Thyroid cancer is the most common endocrine malignancy. Differentiated thyroid cancer (DTC) with the two subtypes, papillary thyroid cancer (PTC) and follicular thyroid cancer (FTC), is the most frequent subtype of thyroid cancer; more rare subtypes are medullary thyroid cancer (MTC) and anaplastic thyroid cancer (ATC). The incidence of DTC has increased rapidly in recent years due to the more frequent use of imaging methods such as ultrasound of the neck and fine-needle aspiration (FNA) of thyroid nodules. After total thyroidectomy and radioiodine treatment, DTC remains an indolent and curable disease in most patients, whereas the cure rate in MTC is lower and depends on early diagnosis. Most ATCs are incurable. In recent years, there has been great progress in identifying genetic changes in thyroid cancer, and genetic testing of FNA samples or blood samples provides useful information for clinical decision making. Tumor staging, either postoperatively or by imaging, and measuring the tumor markers thyroglobulin for DTC and calcitonin for MTC, allow for dynamic risk-adapted stratification for follow-up procedures. In advanced metastatic thyroid cancer, molecular targeted therapy using tyrosine kinase receptor inhibitors, including sorafenib, lenvantinib, vandetanib, and cabozantinib, helps control tumor progression and prolongs progression-free survival. Using a dynamic risk-stratified approach to manage thyroid cancer, the outcomes for most thyroid cancer patients are excellent compared with those for other cancers. The major challenge in the future is to identify high-risk patients and to treat and monitor them appropriately. Clin Cancer Res; 22(20); 5012–21. ©2016 AACR.

See all articles in this CCR Focus section, “Endocrine Cancers: Revising Paradigms.”

In the past three decades, the number of people diagnosed with thyroid cancer worldwide has increased dramatically (1–4). In 2011, there were more than 500,000 people with thyroid cancer in the United States, and it is projected that in 2016, there will be more than 64,000 new cases and more than 1,980 thyroid cancer–associated deaths in the United States (5). The yearly incidence of thyroid cancer has risen over the past 40 years from 4.9 per 100,000 in 1975 to 14.3 per 100,000 in 2009 (6), but the age- and sex-adjusted annual death rate of 0.5 per 100,000 has remained stable (7). The increased incidence is due almost entirely to an increase in the incidence of papillary thyroid cancer (PTC; ref. 8); in turn, the PTC increase is due mainly to the increasing use of neck ultrasonography (U.S.), to the improved feasibility of performing US-guided fine-needle aspiration (FNA) biopsy of very small nodules, to the increase in thyroid surgery that reveals occult cancers, and to better histologic analysis of surgical specimens (9, 10). The increased incidence is much more pronounced for small indolent cancers (<2 cm in diameter; ref. 11). In industrialized countries, around 40% of all treated thyroid carcinomas are microcarcinomas (<1 cm in diameter) with excellent long-term prognoses (12), which is in line with autopsy findings that occult papillary thyroid microcarcinomas are present in 4% to 36% of cases (a mean prevalence of 11.5%; ref. 13). The incidence of other types of thyroid cancer, including follicular thryoid cancer (FTC) and medullary thyroid cancer (MTC), and aggressive subtypes such as poorly differentiated follicular and anaplastic carcinomas (ATC), has probably not increased. 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 to 40 years for PTC and FTC, respectively; 65% for MTC after 10 years; less than 20% for poorly differentiated thyroid cancer (DTC) at 5 years; and less than 10% for ATC at 6 months after the initial diagnosis (14).

These trends should be considered when deciding upon the initial treatment and follow-up protocol for patients with thyroid cancer. One major goal is to minimize the potential harm from overtreatment in the majority of patients who are at low risk of disease-specific mortality and morbidity while appropriately treating and monitoring patients who are at higher risk. These changes and the need for new risk-stratified approaches for patients with thyroid cancer have recently prompted new guidelines in many countries (15–22).

Thyroid nodules, which are mostly benign, are very common in the general population. Most are diagnosed as unexpected asymptomatic thyroid tumors that are discovered while investigating unrelated conditions (12, 23). There is a female preponderance and an increase in prevalence with age, reaching 30% to 40% in individuals more than 50 years old. The prevalence of thyroid nodules increases with advancing age, while the risk that such nodules are malignant decreases. Nonetheless, when thyroid cancer is detected in older individuals, a higher risk histologic phenotype is more likely. Older patients are more likely to have higher risk PTC variants, poorly differentiated cancer, or ATC (24).

All patients with a thyroid nodule, regardless of the mode of detection, should undergo a dedicated neck US for quantitative risk stratification. Microcalcifications, irregular margins, and a taller-than-wide shape are the features with the highest specificities (>70%–90%) for thyroid cancer, although the sensitivities are significantly lower for any single feature (25, 26). Notably, no single US feature and no single US feature combination is sensitive or specific enough to identify malignancy (27). However, thyroid US is widely used to stratify the risk of malignancy for thyroid nodules and to aid in decision making about whether FNA is indicated.

Diagnostic FNA should be performed on nodules >1 cm with a suspicious US pattern. Thyroid nodule FNA cytology should be reported using the diagnostic groups outlined in the Bethesda System for Reporting Thyroid Cytopathology, which has six diagnostic categories that range from nondiagnostic to malignant (28, 29). Surgery is generally recommended if the cytology results suggest primary thyroid malignancy. For thyroid nodules that are classified as indeterminate on FNA biopsy, molecular testing can be used to determine whether a nodule is likely to be benign or malignant (30).

Considerable progress has been made in understanding the molecular mechanisms of thyroid cancer in the past 20 years (Fig. 1). Common driver abnormalities in PTC responsible for initiation of thyroid cancer development center around constitutive activation of the MAPK cellular signaling pathway through mutational activation of BRAF, NRAS, and gene rearrangements, including those involving the tyrosine kinase Ret (RET/PTC; refs. 31–33). FTC is frequently linked to activation of the PI3K and MAPK pathways, through loss of PTEN expression, NRAS mutations, rearrangements such as PPARγ/PAX8, and other events (31, 32). ATCs are more genomically complex, frequently harboring multiple abnormalities including genes encoding tyrosine kinase receptors (TKR)—such as VEGFR, MET, EGFR, PDGFR, KIT, and PI3K/AKT pathway kinases, including PIK3CA, PIK3CB, 3 phosphoinositide–dependent protein kinase 1 (PDPK1)—and increased expression of stem cell markers (34). Finally, MTC, a neoplasm arising from the C cells, is caused most frequently by either germline (familial MTC or multiple endocrine neoplasia type 2A) or somatic activating mutations in RET or RAS genes (35–37). RAS mutations did not overlap with mutations in RET in sporadic MTC, which indicates that each type of mutation is an alternative driver event for MTC (38). Despite the many genetic alterations that have been described for thyroid cancer and the most recent efforts to find other activated oncogenes, approximately 5% to 10% of PTCs, 50% to 60% of MTCs, and 10% of ATCs are still negative for all known genetic abnormalities (38, 39).

Figure 1.

Signaling pathways implicated in thyroid carcinogenesis and possible targets for therapeutic interventions. The two pathways (RAS/RAF/MEK/ERK and PI3K/AKT/mTOR) are involved in the propagation of signals from the cell membrane tyrosine kinase receptors (RET, EGF, VEGF, PDGF) into the nucleus. Gene alteration in the RAF/RAS/MEK pathway leads to promotion of cell proliferation, cell growth, and angiogenesis and loss of differentiation, while mutation in the PI3K/AKT/mTOR pathway results in tumor progression. Red arrows show the targets of the therapeutic agents.

Figure 1.

Signaling pathways implicated in thyroid carcinogenesis and possible targets for therapeutic interventions. The two pathways (RAS/RAF/MEK/ERK and PI3K/AKT/mTOR) are involved in the propagation of signals from the cell membrane tyrosine kinase receptors (RET, EGF, VEGF, PDGF) into the nucleus. Gene alteration in the RAF/RAS/MEK pathway leads to promotion of cell proliferation, cell growth, and angiogenesis and loss of differentiation, while mutation in the PI3K/AKT/mTOR pathway results in tumor progression. Red arrows show the targets of the therapeutic agents.

Close modal

Mutations identified may have diagnostic and prognostic implications, and provide an opportunity to develop therapies that are targeted at these potential molecular drivers. BRAF mutations are present in 30% to 67% of PTCs and are associated with locoregional metastases, extrathyroidal extension, and higher AJCC stage at presentation (refs. 40–43; Fig. 2). Both BRAF and TERT promoter mutations, which in one study were present in 13% of 242 PTCs, were associated with the clinicopathologic features of high-risk thyroid cancer (44). These and other genetic mutations and rearrangements, such as those affecting RAS, RET/PTC, and PAX8/PPARϒ, are now used as molecular markers and included in a multigene mutational panel investigated in FNA or surgically resected specimens. Using this gene panel to analyze thyroid nodules with indeterminate cytology (Bethesda system) showed 91% sensitivity and 92% specificity for cancer detection, and a 97% negative predictive value and a 77% positive predictive value (15, 45). Another approach to molecular diagnosis of intermediate thyroid nodules is the gene expression classifier using extracted RNA and analysis of 167 transcripts (46). The gene expression classifier has a sensitivity of 92% and 52% specificity, negative predictive values of 95%, and negative predictive value of 47%, respectively, for detecting benign nodules (46). Additional studies validating the mutation panels are ongoing (47, 48); long-term outcome data from a strategy of using molecular markers in indeterminate FNA specimens to stratify surgical approach are currently lacking (15).

Figure 2.

Main subtypes of thyroid cancer and most prevalent oncogenic driving lesions (in percentages; ref. 110).

Figure 2.

Main subtypes of thyroid cancer and most prevalent oncogenic driving lesions (in percentages; ref. 110).

Close modal

Once thyroid cancer is highly suspected or diagnosed, a decision must be made regarding the extent of surgery. Risk factors must be taken into consideration, like clinical risk factors associated with aggressive tumor behavior, the patient's age and sex, the initial tumor size and location, the presence of lymph node and/or distant metastases, cytologic and mutational data, and patient preferences. A positive test for BRAF mutations means a close to 100% probability of malignancy (49, 50)—this is likely helpful to guide the extent of thyroidectomy.

The treatment for thyroid cancer is predominantly surgical, and total thyroidectomy with preservation of the recurrent laryngeal nerve and parathyroid glands is generally considered standard. This achieves disease clearance and minimizes the risk of thyroid bed recurrence. A second aim of primary surgery is preparing the patient for adjuvant radioactive iodine (RAI) therapy by removing all thyroid tissue. This allows optimal follow-up using thyroglobulin (TG) as a tumor marker and addresses concerns about multifocal disease within the gland. PTC commonly metastasizes to the central neck, followed by the lateral neck. If this is documented in the preoperative work-up, then compartment-oriented lymph node neck dissection is recommended (15).

The decision to use standard aggressive surgical treatment remains controversial due to the excellent outcomes for most patients with DTC, irrespective of the nature of the surgical procedure (refs. 51, 52; Fig. 3). The treatment approaches recommended by the new American Thyroid Association (ATA) guidelines are more conservative (15). High-risk patients are treated aggressively, whereas less-aggressive approaches may be suitable for low-risk patients; indeed, some patients with the lowest risk disease (micropapillary carcinoma distant from the recurrent nerve or trachea) may be candidates for an observational approach (53, 54) or thyroid lobectomy (15). Complication rates associated with lobectomy are roughly half of those reported with total thyroidectomy. By balancing all of the tumor-, clinician-, and patient-related factors, a risk-adapted approach can be used to tailor a treatment plan for each patient to optimize outcomes on a case-by-case basis.

Figure 3.

Surgical and RIA treatment in patients with differentiated thyroid cancer. *, for further details of risk classification, see the American Thyroid Association guidelines (15).

Figure 3.

Surgical and RIA treatment in patients with differentiated thyroid cancer. *, for further details of risk classification, see the American Thyroid Association guidelines (15).

Close modal

Traditionally, RAI treatment has been used in all patients with DTC to ablate residual thyroid tissue and to postoperatively eradicate possible residual cancer, thereby decreasing the long-term risk of recurrent disease (55, 56). It should not be used in patients with ATC, even if they have DTC in addition to ATC in the pathology. It can also be used to identify and treat patients with distant metastatic disease that is sensitive to RAI. Side effects are common with I-131 therapy, including salivary gland dysfunction (>40%), abnormally dry eyes (25%), transient fertility reduction (20%), transient leukopenia, and thrombocytopenia (57). Guidelines now recommend a selective use of RAI, based on a risk-adapted, individualized approach, although RAI is still recommended in patients with aggressive primary lesions or metastatic disease in the neck or beyond. RAI remnant ablation is not recommended (tumor diameter <1 cm) or not routinely recommended (tumor diameter 1–4 cm) after lobectomy or total thyroidectomy for patients with unifocal papillary microcarcinoma in the absence of other adverse features (15).

After initial therapy, all patient data must be considered to determine follow-up treatment, including information obtained prior to surgery and the intra- and postoperative findings. These data are essential components for initial risk stratification. In the future, molecular testing results will be incorporated into this process, as, for example, a TERT mutation is an independent predictor of mortality for all differentiated cancers and for papillary carcinomas (58). Older staging systems, such as EORTC, AGES, AMES, MACES, and MSK, based mainly on the extent of tumor and age shortly after initial therapy, provide good risk stratification, but they fail to predict the risk of recurrence (59–62). The ATA guidelines include the results of postoperative US, postablative whole-body scan (WBS) if done, serum TG measurement, and, in cases where available, analysis of BRAF and/or TERT status for initial risk estimation (15). Patients are classified as low, intermediate, or high risk of recurrence, and this is modified as new data are collected during follow-up (15).

The initial follow-up plan for low-risk patients (inconspicuous US of the neck and serum TG <0.2 pg/mL) includes a visit 6 to 12 months after the initial risk assessment, with a target thyroid-stimulating hormone (TSH) level of 0.5 to 1.5 mIU/L for thyroid hormone therapy. Diagnostic RAI scans are seldom needed in these patients because nearly all recurrences can be identified by serum TG and neck US. The primary goal of early follow-up for low-risk patients is to identify patients that demonstrate an excellent response to therapy (remission) and can quickly be transitioned to a much less-intense follow-up program.

Intermediate-risk patients (TG level >5 ng/mL) are initially followed at 6-month intervals with an US and a target TSH in the 0.1 to 0.5 mIU/L range. A nonstimulated elevated TG level at 6 or more weeks after a completion thyroidectomy alerts the clinician to look for residual thyroid tissue or metastatic disease that remains after surgery (63). Diagnostic RAI scans may be used to characterize the functional status of structural disease identified during follow-up or to localize the source of markedly elevated or rising serum TG levels. Additional imaging studies may be necessary. The goal of follow-up for intermediate-risk patients is to identify the 30% of intermediate-risk patients that rapidly go into remission and can be transitioned to less-intense follow-up and the 70% of patients that do not go into remission and who might benefit from additional observation, imaging, or intervention.

The majority of high-risk patients have persistent disease after initial therapy. Therefore, the dynamics, intensity, and type of imaging used over the course of the first year vary by individual but should occur on average every 3 months. The target TSH level in high-risk patients is usually 0.1 mIU/L. Diagnostic RAI scans are frequently used to follow up these patients, particularly if RAI avid disease was identified previously.

The follow-up management strategy should be designed to optimize follow-up frequency and to determine the extent of additional testing that is needed to identify persistent or recurrent disease in a timely fashion (dynamic risk assessment; 64–67). During follow-up, the response to therapy is classified as an excellent, biochemical incomplete, structural incomplete, or indeterminate response. An excellent response is defined as no biochemical, structural, or functional evidence of disease, with a risk of recurrence of 1% to 2%. Patients with a biochemical incomplete response to therapy have an abnormal serum TG level without structurally identifiable disease. If the serum TG is increasing, then additional imaging is warranted, depending on the TG level and its increase over time; this is often expressed as the TG doubling time (68). Patients with structurally identifiable disease are classified as having a structural incomplete response to therapy with disease-specific prognostic outcomes (65). In such patients, further therapy is necessary and depends on the location of metastases, the rate of progression, the RAI avidity, and the response to previous therapies. Localized treatments can be considered for patients with progressive disease, thereby delaying the need for systemic treatment [e.g., external beam radiation or embolization for bone or liver metastases in patients in whom these are symptomatic or likely to cause morbidity (15)]. Decision making requires that the clinician and the patient carefully weigh the risks and benefits of additional therapy, as the majority of patients will not be disease free, even after additional treatment.

MTC accounts for less than 5% of thyroid cancer and has some special features: it arises from the C cells of the thyroid, which do not accumulate radioiodine; it secretes calcitonin (Ctn), which is used as a tumor marker; and 25% are part of the autosomal-dominant syndrome, multiple endocrine neoplasia type 2 (MEN2), which is caused by germline-activating mutations in the RET proto-oncogene (69). These features allow early recognition of sporadic MTC using Ctn screening in patients with thyroid nodules and preclinical diagnosis of patients with MEN2 by RET gene analysis. The ATA guidelines recommend that physicians decide whether the Ctn screening is useful in the management of patients in their clinics (20). The sensitivity of Ctn measurement for the preoperative diagnosis of MTC is higher than that of FNA, with Ctn showing approximately 100% sensitivity and 95% specificity (70–72). Surgery represents the only curative therapeutic strategy. MTC cure is possible in early-stage disease that is detected by Ctn screening before the tumor has metastasized beyond the thyroid and in MEN2 patients by prophylactic thyroidectomy. Sex-specific cut-off values have been proposed to improve the accuracy of basal Ctn levels for mandating total thyroidectomy (20–30 pg/mL for women, 60–79 pg/mL for men; refs. 73, 74). The recommendations for timing prophylactic thyroidectomy in MEN2 patients are based on a model that utilizes genotype–phenotype correlations to stratify mutations into three risk levels—(i) highest (patients with MEN2B and RET M918T mutation, operated in the first year), (ii) high (patients with MEN2A and RET 634 and 883 mutation, operated before the age of 5 years), and (iii) moderate risk (patients with all other mutations, operated on Ctn levels)—that reflect the aggression level of the MTC (20, 75). For patients with moderate-risk mutations, the decision regarding the age at which prophylactic thyroidectomy should be performed is no longer based upon genotype alone but is driven by additional clinical data, especially basal or stimulated serum Ctn levels (75–79). Surgery may be postponed until the patient has an abnormal basal Ctn level.

After surgery, the presence of residual disease, the localization of metastases, and the presence of progressive disease should be assessed to stratify patients with low-risk versus high-risk MTC (20). Somatic 918 RET mutations are a very strong factor for poor prognosis in MTC. A dynamic risk stratification system that uses a combination of the TNM/AJCC staging system, postoperative nadir of Ctn and CEA, and imaging studies to identify local recurrences or distant metastases allows the stratification of MTC patients into three risk groups (80, 81).

  1. Patients with undetectable postoperative Ctn levels who are likely to be disease-free and have an excellent prognosis (10-year survival >95%). This group includes 60% to 90% of patients with a small tumor and no lymph node involvement, but only 20% of those with lymph node metastases. Long-term observation without any further treatment is sufficient. Serum Ctn becomes detectable during follow-up in only 3% of these patients (82).

  2. Patients with detectable Ctn levels after initial treatment with no initial evidence of disease in routine imaging (biochemical incomplete response). Elevations in serum Ctn <150 pg/mL following total thyroidectomy are usually associated with locoregional disease and, very rarely, with distant metastases (83, 84). These patients might be candidates for a second surgery with curative intent. Unfortunately, many patients with MTC who have regional lymph node metastases also have systemic disease and are not cured biochemically despite aggressive surgery, including bilateral neck dissection (85, 86). In all other asymptomatic patients, watchful waiting and, primarily, careful examination by neck US is sufficient (87). In most cases with comprehensive follow-up examinations, tumor markers increase slowly; local recurrence or small, slow-growing, or stable distant metastases without clinical symptoms are detected by imaging in 40% of cases during 10 years of follow-up. If any treatment is necessary, local treatment may be sufficient. Active surveillance is appropriate in most cases.

  3. Patients in an advanced stage of disease with distant metastases at diagnosis and high tumor marker levels (structural incomplete response). These patients have a poor prognosis, with only 40% surviving for 10 years (88). Imaging is used to document metastasis sites, tumor volume, and progression rate (89–91). The growth rate of selected metastases can be determined by sequential imaging studies every 3 to 6 months using Response Evaluation Criteria in Solid Tumors (RECIST; ref. 92) and by measuring serum levels of Ctn or CEA over time to determine the tumor marker doubling time, as tumor marker and tumor mass are correlated (93, 94). Evaluating symptomatic disease manifestation is crucial for making decisions about therapy, such as palliative surgery of metastases, external beam radiation of bone metastases, chemoembolization of liver metastases or administration of systemic therapy like tyrosine kinase inhibitors (TKI). The clinician and patient should discuss expectations regarding quality of life, and the risks and benefits of therapy to determine the best personalized treatment plan. The treatment decision must, therefore, balance the progression rate of the tumor and the quality of life without treatment against the efficacy and side effects of therapy. As none of the possible treatments are curative, palliative therapy should aim to improve quality of life by relieving symptoms (best supportive care). The goals are to provide locoregional disease control, to palliate symptoms of hormonal excess such as diarrhea, to palliate symptomatic metastases like pain or bone fracture, and to control life-threatening metastases such as bronchial obstruction or spinal cord compression.

Before the advent of targeted therapies, chemotherapy was the only option for treating patients with progressive advanced thyroid cancer, but it had only minor efficiency (95). The oncogene pathway–driven approach to understanding the pathophysiology of thyroid cancer prompted clinical trials to assess the antitumor activity of TKIs that inhibit the RET kinase, VEGFR, and other kinases (Fig. 1). Thus, treatment with TKI is indicated in patients with significant tumor burden and documented tumor progression, or those with disease that is threatening vital structures or causing substantial clinical symptoms. Patients with DTC should have RIA-refractory disease before TKIs are considered. This is important because patients with DTC or MTC often have indolent disease, and localized treatment for local control and palliation should be considered first. In patients with metastatic MTC, progression is documented by RECIST (92) and shortened doubling times (<6 months) of serum Ctn and CEA levels (96). Systemic therapy should not be administered to patients with increasing serum Ctn and CEA levels who do not have documented metastatic disease, to patients with stable low-volume metastatic disease as determined by imaging studies, or patients with serum Ctn and CEA doubling times greater than 2 years. Vandetanib (ZETA trial) and cabozantinib (EXAM trial) are approved for the treatment of MTC (97, 98), and sorafenib (DECISION trial) and lenvatinib (SELECT trial) for progressive radioiodine-refractory PTC and FTC (refs. 99, 100; Table 1). All TKI phase III studies have demonstrated significant improvement in progression-free survival, but not in overall survival. However, in a subgroup analysis, a statistically significant difference in overall survival (44.3 months vs. 18.9 months; HR 0.60; 95% CI, 0.38–0.95) was observed in patients with MTC and somatic RET M918T mutations who received cabozantinib compared with placebo (101). In another subgroup, analysis of older patients (>65 years) from the SELECT trial who were treated with lenvatinib had an improved overall survival compared with placebo (102). In patients with ATC and BRAF V600F mutation, a study with the selective BRAF inhibitor vemurafenib was done (103). Selumetinib, a MEK1/2 inhibitor was found to reverse refractoriness to RAI in patients with metastatic DTC (104).

Table 1.

Tyrosine kinase inhibitors (TKI) used in thyroid cancer

SubstanceDrug targetsCancerNo. of patients, verum vs. placeboPFS (months), verumvs. placeboPR (%), verum vs. placeboRef.
Phase III clinical trials with approved TKI for advanced thyroid cancer 
Sorafenib VEGF1,2,3 RET, BRAF, PDGFR DTC 207 vs. 210 10.8 vs. 5.8 12.2 vs. 0.5 99 
Lenvatinib VEGF1,2,3 RET, PDGFR, FGFR1,2,3,4 DTC 261 vs. 131 18.3 vs. 3.6 63.2 vs. 1.5 100 
Vandetanib VEGFR2, RET, EGFR MTC 231 vs. 100 30.5 vs. 19.3 45 vs. 13 97 
Cabozantinib VEGFR2, RET, MET MTC 219 vs. 111 11.2 vs. 4.0 28 vs. 0 98 
Phase II clinical trials with TKI and mTOR inhibitors for thyroid cancer 
Axitinib VEGFR1,2,3 DTC/MTC 52 16.1 35 111 
Everolimus mTOR MTC 8.3 NA 112 
Gefitinib EGFR DTC 18 3.7 3.7 113 
  ATC NA  
Imatinib Bcr-Abl, c-KIT, PDGFR, RET MTC 15 NA 114 
  MTC NA 115 
  ATC NA 25 116 
Lenvatinib VEGFR, c-KIT, RET, PDGFR, FGFR MTC 59  117 
Motesanib VEGFR, PDGFR, c-KIT DTC 93 10 14 118 
  MTC 91 12 119 
Pazopanib VEGFR, c-KIT PDGFR, RET, FGRF DTC 37 11.7 49 120 
  ATC 15 NA 121 
Selumetinib MEK1,2 DTC 32 122 
Sorafenib VEGFR, RET, PDGFR, FGFR, c-KIT, BRAF DTC 31 14 25 123 
  MTC 16 17.9 124 
  ATC 20 1.9 10 125 
Sunitinib VEGFR, c-KIT PDGFR, RET DTC 29 NA 28 126 
Vemurafenib BRAFV600E PTC BRAFV600E positive 51 NA 38 127 
SubstanceDrug targetsCancerNo. of patients, verum vs. placeboPFS (months), verumvs. placeboPR (%), verum vs. placeboRef.
Phase III clinical trials with approved TKI for advanced thyroid cancer 
Sorafenib VEGF1,2,3 RET, BRAF, PDGFR DTC 207 vs. 210 10.8 vs. 5.8 12.2 vs. 0.5 99 
Lenvatinib VEGF1,2,3 RET, PDGFR, FGFR1,2,3,4 DTC 261 vs. 131 18.3 vs. 3.6 63.2 vs. 1.5 100 
Vandetanib VEGFR2, RET, EGFR MTC 231 vs. 100 30.5 vs. 19.3 45 vs. 13 97 
Cabozantinib VEGFR2, RET, MET MTC 219 vs. 111 11.2 vs. 4.0 28 vs. 0 98 
Phase II clinical trials with TKI and mTOR inhibitors for thyroid cancer 
Axitinib VEGFR1,2,3 DTC/MTC 52 16.1 35 111 
Everolimus mTOR MTC 8.3 NA 112 
Gefitinib EGFR DTC 18 3.7 3.7 113 
  ATC NA  
Imatinib Bcr-Abl, c-KIT, PDGFR, RET MTC 15 NA 114 
  MTC NA 115 
  ATC NA 25 116 
Lenvatinib VEGFR, c-KIT, RET, PDGFR, FGFR MTC 59  117 
Motesanib VEGFR, PDGFR, c-KIT DTC 93 10 14 118 
  MTC 91 12 119 
Pazopanib VEGFR, c-KIT PDGFR, RET, FGRF DTC 37 11.7 49 120 
  ATC 15 NA 121 
Selumetinib MEK1,2 DTC 32 122 
Sorafenib VEGFR, RET, PDGFR, FGFR, c-KIT, BRAF DTC 31 14 25 123 
  MTC 16 17.9 124 
  ATC 20 1.9 10 125 
Sunitinib VEGFR, c-KIT PDGFR, RET DTC 29 NA 28 126 
Vemurafenib BRAFV600E PTC BRAFV600E positive 51 NA 38 127 

Abbreviations: NA, not available; PFS, progression-free survival; PR, partial response.

Details of clinical trials in thyroid cancer are summarized in Table 1 and can be found elsewhere (105, 106). A limitation of targeted therapy is the development of an escape mechanism. This phenomenon of resistance to treatment is almost always present, independent of the type of TKI used and the type of human tumor treated (107, 108). TKIs have substantial and unique toxicity profiles, and the doses must be reduced or treatment halted in a significant proportion of patients (Table 2). Common adverse effects associated are hand–foot skin reaction, hypertension, diarrhea, rash, fatigue, weight loss, and QTc prolongation (109). Few data are available on their long-term toxicity.

Table 2.

Any grade of common adverse events associated with different TKI

Sorafinib (99)Lenvatinib (100)Vandetanib (97)Cabozantinib (98)
Hand–foot syndrome 75% Hypertension 68% Diarrhea 56% Diarrhea 63% 
Diarrhea 69% Diarrhea 60% Rash 45% Hand–foot syndrome 50% 
Alopecia 67% Fatigue 59% Nausea 33% Decreased weight 48% 
Rash 50% Decreased appetite 50% Hypertension 32% Decreased appetite 46% 
Weight loss 47% Decreased weight 46% Fatigue 24% Nausea 43% 
Hypertension 41% Nausea 41% Headache 26% Fatigue 41% 
Anorexia 32% Stomatitis 36% Decreased appetite 21% Dysgeusia 34% 
Oral mucositis 23% Hand–foot syndrome 32% Acne 20% Hypertension 33% 
Sorafinib (99)Lenvatinib (100)Vandetanib (97)Cabozantinib (98)
Hand–foot syndrome 75% Hypertension 68% Diarrhea 56% Diarrhea 63% 
Diarrhea 69% Diarrhea 60% Rash 45% Hand–foot syndrome 50% 
Alopecia 67% Fatigue 59% Nausea 33% Decreased weight 48% 
Rash 50% Decreased appetite 50% Hypertension 32% Decreased appetite 46% 
Weight loss 47% Decreased weight 46% Fatigue 24% Nausea 43% 
Hypertension 41% Nausea 41% Headache 26% Fatigue 41% 
Anorexia 32% Stomatitis 36% Decreased appetite 21% Dysgeusia 34% 
Oral mucositis 23% Hand–foot syndrome 32% Acne 20% Hypertension 33% 

Characterization of the molecular mechanisms and mutations that affect key signaling pathways in thyroid cancer pathogenesis is now being translated into clinical practice, with increasing effects on patient care and more specifically on cancer diagnosis, prognostication, and targeted therapies. TERT promoter mutations, RET, and BRAF mutations are major molecular biomarkers of prognosis. Mutation analysis of thyroid nodule FNA samples helps determine the initial treatment and affects postoperative risk stratification in patients with thyroid cancer.

F. Raue is a consultant/advisory board member for AstraZeneca, Eisai, Sanofi Genzyme, and Swedish Orphan Biovitrum. K. Frank-Raue is a consultant/advisory board member for AstraZeneca, Sanofi Genzyme, and Swedish Orphan Biovitrum. No other potential conflicts of interest were disclosed.

Conception and design: F. Raue, K. Frank-Raue

Development of methodology: F. Raue, K. Frank-Raue

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): F. Raue

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): F. Raue, K. Frank-Raue

Writing, review, and/or revision of the manuscript: F. Raue, K. Frank-Raue

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): F. Raue

Study supervision: F. Raue, K. Frank-Raue

1.
Ito
Y
,
Nikiforov
YE
,
Schlumberger
M
,
Vigneri
R
. 
Increasing incidence of thyroid cancer: controversies explored
.
Nat Rev Endocrinol
2013
;
9
:
178
84
.
2.
Leenhardt
L
,
Grosclaude
P
,
Cherie-Challine
L
, Thyroid Cancer Committee. 
Increased incidence of thyroid carcinoma in France: a true epidemic or thyroid nodule management effects? Report from the French Thyroid Cancer Committee
.
Thyroid
2004
;
14
:
1056
60
.
3.
Burgess
JR
. 
Temporal trends for thyroid carcinoma in Australia: an increasing incidence of papillary thyroid carcinoma (1982–1997)
.
Thyroid
2002
;
12
:
141
9
.
4.
Ahn
HS
,
Kim
HJ
,
Welch
HG
. 
Korea's thyroid-cancer "epidemic"–screening and overdiagnosis
.
N Engl J Med
2014
;
371
:
1765
7
.
5.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2015
.
CA Cancer J Clin
2015
;
65
:
5
29
.
6.
Davies
L
,
Welch
HG
. 
Current thyroid cancer trends in the United States
.
JAMA Otolaryngol Head Neck Surg
2014
;
140
:
317
22
.
7.
Howlader
N
,
Noone
AM
,
Krapcho
M
,
Miller
D
,
Bishop
K
,
Altekruse
SF
, et al
,
editors
.
SEER cancer statistics review, 1975–2013 [monograph on the Internet]
.
Bethesda (MD)
:
National Cancer Institute
; 
2016
. Available from: http://seer.cancer.gov/csr/1975_2013/.
8.
McLeod
DS
,
Sawka
AM
,
Cooper
DS
. 
Controversies in primary treatment of low-risk papillary thyroid cancer
.
Lancet
2013
;
381
:
1046
57
.
9.
Brito
JP
,
Al Nofal
A
,
Montori
VM
,
Hay
ID
,
Morris
JC
. 
The impact of subclinical disease and mechanism of detection on the rise in thyroid cancer incidence: population-based study in Olmsted County, Minnesota during 1935 through 2012
.
Thyroid
2015
;
25
:
999
1007
.
10.
Udelsman
R
,
Zhang
Y
. 
The epidemic of thyroid cancer in the United States: the role of endocrinologists and ultrasounds
.
Thyroid
2014
;
24
:
472
9
.
11.
Morris
LG
,
Sikora
AG
,
Tosteson
TD
,
Davies
L
. 
The increasing incidence of thyroid cancer: the influence of access to care
.
Thyroid
2013
;
23
:
885
91
.
12.
Tuttle
RM
,
Ball
DW
,
Byrd
D
,
Dilawari
RA
,
Doherty
GM
,
Duh
QY
, et al
Thyroid carcinoma
.
J Natl Compr Canc Netw
2010
;
8
:
1228
74
.
13.
Lee
YS
,
Lim
H
,
Chang
HS
,
Park
CS
. 
Papillary thyroid microcarcinomas are different from latent papillary thyroid carcinomas at autopsy
.
J Korean Med Sci
2014
;
29
:
676
9
.
14.
Elisei
R
,
Pinchera
A
. 
Advances in the follow-up of differentiated or medullary thyroid cancer
.
Nat Rev Endocrinol
2012
;
8
:
466
75
.
15.
Haugen
BR
,
Alexander
EK
,
Bible
KC
,
Doherty
GM
,
Mandel
SJ
,
Nikiforov
YE
, et al
2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer
.
Thyroid
2016
;
26
:
1
133
.
16.
Perros
P
,
Boelaert
K
,
Colley
S
,
Evans
C
,
Evans
RM
,
Gerrard Ba
G
, et al
Guidelines for the management of thyroid cancer
.
Clin Endocrinol
2014
;
81
Suppl 1
:
1
122
.
17.
Luster
M
,
Clarke
SE
,
Dietlein
M
,
Lassmann
M
,
Lind
P
,
Oyen
WJ
, et al
Guidelines for radioiodine therapy of differentiated thyroid cancer
.
Eur J Nucl Med Mol Imaging
2008
;
35
:
1941
59
.
18.
Takami
H
,
Ito
Y
,
Okamoto
T
,
Onoda
N
,
Noguchi
H
,
Yoshida
A
. 
Revisiting the guidelines issued by the Japanese Society of Thyroid Surgeons and Japan Association of Endocrine Surgeons: a gradual move towards consensus between Japanese and western practice in the management of thyroid carcinoma
.
World J Surg
2014
;
38
:
2002
10
.
19.
Pacini
F
,
Schlumberger
M
,
Dralle
H
,
Elisei
R
,
Smit
JW
,
Wiersinga
W
, et al
European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium
.
Eur J Endocrinol
2006
;
154
:
787
803
.
20.
Wells
SA
 Jr
,
Asa
SL
,
Dralle
H
,
Elisei
R
,
Evans
DB
,
Gagel
RF
, et al
Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma
.
Thyroid
2015
;
25
:
567
610
.
21.
Dralle
H
,
Musholt
TJ
,
Schabram
J
,
Steinmuller
T
,
Frilling
A
,
Simon
D
, et al
German Association of Endocrine Surgeons practice guideline for the surgical management of malignant thyroid tumors
.
Langenbecks Arch Surg
2013
;
398
:
347
75
.
22.
NCCN Clinical Practice Guidelines in Oncology: Thyroid Carcinoma [database on the Internet]
.
Fort Washington, PA
:
National Comprehensive Cancer Network
; 
2014
. Available from: https://www.nccn.org/professionals/physician_gls/f_guidelines.asp.
23.
Gharib
H
,
Papini
E
,
Paschke
R
,
Duick
DS
,
Valcavi
R
,
Hegedus
L
, et al
American Association of Clinical Endocrinologists, Associazione Medici Endocrinologi, and European Thyroid Association Medical guidelines for clinical practice for the diagnosis and management of thyroid nodules: executive summary of recommendations
.
Endocr Pract
2010
;
16
:
468
75
.
24.
Kwong
N
,
Medici
M
,
Angell
TE
,
Liu
X
,
Marqusee
E
,
Cibas
ES
, et al
The influence of patient age on thyroid nodule formation, multinodularity, and thyroid cancer risk
.
J Clin Endocrinol Metab
2015
;
100
:
4434
40
.
25.
Kwak
JY
,
Han
KH
,
Yoon
JH
,
Moon
HJ
,
Son
EJ
,
Park
SH
, et al
Thyroid imaging reporting and data system for US features of nodules: a step in establishing better stratification of cancer risk
.
Radiology
2011
;
260
:
892
9
.
26.
Frates
MC
,
Benson
CB
,
Doubilet
PM
,
Kunreuther
E
,
Contreras
M
,
Cibas
ES
, et al
Prevalence and distribution of carcinoma in patients with solitary and multiple thyroid nodules on sonography
.
J Clin Endocrinol Metab
2006
;
91
:
3411
7
.
27.
Remonti
LR
,
Kramer
CK
,
Leitao
CB
,
Pinto
LC
,
Gross
JL
. 
Thyroid ultrasound features and risk of carcinoma: a systematic review and meta-analysis of observational studies
.
Thyroid
2015
;
25
:
538
50
.
28.
Crippa
S
,
Mazzucchelli
L
,
Cibas
ES
,
Ali
SZ
. 
The Bethesda System for reporting thyroid fine-needle aspiration specimens
.
Am J Clin Pathol
2010
;
134
:
343
4
.
29.
Cibas
ES
,
Ali
SZ
;
NCI Thyroid FNA State of the Science Conference
. 
The Bethesda System for Reporting Thyroid Cytopathology
.
Am J Clin Pathol
2009
;
132
:
658
65
.
30.
Nikiforov
YE
,
Yip
L
,
Nikiforova
MN
. 
New strategies in diagnosing cancer in thyroid nodules: impact of molecular markers
.
Clin Cancer Res
2013
;
19
:
2283
8
.
31.
Nikiforov
YE
,
Nikiforova
MN
. 
Molecular genetics and diagnosis of thyroid cancer
.
Nat Rev Endocrinol
2011
;
7
:
569
80
.
32.
Xing
M
. 
Molecular pathogenesis and mechanisms of thyroid cancer
.
Nat Rev Cancer
2013
;
13
:
184
99
.
33.
Cancer Genome Atlas Research Network
. 
Integrated genomic characterization of papillary thyroid carcinoma
.
Cell
2014
;
159
:
676
90
.
34.
Carina
V
,
Zito
G
,
Pizzolanti
G
,
Richiusa
P
,
Criscimanna
A
,
Rodolico
V
, et al
Multiple pluripotent stem cell markers in human anaplastic thyroid cancer: the putative upstream role of SOX2
.
Thyroid
2013
;
23
:
829
37
.
35.
Boichard
A
,
Croux
L
,
Al Ghuzlan
A
,
Broutin
S
,
Dupuy
C
,
Leboulleux
S
, et al
Somatic RAS mutations occur in a large proportion of sporadic RET-negative medullary thyroid carcinomas and extend to a previously unidentified exon
.
J Clin Endocrinol Metab
2012
;
97
:
E2031
5
.
36.
Ciampi
R
,
Mian
C
,
Fugazzola
L
,
Cosci
B
,
Romei
C
,
Barollo
S
, et al
Evidence of a low prevalence of RAS mutations in a large medullary thyroid cancer series
.
Thyroid
2013
;
23
:
50
7
.
37.
Wells
SA
 Jr
,
Pacini
F
,
Robinson
BG
,
Santoro
M
. 
Multiple endocrine neoplasia type 2 and familial medullary thyroid carcinoma: an update
.
J Clin Endocrinol Metab
2013
;
98
:
3149
64
.
38.
Ji
JH
,
Oh
YL
,
Hong
M
,
Yun
JW
,
Lee
HW
,
Kim
D
, et al
Identification of driving ALK fusion genes and genomic landscape of medullary thyroid cancer
.
PLoS Genet
2015
;
11
:
e1005467
.
39.
Giordano
TJ
,
Beaudenon-Huibregtse
S
,
Shinde
R
,
Langfield
L
,
Vinco
M
,
Laosinchai-Wolf
W
, et al
Molecular testing for oncogenic gene mutations in thyroid lesions: a case-control validation study in 413 postsurgical specimens
.
Hum Pathol
2014
;
45
:
1339
47
.
40.
Zheng
X
,
Wei
S
,
Han
Y
,
Li
Y
,
Yu
Y
,
Yun
X
, et al
Papillary microcarcinoma of the thyroid: clinical characteristics and BRAF(V600E) mutational status of 977 cases
.
Ann Surg Oncol
2013
;
20
:
2266
73
.
41.
Kim
TY
,
Kim
WB
,
Rhee
YS
,
Song
JY
,
Kim
JM
,
Gong
G
, et al
The BRAF mutation is useful for prediction of clinical recurrence in low-risk patients with conventional papillary thyroid carcinoma
.
Clin Endocrinol
2006
;
65
:
364
8
.
42.
Bastos
AU
,
Oler
G
,
Nozima
BH
,
Moyses
RA
,
Cerutti
JM
. 
BRAF V600E and decreased NIS and TPO expression are associated with aggressiveness of a subgroup of papillary thyroid microcarcinoma
.
Eur J Endocrinol
2015
;
173
:
525
40
.
43.
Li
F
,
Chen
G
,
Sheng
C
,
Gusdon
AM
,
Huang
Y
,
Lv
Z
, et al
BRAFV600E mutation in papillary thyroid microcarcinoma: a meta-analysis
.
Endocr Relat Cancer
2015
;
22
:
159
68
.
44.
Lee
SE
,
Hwang
TS
,
Choi
YL
,
Han
HS
,
Kim
WS
,
Jang
MH
, et al
Prognostic significance of TERT promoter mutations in papillary thyroid carcinomas in a BRAFV600E mutation-prevalent population
.
Thyroid
2016
;
26
:
901
10
.
45.
Nikiforov
YE
,
Carty
SE
,
Chiosea
SI
,
Coyne
C
,
Duvvuri
U
,
Ferris
RL
, et al
Impact of the multi-gene ThyroSeq next-generation sequencing assay on cancer diagnosis in thyroid nodules with atypia of undetermined significance/follicular lesion of undetermined significance cytology
.
Thyroid
2015
;
25
:
1217
23
.
46.
Alexander
EK
,
Kennedy
GC
,
Baloch
ZW
,
Cibas
ES
,
Chudova
D
,
Diggans
J
, et al
Preoperative diagnosis of benign thyroid nodules with indeterminate cytology
.
N Engl J Med
2012
;
367
:
705
15
.
47.
Marti
JL
,
Avadhani
V
,
Donatelli
LA
,
Niyogi
S
,
Wang
B
,
Wong
RJ
, et al
Wide inter-institutional variation in performance of a molecular classifier for indeterminate thyroid nodules
.
Ann Surg Oncol
2015
;
22
:
3996
4001
.
48.
Wu
JX
,
Young
S
,
Hung
ML
,
Li
N
,
Yang
SE
,
Cheung
DS
, et al
Clinical factors influencing the performance of gene expression classifier testing in indeterminate thyroid nodules
.
Thyroid
2016
;
26
:
916
22
.
49.
Xing
M
,
Haugen
BR
,
Schlumberger
M
. 
Progress in molecular-based management of differentiated thyroid cancer
.
Lancet
2013
;
381
:
1058
69
.
50.
Kim
SK
,
Hwang
TS
,
Yoo
YB
,
Han
HS
,
Kim
DL
,
Song
KH
, et al
Surgical results of thyroid nodules according to a management guideline based on the BRAF(V600E) mutation status
.
J Clin Endocrinol Metab
2011
;
96
:
658
64
.
51.
Haigh
PI
,
Urbach
DR
,
Rotstein
LE
. 
Extent of thyroidectomy is not a major determinant of survival in low- or high-risk papillary thyroid cancer
.
Ann Surg Oncol
2005
;
12
:
81
9
.
52.
Barney
BM
,
Hitchcock
YJ
,
Sharma
P
,
Shrieve
DC
,
Tward
JD
. 
Overall and cause-specific survival for patients undergoing lobectomy, near-total, or total thyroidectomy for differentiated thyroid cancer
.
Head Neck
2011
;
33
:
645
9
.
53.
Ito
Y
,
Miyauchi
A
,
Kihara
M
,
Higashiyama
T
,
Kobayashi
K
,
Miya
A
. 
Patient age is significantly related to the progression of papillary microcarcinoma of the thyroid under observation
.
Thyroid
2014
;
24
:
27
34
.
54.
Oda
H
,
Miyauchi
A
,
Ito
Y
,
Yoshioka
K
,
Nakayama
A
,
Sasai
H
, et al
Incidences of unfavorable events in the management of low-risk papillary microcarcinoma of the thyroid by active surveillance versus immediate surgery
.
Thyroid
2016
;
26
:
150
5
.
55.
Wartofsky
L
,
Van Nostrand
D
. 
Radioiodine treatment of well-differentiated thyroid cancer
.
Endocrine
2012
;
42
:
506
13
.
56.
Schlumberger
M
,
Catargi
B
,
Borget
I
,
Deandreis
D
,
Zerdoud
S
,
Bridji
B
, et al
Strategies of radioiodine ablation in patients with low-risk thyroid cancer
.
N Engl J Med
2012
;
366
:
1663
73
.
57.
Iyer
NG
,
Morris
LG
,
Tuttle
RM
,
Shaha
AR
,
Ganly
I
. 
Rising incidence of second cancers in patients with low-risk (T1N0) thyroid cancer who receive radioactive iodine therapy
.
Cancer
2011
;
117
:
4439
46
.
58.
Melo
M
,
da Rocha
AG
,
Vinagre
J
,
Batista
R
,
Peixoto
J
,
Tavares
C
, et al
TERT promoter mutations are a major indicator of poor outcome in differentiated thyroid carcinomas
.
J Clin Endocrinol Metab
2014
;
99
:
E754
65
.
59.
Byar
DP
,
Green
SB
,
Dor
P
,
Williams
ED
,
Colon
J
,
van Gilse
HA
, et al
A prognostic index for thyroid carcinoma. A study of the E.O.R.T.C. Thyroid Cancer Cooperative Group
.
Eur J Cancer
1979
;
15
:
1033
41
.
60.
Hay
ID
,
Grant
CS
,
Taylor
WF
,
McConahey
WM
. 
Ipsilateral lobectomy versus bilateral lobar resection in papillary thyroid carcinoma: a retrospective analysis of surgical outcome using a novel prognostic scoring system
.
Surgery
1987
;
102
:
1088
95
.
61.
Cady
B
,
Rossi
R
. 
An expanded view of risk-group definition in differentiated thyroid carcinoma
.
Surgery
1988
;
104
:
947
53
.
62.
Shaha
AR
,
Loree
TR
,
Shah
JP
. 
Intermediate-risk group for differentiated carcinoma of thyroid
.
Surgery
1994
;
116
:
1036
40
.
63.
Momesso
DP
,
Tuttle
RM
. 
Update on differentiated thyroid cancer staging
.
Endocrinol Metab Clin North Am
2014
;
43
:
401
21
.
64.
Castagna
MG
,
Maino
F
,
Cipri
C
,
Belardini
V
,
Theodoropoulou
A
,
Cevenini
G
, et al
Delayed risk stratification, to include the response to initial treatment (surgery and radioiodine ablation), has better outcome predictivity in differentiated thyroid cancer patients
.
Eur J Endocrinol
2011
;
165
:
441
6
.
65.
Vaisman
F
,
Tala
H
,
Grewal
R
,
Tuttle
RM
. 
In differentiated thyroid cancer, an incomplete structural response to therapy is associated with significantly worse clinical outcomes than only an incomplete thyroglobulin response
.
Thyroid
2011
;
21
:
1317
22
.
66.
Pitoia
F
,
Bueno
F
,
Urciuoli
C
,
Abelleira
E
,
Cross
G
,
Tuttle
RM
. 
Outcomes of patients with differentiated thyroid cancer risk-stratified according to the American thyroid association and Latin American thyroid society risk of recurrence classification systems
.
Thyroid
2013
;
23
:
1401
7
.
67.
Tuttle
RM
,
Tala
H
,
Shah
J
,
Leboeuf
R
,
Ghossein
R
,
Gonen
M
, et al
Estimating risk of recurrence in differentiated thyroid cancer after total thyroidectomy and radioactive iodine remnant ablation: using response to therapy variables to modify the initial risk estimates predicted by the new American Thyroid Association staging system
.
Thyroid
2010
;
20
:
1341
9
.
68.
Miyauchi
A
,
Kudo
T
,
Miya
A
,
Kobayashi
K
,
Ito
Y
,
Takamura
Y
, et al
Prognostic impact of serum thyroglobulin doubling-time under thyrotropin suppression in patients with papillary thyroid carcinoma who underwent total thyroidectomy
.
Thyroid
2011
;
21
:
707
16
.
69.
Raue
F
,
Frank-Raue
K
. 
Epidemiology and clinical presentation of medullary thyroid carcinoma
.
Recent Results Cancer Res
2015
;
204
:
61
90
.
70.
Bugalho
MJ
,
Santos
JR
,
Sobrinho
L
. 
Preoperative diagnosis of medullary thyroid carcinoma: fine needle aspiration cytology as compared with serum calcitonin measurement
.
J Surg Oncol
2005
;
91
:
56
60
.
71.
Papi
G
,
Corsello
SM
,
Cioni
K
,
Pizzini
AM
,
Corrado
S
,
Carapezzi
C
, et al
Value of routine measurement of serum calcitonin concentrations in patients with nodular thyroid disease: a multicenter study
.
J Endocrinol Invest
2006
;
29
:
427
37
.
72.
Hasselgren
M
,
Hegedus
L
,
Godballe
C
,
Bonnema
SJ
. 
Benefit of measuring basal serum calcitonin to detect medullary thyroid carcinoma in a Danish population with a high prevalence of thyroid nodules
.
Head Neck
2010
;
32
:
612
8
.
73.
Mian
C
,
Perrino
M
,
Colombo
C
,
Cavedon
E
,
Pennelli
G
,
Ferrero
S
, et al
Refining calcium test for the diagnosis of medullary thyroid cancer: cutoffs, procedures, and safety
.
J Clin Endocrinol Metab
2014
;
99
:
1656
64
.
74.
Machens
A
,
Hoffmann
F
,
Sekulla
C
,
Dralle
H
. 
Importance of gender-specific calcitonin thresholds in screening for occult sporadic medullary thyroid cancer
.
Endocr Relat Cancer
2009
;
16
:
1291
8
.
75.
Frank-Raue
K
,
Raue
F
. 
Hereditary medullary thyroid cancer genotype-phenotype correlation
.
Recent Results Cancer Res
2015
;
204
:
139
56
.
76.
Machens
A
,
Niccoli-Sire
P
,
Hoegel
J
,
Frank-Raue
K
,
van Vroonhoven
TJ
,
Roeher
HD
, et al
Early malignant progression of hereditary medullary thyroid cancer
.
N Engl J Med
2003
;
349
:
1517
25
.
77.
Frank-Raue
K
,
Buhr
H
,
Dralle
H
,
Klar
E
,
Senninger
N
,
Weber
T
, et al
Long-term outcome in 46 gene carriers of hereditary medullary thyroid carcinoma after prophylactic thyroidectomy: impact of individual RET genotype
.
Eur J Endocrinol
2006
;
155
:
229
36
.
78.
Skinner
MA
,
Moley
JA
,
Dilley
WG
,
Owzar
K
,
Debenedetti
MK
,
Wells
SA
 Jr
. 
Prophylactic thyroidectomy in multiple endocrine neoplasia type 2A
.
N Engl J Med
2005
;
353
:
1105
13
.
79.
Niccoli-Sire
P
,
Murat
A
,
Baudin
E
,
Henry
JF
,
Proye
C
,
Bigorgne
JC
, et al
Early or prophylactic thyroidectomy in MEN 2/FMTC gene carriers: results in 71 thyroidectomized patients. The French Calcitonin Tumours Study Group (GETC)
.
Eur J Endocrinol
1999
;
141
:
468
74
.
80.
Lindsey
SC
,
Ganly
I
,
Palmer
F
,
Tuttle
RM
. 
Response to initial therapy predicts clinical outcomes in medullary thyroid cancer
.
Thyroid
2015
;
25
:
242
9
.
81.
Raue
F
,
Frank-Raue
K
. 
Long-term follow-up in medullary thyroid carcinoma
.
Recent Results Cancer Res
2015
;
204
:
207
25
.
82.
Franc
S
,
Niccoli-Sire
P
,
Cohen
R
,
Bardet
S
,
Maes
B
,
Murat
A
, et al
Complete surgical lymph node resection does not prevent authentic recurrences of medullary thyroid carcinoma
.
Clin Endocrinol
2001
;
55
:
403
9
.
83.
Yen
TW
,
Shapiro
SE
,
Gagel
RF
,
Sherman
SI
,
Lee
JE
,
Evans
DB
. 
Medullary thyroid carcinoma: results of a standardized surgical approach in a contemporary series of 80 consecutive patients
.
Surgery
2003
;
134
:
890
9
.
84.
Pellegriti
G
,
Leboulleux
S
,
Baudin
E
,
Bellon
N
,
Scollo
C
,
Travagli
JP
, et al
Long-term outcome of medullary thyroid carcinoma in patients with normal postoperative medical imaging
.
Br J Cancer
2003
;
88
:
1537
42
.
85.
Machens
A
,
Dralle
H
. 
Prognostic impact of N staging in 715 medullary thyroid cancer patients: proposal for a revised staging system
.
Ann Surg
2013
;
257
:
323
9
.
86.
Machens
A
,
Dralle
H
. 
Biomarker-based risk stratification for previously untreated medullary thyroid cancer
.
J Clin Endocrinol Metab
2010
;
95
:
2655
63
.
87.
Kouvaraki
MA
,
Shapiro
SE
,
Fornage
BD
,
Edeiken-Monro
BS
,
Sherman
SI
,
Vassilopoulou-Sellin
R
, et al
Role of preoperative ultrasonography in the surgical management of patients with thyroid cancer
.
Surgery
2003
;
134
:
946
54
.
88.
Roman
S
,
Lin
R
,
Sosa
JA
. 
Prognosis of medullary thyroid carcinoma: demographic, clinical, and pathologic predictors of survival in 1252 cases
.
Cancer
2006
;
107
:
2134
42
.
89.
Giraudet
AL
,
Vanel
D
,
Leboulleux
S
,
Auperin
A
,
Dromain
C
,
Chami
L
, et al
Imaging medullary thyroid carcinoma with persistent elevated calcitonin levels
.
J Clin Endocrinol Metab
2007
;
92
:
4185
90
.
90.
Koopmans
KP
,
de Groot
JW
,
Plukker
JT
,
de Vries
EG
,
Kema
IP
,
Sluiter
WJ
, et al
18F-dihydroxyphenylalanine PET in patients with biochemical evidence of medullary thyroid cancer: relation to tumor differentiation
.
J Nucl Med
2008
;
49
:
524
31
.
91.
Rubello
D
,
Rampin
L
,
Nanni
C
,
Banti
E
,
Ferdeghini
M
,
Fanti
S
, et al
The role of 18F-FDG PET/CT in detecting metastatic deposits of recurrent medullary thyroid carcinoma: a prospective study
.
Eur J Surg Oncol
2008
;
34
:
581
6
.
92.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
93.
Laure Giraudet
A
,
Al Ghulzan
A
,
Auperin
A
,
Leboulleux
S
,
Chehboun
A
,
Troalen
F
, et al
Progression of medullary thyroid carcinoma: assessment with calcitonin and carcinoembryonic antigen doubling times
.
Eur J Endocrinol
2008
;
158
:
239
46
.
94.
Barbet
J
,
Campion
L
,
Kraeber-Bodere
F
,
Chatal
JF
;
GTE Study Group
. 
Prognostic impact of serum calcitonin and carcinoembryonic antigen doubling-times in patients with medullary thyroid carcinoma
.
J Clin Endocrinol Metab
2005
;
90
:
6077
84
.
95.
Albero
A
,
Lopez
JE
,
Torres
A
,
de la Cruz
L
,
Martin
T
. 
Effectiveness of chemotherapy in advanced differentiated thyroid cancer: a systematic review
.
Endocr Relat Cancer
2016
;
23
:
R71
84
.
96.
Calctonin and carcinoembryonic antigen (CEA) doubling time calculator [about 1 screen]. [cited 2016 Aug 22].
Available from: http://www.thyroid.org/professionals/calculators/thyroid-cancer-carcinoma/2016.
97.
Wells
SA
 Jr
,
Robinson
BG
,
Gagel
RF
,
Dralle
H
,
Fagin
JA
,
Santoro
M
, et al
Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial
.
J Clin Oncol
2012
;
30
:
134
41
.
98.
Elisei
R
,
Schlumberger
MJ
,
Muller
SP
,
Schoffski
P
,
Brose
MS
,
Shah
MH
, et al
Cabozantinib in progressive medullary thyroid cancer
.
J Clin Oncol
2013
;
31
:
3639
46
.
99.
Brose
MS
,
Nutting
CM
,
Jarzab
B
,
Elisei
R
,
Siena
S
,
Bastholt
L
, et al
Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial
.
Lancet
2014
;
384
:
319
28
.
100.
Schlumberger
M
,
Tahara
M
,
Wirth
LJ
,
Robinson
B
,
Brose
MS
,
Elisei
R
, et al
Lenvatinib versus placebo in radioiodine-refractory thyroid cancer
.
N Engl J Med
2015
;
372
:
621
30
.
101.
Schlumberger
M
,
Elisei
R
,
Müller
S
,
Schöffski
P
,
Brose
MS
,
Shah
LF
, et al
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
. J Clin Oncol 33, 2015 (suppl; abstr 6012).
102.
Brose
MS
,
Schlumberger
M
,
Tahara
M
,
Wirth
LJ
,
Robinson
B
,
Elisei
R
, et al
Effect of age and lenvatinib treatment on overall survival for patients with 131-I-refractory differentiated thyroid cancer in SELECT
. J Clin Oncol 33, 2015 (suppl; abstr 6048).
103.
Rosove
MH
,
Peddi
PF
,
Glaspy
JA
. 
BRAF V600E inhibition in anaplastic thyroid cancer
.
N Engl J Med
2013
;
368
:
684
5
.
104.
Ho
AL
,
Grewal
RK
,
Leboeuf
R
,
Sherman
EJ
,
Pfister
DG
,
Deandreis
D
, et al
Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer
.
N Engl J Med
2013
;
368
:
623
32
.
105.
Viola
D
,
Valerio
L
,
Molinaro
E
,
Agate
L
,
Bottici
V
,
Biagini
A
, et al
Treatment of advanced thyroid cancer with targeted therapies: ten years of experience
.
Endocr Relat Cancer
2016
;
23
:
R185
205
.
106.
Spitzweg
C
,
Morris
JC
,
Bible
KC
. 
New drugs for medullary thyroid cancer: new promises?
Endocr Relat Cancer
2016
;
23
:
R287
97
.
107.
Arao
T
,
Matsumoto
K
,
Furuta
K
,
Kudo
K
,
Kaneda
H
,
Nagai
T
, et al
Acquired drug resistance to vascular endothelial growth factor receptor 2 tyrosine kinase inhibitor in human vascular endothelial cells
.
Anticancer Res
2011
;
31
:
2787
96
.
108.
Finke
J
,
Ko
J
,
Rini
B
,
Rayman
P
,
Ireland
J
,
Cohen
P
. 
MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy
.
Int Immunopharmacol
2011
;
11
:
856
61
.
109.
Klein Hesselink
EN
,
Steenvoorden
D
,
Kapiteijn
E
,
Corssmit
EP
,
van der Horst-Schrivers
AN
,
Lefrandt
JD
, et al
Therapy of endocrine disease: response and toxicity of small-molecule tyrosine kinase inhibitors in patients with thyroid carcinoma: a systematic review and meta-analysis
.
Eur J Endocrinol
2015
;
172
:
R215
25
.
110.
Wells
SA
 Jr
,
Santoro
M
. 
Update: the status of clinical trials with kinase inhibitors in thyroid cancer
.
J Clin Endocrinol Metab
2014
;
99
:
1543
55
.
111.
Locati
LD
,
Licitra
L
,
Agate
L
,
Ou
SH
,
Boucher
A
,
Jarzab
B
, et al
Treatment of advanced thyroid cancer with axitinib: Phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments
.
Cancer
2014
;
120
:
2694
703
.
112.
Schneider
TC
,
de Wit
D
,
Links
TP
,
van Erp
NP
,
van der Hoeven
JJ
,
Gelderblom
H
, et al
Beneficial effects of the mTOR inhibitor everolimus in patients with advanced medullary thyroid carcinoma: subgroup results of a phase II trial
.
Int J Endocrinol
2015
;
2015
:
348124
.
113.
Pennell
NA
,
Daniels
GH
,
Haddad
RI
,
Ross
DS
,
Evans
T
,
Wirth
LJ
, et al
A phase II study of gefitinib in patients with advanced thyroid cancer
.
Thyroid
2008
;
18
:
317
23
.
114.
de Groot
JW
,
Zonnenberg
BA
,
van Ufford-Mannesse
PQ
,
de Vries
MM
,
Links
TP
,
Lips
CJ
, et al
A phase II trial of imatinib therapy for metastatic medullary thyroid carcinoma
.
J Clin Endocrinol Metab
2007
;
92
:
3466
9
.
115.
Frank-Raue
K
,
Fabel
M
,
Delorme
S
,
Haberkorn
U
,
Raue
F
. 
Efficacy of imatinib mesylate in advanced medullary thyroid carcinoma
.
Eur J Endocrinol
2007
;
157
:
215
20
.
116.
Ha
HT
,
Lee
JS
,
Urba
S
,
Koenig
RJ
,
Sisson
J
,
Giordano
T
, et al
A phase II study of imatinib in patients with advanced anaplastic thyroid cancer
.
Thyroid
2010
;
20
:
975
80
.
117.
Schlumberger
M
,
Jarzab
B
,
Cabanillas
ME
,
Robinson
B
,
Pacini
F
,
Ball
DW
, et al
A phase II trial of the multitargeted tyrosine kinase inhibitor lenvatinib (E7080) in advanced medullary thyroid cancer
.
Clin Cancer Res
2016
;
22
:
44
53
.
118.
Sherman
SI
,
Wirth
LJ
,
Droz
JP
,
Hofmann
M
,
Bastholt
L
,
Martins
RG
, et al
Motesanib diphosphate in progressive differentiated thyroid cancer
.
N Engl J Med
2008
;
359
:
31
42
.
119.
Schlumberger
MJ
,
Elisei
R
,
Bastholt
L
,
Wirth
LJ
,
Martins
RG
,
Locati
LD
, et al
Phase II study of safety and efficacy of motesanib in patients with progressive or symptomatic, advanced or metastatic medullary thyroid cancer
.
J Clin Oncol
2009
;
27
:
3794
801
.
120.
Bible
KC
,
Suman
VJ
,
Molina
JR
,
Smallridge
RC
,
Maples
WJ
,
Menefee
ME
, et al
Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study
.
Lancet Oncol
2010
;
11
:
962
72
.
121.
Bible
KC
,
Suman
VJ
,
Menefee
ME
,
Smallridge
RC
,
Molina
JR
,
Maples
WJ
, et al
A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer
.
J Clin Endocrinol Metab
2012
;
97
:
3179
84
.
122.
Hayes
DN
,
Lucas
AS
,
Tanvetyanon
T
,
Krzyzanowska
MK
,
Chung
CH
,
Murphy
BA
, et al
Phase II efficacy and pharmacogenomic study of selumetinib (AZD6244; ARRY-142886) in iodine-131 refractory papillary thyroid carcinoma with or without follicular elements
.
Clin Cancer Res
2012
;
18
:
2056
65
.
123.
Schneider
TC
,
Abdulrahman
RM
,
Corssmit
EP
,
Morreau
H
,
Smit
JW
,
Kapiteijn
E
. 
Long-term analysis of the efficacy and tolerability of sorafenib in advanced radio-iodine refractory differentiated thyroid carcinoma: final results of a phase II trial
.
Eur J Endocrinol
2012
;
167
:
643
50
.
124.
Lam
ET
,
Ringel
MD
,
Kloos
RT
,
Prior
TW
,
Knopp
MV
,
Liang
J
, et al
Phase II clinical trial of sorafenib in metastatic medullary thyroid cancer
.
J Clin Oncol
2010
;
28
:
2323
30
.
125.
Savvides
P
,
Nagaiah
G
,
Lavertu
P
,
Fu
P
,
Wright
JJ
,
Chapman
R
, et al
Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid
.
Thyroid
2013
;
23
:
600
4
.
126.
Carr
LL
,
Mankoff
DA
,
Goulart
BH
,
Eaton
KD
,
Capell
PT
,
Kell
EM
, 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
2010
;
16
:
5260
8
.
127.
Brose
MS
,
Cabanillas
ME
,
Cohen
EE
,
Wirth
LJ
,
Riehl
T
,
Yue
H
, et al
Vemurafenib in patients with BRAFV600E-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: a non-randomised, multicentre, open-label, phase 2 trial
.
Lancet Oncol.
2016
Jul 22.
[Epub ahead of print]
.