Purpose:

Restoration of iodine incorporation (redifferentiation) by MAPK inhibition was achieved in previously radioiodine-refractory, unresectable thyroid carcinoma (RR-TC). However, results were unsatisfactory in BRAFV600E-mutant (BRAF-MUT) RR-TC. Here we assess safety and efficacy of redifferentiation therapy through genotype-guided MAPK-modulation in patients with BRAF-MUT or wildtype (BRAF-WT) RR-TC.

Patients and Methods:

In this prospective single-center, two-arm phase II study, patients received trametinib (BRAF-WT) or trametinib + dabrafenib (BRAF-MUT) for 21 ± 3 days. Redifferentiation was assessed by 123I-scintigraphy. In case of restored radioiodine uptake, 124I-guided 131I therapy was performed. Primary endpoint was the redifferentiation rate. Secondary endpoints were treatment response (thyroglobulin, RECIST 1.1) and safety. Parameters predicting successful redifferentiation were assessed using a receiver operating characteristic analysis and Youden J statistic.

Results:

Redifferentiation was achieved in 7 of 20 (35%) patients, 2 of 6 (33%) in the BRAF-MUT and 5 of 14 (36%) in the BRAF-WT arm. Patients received a mean (range) activity of 300.0 (273.0–421.6) mCi for 131I therapy. Any thyroglobulin decline was seen in 57% (4/7) of the patients, RECIST 1.1 stable/partial response/progressive disease in 71% (5/7)/14% (1/7)/14% (1/7). Peak standardized uptake value (SUVpeak) < 10 on 2[18F]fluoro-2-deoxy-D-glucose (FDG)-PET was associated with successful redifferentiation (P = 0.01). Transient pyrexia (grade 3) and rash (grade 4) were noted in one patient each.

Conclusions:

Genotype-guided MAPK inhibition was safe and resulted in successful redifferentiation in about one third of patients in each arm. Subsequent 131I therapy led to a thyroglobulin (Tg) decline in more than half of the treated patients. Low tumor glycolytic rate as assessed by FDG-PET is predictive of redifferentiation success.

See related commentary by Cabanillas et al., p. 4164

Translational Relevance

MAPK pathway inhibition can restore radioiodine uptake in previously radioiodine-refractory thyroid carcinoma (RR-TC) and thereby enable subsequent radioiodine therapy. However, redifferentiation rates were unsatisfactory in BRAFV600E-mutant (BRAF-MUT) RR-TC thus far. In our cohort of 20 patients with unresectable RR-TC, treatment with trametinib [BRAFV600E-wildtype (BRAF-WT)] or trametinib + dabrafenib (BRAF-MUT RR-TC) for 3 weeks restored radioiodine uptake in 7 of 20 (35%) patients and showed a favorable safety profile. Redifferentiation rates were comparable between patients with BRAF-WT and those with BRAF-MT RR-TC. Low glycolytic activity as assessed by 2[18F]fluoro-2-deoxy-D-glucose (FDG)-PET predicted successful restoration of radioiodine uptake. Genotype-guided MAPK inhibition is feasible and holds promise to delay treatment with tyrosine kinase inhibitors for BRAF-WT and -MUT RR-TC.

Radioiodine is the first-line treatment in patients with unresectable and/or metastatic thyroid carcinoma of follicular origin (i.e., follicular or papillary carcinoma; ref. 1). However, 60% to 70% of these patients are or eventually become refractory to radioiodine, a state that is associated with a considerable reduction in overall survival (25).

On a molecular level, radioiodine-refractoriness has been linked to mutually exclusive genetic alterations in genes encoding the growth factors RET, NTRK1, RAS, and BRAF. Constitutive activation of these oncoproteins stimulates MAPK signaling to activate downstream kinases MEK and ERK resulting in a disruption of follicular cell differentiation (6–9). This leads to upregulation of proliferation and suppression of key genes involved in iodine metabolism including the sodium iodide symporter (NIS), a protein critical for mediating iodide uptake into thyroid cells (10–15).

Preclinical and clinical studies have shown that oncogenic BRAF-induced gene patterns can be reversed by abrogation of MAPK signaling leading to a restoration of iodine uptake into thyroid carcinoma cells (16, 17). However, in the first translational study using the MEK inhibitor selumetinib, despite convincing results in BRAFV600E-wildtype (BRAF-WT) patients, only 1 in 9 patients with a BRAFV600E-mutation (BRAF-MUT) achieved sufficient improvement in iodine incorporation to warrant radioiodine treatment (17). As the degree of iodine restoration has been linked to the extent of MAPK pathway inhibition (18), the findings imply that a more potent inhibition may improve NIS restoration in BRAF-MUT patients. Subsequent studies on these patients revealed improved radioiodine uptake by using the BRAF-inhibitors vemurafenib (18) and dabrafenib (19). Combined targeting of BRAF and downstream MEK kinase has been shown to lead to enhanced MAPK inhibition in a preclinical setting (20) resulting in a more profound expression of NIS.

These studies provide the rationale for our prospective study, in which we evaluate the efficacy and safety of radioiodine uptake restoration through genotype-guided MAPK modulation: MEK-targeted treatment with trametinib was performed for BRAF-WT, whereas combined MEK- and BRAF-inhibition with trametinib and dabrafenib was given in BRAF-MUT patients with radioiodine-refractory metastatic thyroid carcinoma (RR-TC). Redifferentiation success was quantified by baseline and follow-up 124I PET with subsequent dosimetry-based 131I therapy (21, 22).

Patients

Patients were eligible if they had unresectable and/or RR-TC of follicular origin. Radioiodine-refractoriness was defined as the presence of at least one metastatic lesion without therapeutically sufficient radioiodine accumulation on a diagnostic or therapeutic radioiodine scan performed less than 1 year prior to enrollment. Further requirements were the presence of at least one measurable lesion ≥15 mm (short-axis diameter for nodal lesions, long-axis diameter for nonnodal lesions) and confirmation of BRAF V600E mutation status. For all inclusion and exclusion criteria, see “Criteria for subject eligibility” in the supplementary appendix.

The study was registered as a prospective trial (clinicaltrials.gov, NCT04619316) and approved by the local institutional review board (IRB; University of Duisburg-Essen, Medical Faculty; Essen, Germany; protocol number: 17–7323-AF) and federal administrations (BfArM, BfS). The study was performed in concordance with the Declaration of Helsinki and all patients gave written informed consent for enrollment.

Study design and safety

The protocol design is shown in Fig. 1. Patients underwent 124I-PET for baseline assessment of lesion size and iodine-avidity on clinical indication before entering the study. Treatment with 2 mg trametinib daily (and additional dabrafenib 75 mg twice daily in BRAF-MUT patients) was given for 21 days. After redifferentiation treatment, iodine uptake was assessed by 123I scintigraphy and repeat 124I-PET outside of the protocol. Patients in whom the posttreatment 123I single-photon emission computed tomography (SPECT)/CT demonstrated a regional target/background ratio of more than 4 and a 2-fold higher iodine uptake than the mean uptake in liver parenchyma (in at least one tumor lesion) by visual assessment were considered responders, and 131I therapy was performed. Images were assessed by single-session consensus of two Nuclear Medicine Physicians. Personalized 131I therapy activity was derived from lung, blood, and lesion dosimetry. In all responders, redifferentiation treatment was continued until 2 days after 131I administration and a posttherapy whole-body scintigraphy was performed 7 days after 131I administration. In nonresponders the study medication was discontinued immediately and no 131I therapy was performed.

Figure 1.

Protocol design. QD, once daily; BID, twice daily; d/c, discontinue; WBS, whole body scintigraphy. Off-study examinations are marked with an asterisk.

Figure 1.

Protocol design. QD, once daily; BID, twice daily; d/c, discontinue; WBS, whole body scintigraphy. Off-study examinations are marked with an asterisk.

Close modal

Adverse events were monitored until 28 days after discontinuation of the study drugs and graded according to Common Terminology Criteria for Adverse Events (CTCAE; version 4.0) criteria.

Efficacy

In patients who underwent 131I therapy, 2[18F]fluoro-2-deoxy-D-glucose (FDG)-PET/CT imaging was performed between 3 and 12 months after 131I therapy and best imaging response based on RECIST 1.1 (23) and PET response criteria in solid tumors (PERCIST) 1.0 (24) criteria was assessed by single-session consensus of two readers.

Serum thyrotropin, thyroglobulin, and thyroglobulin antibody levels were monitored and the best thyroglobulin response within 1 year after 131I therapy was assessed.

Iodine imaging studies and dosimetry

Before any iodine imaging, a minimum interval of 2 months after the last administration of iodinated contrast-medium was mandated, and patients were advised to follow a low-iodine diet for at least 5 days. A mean of 3.9 (interquartile range: 3.9–4.2) mCi [equivalent to 144.5 (interquartile range: 143–157) MBq] 123I were administered after intramuscular injection of 0.9 mg recombinant thyrotropin on 2 consecutive days. After 4 hours, planar scintigraphy and–in the case of suspicious findings–additional SPECT/CT was performed. On the same day after 123I scintigraphy, 1.0 (range: 0.8–1.1) mCi [equivalent to 36.3 (range: 31–40) MBq] 124I were administered orally and PET/CT was acquired from skull to mid-thigh 20 and 120 hours after tracer ingestion.

Optimal 131I therapy activity was determined using CT-derived lesion volume and the lesion activity concentration on 124I PET for lesion dosimetry and repeated blood measurements for blood dosimetry in accordance with prior publications (25, 26).

Statistical analysis

The primary endpoint was the proportion of patients with successful redifferentiation on the 123I SPECT/CT scan. Secondary endpoints were thyroglobulin response and safety. RECIST/PERCIST response was assessed posthoc. Posthoc analysis of the statistical association with redifferentiation rate was assessed for a maximum of six parameters that were previously associated with patient outcome or redifferentiation success [i.e., histopathology, cumulative previously administered radioiodine activity, BRAF mutation status, FDG-PET peak standardized uptake value (SUVpeak), unstimulated thyroglobulin (Tg) pre-redifferentiation] using Fisher exact test (17, 18, 27–29). Continuous variables were dichotomized by receiver operating characteristic analyses; Youden J was used for optimal cut-off point determination. Mean pre- and post-redifferentiation mean absorbed doses per administered activity were assessed using a Wilcoxon signed-rank test. P values <0.05 were regarded as statistically significant. Data were collected using electronic case report forms in a Research Electronic Data Capture (REDCap) database (30) hosted at University Hospital Essen (University Duisburg-Essen, Essen, Germany). All statistical analyses were performed using R v4.0.3 (The R Foundation, r-project.org).

Data availability statement

The data generated in this study are not publicly available due to privacy concerns but are available upon reasonable request from the corresponding author.

Study population

Between March 2018 and March 2021, 40 patients were referred and screened for the study. 20 patients were eligible. The flow of patients is shown in Supplementary Fig. S1.

Baseline characteristics of the included patients are provided in Table 1. 10 (50%) patients had papillary thyroid carcinoma (PTC), 7 (35%) had follicular thyroid carcinoma (FTC), and 3 (15%) had poorly differentiated thyroid carcinoma (PDTC). 14 (70%) BRAF-WT and 6 (30%) BRAF-MUT patients were included. Sufficient histopathologic tissue for further assessment of the molecular profile was available in 15 of 20 (75%) patients. Using a previously published methodology (31), a solid tumor panel analysis of 26 different genes was performed (Supplementary Table S1). Telomerase reverse transcriptase (TERT) promoter mutations were detected in 10 of 15 (67%) patients, PTEN-mutations in 3 of 15 (20%) patients, and NRAS-mutations in 2 of 15 (13%) patients. 1 of 15 (7%) patients each had KRAS, AKT1, and APC (adenomatous polyposis coli) mutations, respectively.

Table 1.

Patient characteristics.

Age, years  
 Mean (range) 65.2 (48–83) 
Sex, n (%) 
 Male 9 (45) 
 Female 11 (55) 
Tumor type, n (%) 
 PTC 10 (50) 
 FTC 7 (35) 
 PDTC 3 (15) 
Unstimulated serum Tg (ng/mL) 
 Mean (range) median 4,213 (0–74,000) 
 Median 111 
BRAFV600E mutation status, n (%) 
 WT 14 (70) 
 MUT 6 (30) 
Further mutation status, n (%) 
 TERT promoter 10 (67) 
 PTEN 3 (20) 
 NRAS 2 (13) 
 KRAS 1 (7) 
 APC 1 (7) 
 AKT1 1 (7) 
Prior treatments 
 Cumulative 131I activity in mCi, mean (range) 383.8 (162.2–805) 
 Cumulative 131I activity in GBq, mean (range) 14.2 (6.0–29.8) 
 EBRT, n (%) 7 (35) 
Tumor sites, n (%) 
 Local 6 (30) 
 Cervical lymph nodes 9 (45) 
 Other lymph nodes 7 (35) 
 Lung 19 (95) 
 Bone 13 (65) 
 Other 3 (15) 
Age, years  
 Mean (range) 65.2 (48–83) 
Sex, n (%) 
 Male 9 (45) 
 Female 11 (55) 
Tumor type, n (%) 
 PTC 10 (50) 
 FTC 7 (35) 
 PDTC 3 (15) 
Unstimulated serum Tg (ng/mL) 
 Mean (range) median 4,213 (0–74,000) 
 Median 111 
BRAFV600E mutation status, n (%) 
 WT 14 (70) 
 MUT 6 (30) 
Further mutation status, n (%) 
 TERT promoter 10 (67) 
 PTEN 3 (20) 
 NRAS 2 (13) 
 KRAS 1 (7) 
 APC 1 (7) 
 AKT1 1 (7) 
Prior treatments 
 Cumulative 131I activity in mCi, mean (range) 383.8 (162.2–805) 
 Cumulative 131I activity in GBq, mean (range) 14.2 (6.0–29.8) 
 EBRT, n (%) 7 (35) 
Tumor sites, n (%) 
 Local 6 (30) 
 Cervical lymph nodes 9 (45) 
 Other lymph nodes 7 (35) 
 Lung 19 (95) 
 Bone 13 (65) 
 Other 3 (15) 

Abbreviation: EBRT, external beam radiotherapy.

Efficacy

Seven of 20 (35%) patients demonstrated successful redifferentiation on study medication; 5 of 14 (36%) in the BRAF-WT, 2 of 6 (33%) in the BRAF-MUT arm. Four of 7 (57%) patients had shown some iodine uptake at baseline. A comparison of pre- and post-redifferentiation 124I PET images of patients with successful restoration of radioiodine uptake are provided in Supplementary Fig. S2.

In patients with successful redifferentiation, dosimetry-guided 131I therapy with a mean (range) activity of 300.0 (273.0–421.6) mCi [equivalent to 11.1 (range: 10.1–15.6) GBq] 131I was performed.

Mean ± SD absorbed dose per administered activity was 0.03 ± 0.04 Gy/mCi (equivalent to 0.9 ± 1.2 Gy/GBq) before and 0.28 ± 0.22 Gy/mCi (equivalent to 7.6 ± 5.9 Gy/GBq) after successful redifferentiation (Fig. 3A).

Redifferentiation and 131I therapy led to any decrease in thyroglobulin level in 4 of 7 (57%) patients (Fig. 2A). Mean thyroglobulin decline was 58% (range: 36%–93%), respectively.

Figure 2.

Efficacy of redifferentiation therapy. Treatment response of patients with successful restoration of radioiodine uptake by change in (A) Tg level, (B) RECIST summed diameter (asterisk: occurrence of new lesions), (C) PERCIST SUVpeak (asterisk: occurrence of new lesions), and (D) mean absorbed dose on a per-patient level. Waterfall plots of individual patients are sorted by best Tg response. Of note, persistence of iodine-negative lesions after redifferentiation was observed in patients 5, 6, and 7 with a consecutive increase in Tg levels at follow-up. Pre-rediff, pre-redifferentiation; post-rediff, post-redifferentiation.

Figure 2.

Efficacy of redifferentiation therapy. Treatment response of patients with successful restoration of radioiodine uptake by change in (A) Tg level, (B) RECIST summed diameter (asterisk: occurrence of new lesions), (C) PERCIST SUVpeak (asterisk: occurrence of new lesions), and (D) mean absorbed dose on a per-patient level. Waterfall plots of individual patients are sorted by best Tg response. Of note, persistence of iodine-negative lesions after redifferentiation was observed in patients 5, 6, and 7 with a consecutive increase in Tg levels at follow-up. Pre-rediff, pre-redifferentiation; post-rediff, post-redifferentiation.

Close modal

Redifferentiation and 131I therapy led to any decrease in RECIST summed lesion diameter in 6 of 7 (86%) and a 30% or higher decrease in 1 of 7 (14%) patients, respectively (Fig. 2B). In 1 of these patients, the occurrence of new lesions was observed. Thus, best RECIST 1.1 response within 1 year after radioiodine treatment was partial response (PR; n = 1, 14%), stable disease (n = 5, 71%), and progressive disease (n = 1, 14%), respectively.

Any decrease in mean SUVpeak on FDG-PET for five lesions with the highest uptake was observed in 3 of 6 (50%) patients, an increase was observed in 3 of 6 (50%). All changes in mean SUVpeak were ≥30%. The best PERCIST response was partial metabolic response (N = 3, 50%), and progressive metabolic disease (N = 3, 50%; Fig. 2C); in 1 patient PERCIST assessment was not possible due to missing baseline FDG-PET/CT. Changes in per-lesion absorbed dose are provided in Fig. 2D for RECIST 1.1 target lesions and in Fig. 3A across all lesions.

Figure 3.

Association of lesion absorbed doses as assessed by 124I PET/CT and FDG-PET. A, Box plot showing pre- and post-redifferentiation therapy lesion-based mean absorbed dose per administered 131I activity (statistical significance in Wilcoxon test indicated by an asterisk; P < 0.00001) for all patients with successful restoration of radioiodine uptake. B, Dot plot showing the per-lesion association of post-redifferentiation absorbed dose and pre-redifferentiation FDG uptake on a per-lesion level for all patients with available FDG-PET (n = 16).

Figure 3.

Association of lesion absorbed doses as assessed by 124I PET/CT and FDG-PET. A, Box plot showing pre- and post-redifferentiation therapy lesion-based mean absorbed dose per administered 131I activity (statistical significance in Wilcoxon test indicated by an asterisk; P < 0.00001) for all patients with successful restoration of radioiodine uptake. B, Dot plot showing the per-lesion association of post-redifferentiation absorbed dose and pre-redifferentiation FDG uptake on a per-lesion level for all patients with available FDG-PET (n = 16).

Close modal

Between baseline and week 3 imaging RECIST 1.1 stable disease was observed in 18 of 20 (90%) patients and PR in 2of 20 (10%) patients. PR occurred in BRAF-MUT patients without restoration of radioiodine uptake.

Safety

A total number of 40 adverse events were observed in 17 of 20 (85%) patients. 38 of 40 (95%) events were graded as G1/2. Grade 3/4 events were observed in 2 (10%) patients: G3 pyrexia in 1 BRAF-MUT patient resolved after temporary discontinuation of trametinib + dabrafenib for 2 days and did not reoccur thereafter. Transient G4 rash in 1 BRAF-WT patient occurred 3 days after the termination of the study drug and occurred after the start of doxycycline intake. Table 2 lists safety details.

Table 2.

Adverse events stratified by BRAF status from treatment start until follow-up 28 days after discontinuation of the study medication.

BRAF-WT (n = 14)BRAF-MUT (n = 6)
Side effectAll grades, n (%)Grade 3/4, n (%)All grades, n (%)Grade 3/4, n (%)
Rash 14 (100.0) 1 (7.1) 1 (16.7) – 
Diarrhea 4 (28.6) – – – 
Oral mucositis 2 (14.3) – – – 
Hypertension 2 (14.3) – 1 (16.7) – 
Dyspepsia 1 (7.1) – – – 
Nausea 1 (7.1) – 2 (33.3) – 
Oral pain 1 (7.1) – – – 
Vomiting 1 (7.1) – 1 (16.7) – 
Fatigue 1 (7.1) – – – 
Dizziness 1 (7.1) – – – 
Syncope 1 (7.1) – – – 
Epistaxis 1 (7.1) – – – 
Scalp pain 1 (7.1) – – – 
Erythema – – 1 (16.7) – 
Palpitations – – 1 (16.7) – 
Pyrexia – – 2 (33.3) 1 (16.7) 
BRAF-WT (n = 14)BRAF-MUT (n = 6)
Side effectAll grades, n (%)Grade 3/4, n (%)All grades, n (%)Grade 3/4, n (%)
Rash 14 (100.0) 1 (7.1) 1 (16.7) – 
Diarrhea 4 (28.6) – – – 
Oral mucositis 2 (14.3) – – – 
Hypertension 2 (14.3) – 1 (16.7) – 
Dyspepsia 1 (7.1) – – – 
Nausea 1 (7.1) – 2 (33.3) – 
Oral pain 1 (7.1) – – – 
Vomiting 1 (7.1) – 1 (16.7) – 
Fatigue 1 (7.1) – – – 
Dizziness 1 (7.1) – – – 
Syncope 1 (7.1) – – – 
Epistaxis 1 (7.1) – – – 
Scalp pain 1 (7.1) – – – 
Erythema – – 1 (16.7) – 
Palpitations – – 1 (16.7) – 
Pyrexia – – 2 (33.3) 1 (16.7) 

Predictors of successful redifferentiation

An overview of factors and redifferentiation groups is provided in Table 3.

Table 3.

Predictors of successful redifferentiation.

Successful redifferentiation, n (%)
ParameterNoYesP
Histopathology PTC/FTC 11 (65) 6 (35) 1.00 
 PDTC 2 (67) 1 (33)  
Cumulative previous radioiodine activity ≤432 mCi (∼16 GBq) 12 (75) 4 (25) 0.10 
 >432 mCi (∼16 GBq) 1 (25) 3 (75)  
BRAFV600E mutation No 9 (64) 5 (36) 1.00 
 Yes 4 (67) 2 (33)  
SUVpeak (FDG-PET)a ≤10 3 (33) 6 (67) 0.01b 
 >10 7 (100) 0 (0)  
Unstimulated Tg pre-redifferentiation (ng/mL) ≤66 6 (100) 0 (0) 0.05 
 >66 7 (50) 7 (50)  
Histopathology/BRAFV600E mutationc FTC/no 4 (57) 3 (43) 1.00 
 PTC/no 3 (60) 2 (40)  
Successful redifferentiation, n (%)
ParameterNoYesP
Histopathology PTC/FTC 11 (65) 6 (35) 1.00 
 PDTC 2 (67) 1 (33)  
Cumulative previous radioiodine activity ≤432 mCi (∼16 GBq) 12 (75) 4 (25) 0.10 
 >432 mCi (∼16 GBq) 1 (25) 3 (75)  
BRAFV600E mutation No 9 (64) 5 (36) 1.00 
 Yes 4 (67) 2 (33)  
SUVpeak (FDG-PET)a ≤10 3 (33) 6 (67) 0.01b 
 >10 7 (100) 0 (0)  
Unstimulated Tg pre-redifferentiation (ng/mL) ≤66 6 (100) 0 (0) 0.05 
 >66 7 (50) 7 (50)  
Histopathology/BRAFV600E mutationc FTC/no 4 (57) 3 (43) 1.00 
 PTC/no 3 (60) 2 (40)  

aMean of five lesions with highest uptake.

bP < 0.05 by Fisher exact test.

cThe lower row presents a subgroup analysis for BRAF-WT PTC versus FTC.

A mean SUVpeak <10 on FDG-PET for five lesions with the highest uptake was determined as the optimal cut-off value for discrimination of redifferentiation success. Baseline SUVpeak significantly predicted redifferentiation rate: Redifferentiation was successful in 6 of 9 (66.7%) patients with a mean SUVpeak <10 and unsuccessful in all 11 patients with a higher SUVpeak (P = 0.01). Redifferentiation was not successful in any (n = 6) of the patients with an unstimulated Tg < median (66 ng/mL) but statistical significance was not reached (P = 0.05). A comparison of pre-redifferentiation SUVpeak and lesion absorbed dose is shown in Fig. 3B.

Histopathology (FTC/PTC vs. PDTC), cumulative previously administered radioiodine activity, BRAF-mutation status, and unstimulated thyroglobulin pre-redifferentiation were not significantly associated with redifferentiation success.

In a subgroup analysis comparing FTC with BRAF-WT PTC no association with redifferentiation success was observed (Table 3).

Here we demonstrate successful restoration of radioiodine uptake in about one third of patients with similar redifferentiation rate among patients with BRAF-MUT and BRAF-WT thyroid cancer. Subsequent 131I therapy led to decreases in thyroglobulin levels in more than half of the 131I-treated patients. Previous redifferentiation attempts using other substance classes, such as retinoids, resulted in only modest success (32–37). Further insight into molecular drivers of carcinogenesis and dedifferentiation led to improved redifferentiation outcomes by the use of MAPK pathway-targeting agents. In this redifferentiation trial, two arms were designed to assess genotype-guided MAPK inhibition with dual MEK and BRAF targeting for BRAF-MUT and single MEK targeting for BRAF-WT candidates. It is the first prospective study to employ combined BRAF- and MEK-targeting in BRAF-MUT unresectable RR-TC. Combination therapy demonstrated superior efficacy and safety when compared with monotherapy with BRAF small molecule inhibitors in BRAF-mutant melanoma patients (38, 39) and may be advantageous in the redifferentiation of RR-TC as well. Our study is the first redifferentiation trial using MAPK pathway-targeting that included a significant number of patients with FTC.

Successful redifferentiation was achieved by trametinib (BRAF-WT) or trametinib plus dabrafenib (BRAF-MUT) in about one third of patients in each group. The results of our study are in line with the findings of Ho and colleagues (17), Rothenberg and colleagues (19), Jaber and colleagues (40), and Dunn and colleagues (18), albeit with a lower redifferentiation rate.

Redifferentiation 131I therapy led to thyroglobulin decline in more than half of successfully redifferentiated patients, RECIST 1.1 partial response in 1 patient, and stable disease in more than two thirds of patients, as well as PERCIST partial response in half of patients. Response rates were similar in both arms, which indicates additional value of combined therapy in BRAF-MUT patients.

An overview of redifferentiation trials involving MAPK and BRAF inhibitors is provided in Table 4. Treatment response to 131I therapy was lower than in the trials of Rothenberg and colleagues (19), Ho and colleagues (17), Jaber and colleagues (40), Iravani and colleagues (41), and Dunn and colleagues (18), where partial responses were reported in 33%, 63%, 33%, 75%, and 50% of patients treated with radioiodine, respectively. Potential explanations include a lower rate of patients with radioiodine uptake present at baseline compared with Ho and colleagues (17), different definitions as to what constitutes radioiodine-refractoriness, and a shorter time span for redifferentiation treatment compared with Ho and colleagues (17), Rothenburg and colleagues (19), Iravani and colleagues (41), and Jaber and colleagues (40). In addition, in our study a minimum lesion size of 1.5 cm (short-axis diameter for nodal lesions, long-axis diameter for nonnodal lesions) was required for inclusion, whereas no size criteria were imposed in the other studies leading to the systematic exclusion of patients with lower tumor burden in our cohort. This may have negatively affected response rates, as small lesion size has previously been shown to be a positive predictor of response to 131I therapy, partly because large lesions are more likely to show a nonuniform dose distribution (25, 26).

Table 4.

Comparison of studies on redifferentiation involving MAPK and/or BRAF inhibitors identified via a PubMed database search using the search strings “Redifferentiation MEK” and “Redifferentiation BRAF” and the initial Ho and colleagues trial.

StudyCohortBRAF statusStudy drugsDrug intake until iodine imaging131I treatment, n (%)RECIST response
Ho (1713 PTC, 7 PDTC 9 BRAF-MUT, 11 BRAF-WT Selumetinib 4 weeks 8/20 (40) 5 PR, 3 SD 
Dunn (1810 PTC 10 BRAF-MUT Vemurafenib 4 weeks 4/10 (40) 2 PR, 2 SD 
Jaber (4010 PTC, 1 FTC, 2 PDTC 9 BRAF-MUT, 4 BRAF-WT Dabrafenib, vemurafenib, trametinib, investigational MEK inhibitor 14.3 months 9/13 (69) 3 PR, 6 SD 
Rothenberg (1910 PTC 10 BRAF-MUT Dabrafenib 25 days 6 (60) 2 PR, 4 SD 
Iravani (413 PTC, 2 FTC, 1 PDTC 3 BRAF-MUT, 3 BRAF-WT Dabrafenib + trametinib, trametinib 4 weeks 4 (66) 3 PR, 1 SD 
StudyCohortBRAF statusStudy drugsDrug intake until iodine imaging131I treatment, n (%)RECIST response
Ho (1713 PTC, 7 PDTC 9 BRAF-MUT, 11 BRAF-WT Selumetinib 4 weeks 8/20 (40) 5 PR, 3 SD 
Dunn (1810 PTC 10 BRAF-MUT Vemurafenib 4 weeks 4/10 (40) 2 PR, 2 SD 
Jaber (4010 PTC, 1 FTC, 2 PDTC 9 BRAF-MUT, 4 BRAF-WT Dabrafenib, vemurafenib, trametinib, investigational MEK inhibitor 14.3 months 9/13 (69) 3 PR, 6 SD 
Rothenberg (1910 PTC 10 BRAF-MUT Dabrafenib 25 days 6 (60) 2 PR, 4 SD 
Iravani (413 PTC, 2 FTC, 1 PDTC 3 BRAF-MUT, 3 BRAF-WT Dabrafenib + trametinib, trametinib 4 weeks 4 (66) 3 PR, 1 SD 

Abbreviations: PR, partial response; SD, stable disease.

Redifferentiation treatment in both arms was safe with only two grade 3/4 events that were both manageable. Successful redifferentiation through genotype-guided MAPK modulation and subsequent 131I therapy may delay the onset of tyrosine kinase inhibitors (TKI), especially in patients with low FDG avidity. Delayed onset of TKIs comes with potential benefits for safety, quality of life, and economic burden. In 1 patient successful redifferentiation was repeated a second time after 2 years leading to further decreases in tumor size and thyroglobulin levels (Supplementary Fig. S3).

The implementation of pre-therapeutic 124I PET in this study enabled dosimetry of tumor lesions and organs-at-risk leading to the administration of higher activities than in previous redifferentiation trials. High 124I PET-derived tumor absorbed doses after 131I therapy were associated with thyroglobulin decline (Fig. 2D). In addition, in patients with extensive prior 131I therapy pretherapeutic 124I PET dosimetry allows for a deescalation of treatment activity to reduce the risk of side effects.

In a posthoc analysis, we assessed predictors of therapy response. Low tumor glycolytic rate by FDG-PET was a significant predictor of redifferentiation success, which is likely due to the known association between FDG uptake and dedifferentiation in thyroid carcinoma (42, 43) and a variety of other tumor entities (44–46). As previously shown (18), low Tg values as a biochemical sign of dedifferentiation might be associated with a lower redifferentiation rate.

Our study benefits from systematic baseline and follow-up 124I PET dosimetry and additional FDG-PET assessment. However, several limitations are noted:

The small number of included patients may affect the reliability of the study results. In addition, a larger range for imaging follow-up of 3 to 12 months may have reduced the consistency of RECIST/PERCIST assessments. In addition, the follow-up period does not yet allow drawing reliable conclusions on late toxicity, such as radiation fibrosis and hematopoietic disorders, as well as the extent to which systemic treatment can be delayed. Due to the trial design, no comparison with standalone MEK- and BRAF-targeting without subsequent 131I therapy or TKI treatment was performed. Of note, 2 of 6 BRAF-MUT patients demonstrated partial response after dabrafenib + trametinib treatment without sufficient restoration of radioiodine uptake. This suggests that combined MAK- and BRAF-targeting itself may have been efficacious, which is in line with the findings of performed trials (47, 48).

Furthermore, BRAF mutation status was only evaluated in one lesion per patient potentially leading to misclassification of patients with a discordant molecular profile among tumor lesions. However, the published literature implies a high concordance of BRAF mutation status between primary tumor, lymph node metastases, and remote metastases (49 51).

Conclusion

Genotype-guided MAPK inhibition was safe and resulted in successful redifferentiation in about one third of patients. The redifferentiation rate was similar in BRAF-MUT and -WT arms indicating additional value of combination therapy for BRAF-MUT cancer. Following successful redifferentiation, dosimetry-guided 131I therapy resulted in Tg decline in more than half of patients and morphologic and metabolic disease control was noted. Low tumor glycolytic rate by FDG-PET, i.e., SUVpeak of 10 or less, was a predictor of redifferentiation success.

M. Weber reports personal fees from Boston Scientific, Terumo, Eli Lilly and Company, and Advanced Accelerator Applications outside the submitted work. D. Kersting reports grants from Pfizer outside the submitted work as well as support by the clinician scientist program of University Medicine Essen Clinician Scientist Academy (UMEA), sponsored by Faculty of Medicine (University of Duisburg-Essen) and German Research Foundation [Deutsche Forschungsgemeinschaft (DFG)]. T. Brandenburg reports personal fees from Eli Lilly and Company, Eisai, Bayer Pharmaceuticals, and Liberum and grants from UMEA Clinician Scientist Program (DGE) University Hospital Essen (University of Duisburg-Essen) outside the submitted work. D. Führer-Sakel reports personal fees from Eisai outside the submitted work. M.C. Kreissl reports personal fees and nonfinancial support from Eisai, GE Healthcare, Bayer HealthCare; AAA/Novartis, Takeda, Eli Lilly and Company, Sanofi-Aventis and Liam Gmbh and personal fees from Ipsen, Exelixis, Roche, Pfizer, OPEN Health, and Onkowissen outside the submitted work. K. Herrmann reports personal fees from SOFIE Biosciences, Bayer HealthCare, Adacap/Novartis, Curium, ITG/ITM, BSCI, Ipsen, Siemens Healthineers, GE Healthcare, Amgen, Y-mAbs Therapeutics, Inc., Aktis Oncology, Theragnostics, and Pharma15 outside the submitted work. C. Rischpler reports grants and personal fees from Pfizer and personal fees from Adacap, Alnylam, BTG, GE Healthcare, and Siemens Healthineers outside the submitted work. J. Nagarajah reports nonfinancial support from Novartis, personal fees from Curium and Bayer HealthCare, and grants from ABX outside the submitted work. W.P. Fendler reports grants from SOFIE Biosciences; personal fees from Janssen, Calyx, and Parexel; and grants and personal fees from Bayer HealthCare outside the submitted work. No disclosures were reported by the other authors.

M. Weber: Resources, data curation, formal analysis, methodology, writing–original draft, writing–review and editing. D. Kersting: Data curation, software, formal analysis, validation, visualization, writing–original draft, writing–review and editing. B. Riemann: Resources, writing–review and editing. T. Brandenburg: Writing–review and editing. D. Führer-Sakel: Resources, writing–review and editing. F. Grünwald: Resources, writing–review and editing. M.C. Kreissl: Resources, writing–review and editing. H. Dralle: Resources, writing–review and editing. F. Weber: Resources, writing–review and editing. K.W. Schmid: Resources, writing–review and editing. K. Herrmann: Supervision, investigation, writing–review and editing. W. Jentzen: Formal analysis, investigation, writing–review and editing. H. Grafe: Data curation, supervision, writing–review and editing. C. Rischpler: Supervision, investigation, writing–review and editing. S. Theurer: Data curation, investigation, writing–review and editing. A. Bockisch: Conceptualization, resources, supervision, investigation, methodology, writing–review and editing. J. Nagarajah: Conceptualization, data curation, investigation, methodology, writing–review and editing. W.P. Fendler: Data curation, supervision, validation, investigation, methodology, writing–original draft, project administration, writing–review and editing.

We acknowledge the substantial contribution by our colleague Ina Binse (MD), who was deeply involved in the conduction of the trial, but sadly passed before completion.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
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
.
2.
Fugazzola
L
,
Elisei
R
,
Fuhrer
D
,
Jarzab
B
,
Leboulleux
S
,
Newbold
K
, et al
.
2019 European thyroid association guidelines for the treatment and follow-up of advanced radioiodine-refractory thyroid cancer
.
Eur Thyroid J
2019
;
8
:
227
45
.
3.
Busaidy
NL
,
Cabanillas
ME
.
Differentiated thyroid cancer: management of patients with radioiodine nonresponsive disease
.
J Thyroid Res
2012
;
2012
:
618985
.
4.
Durante
C
,
Haddy
N
,
Baudin
E
,
Leboulleux
S
,
Hartl
D
,
Travagli
JP
, et al
.
Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy
.
J Clin Endocrinol Metab
2006
;
91
:
2892
9
.
5.
Schlumberger
M
,
Brose
M
,
Elisei
R
,
Leboulleux
S
,
Luster
M
,
Pitoia
F
, et al
.
Definition and management of radioactive iodine-refractory differentiated thyroid cancer
.
Lancet Diabetes Endocrinol
2014
;
2
:
356
8
.
6.
Cohen
Y
,
Xing
M
,
Mambo
E
,
Guo
Z
,
Wu
G
,
Trink
B
, et al
.
BRAF mutation in papillary thyroid carcinoma
.
J Natl Cancer Inst
2003
;
95
:
625
7
.
7.
Kimura
ET
,
Nikiforova
MN
,
Zhu
Z
,
Knauf
JA
,
Nikiforov
YE
,
Fagin
JA
.
High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma
.
Cancer Res
2003
;
63
:
1454
7
.
8.
Knauf
JA
,
Fagin
JA
.
Role of MAPK pathway oncoproteins in thyroid cancer pathogenesis and as drug targets
.
Curr Opin Cell Biol
2009
;
21
:
296
303
.
9.
Soares
P
,
Trovisco
V
,
Rocha
AS
,
Lima
J
,
Castro
P
,
Preto
A
, et al
.
BRAF mutations and RET/PTC rearrangements are alternative events in the etiopathogenesis of PTC
.
Oncogene
2003
;
22
:
4578
80
.
10.
Knauf
JA
,
Kuroda
H
,
Basu
S
,
Fagin
JA
.
RET/PTC-induced dedifferentiation of thyroid cells is mediated through Y1062 signaling through SHC-RAS-MAP kinase
.
Oncogene
2003
;
22
:
4406
12
.
11.
Durante
C
,
Puxeddu
E
,
Ferretti
E
,
Morisi
R
,
Moretti
S
,
Bruno
R
, et al
.
BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism
.
J Clin Endocrinol Metab
2007
;
92
:
2840
3
.
12.
Liu
D
,
Hu
S
,
Hou
P
,
Jiang
D
,
Condouris
S
,
Xing
M
.
Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant
.
Clin Cancer Res
2007
;
13
:
1341
9
.
13.
Mitsutake
N
,
Knauf
JA
,
Mitsutake
S
,
Mesa
C
,
Zhang
L
,
Fagin
JA
.
Conditional BRAFV600E expression induces DNA synthesis, apoptosis, dedifferentiation, and chromosomal instability in thyroid PCCL3 cells
.
Cancer Res
2005
;
65
:
2465
73
.
14.
Riesco-Eizaguirre
G
,
Gutiérrez-Martínez
P
,
García-Cabezas
MA
,
Nistal
M
,
Santisteban
P
.
The oncogene BRAF V600E is associated with a high risk of recurrence and less differentiated papillary thyroid carcinoma due to the impairment of Na+/I- targeting to the membrane
.
Endocr Relat Cancer
2006
;
13
:
257
69
.
15.
Buffet
C
,
Wassermann
J
,
Hecht
F
,
Leenhardt
L
,
Dupuy
C
,
Groussin
L
, et al
.
Redifferentiation of radioiodine-refractory thyroid cancers
.
Endocr Relat Cancer
2020
;
27
:
R113
32
.
16.
Chakravarty
D
,
Santos
E
,
Ryder
M
,
Knauf
JA
,
Liao
X-H
,
West
BL
, et al
.
Small-molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation
.
J Clin Invest
2011
;
121
:
4700
11
.
17.
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
.
18.
Dunn
LA
,
Sherman
EJ
,
Baxi
SS
,
Tchekmedyian
V
,
Grewal
RK
,
Larson
SM
, et al
.
Vemurafenib Redifferentiation of BRAF Mutant, RAI-Refractory Thyroid Cancers
.
J Clin Endocrinol Metab
2019
;
104
:
1417
28
.
19.
Rothenberg
SM
,
Mcfadden
DG
,
Palmer
EL
,
Daniels
GH
,
Wirth
LJ
.
Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib
.
Clin Cancer Res
2015
;
21
:
1028
35
.
20.
Nagarajah
J
,
Le
M
,
Knauf
JA
,
Ferrandino
G
,
Montero-Conde
C
,
Pillarsetty
N
, et al
.
Sustained ERK inhibition maximizes responses of BrafV600E thyroid cancers to radioiodine
.
J Clin Invest
2016
;
126
:
4119
24
.
21.
Pentlow
KS
,
Graham
MC
,
Lambrecht
RM
,
Daghighian
F
,
Bacharach
SL
,
Bendriem
B
, et al
.
Quantitative imaging of iodine-124 with PET
.
J Nucl Med
1996
;
37
:
1557
62
.
22.
Sgouros
G
,
Kolbert
KS
,
Sheikh
A
,
Pentlow
KS
,
Mun
EF
,
Barth
A
, et al
.
Patient-specific dosimetry for 131I thyroid cancer therapy using 124I PET and 3-dimensional-internal dosimetry (3D-ID) software
.
J Nucl Med
2004
;
45
:
1366
72
.
23.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
.
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
24.
Wahl
RL
,
Jacene
H
,
Kasamon
Y
,
Lodge
MA
.
From RECIST to PERCIST: Evolving Considerations for PET response criteria in solid tumors
.
J Nucl Med
2009
;
50
Suppl 1
:
122S
50S
.
25.
Jentzen
W
,
Freudenberg
L
,
Bockisch
A
.
Quantitative imaging of (124)I with PET/CT in pretherapy lesion dosimetry. Effects impairing image quantification and their corrections
.
Q J Nucl Med Mol Imaging
2011
;
55
:
21
43
.
26.
Jentzen
W
,
Hoppenbrouwers
J
,
Van Leeuwen
P
,
Van Der Velden
D
,
Van De Kolk
R
,
Poeppel
TD
, et al
.
Assessment of lesion response in the initial radioiodine treatment of differentiated thyroid cancer using 124I PET imaging
.
J Nucl Med
2014
;
55
:
1759
65
.
27.
Deandreis
D
,
Al Ghuzlan
A
,
Leboulleux
S
,
Lacroix
L
,
Garsi
JP
,
Talbot
M
, et al
.
Do histological, immunohistochemical, and metabolic (radioiodine and fluorodeoxyglucose uptakes) patterns of metastatic thyroid cancer correlate with patient outcome?
Endocr Relat Cancer
2011
;
18
:
159
69
.
28.
Ibrahimpasic
T
,
Ghossein
R
,
Shah
JP
,
Ganly
I
.
Poorly differentiated carcinoma of the thyroid gland: current status and future prospects
.
Thyroid
2019
;
29
:
311
21
.
29.
Martins-Filho
R
,
Ward
LS
,
Amorim
BJ
,
Santos
AO
,
Lima
MCLD
,
Ramos
CD
, et al
.
Cumulative doses of radioiodine in the treatment of differentiated thyroid carcinoma: knowing when to stop
.
Arq Bras Endocrinol Metabol
2010
;
54
:
807
12
.
30.
Harris
PA
,
Taylor
R
,
Thielke
R
,
Payne
J
,
Gonzalez
N
,
Conde
JG
.
Research electronic data capture (REDCap)–a metadata-driven methodology and workflow process for providing translational research informatics support
.
J Biomed Inform
2009
;
42
:
377
81
.
31.
Theurer
S
,
Ingenwerth
M
,
Herold
T
,
Herrmann
K
,
Schmid
KW
.
Immunohistochemical profile and 47-gene next-generation sequencing (NGS) solid tumor panel analysis of a series of 13 strumal carcinoids
.
Endocr Pathol
2020
;
31
:
101
7
.
32.
Short
SC
,
Suovuori
A
,
Cook
G
,
Vivian
G
,
Harmer
C
.
A phase II study using retinoids as redifferentiation agents to increase iodine uptake in metastatic thyroid cancer
.
Clin Oncol (R Coll Radiol)
2004
;
16
:
569
74
.
33.
Simon
D
,
Koehrle
J
,
Reiners
C
,
Boerner
AR
,
Schmutzler
C
,
Mainz
K
, et al
.
Redifferentiation therapy with retinoids: therapeutic option for advanced follicular and papillary thyroid carcinoma
.
World J Surg
1998
;
22
:
569
74
.
34.
Kim
WG
,
Kim
EY
,
Kim
TY
,
Ryu
J-S
,
Hong
SJ
,
Kim
WB
, et al
.
Redifferentiation therapy with 13-cis retinoic acids in radioiodine-resistant thyroid cancer
.
Endocr J
2009
;
56
:
105
12
.
35.
Rosenbaum-Krumme
SJ
,
Bockisch
A
,
Nagarajah
J
.
Pioglitazone therapy in progressive differentiated thyroid carcinoma
.
Nuklearmedizin
2012
;
51
:
111
5
.
36.
Rosenbaum-Krumme
SJ
,
Freudenberg
LS
,
Jentzen
W
,
Bockisch
A
,
Nagarajah
J
.
Effects of rosiglitazone on radioiodine negative and progressive differentiated thyroid carcinoma as assessed by (1)(2)(4)I PET/CT imaging
.
Clin Nucl Med
2012
;
37
:
e47
52
.
37.
Tepmongkol
S
,
Keelawat
S
,
Honsawek
S
,
Ruangvejvorachai
P
.
Rosiglitazone effect on radioiodine uptake in thyroid carcinoma patients with high thyroglobulin but negative total body scan: a correlation with the expression of peroxisome proliferator-activated receptor-gamma
.
Thyroid
2008
;
18
:
697
704
.
38.
Flaherty
KT
,
Infante
JR
,
Daud
A
,
Gonzalez
R
,
Kefford
RF
,
Sosman
J
, et al
.
Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations
.
N Engl J Med
2012
;
367
:
1694
703
.
39.
Long
GV
,
Flaherty
KT
,
Stroyakovskiy
D
,
Gogas
H
,
Levchenko
E
,
De Braud
F
, et al
.
Dabrafenib plus trametinib versus dabrafenib monotherapy in patients with metastatic BRAF V600E/K-mutant melanoma: long-term survival and safety analysis of a phase 3 study
.
Ann Oncol
2017
;
28
:
1631
9
.
40.
Jaber
T
,
Waguespack
SG
,
Cabanillas
ME
,
Elbanan
M
,
Vu
T
,
Dadu
R
, et al
.
Targeted therapy in advanced thyroid cancer to resensitize tumors to radioactive iodine
.
J Clin Endocrinol Metab
2018
;
103
:
3698
705
.
41.
Iravani
A
,
Solomon
B
,
Pattison
DA
,
Jackson
P
,
Ravi Kumar
A
,
Kong
G
, et al
.
Mitogen-activated protein kinase pathway inhibition for redifferentiation of radioiodine refractory differentiated thyroid cancer: an evolving protocol
.
Thyroid
2019
;
29
:
1634
45
.
42.
Gild
ML
,
Topliss
DJ
,
Learoyd
D
,
Parnis
F
,
Tie
J
,
Hughes
B
, et al
.
Clinical guidance for radioiodine refractory differentiated thyroid cancer
.
Clin Endocrinol (Oxf)
2018
;
88
:
529
37
.
43.
Lin
Y
,
Wang
C
,
Gao
W
,
Cui
R
,
Liang
J.
Overwhelming rapid metabolic and structural response to apatinib in radioiodine refractory differentiated thyroid cancer
.
Oncotarget
2017
;
8
:
42252
61
.
44.
Flavell
RR
,
Naeger
DM
,
Mari Aparici
C
,
Hawkins
RA
,
Pampaloni
MH
,
Behr
SC
.
Malignancies with low fluorodeoxyglucose uptake at PET/CT: pitfalls and prognostic importance: resident and fellow education feature
.
Radiographics
2016
;
36
:
293
4
.
45.
Folpe
AL
,
Sprouse
JT
,
Conrad
EU
3rd
,
Eary
JF
. (
F-18) fluorodeoxyglucose positron emission tomography as a predictor of pathologic grade and other prognostic variables in bone and soft tissue sarcoma
.
Clin Cancer Res
2000
;
6
:
1279
87
.
46.
Binderup
T
,
Knigge
U
,
Loft
A
,
Federspiel
B
,
Kjaer
A
.
18F-fluorodeoxyglucose positron emission tomography predicts survival of patients with neuroendocrine tumors
.
Clin Cancer Res
2010
;
16
:
978
85
.
47.
Falchook
GS
,
Millward
M
,
Hong
D
,
Naing
A
,
Piha-Paul
S
,
Waguespack
SG
, et al
.
BRAF inhibitor dabrafenib in patients with metastatic BRAF-mutant thyroid cancer
.
Thyroid
2015
;
25
:
71
77
.
48.
Brose
MS
,
Cabanillas
ME
,
Cohen
EEW
,
Wirth
LJ
,
Riehl
T
,
Yue
H
, et al
.
Vemurafenib in patients with BRAF(V600E)-positive metastatic or unresectable papillary thyroid cancer refractory to radioactive iodine: a non-randomised, multicentre, open-label, phase 2 trial
.
Lancet Oncol
2016
;
17
:
1272
82
.
49.
Melo
M
,
Gaspar Da Rocha
A
,
Batista
R
,
Vinagre
J
,
Martins
MJ
,
Costa
G
, et al
.
TERT, BRAF, and NRAS in primary thyroid cancer and metastatic disease
.
J Clin Endocrinol Metab
2017
;
102
:
1898
907
.
50.
Fakhruddin
N
,
Jabbour
M
,
Novy
M
,
Tamim
H
,
Bahmad
H
,
Farhat
F
, et al
.
BRAF and NRAS mutations in papillary thyroid carcinoma and concordance in BRAF mutations between primary and corresponding lymph node metastases
.
Sci Rep
2017
;
7
:
4666
.
51.
Sohn
SY
,
Park
WY
,
Shin
HT
,
Bae
JS
,
Ki
CS
,
Oh
YL
, et al
.
Highly concordant key genetic alterations in primary tumors and matched distant metastases in differentiated thyroid cancer
.
Thyroid
2016
;
26
:
672
82
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

Supplementary data