumors. Archivves of Pathology and Laboratory Medicine 135 569–577. (doi:10.1043/2010-0664-RAIR.1)
Nikiforov YE, Rowland JM, Bove KE, Monforte-Munoz H & Fagin JA 1997 Distinct pattern of ret oncogene rearrangements in morphological variants of radiation-induced and sporadic thyroid papillary carcinomas in children. Cancer Research 57 1690–1694.
Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW, Knauf JA, Basolo F, Zhu Z, Giannini R, Salvatore G & Fusco A et al. 2003 BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. Journal of Clinical Endocrinology and Metabolism 88 5399–5404. (doi:10.1210/jc.2003-030838)
Ohashi K, Sequist LV, Arcila ME, Moran T, Chmielecki J, Lin YL, Pan Y, Wang L, de Stanchina E & Shien K et al. 2012 Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF gene mutations but lack mutations in KRAS, NRAS, or MEK1. PNAS 109 E2127–E2133. (doi:10.1073/pnas.1203530109)
Orlandi F, Caraci P, Berruti A, Puligheddu B, Pivano G, Dogliotti L & Angeli A 1994 Chemotherapy with dacarbazine and 5-fluorouracil in advanced medullary thyroid cancer. Annals of Oncology 5 763–765.
Ott RA, Hofmann C, Oslapas R, Nayyar R & Paloyan E 1987 Radioiodine sensitivity of parafollicular C cells in aged long-evans rats. Surgery 102 1043–1048.
Pacini F, Basolo F, Elisei R, Fugazzola L, Cola A & Pinchera A 1991 Medullary thyroid cancer. An immunohistochemical and humoral study using six separate antigens. American Journal of Clinical Pathology 95 300–308. (doi:10.1093/ajcp/95.3.300)
Pacini F, Molinaro E, Lippi F, Castagna MG, Agate L, Ceccarelli C, Taddei D, Elisei R, Capezzone M & Pinchera A 2001 Prediction of disease status by recombinant human TSH-stimulated serum Tg in the postsurgical follow-up of differentiated thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 86 5686–5690. (doi:10.1210/jcem.86.12.8065)
Papotti M, Olivero M, Volante M, Negro F, Prat M, Comoglio PM & DiRenzo MF 2000 Expression of hepatocyte growth factor (HGF) and its receptor (MET) in medullary carcinoma of the thyroid. Endocrine Pathology 11 19–30. (doi:10.1385/EP:11:1)
Pitoia F 2014 Response to sorafenib treatment in advanced metastatic thyroid cancer. Arquivos Brasileiros de Endocrinologia and Metabologia 58 37–41. (doi:10.1590/0004-2730000002839)
Pitoia F, Bueno F, Schmidt A, Lucas S & Cross G 2015 Rapid response of hypercortisolism to vandetanib treatment in a patient with advanced medullary thyroid cancer and ectopic Cushing syndrome. Archives of Endocrinology and Metabolism 59 343–346. (doi:10.1590/2359-3997000000057)
Ramirez R, Hsu D, Patel A, Fenton C, Dinauer C, Tuttle RM & Francis GL 2000 Over-expression of hepatocyte growth factor/scatter factor (HGF/SF) and the HGF/SF receptor (cMET) are associated with a high risk of metastasis and recurrence for children and young adults with papillary thyroid carcinoma. Clinical Endocrinology (Oxford) 53 635–644. (doi:10.1046/j.1365-2265.2000.01124.x)
Riesco-Eizaguirre G, Gutierrez-Martinez P, Garcia-Cabezas MA, Nistal M & Santisteban P 2006 The oncogene BRAFV600E is associated with a high risk of recurrence and less differentiated papillary thyroid carcinoma due to the impairment of Na+/I- targeting to the membrane. Endocrine-Related Cancer 13 257–269. (doi:10.1677/erc.1.01119)
Rini BI, Cohen DP, Lu DR, Chen I, Hariharan S, Gore ME, Figlin RA, Baum MS & Motzer RJ 2011 Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. Journal of National Cancer Institute 103 763–773. (doi:10.1093/jnci/djr128)
Robbins RJ, Wan Q, Grewal RK, Reibke R, Gonen M, Strauss HW, Tuttle RM, Drucker W & Larson SM 2006 Real-time prognosis for metastatic thyroid carcinoma based on 2-[18F]fluoro-2-deoxy-D-glucose-positron emission tomography scanning. Journal of Clinical Endocrinology and Metabolism 91 498–505. (doi:10.1210/jc.2005-1534)
Romei C, Fugazzola L, Puxeddu E, Frasca F, Viola D, Muzza M, Moretti S, Nicolosi ML, Giani C & Cirello V et al. 2012 Modifications in the papillary thyroid cancer gene profile over the last 15 years. Journal of Clinical Endocrinology and Metabolism 97 E1758–E1765. (doi:10.1210/jc.2012-1269)
Rong S, Segal S, Anver M, Resau JH & Vande Woude GF 1994 Invasiveness and metastasis of NIH 3T3 cells induced by Met-hepatocyte growth factor/scatter factor autocrine stimulation. PNAS 91 4731–4735. (doi:10.1073/pnas.91.11.4731)
Salvatore G, De Falco V, Salerno P, Nappi TC, Pepe S, Troncone G, Carlomagno F, Melillo RM, Wilhelm SM & Santoro M 2006 BRAF is a therapeutic target in aggressive thyroid carcinoma. Clinical Cancer Research 12 1623–1629. (doi:10.1158/1078-0432.CCR-05-2378)
Santarpia L, El-Naggar AK, Cote GJ, Myers JN & Sherman SI 2008 Phosphatidylinositol 3-kinase/akt and ras/raf-mitogen-activated protein kinase pathway mutations in anaplastic thyroid cancer. Journal of Clinical Endocrinology and Metabolism 93 278–284. (doi:10.1210/jc.2007-1076)
Savvides P, Nagaiah G, Lavertu P, Fu P, Wright JJ, Chapman R, Wasman J, Dowlati A & Remick SC 2013 Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid 23 600–604. (doi:10.1089/thy.2012.0103)
Scarpino S, Cancellario d'Alena F, Di Napoli A, Pasquini A, Marzullo A & Ruco LP 2004 Increased expression of Met protein is associated with up-regulation of hypoxia inducible factor-1 (HIF-1) in tumour cells in papillary carcinoma of the thyroid. Journal of Pathology 202 352–358. (doi:10.1002/(ISSN)1096-9896)
Schlumberger M, Bastholt L, Dralle H, Jarzab B, Pacini F & Smit JW 2012 European thyroid association guidelines for metastatic medullary thyroid cancer. European Thyroid Journal 1 5–14. (doi:10.1159/000336977)
Schlumberger M, Brose M, Elisei R, Leboulleux S, Luster M, Pitoia F & Pacini F 2014a Definition and management of radioactive iodine-refractory differentiated thyroid cancer. Lancet Diabetes and Endocrinology 2 356–358. (doi:10.1016/S2213-8587(13)70215-8)
Schlumberger M, Nutting C, Jarzab B, Elisei R, Siena S, Bastholt L, De la Fourchardiere C, Pacini F, Paschke R & Shong Y et al. 2014b Exploratory analysis of outcomes for patients with locally advanced or metastatic radioactive iodine-refractory differentiated thyroid cancer (RAI-RDTC) receiving open-label sorafenib post-progression on the phase III DECISION trial. Abstract OP87 presented at the European Thyroid Congress, September 2014, Santiago de Compostela, Spain. Basel, Switzerland: Karger. (doi:10.1159/000365244)
Schlumberger M, Jarzab B, Cabanillas ME, Robinson B, Pacini F, Ball DW, McCaffrey J, Newbold K, Allison R & Martins RG et al. 2015a A phase II trial of the multitargeted tyrosine kinase inhibitor lenvatinib (E7080) in advanced medullary thyroid cancer. Clinical Cancer Research 22 44–53 (doi:10.1158/1078-0432.CCR-15-1127).
Schlumberger M, Müller S, Elisei R, Schöffski P, Brose MS, Shah MH, Licitra LF, Jarzab B, Medvedev V & Kreissl M et al. 2015b Final overall survival analysis of EXAM, an international, double-blind, randomized, placebo-controlled phase III trial of cabozantinib (Cabo) in medullary thyroid carcinoma (MTC) patients with documented RECIST progression at baseline. Journal of Clinical Oncology 33 (Supplement 15) abstract 6012. (available at: http://meeting.ascopubs.org/cgi/content/abstract/33/15_suppl/6012).
Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, Habra MA, Newbold K, Shah MH & Hoff AO et al. 2015c Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. New England Journal of Medicine 372 621–630. (doi:10.1056/NEJMoa1406470)
Schlumberger MJ, Elisei R, Bastholt L, Wirth LJ, Martins RG, Locati LD, Jarzab B, Pacini F, Daumerie C & Droz JP et al. 2009 Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer. Journal of Clinical Oncology 27 3794–3801. (doi:10.1200/JCO.2008.18.7815)
Shah DR, Shah RR & Morganroth J 2013 Tyrosine kinase inhibitors: their on-target toxicities as potential indicators of efficacy. Drug Safety 36 413–426. (doi:10.1007/s40264-013-0050-x)
Sherman SI, Wirth LJ, Droz JP, Hofmann M, Bastholt L, Martins RG, Licitra L, Eschenberg MJ, Sun YN & Juan T et al. 2008 Motesanib diphosphate in progressive differentiated thyroid cancer. New England Journal of Medicine 359 31–42. (doi:10.1056/NEJMoa075853)
Smallridge RC, Marlow LA & Copland JA 2009 Anaplastic thyroid cancer: molecular pathogenesis and emerging therapies. Endocrine Related Cancer 16 17–44. (doi:10.1677/ERC-08-0154)
Smallridge RC, Copland JA, Brose MS, Wadsworth JT, Houvras Y, Menefee ME, Bible KC, Shah MH, Gramza AW & Klopper JP et al. 2013 Efatutazone, an oral PPAR-gamma agonist, in combination with paclitaxel in anaplastic thyroid cancer: results of a multicenter phase 1 trial. Journal of Clinical Endocrinology and Metabolism 98 2392–2400. (doi:10.1210/jc.2013-1106)
Smyth P, Finn S, Cahill S, O'Regan E, Flavin R, O'Leary JJ & Sheils O 2005 ret/PTC and BRAF act as distinct molecular, time-dependant triggers in a sporadic Irish cohort of papillary thyroid carcinoma. International Journal of Surgical Pathology 13 1–8. (doi:10.1177/106689690501300101)
Soares P, Lima J, Preto A, Castro P, Vinagre J, Celestino R, Couto JP, Prazeres H, Eloy C & Maximo V et al. 2011 Genetic alterations in poorly differentiated and undifferentiated thyroid carcinomas. Current Genomics 12 609–617. (doi:10.2174/138920211798120853)
Sobrinho-Simoes M, Maximo V, Rocha AS, Trovisco V, Castro P, Preto A, Lima J & Soares P 2008 Intragenic mutations in thyroid cancer. Endocrinology and Metabolism Clinics of North America 37 333–362, viii. (doi:10.1016/j.ecl.2008.02.004)
Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C, Marur S, Gramza A, Yosef RB & Gitlitz B et al. 2014 Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid 24 232–240. (doi:10.1089/thy.2013.0078)
Stein B & Smith BD 2010 Treatment options for patients with chronic myeloid leukemia who are resistant to or unable to tolerate imatinib. Clinical Therapeutics 32 804–820. (doi:10.1016/j.clinthera.2010.05.003)
Tallini G & Asa SL 2001 RET oncogene activation in papillary thyroid carcinoma. Advances in Anatomic Pathology 8 345–354. (doi:10.1097/00125480-200111000-00005)
Torlontano M, Crocetti U, Augello G, D'Aloiso L, Bonfitto N, Varraso A, Dicembrino F, Modoni S, Frusciante V & Di Giorgio A et al. 2006 Comparative evaluation of recombinant human thyrotropin-stimulated thyroglobulin levels, 131I whole-body scintigraphy, and neck ultrasonography in the follow-up of patients with papillary thyroid microcarcinoma who have not undergone radioiodine therapy. Journal of Clinical Endocrinology and Metabolism 91 60–63.
Veiga LH, Neta G, Aschebrook-Kilfoy B, Ron E & Devesa SS 2013 Thyroid cancer incidence patterns in Sao Paulo, Brazil, and the U.S. SEER program, 1997–2008. Thyroid 23 748–757. (doi:10.1089/thy.2012.0532)
Wang S, Pashtan I, Tsutsumi S, Xu W & Neckers L 2009 Cancer cells harboring MET gene amplification activate alternative signaling pathways to escape MET inhibition but remain sensitive to Hsp90 inhibitors. Cell Cycle 8 2050–2056. (doi:10.4161/cc.8.13.8861)
Wells SA Jr., Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, Baudin E, Elisei R, Jarzab B & Vasselli JR et al. 2012 Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. Journal of Clinical Oncology 30 134–141. (doi:10.1200/JCO.2011.35.5040)
Worden F, Fassnacht M, Shi Y, Hadjieva T, Bonichon F, Gao M, Fugazzola L, Ando Y, Hasegawa Y & Park do J et al. 2015 Safety and tolerability of sorafenib in patients with radioiodine-refractory thyroid cancer. Endocrine-Related Cancer 22 877–887. (doi:10.1530/ERC-15-0252)
Xing M 2007 Gene methylation in thyroid tumorigenesis. Endocrinology 148 948–953.
Xing M 2013 Molecular pathogenesis and mechanisms of thyroid cancer. Nature Review Cancer 13 184–199. (doi:10.1038/nrc3431)
Xing M, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F & Tuttle RM et al. 2013 Association between BRAFV600E mutation and mortality in patients with papillary thyroid cancer. JAMA 309 1493–1501. (doi:10.1001/jama.2013.3190)
Xing M, Alzahrani AS, Carson KA, Shong YK, Kim TY, Viola D, Elisei R, Bendlova B, Yip L & Mian C et al. 2015 Association between BRAFV600E mutation and recurrence of papillary thyroid cancer. Journal of Clinical Oncology 33 42–50. (doi:10.1200/JCO.2014.56.8253)
Yaish P, Gazit A, Gilon C & Levitzki A 1988 Blocking of EGF-dependent cell proliferation by EGF receptor kinase inhibitors. Science 242 933–935. (doi:10.1126/science.3263702)
Zerilli M, Zito G, Martorana A, Pitrone M, Cabibi D, Cappello F, Giordano C & Rodolico V 2010 BRAF(V600E) mutation influences hypoxia-inducible factor-1alpha expression levels in papillary thyroid cancer. Modern Pathology 23 1052–1060. (doi:10.1038/modpathol.2010.86)