2,845
Views
39
CrossRef citations to date
0
Altmetric
Review

Radioactive iodine-refractory differentiated thyroid cancer: unmet needs and future directions

, , , , &
Pages 541-554 | Published online: 10 Jan 2014

Abstract

Approximately 90% of thyroid cancers are differentiated (DTCs) and have papillary, follicular or Hürthle cell morphology. Although treatment with surgery and radioactive iodine (I-131; RAI), as appropriate, is associated with significant cure rates and survival benefits, clonal disease progression with development of refractoriness to RAI poses a major therapeutic challenge in about 15% of patients. Traditional chemotherapeutic agents are relatively ineffective and are associated with significant toxicities. Molecular studies have demonstrated that the development and progression of DTC are associated with a series of consistent abnormalities in pathways such as MAPK/ERK and PI3/Akt, which govern cellular growth, proliferation, apoptosis and angiogenesis. Small molecular inhibitors that target these pathogenic pathways, without many of the impairments associated with cytotoxic chemotherapy, have demonstrated efficacy in a variety of malignancies, including renal cell carcinoma, hepatocellular carcinoma, non-small-cell lung cancer and chronic myelogenous leukemia. Several targeted therapeutic agents are in development for the treatment of RAI-refractory DTC. Sorafenib and lenvatinib are being studied in placebo-controlled Phase III trials based on encouraging efficacy results observed in single-arm Phase II studies.

Figure 1. Growth factor receptors and the MAPK/ERK pathway.

EGFR: EGF receptor; VEGFR: VEGF receptor.Adapted with permission from Citation[95].

Figure 1. Growth factor receptors and the MAPK/ERK pathway.EGFR: EGF receptor; VEGFR: VEGF receptor.Adapted with permission from Citation[95].

Globally, the age-standardized incidence of thyroid cancer in the general population is 16.4/100,000, with a mortality rate of 1.7/100,000 Citation[1]. Its prevalence is approximately threefold higher in developed than in the less-developed countries. In the USA, for example, approximately 45,000 patients were newly diagnosed with thyroid cancer in 2010 Citation[2].

Cancers of the follicular epithelium of the thyroid are generally categorized as papillary (80%), follicular (11%), Hürthle cell (3%) and anaplastic (2%) Citation[3]. The first three types are categorized as differentiated thyroid cancers (DTCs) Citation[201]. Anaplastic thyroid cancer generally arises from DTC, with approximately half of these patients having a previous or coexisting DTC at the time of diagnosis Citation[4,5]. Medullary thyroid carcinoma (MTC) is derived from parafollicular C cells rather than the follicular epithelium and is observed in approximately 4% of patients. Although these histologic types have different disease manifestations and tumor markers, as well as differences in expression of oncogenes and tumor suppressor genes, patients with DTC are often treated similarly.

As the epithelium of DTC generally retains the ability of the parent tissue to take up iodine from the blood, radioactive iodine (I-131) is often used to locate and ablate residual tissue, reduce recurrence and treat metastatic disease. The authors review the treatment of DTC with I-131, its limitations and the management of patients with I-131 refractory disease. They also examine the molecular pathways implicated in the development of DTC and describe the principle of targeted therapy and the investigational agents that may be used in future treatment.

Current treatments of DTC & their limitations

DTCs are clinicopathologically staged to evaluate prognosis and make treatment decisions. Staging schemes can vary among institutions and geographic regions. Although these staging schemes can adequately predict DTC-specific mortality, they generally cannot predict recurrence for individual patients. Examples of staging systems include the International Union Against Cancer/American Joint Committee on Cancer tumor node metastasis system, the European Organization for Research on Treatment of Cancer, the National Comprehensive Cancer Network, the National Thyroid Cancer Treatment Cooperative Study, the American Thyroid Association, and the two scoring systems. Intra-operative age, metastases, extent, size; and metastasis, age, completeness of resection, invasion and size Citation[6–11,202]. Factors common to these staging systems include age at diagnosis, the size of the primary tumor, histology, evidence of vascular tumor invasion, extent of spread and presence of distant metastases.

Although treatment guidelines for DTC differ regarding the extent of surgery and I-131 dosing, care is similar between countries Citation[9,12–15; Brose MS et al., rationale and design of decision: a double-blind, randomized, placebo-controlled phase iii trial evaluating the efficacy and safety of sorafenib in patients with locally advanced or metastatic radioactive iodine (rai)-refractory, differentiated thyroid cancer (2011), Submitted]. Surgery is the primary treatment for any DTC, except for low-risk papillary microcarcinomas Citation[16,17], and total thyroidectomy is the treatment of choice, followed by I-131 ablation. The risk of recurrent disease and death are lower in patients who undergo total or near-total thyroidectomy than in those who undergo lobectomy Citation[18–21]. Lymph node dissection may be prophylactic or therapeutic. If there is clinical or ultrasonic evidence of lymph node involvement, therapeutic dissection should be performed. The indications for prophylactic dissection remain unclear, but it should be considered in patients with DTCs showing high-risk features such as papillary carcinomas >4 cm and/or extension outside the capsule Citation[9] or in a particular population at risk; such as those affected by the Chernobyl nuclear reactor accident Citation[22]. Limitations of surgery include residual or metastatic disease and risks of hypoparathyroidism and injury to the recurrent laryngeal nerve or other neurovascular structures of the neck Citation[23,24], but reoperation after recurrence to the central compartment is more difficult and can induce these complications more frequently than central node dissection at initial surgery.

Since over 90% of patients show I-131 uptake in the thyroid bed after thyroidectomy Citation[25], I-131 is used to ablate residual thyroid tissue and act as adjuvant treatment for DTC Citation[9]. Retrospective studies and meta-analyses have demonstrated that I-131 ablation of the thyroid can decrease the 10-year risk of recurrence up to 50%, lower the risk of metastatic disease by 3% and reduce mortality Citation[19,26–28]. Although the optimal I-131 dosage required to successfully ablate all residual thyroid tissue remains unclear Citation[29], recent evidence suggests that low activities are as effective as high activities, even in patients with extrathyroidal disease, whether performed after thyroid hormone withdrawal or after treatment with recombinant human thyroid-stimulating hormone (rhTSH) Citation[30,31].

Although I-131 is generally effective in treating subclinical micrometastatic disease, as well as residual and metastatic tumors, its potential for success is lower in patients with bulky disease Citation[32]. Optimal preparation for I-131 therapy is traditionally based on thyroid hormone withdrawal, although preparation with rhTSH was recently shown to be equally effective Citation[9,22,33–35].

I-131 refractory DTC

I-131 refractory DTC can be defined as an inability of malignant/metastatic tissue to take up I-131 from the start of treatment or loss of ability to take up I-131 after previous evidence of uptake. It is detected by a combination of imaging modalities, including an I-131 whole body scan showing at least one lesion that does not take up I-131, or clinical evidence that I-131 is no longer providing benefit to the patient, for example, at cumulative I-131 > 600 mCi (222,000 MBq Citation[36]).

A registry in the USA reported that 2204 out of 2936 patients (75%) were considered disease-free after near-total thyroidectomy, I-131 ablation and aggressive thyroid hormone suppression Citation[37]. Of patients considered disease-free after primary therapy, 10% had recurrent disease after a median of 1.5 years; of the latter, 71% had local or regional tumor recurrence and 18% had distant metastases, whereas, in the other 11%, the site of tumor recurrence was not reported. Although the prognosis of patients with DTC is usually favorable, treatment of recurrent or metastatic disease is problematic, especially when these foci have become refractory to I-131, with most of these patients dying within 3 years Citation[38].

To optimize care, it is important to reach a common definition of I-131 refractory disease, both for clinical trials and in community practice. At a minimum, I-131 refractory disease should be defined as the presence of a lesion detected at imaging that does not take up I-131 or clinical evidence that I-131 is no longer beneficial despite visible uptake Citation[36]. In the latter, tumor progression should also be confirmed by imaging 6–14 months later according to Response Evaluation Criteria in Solid Tumors Citation[39]. As tumor markers such as thyroglobulin reflect risk, not tumor burden, increasing concentrations of these markers should be considered suspicious but not diagnostic of progressive disease, as should the results of F-18 fluorodeoxyglucose (FDG) PET/computerized tomography. To date, it remains unclear whether empiric I-131 therapy reduces morbidity or mortality in patients with increased serum thyroglobulin concentration, but negative on diagnostic I-131 whole body scan Citation[202]. Although not confirmed in randomized trials, patients with a mixed response to I-131 may have resistant clones requiring a different therapeutic intervention.

External beam radiation therapy

External beam radiation therapy (EBRT) has been used successfully in patients with thyroid tumors, including DTC, as the first-line treatment in patients with unresectable tumors, as adjuvant therapy in patients considered at high risk for recurrence following resection and as treatment of I-131-negative recurrences and metastases. Potential risk factors for recurrent disease include older age, grossly visible extrathyroidal tumor extension at the time of surgery, a high likelihood of microscopic residual disease and/or gross residual tumor considered unresectable or resistant to I-131 Citation[9].

A retrospective study in 113 patients with DTC showed that megavoltage EBRT was effective, but that benefits were related to tumor burden Citation[40]. The 5-year survival rates of patients with residual microscopic disease and gross residual disease were 85 and 27%, respectively, and the 4-year relapse-free, disease-specific and overall survival (OS) rates following EBRT in patients with DTC were each at least 73% Citation[41]. Treatment-related morbidities included esophageal stricture, chronic dysphagia requiring a feeding tube and laryngeal stenosis or edema, although these adverse events (AEs) can be minimized by intensity-modulated radiotherapy.

Cytotoxic chemotherapy

Although chemotherapy has significantly benefited patients with hematologic neoplasms, lymphomas, small-cell-lung cancer and other solid tumors, it is much less effective in patients with I-131 refractory DTC, with response rates to bleomycin, methotrexate, melphalan, mitoxantrone and etoposide each being ≤25% Citation[42]. Patients with I-131 refractory DTC have shown low partial response (PR) rates to doxorubicin monotherapy, with these responses being short-lasting and associated with high toxicity rates Citation[43–46]. Response rates were highest in patients with a high performance status and those with lung metastases. Complications include cytopenia, nausea, vomiting, hair loss and potential cardiac toxicity. Although some patients have shown durable responses, no survival benefit has been reported. Owing to the low response rates to doxorubicin monotherapy, this agent has been combined with other chemotherapeutic agents, including cisplatin, carboplatin and epirubicin, but complete or PR rates were similar to or worse than those with doxorubicin monotherapy Citation[47–49].

Taxane-based regimens have also been evaluated, based on their systemic activity against anaplastic carcinomas of the thyroid. Twice-weekly docetaxel was reported to produce disease stabilization for 14–18 months, but no objective responses, in three patients with I-131 refractory DTC Citation[50], whereas nine patients treated with paclitaxel plus gemcitabine showed no response Citation[51].

Alternative treatment modalities have included the combination of external radiotherapy and chemotherapy, but the results have been only marginally superior to those of each one alone, excluding some patients with anaplastic carcinoma. In selected cases, liver and bone metastases have been treated with local therapies such as arterial embolizaion or chemoembolization. Although some lesions may show significant tumor reduction, these procedures are never curative and have no impact on final survival.

Signaling pathways implicated in the development of DTC

Owing to the poor results with the above-mentioned treatment modalities, the molecular pathways involved in the pathogenesis and progression of DTC have been assessed to identify elements of receptor-signaling pathways that may serve as targets for innovative treatments.

The genotype of specific cancers determines their behavior and may determine their response to treatment Citation[52]. Some mutations may also provide significant prognostic information on these patients. A key molecular element in the biology of DTC is the tyrosine kinase RAS/RAF/MEK MAPK/ERK pathway. Normally, activation of a tyrosine kinase leads to a cascade of sequential phosphorylation of RAS, BRAF, MEK and then MAPK Citation[53]. Physiologically, signaling through these pathways leads to the activation of transcription factors involved in cell cycle regulation and cellular differentiation Citation[54]. Constitutive activation of this and other pathways has been observed in patients with DTC.

The PI3/Akt pathway is also involved in the pathogenesis of DTC and other thyroid cancers Citation[55]. When activated and free of the inhibitory effects of the tumor-suppressor gene product, PTEN, Akt phosphorylates elements of the mTOR pathway; a key element in cell growth, proliferation, and survival, and a pathway crucial in cell growth, proliferation and angiogenesis. In addition to its upregulation of MAPK/ERK, Ras can upregulate the PI3/Akt pathway. Moreover, poorly differentiated and anaplastic thyroid cancers are characterized by an additional genetic event, the loss of the tumor suppressor gene, p53 Citation[52].

Papillary carcinoma

RET and NTRK1 are involved in the pathogenesis of papillary thyroid carcinoma (PTC). Both genes code for nerve growth factor receptors with tyrosine kinase activity Citation[56,57]. Approximately 20–40% of PTCs have rearrangements of one of these genes with other genes, forming chimeric oncogenes, including RET/PTC in tumors with RET rearrangements, with RET alterations being approximately threefold more common than NTRK1 rearrangements Citation[58,59].

The RAS family of proto-oncogenes includes KrasA, KrasB and Nras, all of which are membrane-bound GTP/GDP-binding (G) molecules Citation[60]. These proteins transfer signals from activated tyrosine receptors in the cell membrane to pathways that lead to the synthesis of key proteins involved in cell growth, proliferation and survival. Although Ras mutations have been observed in 10–15% of PTCs, they are more commonly associated with follicular carcinoma and the follicular variant of papillary cancer Citation[52,61]. ARaf, BRaf and CRaf are isoforms of the mammalian Raf serine–threonine kinase that participate in the RAF/MEK/MAPK pathway. BRAF mutations, observed in nearly 40% of PTCs Citation[62], have been associated with a more aggressive course of disease Citation[63,64], although a study from Japan, involving over 600 patients, reported a negative relationship between BRAF mutation and patient prognosis Citation[65]. The presence of RET/PTC and BRAF mutations in microcarcinomas of the thyroid suggests that these mutations are early events in thyroid carcinogenesis and contribute significantly to the initiation and progression of PTCs Citation[66–68]. Moreover, the presence of BRAF and RET mutations in the same tumor, although extremely rare, is associated with an increased risk of recurrence Citation[69].

Among the other genetic anomalies identified in patients with PTC are single-nucleotide polymorphisms in genes that regulate transcription, upregulation of microRNAs that modify gene expression and increased methylation of tumor suppressor genes Citation[70–73]. In addition, familial PTC has been associated with abnormalities in the short arm of chromosome 19 and the long arms of chromosomes 1, 2 and 8 Citation[74–76], as well as with short telomeres in the germline Citation[77].

Follicular carcinoma

A translocation involving the PAX8 promotor gene on the long arm of chromosome 2 and the PPAR-γ-1 gene on the short arm of chromosome 3 [t(2;3)(q13;p25)] has been observed in follicular carcinoma cells, resulting in the fusion of their DNA-binding domains. PAX8 is a critical regulator of the development and differentiation of follicular epithelium Citation[78], whereas PPAR-γ is a nuclear hormone receptor, not normally expressed at high concentrations in thyroid, which is involved in carbohydrate and lipid metabolism and inflammation Citation[79]. The exact mechanism by which this fusion protein exerts its effects is unknown. In one study, this cytogenetic abnormality was observed in five out of eight follicular carcinomas but in zero out of 20 follicular adenomas Citation[80], suggesting that it may be a marker of follicular carcinoma in fine-needle aspirates, distinguishing between well-differentiated follicular carcinomas without microscopic vascular tumor invasion and follicular adenomas in surgical specimens.

Mutations of Nras, Hras and Kras have been observed in both benign and malignant thyroid follicular lesions Citation[81–83]. Similar to the overexpression of c-myc and c-fos in these lesions, these RAS mutations probably promote tumor growth rather than transformation. The PIK3/Akt pathway is also upregulated in DTCs, most often in follicular carcinomas Citation[52], possibly by amplification of the PIK3CA gene Citation[84,85]. Although hypermethylation of the tumor suppressor gene, RASSF1A, has been observed in 75% of follicular carcinomas, it has also been detected in PTCs and follicular adenomas Citation[86].

Hürthle cell carcinoma

Hürthle cells are characterized on light and electron microscopy by their oncocytic and mitochondria-rich cytoplasm, respectively Citation[52]. These tumors have abnormalities in both mitochondrial and nuclear genes, which may disrupt oxidative phosphorylation. Specific molecular pathways that may be involved in Hürthle cell carcinomas are currently being investigated.

Overview of a targeted approach to I-131 refractory DTC

Historically, treatment options have been limited in patients with I-131 refractory DTC. Molecular abnormalities common to DTCs may provide the opportunity to target specific pathways involved in tumor cell growth, metastasis and cancer-induced angiogenesis without the collateral damage to the hematopoietic, gastrointestinal and other organ systems resulting from standard chemotherapy.

Kinase receptors and elements of the MAPK/ERK pathway are centrally involved in the growth and progression of DTC Citation[87]. RET mutations can activate its encoded tyrosine kinase independent of ligand, upregulate the MAPK/ERK pathway, inhibit apoptosis and protect cells from the cytotoxic actions of doxorubicin Citation[88,89]. RAS and RAF mutations (e.g., in BRAF) can result in the constitutive activation of MAPK/ERK. BRAF mutations are associated with resistance to I-131 Citation[90]. VEGF stimulates endothelial cell growth and migration and promotes the formation of collateral blood vessels. VEGF and other growth factors signal through the PI3/Akt pathway. Serum VEGF concentrations are elevated in patients with metastatic DTC and may represent an important potential target Citation[91]. Owing to the fact that most thyroid cancers have multiple mutations that modulate different pathways, agents targeting one or more of these pathways may improve outcomes in patients with I-131 refractory DTC Citation[92].

A number of small molecular weight inhibitors of these signaling kinase pathways have been developed. The proof-of-concept of targeted therapy for cancer has become well established. Receptor kinase pathway inhibitors have been successfully used to treat patients with a variety of different tumor types, including gastrointestinal stromal tumors, non-small-cell lung cancer, renal cell carcinoma, hepatocellular carcinoma and melanoma Citation[93]. Since activation of these signaling pathways is associated with the pathogenesis, growth, invasion, metastasis, survival and angiogenesis of DTC, inhibiting these pathways may represent a significant therapeutic approach to patients with thyroid cancer Citation[36,87,94,95]. Response rates to these inhibitors have suggested that they may be successful in treating patients with I-131 refractory DTC, and thus targeted therapies have been included in recent guidelines. Although these compounds are generally well tolerated, dose-limiting toxicities have been observed Citation[87]. The efficacy of these agents may depend on the site of disease, suggesting that combinations of inhibitors that target different pathways may be more beneficial than monotherapy Citation[96].

Trial design issues for studies in I-131 refractory DTC

Patient selection and trial end points should be the prime considerations in designing trials of patients with I-131 refractory DTC. As noted earlier, I-131 refractory DTC can be defined as an inability to take up I-131, either upon initial exposure or upon loss of prior evidence of uptake. Such determinations should be made under conditions of low-iodine diet and adequate endogenous TSH elevation or rhTSH stimulation. However, the threshold of iodine uptake, the definition of progressive disease and the cumulative amount of I-131 received should be defined in advance. Moreover, the heterogeneity in disease progression within this patient population suggests the importance of enrolling patients with comparable rates of disease progression. Up to now, most of the patients enrolled in clinical trials had measurable node or distant metastases (according to RECIST criteria), while local infiltration in the neck cannot be clearly assessed and thus is excluded from the studies. This is an important issue that should be addressed in the future.

Most of the drugs being developed for the treatment of I-131 refractory DTC are agents targeted to molecular pathways thought to be critical for tumor growth. Whereas traditional chemotherapeutic agents were generally approved based on tumor shrinkage, targeted agents are more commonly approved based on improvements in progression-free survival (PFS) or time to tumor progression. This is due in part to the recognition that anti-angiogenic activity is important for the clinical efficacy of many of these targeted agents, such that stable disease (SD) rather than tumor shrinkage may be the principal outcome. In 2007, the US FDA issued guidelines stating that PFS prolongation may be a surrogate end point for clinical benefits Citation[97]. Although OS remains the preferred end point for oncology trials, the reality of patient crossover and the availability and subsequent use of alternative therapies upon progression tends to confound OS end points. For this reason, the approval of sorafenib, sunitinib, pazopanib, everolimus and bevacizumab in the treatment of patients with advanced refractory DTC was based on studies showing improvements in PFS. In addition, the recent approval of vandetanib to treat MTC was based on a Phase III trial whose primary end point was an improvement in PFS Citation[98].

Targeted agents in Phase III development for treatment of DTC

Two targeted agents, sorafenib and lenvatinib, are currently in Phase III trials in patients with I-131 refractory DTC .

Sorafenib

Sorafenib is a multiple kinase inhibitor, targeting CRAF, BRAF, VEGF receptor (VEGFR)-1, -2, -3, PDGF receptor (PDGFR)-β, RET, c-kit and Flt-3 Citation[99]. As a multifunctional inhibitor, sorafenib inhibits tumor growth, progression, metastasis and angiogenesis, as well as downregulating mechanisms that protect tumors from apoptosis. These findings have suggested that sorafenib may significantly improve outcomes in patients with DTC.

Several Phase II trials have assessed the effects of sorafenib monotherapy in over 200 patients with thyroid cancer, most with DTC, with other patients having medullary and anaplastic carcinomas Citation[100–104]. The median PFS ranged from 58 to 96 weeks, with PR rates as high as 38% and disease control rates (defined as SD plus PR) of 59–100%. Median OS was reported to be as long as 141 weeks. Approximately 62% of patients required dose reductions due to adverse events (AEs) that were mostly grade 1 or 2. The most common AEs have been hand–foot skin reactions and other skin toxicities, fatigue, weight loss, diarrhea and musculoskeletal pain/arthralgias.

In addition, a recently reported retrospective trial reported the results of sorafenib treatment in 34 patients with metastatic thyroid cancer, including 16 with DTC Citation[105]. Of the 34 patients, 11 (32%) achieved PR, including three out of 16 (19%) with DTC, and 14 (41%) achieved SD for >6 months, including 8 (50%) with DTC. At a median follow-up of 11.5 months, the median PFS was 10.5 months (95% CI: 6.13–14.87 months) in all patients and 13.3 months (95% CI: 6.28–20.45 months) in patients with DTC; and the median OS was 23.6 months for all patients and for those with DTC.

These encouraging results and the need for improvement in the treatment of I-131 refractory DTC have led to the designing of a double-blind, multicenter, multinational, randomized Phase III trial of sorafenib in 380 patients with locally advanced or metastatic I-131 refractory DTC (DECISION) Citation[106]. Briefly, patients should have evidence of progressive disease based on RECIST criteria within the previous 14 months. I-131 refractoriness in this trial is defined as a lack of I-131 uptake by a qualified target lesion in patients with low-iodine diets and adequate endogenous TSH elevation or rhTSH stimulation. In addition, patients with some iodine uptake (i.e., those who received I-131 within the last 16 months and whose target lesions had progressed, and those who received multiple I-131 treatments and whose target lesions had progressed after each of two I-131 treatments within 16 months), as well as patients who had received cumulative I-131 ≥ 22.2 GBq (600 mCi), are eligible for inclusion. The primary efficacy end point is PFS and treatment groups will be compared using a one-sided log-rank test with an overall one-sided a of 0.01 stratified by age (< 60 years vs ≥ 60 years) and geographic region. Secondary outcome measures will include OS, time to tumor progression, duration of response, disease control rates, complete response, PR and SD defined by RECIST criteria. Patient-reported quality-of-life outcomes will be measured using the FACT-G and EQ-5D questionnaires. No interim analysis for efficacy or futility is planned. Sorafenib 400 mg or matching placebo will be administered orally twice daily until radiologically documented progression of disease or unacceptable toxicity. At the investigators’ discretion, patients who develop progressive disease while on placebo will be allowed to cross over to sorafenib. This trial has completed enrollment, and results are expected to be reported in 2012. Sorafenib is also being evaluated in combination with the mTOR inhibitor temsirolimus (rapamycin) and its derivative, everolimus, in patients with DTC. Other combination regimens being investigated include sorafenib/everolimus/pasireotide and sorafenib/cediranib/lenalidomide.

Lenvatinib

Lenvatinib, previously known as E7080, inhibits RET, VEGFR 1, -2, -3, PDGFR, c-kit and FGF receptor 1, 2, 3 and 4 Citation[107]. A Phase II trial of this multikinase inhibitor in 58 patients with I-131 refractory DTC and disease progression over the previous 12 months by RECIST criteria showed that 50% of patients achieved a PR, with a median PFS of 12.6 months Citation[108]. Most AEs were grade 1–2, with only 8.6% of patients experiencing a grade 4 event. AEs included hypertension, fatigue, diarrhea, weight loss, anorexia and proteinuria.

In 2011, a multicenter, randomized, double-blind, placebo-controlled, Phase III study of lenvatinib (24 mg once daily) was initiated in patients with I-131 refractory DTC and radiographic evidence of disease progression within the prior 12 months; prior targeted therapy is not an exclusion criterion Citation[203]. The primary end point of the trial is PFS and the secondary end point is OS.

Other targeted agents for the treatment of DTC

Clinical trials have been conducted with several other targeted agents and some are in various stages of clinical development in the treatment of patients with DTC .

Sunitinib

Sunitinib is a selective, orally administered tyrosine kinase receptor inhibitor that targets all three VEGF receptors, RET and RET/PTC Citation[109]. A Phase I trial in two patients with metastatic, progressive PTC or follicular carcinoma resulted in clinical responses sustained for 4 years Citation[110]. Preliminary results from two Phase II trials of patients with I-131 refractory DTC support additional studies of sunitinib. In an open-label, Phase II trial of 43 subjects, (37 with DTC), sunitinib produced PRs in 13% and SD in 68% Citation[111]. AEs included fatigue, diarrhea, palmar–plantar erythrodysesthesia, neutropenia and hypertension. Interim results from a second Phase II trial showed that sunitinib induced PR or SD for more than 12 weeks in two out of 12 patients with DTC Citation[112]. Continuous dosing of sunitinib in patients with F-18 FDG/PET-avid advanced thyroid cancer, 26 with DTC and seven with medullary carcinoma, showed complete response, PR and SD rates of 7, 25, and 48%, respectively Citation[113]. FDG/PET responses were reported in 36% of these patients, 88% of whom had disease control by RECIST criteria.

Pazopanib

Pazopanib is an orally bioavailable tyrosine kinase receptor inhibitor with activity at low nanomolar concentrations against VEGFR, PDGFR and c-kit Citation[114]. It inhibits the growth of a wide range of human tumor xenografts in a murine model. In a Phase II study, 39 patients with I-131 refractory DTC were treated with 800 mg/day of pazopanib in 4-week cycles, with a median 12 cycles received by patients Citation[115]. Nineteen patients (49%) had a confirmed PR, and Kaplan–Meier analysis showed a 1-year response rate of 66%.

Vemurafenib

Vemurafenib (PLX4032, RG7204 or RO5185426) is a selective inhibitor of the V600E mutant BRAF and was recently approved by the FDA for treatment of patients with unresectable or metastatic melanoma with the BRAF V600E mutation Citation[116]. Preclinical studies have shown that thyroid cancer cells are sensitive to this agent Citation[117]. An ongoing Phase II trial is testing vemurafenib in patients with papillary thyroid cancer who are positive for BRAF V600E mutations and resistant to I-131 (NCT01286753) .

Gefitinib

Gefitinib is an orally active inhibitor of epidermal growth factor signaling that blocks EGF receptor (EGFR) autophosphorylation in several EGFR-expressing human tumor cell lines Citation[118]. It is currently licensed for the treatment of patients with non-small cell lung cancer after they become refractory to platinum-based compounds and taxanes. A Phase II study of gefitinib in patients with advanced thyroid cancer, 67% of whom had DTC, showed no objective responses, but 32% of patients had small reductions in tumor volume, and five patients with SD showed reductions in serum thyroglobulin concentrations to <90% of normal Citation[119].

Axitinib

Axitinib is an oral inhibitor of VEGF but not RET. A multicenter, open-label, Phase II trial in 60 patients with I-131 resistant thyroid cancer of various histologic types showed an overall PR rate of 30% (including 31% of the patients with DTC), with a median PFS of 18 months Citation[120].

Vandetanib

Vandetanib is a VEGFR, EGFR and RET tyrosine kinase inhibitor (TKIs) and has been approved for treatment of MTC Citation[98]. It is currently being studied in patients with papillary or follicular thyroid cancer (NCT00537095) .

Everolimus

The mTOR is a serine–threonine protein kinase that regulates cellular metabolism, growth and proliferation and may play a role in thyroid carcinogenesis. Everolimus is a mTOR inhibitor and is currently being studied in a Phase II trial in patients with locally advanced or metastatic thyroid cancer (medullary, differentiated or anaplastic; NCT00936858) .

Others

A number of other agents are in various stages of development as potential treatment for patients with thyroid cancer: motesanib, axitinib, XL281, panobinostat, pasireotide and cabozantinib.

Motesanib is an oral tyrosine kinase receptor inhibitor that targets VEGF-1, -2 and -3. In a Phase II open-label trial that included 93 patients with DTC, 13 patients (14%) achieved a PR and 33 (35%) had SD Citation[121].

XL281 is an inhibitor of wild-type and mutant RAF kinase that has shown activity in a Phase I trial of patients with multiple tumor types, including PTC Citation[122].

Panobinostat (LBH589) is an inhibitor of histone deacetylase Citation[123]. Patients with DTC and medullary carcinoma of the thyroid are currently being recruited for a Phase II trial of panobinostat.

Pasireotide (SOM230) is a novel, multireceptor-targeted somatostatin analog with high-binding affinity for somatostatin receptors 1, 2, 3 and 5, suggesting it has antisecretory and potential antitumor properties Citation[123]. It is currently being studied as monotherapy and in combination with everolimus in patients with I-131 refractory DTC and in those with medullary carcinoma.

Cabozantinib (XL184) is a potent inhibitor of the tyrosine kinases MET and VEGFR-2. Preliminary results of a Phase III trial of cabozantinib in patients with medullary thyroid cancer reported that this drug met its primary outcome, improved PFS Citation[124,125], suggesting that testing of this agent may be warranted in patients with I-131 refractory DTC.

Management of AEs

TKIs are generally quite well tolerated and associated with much less toxicity than chemotherapy. AEs are evaluated, by convention, using the National Cancer Institute’s Common Terminology Criteria for Adverse Event, in which severity of each event can be classified in various grades and, according to these, the study drug is temporary or permanently withdrawn or the dosage is reduced. The most common AEs are represented by constitutional symptoms such fatigue, weight loss, diarrhea and nausea, although only rarely do they reach grade 3 or 4 severity, for which temporary discontinuation or dosage reduction is required. Hypertension is a common effect of all VEGF inhibitors; total incidence varies from 17 to 56% with grade ≥3 between 2 and 25%. Common to almost all TKIs is the occurrence of a series of cutaneous AEs including hand–foot skin reaction, mucositis, papulopustolar rash, alopecia and xerosis. Skin reaction may be attenuated or prevented using the application of cream or other topical agents.

Another common side effect with some TKIs is the increase of serum TSH that often requires an adjustment of l-tyroxine therapy. The intimate mechanism of this alteration is still unknown but an interference in thyroid hormone metabolism or a decrease in l-tyroxine absorption has been postulated.

Future directions

The paucity of current treatment options for patients with I-131 refractory DTC, together with the number of investigational targeted therapies, make study design and definitions keys to optimizing treatment with these agents. It is important to clearly define patient inclusion and exclusion criteria and to select appropriate outcome measures. Significant issues include the definitions of I-131 refractory disease, differentiation between indolent and progressive disease, and primary and secondary outcome variables . In addition, among patients with progressive disease, it is important to distinguish those who may benefit from a TKI from those for whom watchful waiting may be the best approach. Most studies of I-131 refractory patients use RECIST criteria to assess targeted and other antineoplastic strategies, although limitations associated with this morphologic assessment should be considered (e.g., necrosis and edema) Citation[39].

At present, there is sufficient evidence for the efficacy of sorafenib and lenvatinib to support Phase III trials. Future studies of these and other novel agents will include different dosing regimens and combinations with other kinase inhibitors involved in cell growth, proliferation and metastasis, such as mTOR inhibitors, somatostatin analogs and thalidomide derivatives.

Conclusion

The global incidence of thyroid cancer is increasing. Although most patients with DTC can be successfully treated by surgery and adjuvant I-131, some thyroid cancers develop resistance to I-131. In these patients, therapeutic options have been limited, are relatively ineffective and associated with significant and treatment-limiting toxicities and impaired quality of life. Therefore, patients with I-131 refractory DTC are candidates for clinical trials of various interventions, which may improve outcomes.

Advances in molecular biology have been accompanied by significant improvements in understanding the cellular alterations associated with transformation and progression of thyroid cancers. Knowledge of signaling pathways in DTC has led to the development of small molecular weight inhibitors of those pathways. Kinase inhibitors have significantly improved outcomes in patients with non-small-cell lung cancer, chronic myelogenous leukemia, hepatocellular carcinoma and renal cell carcinoma. Phase I and II trials have suggested that kinase inhibitors, either as monotherapy or in combination with other agents, may improve outcomes in patients with I-131-resistant DTC. Targeted therapies also have a narrower spectrum of toxicities than cytotoxic chemotherapy, thus improving the patients’ quality of life.

Expert commentary

The involvement of signaling pathways in the development of DTC has suggested that agents targeting these pathways may be effective in treating patients with I-131 refractory DTC. Several TKIs targeting these signal pathways have shown efficacy in the treatment of these patients. Two targeted agents, sorafenib and lenvatinib, are currently in Phase III trials in patients with I-131 refractory DTC, and several other agents are currently being tested in Phase II trials. Expansion of the therapeutic armamentarium in the treatment of I-131 refractory DTC may enhance survival of these patients.

Five-year view

Evidence to date supports Phase III trials of sorafenib and lenvatinib in patients with I-131 refractory DTC, and several other TKIs now being tested may also be shown to be effective. Future developments are directed toward the definition of the most appropriate dosage in the intent of reducing AEs and side effects, which are currently the main limitation. In addition, the problem of resistance to the drug after a period of favorable response is one of the key issues that needs to be addressed in the years to come.

Table 1. Phase II trials of sorafenib in differentiated thyroid cancers.

Table 2. Targeted therapeutic agents in late-stage clinical trials for differentiated thyroid cancer.

Table 3. Key terms and outcome measures for clinical trials of targeted therapies in patients with differentiated thyroid cancer.

Financial & competing interests disclosure

The authors were supported by Bayer HealthCare Pharmaceuticals and Onyx Pharmaceuticals, Inc. F Pitoia is a consultant to and member of the speaker’s bureau of Genzyme Corp. B Robinson is a consultant or advisor with Eisai Corporation. L Wirth is a consultant or advisor with AstraZeneca and Boehringer Ingelheim Corporations. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

The authors thank BelMed Professional Resources, NY, USA for editorial support, with funding provided by Bayer HealthCare Pharmaceuticals and Onyx Pharmaceuticals, Inc.

References

  • Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA. Cancer J. Clin. 61(2), 69–90 (2011).
  • Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA. Cancer J. Clin. 60(5), 277–300 (2010).
  • Hundahl SA, Fleming ID, Fremgen AM, Menck HR. A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995. Cancer 83(12), 2638–2648 (1998).
  • Venkatesh YS, Ordonez NG, Schultz PN, Hickey RC, Goepfert H, Samaan NA. Anaplastic carcinoma of the thyroid. A clinicopathologic study of 121 cases. Cancer 66(2), 321–330 (1990).
  • McIver B, Hay ID, Giuffrida DF et al. Anaplastic thyroid carcinoma: a 50-year experience at a single institution. Surgery 130(6), 1028–1034 (2001).
  • Byar DP, Green SB, Dor P et al. A prognostic index for thyroid carcinoma. A study of the E.O.R.T.C. Thyroid Cancer Cooperative Group. Eur. J. Cancer 15(8), 1033–1041 (1979).
  • Cady B, Rossi R. An expanded view of risk-group definition in differentiated thyroid carcinoma. Surgery 104(6), 947–953 (1988).
  • Sherman SI, Brierley JD, Sperling M et al. Prospective multicenter study of thyroiscarcinoma treatment: initial analysis of staging and outcome. National Thyroid Cancer Treatment Cooperative Study Registry Group. Cancer 83(5), 1012–1021 (1998).
  • Cooper DS, Doherty GM, Haugen BR et al; American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 19(11), 1167–1214 (2009).
  • Edge SB; American Joint Committee on Cancer. AJCC Cancer Staging Manual. Springer, New York, NY, USA (2010).
  • Hay ID, Bergstralh EJ, Goellner JR, Ebersold JR, Grant CS. Predicting outcome in papillary thyroid carcinoma: development of a reliable prognostic scoring system in a cohort of 1779 patients surgically treated at one institution during 1940 through 1989. Surgery 114(6), 1050–1057; discussion 1057 (1993).
  • Pacini F, Schlumberger M, Dralle H, Elisei R, Smit JW, Wiersinga W; European Thyroid Cancer Taskforce. European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur. J. Endocrinol. 154(6), 787–803 (2006).
  • Luster M, Clarke SE, Dietlein M et al.; European Association of Nuclear Medicine (EANM). Guidelines for radioiodine therapy of differentiated thyroid cancer. Eur. J. Nucl. Med. Mol. Imaging 35(10), 1941–1959 (2008).
  • Pitoia F, Ward L, Wohllk N et al. Recommendations of the Latin American Thyroid Society on diagnosis and management of differentiated thyroid cancer. Arq. Bras. Endocrinol. Metabol. 53(7), 884–887 (2009).
  • Takami H, Ito Y, Okamoto T, Yoshida A. Therapeutic strategy for differentiated thyroid carcinoma in Japan based on a newly established guideline managed by Japanese Society of Thyroid Surgeons and Japanese Association of Endocrine Surgeons. World J. Surg. 35(1), 111–121 (2011).
  • Ito Y, Uruno T, Nakano K et al. An observation trial without surgical treatment in patients with papillary microcarcinoma of the thyroid. Thyroid 13(4), 381–387 (2003).
  • Sugitani I, Toda K, Yamada K, Yamamoto N, Ikenaga M, Fujimoto Y. Three distinctly different kinds of papillary thyroid microcarcinoma should be recognized: our treatment strategies and outcomes. World J. Surg. 34(6), 1222–1231 (2010).
  • Mazzaferri EL, Young RL. Papillary thyroid carcinoma: a 10 year follow-up report of the impact of therapy in 576 patients. Am. J. Med. 70(3), 511–518 (1981).
  • DeGroot LJ, Kaplan EL, McCormick M, Straus FH. Natural history, treatment, and course of papillary thyroid carcinoma. J. Clin. Endocrinol. Metab. 71(2), 414–424 (1990).
  • Samaan NA, Schultz PN, Hickey RC et al. The results of various modalities of treatment of well differentiated thyroid carcinomas: a retrospective review of 1599 patients. J. Clin. Endocrinol. Metab. 75(3), 714–720 (1992).
  • Bilimoria KY, Bentrem DJ, Ko CY et al. Extent of surgery affects survival for papillary thyroid cancer. Ann. Surg. 246(3), 375–81; discussion 381 (2007).
  • Tuttle RM, Vaisman F, Tronko MD. Clinical presentation and clinical outcomes in Chernobyl-related paediatric thyroid cancers: what do we know now? What can we expect in the future? Clin. Oncol. (R. Coll. Radiol). 23(4), 268–275 (2011).
  • Udelsman R, Lakatos E, Ladenson P. Optimal surgery for papillary thyroid carcinoma. World J. Surg. 20(1), 88–93 (1996).
  • Pattou F, Combemale F, Fabre S et al. Hypocalcemia following thyroid surgery: incidence and prediction of outcome. World J. Surg. 22(7), 718–724 (1998).
  • Salvatori M, Raffaelli M, Castaldi P et al. Evaluation of the surgical completeness after total thyroidectomy for differentiated thyroid carcinoma. Eur. J. Surg. Oncol. 33(5), 648–654 (2007).
  • Wong JB, Kaplan MM, Meyer KB, Pauker SG. Ablative radioactive iodine therapy for apparently localized thyroid carcinoma. A decision analytic perspective. Endocrinol. Metab. Clin. North Am. 19(3), 741–760 (1990).
  • Sawka AM, Thephamongkhol K, Brouwers M, Thabane L, Browman G, Gerstein HC. Clinical review 170: a systematic review and metaanalysis of the effectiveness of radioactive iodine remnant ablation for well-differentiated thyroid cancer. J. Clin. Endocrinol. Metab. 89(8), 3668–3676 (2004).
  • Verburg FA, Stokkel MP, Düren C et al. No survival difference after successful (131)I ablation between patients with initially low-risk and high-risk differentiated thyroid cancer. Eur. J. Nucl. Med. Mol. Imaging 37(2), 276–283 (2010).
  • Hackshaw A, Harmer C, Mallick U, Haq M, Franklyn JA. 131I activity for remnant ablation in patients with differentiated thyroid cancer: a systematic review. J. Clin. Endocrinol. Metab. 92(1), 28–38 (2007).
  • Pilli T, Brianzoni E, Capoccetti F et al. A comparison of 1850 (50 mCi) and 3700 MBq (100 mCi) 131-iodine administered doses for recombinant thyrotropin-stimulated postoperative thyroid remnant ablation in differentiated thyroid cancer. J. Clin. Endocrinol. Metab. 92(9), 3542–3546 (2007).
  • Rosario PW, Xavier AC. Recombinant human thyroid stimulating hormone in thyroid remnant ablation with 1.1 GBq 131iodine in low-risk patients. Am. J. Clin. Oncol. 35(2), 101–104 (2012).
  • Maxon HR 3rd, Englaro EE, Thomas SR et al. Radioiodine-131 therapy for well-differentiated thyroid cancer – a quantitative radiation dosimetric approach: outcome and validation in 85 patients. J. Nucl. Med. 33(6), 1132–1136 (1992).
  • Liel Y. Preparation for radioactive iodine administration in differentiated thyroid cancer patients. Clin. Endocrinol. (Oxf) 57(4), 523–527 (2002).
  • Robbins RJ, Larson SM, Sinha N et al. A retrospective review of the effectiveness of recombinant human TSH as a preparation for radioiodine thyroid remnant ablation. J. Nucl. Med. 43(11), 1482–1488 (2002).
  • Matovic MD, Jankovic SM, Jeremic M, Tasic Z, Vlajkovic M. Unexpected effect of furosemide on radioiodine urinary excretion in patients with differentiated thyroid carcinomas treated with iodine 131. Thyroid 19(8), 843–848 (2009).
  • Schlumberger M, Sherman SI. Approach to the patient with advanced differentiated thyroid cancer. Eur. J. Endocrinol. 166(1), 5–11 (2012).
  • Jonklaas J, Sarlis NJ, Litofsky D et al. Outcomes of patients with differentiated thyroid carcinoma following initial therapy. Thyroid 16(12), 1229–1242 (2006).
  • Pfister DG, Fagin JA. Refractory thyroid cancer: a paradigm shift in treatment is not far off. J. Clin. Oncol. 26(29), 4701–4704 (2008).
  • Eisenhauer EA, Therasse P, Bogaerts J et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45(2), 228–247 (2009).
  • O’Connell ME, A’Hern RP, Harmer CL. Results of external beam radiotherapy in differentiated thyroid carcinoma: a retrospective study from the Royal Marsden Hospital. Eur. J. Cancer 30A(6), 733–739 (1994).
  • Schwartz DL, Lobo MJ, Ang KK et al. Postoperative external beam radiotherapy for differentiated thyroid cancer: outcomes and morbidity with conformal treatment. Int. J. Radiat. Oncol. Biol. Phys. 74(4), 1083–1091 (2009).
  • Sherman SI. Cytotoxic chemotherapy for differentiated thyroid carcinoma. Clin. Oncol. (R. Coll. Radiol). 22(6), 464–468 (2010).
  • Carter SK, Blum RH. New chemotherapeutic agents – bleomycin and adriamycin. CA. Cancer J. Clin. 24(6), 322–331 (1974).
  • Gottlieb JA, Hill CS Jr. Chemotherapy of thyroid cancer with adriamycin. Experience with 30 patients. N. Engl. J. Med. 290(4), 193–197 (1974).
  • Haugen BR. Management of the patient with progressive radioiodine non-responsive disease. Semin. Surg. Oncol. 16(1), 34–41 (1999).
  • Matuszczyk A, Petersenn S, Bockisch A et al. Chemotherapy with doxorubicin in progressive medullary and thyroid carcinoma of the follicular epithelium. Horm. Metab. Res. 40(3), 210–213 (2008).
  • Shimaoka K, Schoenfeld DA, DeWys WD, Creech RH, DeConti R. A randomized trial of doxorubicin versus doxorubicin plus cisplatin in patients with advanced thyroid carcinoma. Cancer 56(9), 2155–2160 (1985).
  • Biganzoli L, Gebbia V, Maiorino L, Caraci P, Iirillo A. Thyroid cancer: different outcomes to chemotherapy according to tumour histology. Eur. J. Cancer 31A(13–14), 2423–2424 (1995).
  • Argiris A, Agarwala SS, Karamouzis MV, Burmeister LA, Carty SE. A Phase II trial of doxorubicin and interferon a 2b in advanced, non-medullary thyroid cancer. Invest. New Drugs 26(2), 183–188 (2008).
  • Ikeda M, Tanaka K, Sonoo H et al. Docetaxel administration for radioiodine-resistant patients with metastatic papillary thyroid carcinoma. Gan To Kagaku Ryoho. 34(6), 933–936 (2007).
  • Matuszczyk A, Petersenn S, Voigt W et al. Chemotherapy with paclitaxel and gemcitabine in progressive medullary and thyroid carcinoma of the follicular epithelium. Horm. Metab. Res. 42(1), 61–64 (2010).
  • Fagin JA, Mitsiades N. Molecular pathology of thyroid cancer: diagnostic and clinical implications. Best Pract. Res. Clin. Endocrinol. Metab. 22(6), 955–969 (2008).
  • Ciampi R, Nikiforov YE. RET/PTC rearrangements and BRAF mutations in thyroid tumorigenesis. Endocrinology 148(3), 936–941 (2007).
  • Liu ZM, Wu TT, van Hasselt CA, Chen GG. Carcinogenesis and therapeutic strategies in thyroid cancer. Curr. Drug Targets 11(6), 716–732 (2010).
  • Xing M. Genetic alterations in the phosphatidylinositol-3 kinase/Akt pathway in thyroid cancer. Thyroid 20(7), 697–706 (2010).
  • Indo Y, Mardy S, Tsuruta M, Karim MA, Matsuda I. Structure and organization of the human TRKA gene encoding a high affinity receptor for nerve growth factor. Jpn. J. Hum. Genet. 42(2), 343–351 (1997).
  • Wang H, Hughes I, Planer W et al. The timing and location of glial cell line-derived neurotrophic factor expression determine enteric nervous system structure and function. J. Neurosci. 30(4), 1523–1538 (2010).
  • Santoro M, Grieco M, Melillo RM, Fusco A, Vecchio G. Molecular defects in thyroid carcinomas: role of the RET oncogene in thyroid neoplastic transformation. Eur. J. Endocrinol. 133(5), 513–522 (1995).
  • Bongarzone I, Vigneri P, Mariani L, Collini P, Pilotti S, Pierotti MA. RET/NTRK1 rearrangements in thyroid gland tumors of the papillary carcinoma family: correlation with clinicopathological features. Clin. Cancer Res. 4(1), 223–228 (1998).
  • Adjei AA. Blocking oncogenic Ras signaling for cancer therapy. J. Natl Cancer Inst. 93(14), 1062–1074 (2001).
  • Zhu Z, Gandhi M, Nikiforova MN, Fischer AH, Nikiforov YE. Molecular profile and clinical–pathologic features of the follicular variant of papillary thyroid carcinoma. An unusually high prevalence of ras mutations. Am. J. Clin. Pathol. 120(1), 71–77 (2003).
  • Lupi C, Giannini R, Ugolini C et al. Association of BRAF V600E mutation with poor clinicopathological outcomes in 500 consecutive cases of papillary thyroid carcinoma. J. Clin. Endocrinol. Metab. 92(11), 4085–4090 (2007).
  • Elisei R, Ugolini C, Viola D et al. BRAF(V600E) mutation and outcome of patients with papillary thyroid carcinoma: a 15-year median follow-up study. J. Clin. Endocrinol. Metab. 93(10), 3943–3949 (2008).
  • Xing M. Prognostic utility of BRAF mutation in papillary thyroid cancer. Mol. Cell. Endocrinol. 321(1), 86–93 (2010).
  • Ito Y, Yoshida H, Maruo R et al. BRAF mutation in papillary thyroid carcinoma in a Japanese population: its lack of correlation with high-risk clinicopathological features and disease-free survival of patients. Endocr. J. 56(1), 89–97 (2009).
  • Viglietto G, Chiappetta G, Martinez-Tello FJ et al. RET/PTC oncogene activation is an early event in thyroid carcinogenesis. Oncogene 11(6), 1207–1210 (1995).
  • Giannini R, Ugolini C, Lupi C et al. The heterogeneous distribution of BRAF mutation supports the independent clonal origin of distinct tumor foci in multifocal papillary thyroid carcinoma. J. Clin. Endocrinol. Metab. 92(9), 3511–3516 (2007).
  • Ugolini C, Giannini R, Lupi C et al. Presence of BRAF V600E in very early stages of papillary thyroid carcinoma. Thyroid 17(5), 381–388 (2007).
  • Henderson YC, Shellenberger TD, Williams MD et al. High rate of BRAF and RET/PTC dual mutations associated with recurrent papillary thyroid carcinoma. Clin. Cancer Res. 15(2), 485–491 (2009).
  • He H, Jazdzewski K, Li W et al. The role of microRNA genes in papillary thyroid carcinoma. Proc. Natl Acad. Sci. USA 102(52), 19075–19080 (2005).
  • Hu S, Liu D, Tufano RP et al. Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. Int. J. Cancer 119(10), 2322–2329 (2006).
  • Gudmundsson J, Sulem P, Gudbjartsson DF et al. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nat. Genet. 41(4), 460–464 (2009).
  • Jazdzewski K, de la Chapelle A. Genomic sequence matters: a SNP in microRNA-146a can turn anti-apoptotic. Cell Cycle 8(11), 1642–1643 (2009).
  • Canzian F, Amati P, Harach HR et al. A gene predisposing to familial thyroid tumors with cell oxyphilia maps to chromosome 19p13.2. Am. J. Hum. Genet. 63(6), 1743–1748 (1998).
  • Malchoff CD, Sarfarazi M, Tendler B et al. Papillary thyroid carcinoma associated with papillary renal neoplasia: genetic linkage analysis of a distinct heritable tumor syndrome. J. Clin. Endocrinol. Metab. 85(5), 1758–1764 (2000).
  • McKay JD, Lesueur F, Jonard L et al. Localization of a susceptibility gene for familial nonmedullary thyroid carcinoma to chromosome 2q21. Am. J. Hum. Genet. 69(2), 440–446 (2001).
  • Capezzone M, Cantara S, Marchisotta S et al. Telomere length in neoplastic and nonneoplastic tissues of patients with familial and sporadic papillary thyroid cancer. J. Clin. Endocrinol. Metab. 96(11), e1852–e1856 (2011).
  • Di Palma T, Conti A, de Cristofaro T, Scala S, Nitsch L, Zannini M. Identification of novel Pax8 targets in FRTL-5 thyroid cells by gene silencing and expression microarray analysis. PLoS ONE 6(9), e25162 (2011).
  • Koenig RJ. Detection of the PAX8-PPARγ fusion protein in thyroid tumors. Clin. Chem. 56(3), 331–333 (2010).
  • Marques AR, Espadinha C, Catarino AL et al. Expression of PAX8-PPAR gamma 1 rearrangements in both follicular thyroid carcinomas and adenomas. J. Clin. Endocrinol. Metab. 87(8), 3947–3952 (2002).
  • Terrier P, Sheng ZM, Schlumberger M et al. Structure and expression of c-myc and c-fos proto-oncogenes in thyroid carcinomas. Br. J. Cancer 57(1), 43–47 (1988).
  • Lemoine NR, Mayall ES, Wyllie FS et al. High frequency of ras oncogene activation in all stages of human thyroid tumorigenesis. Oncogene 4(2), 159–164 (1989).
  • Wright PA, Lemoine NR, Mayall ES et al. Papillary and follicular thyroid carcinomas show a different pattern of ras oncogene mutation. Br. J. Cancer 60(4), 576–577 (1989).
  • Wu G, Mambo E, Guo Z et al. Uncommon mutation, but common amplifications, of the PIK3CA gene in thyroid tumors. J. Clin. Endocrinol. Metab. 90(8), 4688–4693 (2005).
  • Liu Z, Hou P, Ji M et al. 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. J. Clin. Endocrinol. Metab. 93(8), 3106–3116 (2008).
  • Xing M, Cohen Y, Mambo E et al. Early occurrence of RASSF1A hypermethylation and its mutual exclusion with BRAF mutation in thyroid tumorigenesis. Cancer Res. 64(5), 1664–1668 (2004).
  • Gild ML, Bullock M, Robinson BG, Clifton-Bligh R. Multikinase inhibitors: a new option for the treatment of thyroid cancer. Nat. Rev. Endocrinol. 7(10), 617–624 (2011).
  • Sherman SI. Tyrosine kinase inhibitors and the thyroid. Best Pract. Res. Clin. Endocrinol. Metab. 23(6), 713–722 (2009).
  • Ye L, Santarpia L, Gagel RF. The evolving field of tyrosine kinase inhibitors in the treatment of endocrine tumors. Endocr. Rev. 31(4), 578–599 (2010).
  • Xing M, Westra WH, Tufano RP et al. BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J. Clin. Endocrinol. Metab. 90(12), 6373–6379 (2005).
  • Tuttle RM, Fleisher M, Francis GL, Robbins RJ. Serum vascular endothelial growth factor levels are elevated in metastatic differentiated thyroid cancer but not increased by short-term TSH stimulation. J. Clin. Endocrinol. Metab. 87(4), 1737–1742 (2002).
  • O’Neill CJ, Oucharek J, Learoyd D, Sidhu SB. Standard and emerging therapies for metastatic differentiated thyroid cancer. Oncologist 15(2), 146–156 (2010).
  • Riley LB, Desai DC. The molecular basis of cancer and the development of targeted therapy. Surg. Clin. North Am. 89(1), 1–15, vii (2009).
  • Sherman SI. Targeted therapies for thyroid tumors. Mod. Pathol. 24(Suppl. 2), S44–S52 (2011).
  • Deshpande HA, Gettinger SN, Sosa JA. Targeted therapy for thyroid cancer: an updated review of investigational agents. Curr. Opin. Investig. Drugs 11(6), 661–668 (2010).
  • Cabanillas ME, Waguespack SG, Bronstein Y et al. Treatment with tyrosine kinase inhibitors for patients with differentiated thyroid cancer: the M. D. Anderson experience. J. Clin. Endocrinol. Metab. 95(6), 2588–2595 (2010).
  • FDA. In: Guidance for industry: clinical trial endpoints for the approval of cancer drugs and biologics. FDA, MD, USA, (2007).
  • Wells SA Jr, Robinson BG, Gagel RF et al. Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind Phase III trial. J. Clin. Oncol. 30(2), 134–141 (2012).
  • Kim A, Balis FM, Widemann BC. Sorafenib and sunitinib. Oncologist 14(8), 800–805 (2009).
  • Gupta-Abramson V, Troxel AB, Nellore A et al. Phase II trial of sorafenib in advanced thyroid cancer. J. Clin. Oncol. 26(29), 4714–4719 (2008).
  • Keefe SM, Troxel AB, Rhee S et al. Phase II trial of sorafenib in patients with advanced thyroid cancer J. Clin. Oncol. 29(Suppl. 15), abstract 5562 (2011).
  • Kloos RT, Ringel MD, Knopp MV et al. Phase II trial of sorafenib in metastatic thyroid cancer. J. Clin. Oncol. 27(10), 1675–1684 (2009).
  • Ahmed M, Barbachano Y, Riddell A et al. Analysis of the efficacy and toxicity of sorafenib in thyroid cancer: a Phase II study in a UK based population. Eur. J. Endocrinol. 165(2), 315–322 (2011).
  • Hoftijzer H, Heemstra KA, Morreau H et al. Beneficial effects of sorafenib on tumor progression, but not on radioiodine uptake, in patients with differentiated thyroid carcinoma. Eur. J. Endocrinol. 161(6), 923–931 (2009).
  • Capdevila J, Iglesias L, Halperin I et al. Sorafenib in metastatic thyroid cancer. Endocr. Relat. Cancer 19(2), 209–216 (2012).
  • Brose MS, Nutting CM, Sherman SI et al. Rationale and design of decision: a double-blind, randomized, placebo-controlled Phase III trial evaluating the efficacy and safety of sorafenib in patients with locally advanced or metastatic radioactive iodine (RAI)-refractory, differentiated thyroid cancer. BMC Cancer 11, 349 (2011).
  • Glen H, Mason S, Patel H, Macleod K, Brunton VG. E7080, a multi-targeted tyrosine kinase inhibitor suppresses tumor cell migration and invasion. BMC Cancer 11, 309 (2011).
  • Sherman SI, Jarzab B, Cabanillas ME et al. A Phase II trial of the multitargeted kinase inhibitor E7080 in advanced radioiodine (RAI)-refractory differentiated thyroid cancer (DTC). J. Clin. Oncol. 29(Suppl. 15), abstract 5503 (2011).
  • Kim DW, Jo YS, Jung HS et al. An orally administered multitarget tyrosine kinase inhibitor, SU11248, is a novel potent inhibitor of thyroid oncogenic RET/papillary thyroid cancer kinases. J. Clin. Endocrinol. Metab. 91(10), 4070–4076 (2006).
  • Dawson SJ, Conus NM, Toner GC et al. Sustained clinical responses to tyrosine kinase inhibitor sunitinib in thyroid carcinoma. Anticancer Drugs 19(5), 547–552 (2008).
  • Cohen EE, Needles BM, Cullen KJ, et al. Phase 2 study of sunitinib in refractory thyroid cancer. J. Clin. Oncol. 26(Suppl. 15), abstract 6025 (2008).
  • Ravaud A, de la Fouchardière C, Asselineau J et al. Efficacy of sunitinib in advanced medullary thyroid carcinoma: intermediate results of Phase II THYSU. Oncologist 15(2), 212–213; author reply 214 (2010).
  • Carr LL, Mankoff DA, Goulart BH et al. Phase II study of daily sunitinib in FDG-PET-positive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation. Clin. Cancer Res. 16(21), 5260–5268 (2010).
  • Kumar R, Knick VB, Rudolph SK et al. Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol. Cancer Ther. 6(7), 2012–2021 (2007).
  • Bible KC, Suman VJ, Molina JR et al.; Endocrine Malignancies Disease Oriented Group; Mayo Clinic Cancer Center; Mayo Phase 2 Consortium. Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a Phase 2 consortium study. Lancet Oncol. 11(10), 962–972 (2010).
  • Chapman PB, Hauschild A, Robert C et al; BRIM-3 Study Group. Improved survival with vemurafenib in melamona with BRAF V600E mutation. N. Engl. J. Med. 364(26), 2507–2616 (2011).
  • Salerno P, De Falco V, Tamburrino A et al. Cytostatic activity of adenosine triphosphate-completitive kinase inhibitors in BRAF mutant thyroid carcinoma cells. J. Clin. Endocrinol. Metab. 95(1), 450–455 (2010).
  • Wakeling AE, Guy SP, Woodburn JR et al. ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res. 62(20), 5749–5754 (2002).
  • Pennell NA, Daniels GH, Haddad RI et al. A Phase II study of gefitinib in patients with advanced thyroid cancer. Thyroid 18(3), 317–323 (2008).
  • Cohen EE, Rosen LS, Vokes EE et al. Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: results from a Phase II study. J. Clin. Oncol. 26(29), 4708–4713 (2008).
  • Sherman SI, Wirth LJ, Droz JP et al.; Motesanib Thyroid Cancer Study Group. Motesanib diphosphate in progressive differentiated thyroid cancer. N. Engl. J. Med. 359(1), 31–42 (2008).
  • Schwartz GK, Robertson S, Shen A et al. A Phase I study of XL281, a selective oral RAF kinase inhibitor, in patients (Pts) with advanced solid tumors. J. Clin. Oncol. 27(Suppl. 15), abstract 3513 (2009).
  • Catalano MG, Pugliese M, Gargantini E et al. Cytotoxic activity of the histone deacetylase inhibitor panobinostat (LBH589) in anaplastic thyroid cancer in vitro and in vivo. Int. J. Cancer 130(3), 694–704 (2012).
  • Kurzrock R, Sherman SI, Ball DW et al. Activity of XL184 (Cabozantinib), an oral tyrosine kinase inhibitor, in patients with medullary thyroid cancer. J. Clin. Oncol. 29(19), 2660–2666 (2011).
  • Houvras Y, Wirth LJ. Cabozantinib in medullary thyroid carcinoma: time to focus the spotlight on this rare disease. J. Clin. Oncol. 29(19), 2616–2618 (2011).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.