Purpose: Targeting mutations leading to PI3K/mTOR/Akt activation are of interest in thyroid cancer. We evaluated the efficacy of everolimus in aggressive, radioactive iodine–refractory (RAIR) thyroid cancer and correlated tumor mutational profiling with response. Exploratory medullary and anaplastic thyroid cancer cohorts were included.

Experimental Design: This single-arm, multi-institutional phase II study was conducted from 2009 to 2013 in patients with incurable RAIR thyroid cancer who had radiographic progression six months prior to enrollment. The primary endpoint was progression-free survival (PFS) with a median follow-up of 31.8 months. The study is closed to enrollment but treatment and follow-up are ongoing. A targeted next-generation sequencing platform was used for mutational analysis.

Results: Thirty-three patients with differentiated thyroid cancer (DTC), 10 with medullary thyroid cancer (MTC), and 7 with anaplastic thyroid cancer (ATC) enrolled. For the DTC cohort, median PFS was 12.9 months (95% CI, 7.3–18.5) with a 2-year PFS of 23.6% (95% CI, 10.5–39.5). Median OS was not reached; 2-year OS was 73.5% (95% CI, 53.8–85.8). Among ATC patients, 1 had a partial response and was progression-free until 17.9 months after study entry and one had disease stability for 26 months, respectively. The genomically profiled cohort enriched for PI3K/mTOR/Akt alterations. PI3K/mTOR/Akt–mutated ATC subgroups appeared to benefit from everolimus. Treatment-related adverse events were as anticipated.

Conclusions: Everolimus has significant antitumor activity in thyroid cancer. While genomic profiling does not currently guide therapeutic selection in thyroid cancer patients, these data have important implications when considering the use of an mTOR inhibitor in an era of precision medicine. Clin Cancer Res; 24(7); 1546–53. ©2018 AACR.

Translational Relevance

What is the clinical efficacy of everolimus in advanced thyroid cancer and which genetic parameters predict response? In this nonrandomized phase II study of 50 adults with prior progression within 6 months, median progression-free survival was 12.5 months with 23.6% of differentiated thyroid cancer (DTC) patients alive and progression-free at 2 years. Patients with PI3K/mTOR/Akt–mutated anaplastic thyroid carcinoma seemed to benefit most. Everolimus demonstrates clinical activity in advanced thyroid cancers, and genomic testing to predict response appears promising.

Of an estimated 64,300 new cases of thyroid cancer diagnosed in 2016, 90% or more were differentiated thyroid cancer (DTC) that comprises both papillary and follicular subtypes, as well as the oncocytic variant of follicular thyroid cancer (FTC): Hürthle cell thyroid carcinoma (HCTC; ref. 1). A subset of DTC patients will eventually progress to radioiodine refractory (RAIR) status with 10-year survival rates approaching 10% (2). Beyond DTC, medullary thyroid cancer (MTC) is a neuroendocrine tumor arising from the parafollicular C-cells of the thyroid gland occasionally linked to heritable cancer syndromes (3). Poorly differentiated TC (PDTC) or anaplastic thyroid cancer (ATC) are rare subtypes with aggressive clinical behavior and unfavorable outcomes (4).

Multitargeted tyrosine kinase inhibitors (TKI) have proved beneficial in the treatment of RAIR DTC and advanced MTC. Both sorafenib and lenvatinib have demonstrated improvements in progression-free survival (PFS) compared with placebo (10.8 vs. 5.8 months and 18 vs. 3.6 months, respectively) in RAIR DTC (5, 6). Similarly, vandetanib and cabozantinib have shown a PFS benefit in MTC (7, 8). However, available treatments for patients with ATC have limited efficacy.

Current multitargeted TKIs target the VEGF receptor (VEGFR) that increases endothelial cell proliferation and promotes angiogenesis. Frequent somatic gene alterations in thyroid cancer involve BRAF (namely the V600E mutation), RAS, or RET that activate the MAPK, but are also linked to PI3K/mTOR/Akt pathways (9). Downstream of RAS, alterations in PI3K/mTOR/Akt have gained interest in thyroid cancer. Upon activation by a receptor tyrosine kinase (RTK), PI3K leads to the subsequent activation of Akt in a RAS-dependent manner, activating mTOR. mTOR promotes cell proliferation and alters apoptosis. Constitutive activation of the pathway can result from activating mutations in PI3K/mTOR/Akt or with loss of PTEN (10). In a group of unselected, early-stage thyroid cancer patients, these alterations appear to be rare (11). Similarly, alterations in RET can lead to PI3K/mTOR/Akt activation (12). These findings suggest that targeting mTOR may be a useful therapeutic strategy in thyroid cancer.

We sought to evaluate the clinical efficacy of everolimus, a rapamycin analogue which allosterically inhibits mTOR and prevents assembly of the mTOR complex 1 (mTORC1), in patients with aggressive, incurable DTC and explore efficacy in MTC and ATC. We also present genomic sequencing data to identify a molecular basis for treatment response.

Participants

Patients were eligible if they were 18 years of age or older with measurable disease, and had a diagnosis of locoregionally advanced or metastatic, incurable thyroid cancer with radiographic evidence of progression by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 within the previous 6 months. DTC patients had to have radioactive iodine refractory (RAIR) disease. Patients could have received up to one prior TKI. No documented disease progression was required for symptomatic MTC or ATC patients. Additional eligibility criteria included: Eastern Cooperative Oncology Group (ECOG) performance status of 2 or less and adequate organ function. Patients with untreated brain metastases were excluded. The study was approved by the institutional review board at each participating site and was conducted in compliance with the Declaration of Helsinki and local regulatory requirements.

Study design and endpoints

In this prospective open-label, nonrandomized, single-arm phase II study, patients were recruited at four centers throughout the United States to receive everolimus 10 mg by mouth daily. The institutional review board at each participating site approved the study and all patients provided written informed consent. Treatment was administered until disease progression, unacceptable adverse event, withdrawal, physician discretion, or death, whichever occurred first. Two dose reductions were permitted for toxicity: everolimus to 5 mg daily and 5 mg every other day. Patients were permitted to reescalate to the prior dose if symptoms were not attributed to treatment. Patients were followed for response with CT or MRI until first disease progression and for survival for 5 years according to the following schedule: at baseline, every 2 months for the first 24 months, and every 3 months beyond 24 months through 5 years. Adverse events were classified and graded according to National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. The primary endpoint of the study was PFS, with response rate and overall survival (OS) as key secondary endpoints.

Genomic sequencing of tumor tissue

Sequencing was performed on DNA extracted from pretreatment, fresh-frozen paraffin-embedded (FFPE) tumor samples. While biopsy just prior to study enrollment was encouraged, archival tumor samples were also permitted. Briefly, total gDNA concentration was determined and samples with sufficient starting material were taken to library construction. Libraries were quantified using qPCR, pooled in equal mass, and captured using the OncoPanel_v2 bait set. Oncopanel_v2 consists of 504 genes and 15 intronic regions with known or potential importance in cancer. Captured libraries underwent paired end 100 (2 × 100 nt) sequencing on a Hiseq 2500 (Illumina Inc.). Read pairs were aligned and demultiplexed using Picard tools. Mutation analysis for single-nucleotide variants was performed using MuTect v1.1.4 in paired mode for tumors with matched germline or single mode for samples without matched germline, and annotated using Oncotator (13). The alignments were further refined using the GATK tool for localized realignment around indel sites. All samples met a minimum requirement that 80% of targets had a minimum coverage of 30×. Structural rearrangements were not reported.

Statistical considerations

The primary objective was to assess PFS in patients with DTC. Thirty-three patients with DTC provided 97% power to rule out a 12% PFS rate and target a 35% PFS rate (two-stage design used with type-1 error rate of 10%, assuming ≥2 alive and progression-free at 6 months among the first 18 eligible patients accrued in the first stage). Ten patients with MTC and 7 patients with ATC were also to be accrued for a total overall accrual of 50 patients. PFS was defined as time from study entry to disease progression or death, whichever occurred first, and otherwise censored at date last known progression-free. Secondary endpoints included response rate (defined according to modified RECIST criteria), overall survival, and assessment of adverse events. Exact binomial confidence intervals were calculated for response. PFS and OS were estimated using the Kaplan–Meier method. Fisher exact test (categorical variables) and Wilcoxon rank-sum test (continuous variables) were used to assess for differences between patients with and without genomic data, with respect to baseline characteristics.

Patient characteristics

Between July 2009 and August 2013, 50 patients were enrolled (n = 33 DTC including 13 HCTCs, n = 10 MTC, n = 7 ATC). The median age at registration was 63 years (range 26–82), the majority were male (66%, 33/50), and PTC was the most common histologic subtype (28%, 14/50). Forty-four percent (22/50) of patients received prior TKI (Table 1).

Table 1.

Demographics and clinical characteristics in patients treated with everolimus for advanced thyroid cancer

Overall (%)aDifferentiatedMTCATC
Histology 50 (100) 33 (66) 10 (20) 7 (4) 
 PTC  14   
 HCTC  13   
 FTC    
 PDTC    
Gender 
 Male 33 (66) 21 (64) 9 (90) 3 (43) 
 Female 17 (34) 12 (36) 1 (10) 4 (57) 
Age at study registration 
 (y, range) 63 (26–82) 63 (45–82) 55 (26–78) 62 (47–75) 
Race 
 White 42 (84) 27 (82) 9 (90) 6 (86) 
 Black 5 (10) 4 (12) 1 (10) 
 Other 3 (6) 2 (6) 1 (14) 
ECOG performance status 
 0 32 (64) 19 (58) 10 (100) 3 (43) 
 1 14 (28) 12 (36) 2 (28) 
 2 4 (8) 2 (6) 2 (28) 
Site of distant metastases 
 None 1 (2) 1 (3)   
 Bone 21 (42) 16 (48) 4 (40) 1 (14) 
 Lung 35 (70) 23 (70) 5 (50) 7 (100) 
 Liver 14 (28) 7 (21) 6 (60) 1 (14) 
 Lymph nodes 26 (52) 16 (48) 6 (60) 4 (57) 
 Other 12 (24) 9 (27) 1 (10) 2 (28) 
Prior treatment(s) 
 Surgery 44 (88) 32 (97) 7 (70) 5 (71) 
 RAI 31 (62) 28 (85) 1 (10) 2 (28) 
 EBRT 30 (60) 21 (63) 5 (50) 4 (57) 
 Chemotherapy 7 (14) 4 (12) 1 (10) 2 (28) 
 TKI 22 (44) 17 (51) 4 (40) 1 (14) 
Overall (%)aDifferentiatedMTCATC
Histology 50 (100) 33 (66) 10 (20) 7 (4) 
 PTC  14   
 HCTC  13   
 FTC    
 PDTC    
Gender 
 Male 33 (66) 21 (64) 9 (90) 3 (43) 
 Female 17 (34) 12 (36) 1 (10) 4 (57) 
Age at study registration 
 (y, range) 63 (26–82) 63 (45–82) 55 (26–78) 62 (47–75) 
Race 
 White 42 (84) 27 (82) 9 (90) 6 (86) 
 Black 5 (10) 4 (12) 1 (10) 
 Other 3 (6) 2 (6) 1 (14) 
ECOG performance status 
 0 32 (64) 19 (58) 10 (100) 3 (43) 
 1 14 (28) 12 (36) 2 (28) 
 2 4 (8) 2 (6) 2 (28) 
Site of distant metastases 
 None 1 (2) 1 (3)   
 Bone 21 (42) 16 (48) 4 (40) 1 (14) 
 Lung 35 (70) 23 (70) 5 (50) 7 (100) 
 Liver 14 (28) 7 (21) 6 (60) 1 (14) 
 Lymph nodes 26 (52) 16 (48) 6 (60) 4 (57) 
 Other 12 (24) 9 (27) 1 (10) 2 (28) 
Prior treatment(s) 
 Surgery 44 (88) 32 (97) 7 (70) 5 (71) 
 RAI 31 (62) 28 (85) 1 (10) 2 (28) 
 EBRT 30 (60) 21 (63) 5 (50) 4 (57) 
 Chemotherapy 7 (14) 4 (12) 1 (10) 2 (28) 
 TKI 22 (44) 17 (51) 4 (40) 1 (14) 

aexcept age.

Abbreviations: ECOG, Eastern Cooperative Oncology Group; RAI, radioactive iodine; EBRT, external beam radiotherapy; TKI, tyrosine kinase inhibitor.

Clinical efficacy

At the time of data cutoff, 46 patients had discontinued protocol treatment [median time on treatment was 10.1 months (range < 1–29.8) with disease progression as the most common reason for stopping]. There were 43 PFS events and 23 patients had died. There were no complete responses (CR), but 3 (6%) partial responses (PR) were observed and 37 patients (74%) experienced stable disease (SD), for a clinical benefit rate (CBR = CR+PR+SD) of 80% (Table 2; Supplementary Table S2). At a median follow-up of 31.8 months, median PFS was 12.5 months (95% CI, 7.2–17.5) with 22.2% of patients alive and progression-free at 2 years. Median OS was 32.7 months (95% CI, 18.7–61.3+) for the entire cohort with 60.7% of patients alive at 2 years.

Table 2.

Clinical response and outcomes to everolimus in patients with ATCs

Overall [n = 50]Differentiated [n = 33]MTC [n = 10]ATC [n = 7]
Best response to therapy 
 CR — — — — 
 PR 3 (6) 1 (3) 1 (10) 1 (14) 
 SD 37 (74) 27 (82) 8 (80) 2 (28) 
 PD 7 (14) 3 (9) 1 (10) 3 (43) 
 Unevaluable 3 (6)a 2 (6) – 1 (14) 
PFS (months) 
 Number of PFS events 43 28 
 Median PFS (95% CI) 12.5 (7.3–17.5) 12.9 (7.3–18.6) 13.1 (1.8–49.2) 2.2 (<1–17.9) 
 2 year PFS (95% CI)b 22.2% (11.6–34.9) 23.6% (10.6–39.5) 22.5% (3.4–51.7) 14.3% (<1–46.5) 
OS (months) 
 Number of deaths 23 11 
 Median OS (95% CI) 32.7 (18.7–61.3+) NR 21.4 (8.5–57+) 4.6 (<1–29.9) 
 2 year OS (95% CI) 60.7% (44.9–73.3) 73.5% (53.8–85.8) 45.7% (14.3–72.9) 28.6% (4–61.2) 
 Median follow-up (range) 31.8 (3.8–61.3) 27.9 (3.8–61.3) 46.5 (11–57.1) 38.7⁁ 
Overall [n = 50]Differentiated [n = 33]MTC [n = 10]ATC [n = 7]
Best response to therapy 
 CR — — — — 
 PR 3 (6) 1 (3) 1 (10) 1 (14) 
 SD 37 (74) 27 (82) 8 (80) 2 (28) 
 PD 7 (14) 3 (9) 1 (10) 3 (43) 
 Unevaluable 3 (6)a 2 (6) – 1 (14) 
PFS (months) 
 Number of PFS events 43 28 
 Median PFS (95% CI) 12.5 (7.3–17.5) 12.9 (7.3–18.6) 13.1 (1.8–49.2) 2.2 (<1–17.9) 
 2 year PFS (95% CI)b 22.2% (11.6–34.9) 23.6% (10.6–39.5) 22.5% (3.4–51.7) 14.3% (<1–46.5) 
OS (months) 
 Number of deaths 23 11 
 Median OS (95% CI) 32.7 (18.7–61.3+) NR 21.4 (8.5–57+) 4.6 (<1–29.9) 
 2 year OS (95% CI) 60.7% (44.9–73.3) 73.5% (53.8–85.8) 45.7% (14.3–72.9) 28.6% (4–61.2) 
 Median follow-up (range) 31.8 (3.8–61.3) 27.9 (3.8–61.3) 46.5 (11–57.1) 38.7⁁ 

Abbreviations: ⁁, 1 patient in the cohort remains alive; +, upper limit not reached, last known censored time used.

aWithdrew and no post baseline scan.

bProportion alive and progression-free at 2 years.

For the DTC cohort, there was 1 PR (3%) and 27 patients (82%) had SD for a CBR of 84.8%. Median PFS was 12.9 months (95% CI, 7.3–18.5) with a 2-year PFS of 23.6%. Median OS was not reached and 2-year OS was 73.5% (Fig. 1). Three patients continued on treatment at 45.1, 47.1, and 57.6 months and 6 remain in follow-up. Among patients with HCTC, median PFS was 11.7 months and 2-year PFS was 16.8%. Serial thyroglobulin (Tg) levels on treatment were available in 23 DTC patients and levels declined in 74% (17/23) of cases. Median PFS was not significantly different for patients who had previously received treatment with a TKI versus those who had not [10.6 (95% CI, 3.3–18.6) vs. 15.2 (95% CI, 9.2–20.6) months, respectively]. Among 10 MTC patients, there was 1 PR (10%) and 8 (80%) had SD. Median PFS was 13.1 months with a 2-year PFS of 22.5%. Median OS in this group was 21.4 months. Among 7 ATC patients, one achieved a near-complete response, found to have a nonsense mutation of the tumor suppressor gene TSC2 (14). Another had ongoing disease stability for 26 months without evidence of progression prior to death (from congestive heart failure), while four progressed within 3 months of study entry.

Figure 1.

Response to everolimus in advanced, aggressive DTC patients. PFS (A) and OS (B) among patients (n = 33) with advanced, aggressive DTC treated on protocol with everolimus.

Figure 1.

Response to everolimus in advanced, aggressive DTC patients. PFS (A) and OS (B) among patients (n = 33) with advanced, aggressive DTC treated on protocol with everolimus.

Close modal

Toxicity

Forty-seven patients had at least one treatment-related adverse event (TRAE). TRAEs were predominantly grade 1 or 2 (50%) of which mucositis, acneiform rash, fatigue, and cough were most common. Anemia, thrombocytopenia and transaminitis were the most frequent grade 1 or 2 laboratory abnormalities. Grade 3 TRAEs occurred in 42% (21/50) of patients and the most common were fatigue, stomatitis, and infections. One patient experienced grade 4 hypercholesterolemia (Table 3). Dose modifications were necessary in 32 patients (62%) with the primary reason being toxicity.

Table 3.

Grade 3 or greater TEAEs in all treated patients

Grade 3Grade 4
Hypercholesterolemia 
Hyperglycemia  
Elevated transaminases  
Infection  
Lymphopenia  
Neutropenia  
Anemia  
Thrombocytopenia  
Fatigue  
Weight loss  
Anorexia  
Mucositis or stomatitis  
Pulmonary / upper respiratory  
Pneumonitis  
Nausea  
Vomiting  
Grade 3Grade 4
Hypercholesterolemia 
Hyperglycemia  
Elevated transaminases  
Infection  
Lymphopenia  
Neutropenia  
Anemia  
Thrombocytopenia  
Fatigue  
Weight loss  
Anorexia  
Mucositis or stomatitis  
Pulmonary / upper respiratory  
Pneumonitis  
Nausea  
Vomiting  

NOTE: Frequency of events are classified using the CTCAE v. 3.0 and categorized by the highest TEAE grade reported. Summary includes most common grade 3 or 4 events classified as possibly, probably, or definitely related to protocol treatment (n = 22 patients experienced at worst grade 3 or 4 TEAEs).

Molecular characterization of the cohort

Genomic profiling data was available for 38 patients (76%). There were no differences with respect to baseline characteristics between those patients with and without genomic profiling data. Over 1,400 unique, nonsynonymous somatic variants were identified (range: 1–60 per sample). A total of 235 unique indels were identified that affected the coding sequence. A total of 766 (766/1406, 54.5%) of these variants have been reported in the Catalog of Somatic Mutations in Cancer (COSMIC) database. The mutational profile, best clinical response to everolimus, and outcomes of sequenced patients are summarized in Fig. 2. Mutations in NOTCH2, ATM, and RET were frequent, with 42.1% of sequenced patients having somatic alterations in PI3K/mTOR/Akt.

Figure 2.

Tumor mutational profiling among aggressive, ATC patients treated with everolimus. A, Mutational landscape among aggressive, advanced thyroid tumors arranged by histologic subtype. Each column represents a single tumor sample, and the pink–blue coding reflects the type of genomic alteration identified. The progression-free survival (PFS) and overall survival (OS) for the corresponding patient is color coded according to the orange heatmap shown. PTC, papillary thyroid cancer; HCTC, Hurthle cell thyroid cancer; FTC, follicular thyroid cancer; MTC, medullary thyroid cancer; PDTC, poorly differentiated thyroid cancer; ATC, anaplastic thyroid cancer. B, Total mutational count among aggressive, ATC patients (N = 38) who underwent targeted next-generation sequencing. Each column represents the total number of mutations identified among the tested tumor samples (not mutually exclusive) in order of frequency. Copy number variation count is also provided.

Figure 2.

Tumor mutational profiling among aggressive, ATC patients treated with everolimus. A, Mutational landscape among aggressive, advanced thyroid tumors arranged by histologic subtype. Each column represents a single tumor sample, and the pink–blue coding reflects the type of genomic alteration identified. The progression-free survival (PFS) and overall survival (OS) for the corresponding patient is color coded according to the orange heatmap shown. PTC, papillary thyroid cancer; HCTC, Hurthle cell thyroid cancer; FTC, follicular thyroid cancer; MTC, medullary thyroid cancer; PDTC, poorly differentiated thyroid cancer; ATC, anaplastic thyroid cancer. B, Total mutational count among aggressive, ATC patients (N = 38) who underwent targeted next-generation sequencing. Each column represents the total number of mutations identified among the tested tumor samples (not mutually exclusive) in order of frequency. Copy number variation count is also provided.

Close modal

Six of 23 sequenced DTC patients (26.1%) had a BRAFV600E mutation (one with HCTC), while PI3K/mTOR/Akt alterations were frequent in those with PTC histology (6/10, 60%) most commonly with TSC2 missense mutations. Figure 3 shows PFS estimates by key mutational status for all histologies in the genomic cohort. DTC patients with only a BRAF mutation (n = 3) demonstrated a longer median PFS compared with all sequenced DTC patients (43.4 vs. 9.2 months). Three sequenced DTC patients (27.3%) had coexisting alterations in BRAF and either TP53 or PI3K/mTOR/Akt, and those with coexisting BRAF+TP53 mutations had a shorter median PFS compared with non-TP53, BRAF-mutated DTC patients (6.8 vs. 43.4 months). Few sequenced FTC patients harbored a PIK3CA alteration. The mutational profile of 10 patients with HCTC was bland with regards to BRAF or PI3K alterations compared with other histologies, although two patients had a TSC2 mutation whose disease progressed at 11.7 and 20.5 months. Median PFS for all sequenced HCTC patients was 12.6 months. There were three TP53-mutated HCTC patients: disease progressed at 2.0 and 11.7 months after enrolling in two patients, and one who died without radiographic progression at 13.5 months.

Figure 3.

Clinical response to everolimus in aggressive, ATC patients by mutational status and prior TKI use. A–C, Progression-free survival (PFS) among all patients treated with everolimus for aggressive, ATC grouped by the presence of key mutations. D, PFS among patients treated with everolimus by prior tyrosine kinase inhibitor (TKI) exposure.

Figure 3.

Clinical response to everolimus in aggressive, ATC patients by mutational status and prior TKI use. A–C, Progression-free survival (PFS) among all patients treated with everolimus for aggressive, ATC grouped by the presence of key mutations. D, PFS among patients treated with everolimus by prior tyrosine kinase inhibitor (TKI) exposure.

Close modal

All but one patient in the sequenced MTC group was RET-mutated. Similar to the HCTC subgroup, coexisting PI3K/mTOR/Akt alterations were uncommon and there were no BRAF mutations. Median PFS for sequenced MTC patients was 14.5 months and was similar in those with only a RET mutation versus those with RET and either PI3K/mTOR/Akt or TP53 alterations (21.4 vs. 10 months). All sequenced MTC patients had evidence of SD with the exception of 1 patient whose best response was a PR who remained progression-free at 55.2 months, and that patient had a RET and NF1 truncating mutation.

The median PFS of those PDTC/ATC patients who were sequenced was 2.8 months (median OS, 8.9 months), the lowest of any subgroup. Two sequenced patients (33.3%) with ATC were BRAF-mutated, while 1 had an NRAS mutation, suggesting dedifferentiation from PTC or FTC histology, respectively. Among PDTC/ATC patients with PI3K/mTOR/Akt mutations, median PFS was 15.2 months. One patient had a TSC truncating mutation and coexisting TP53 mutation but had a best response of PR and remained progression-free until 17.9 months. Another patient had an NF1+TP53 mutation and had SD as best response and was progression-free at 26 months until death at 29.9 months.

The widespread availability of TKIs and their ability to stabilize disease have improved outcomes in thyroid cancer, but therapy eventually fails and additional treatments are needed. Moreover, there are currently no targeted therapies approved for ATC. Our study represents the largest cohort to date showing the clinical benefit of a nonkinase inhibitor in advanced thyroid cancer. While radiographic response rates were low, 74% of patients demonstrated SD with a median PFS of just over a year among rapidly progressing DTC patients, and exceeding 1 year in the MTC subgroup. Our findings are consistent with similar studies that have reported SD rates of 65%–76% in those treated with everolimus (15, 16). These findings are particularly notable given that we selected patients with clinically aggressive disease. It is also important to acknowledge that almost half of patients in our cohort received prior TKI therapy yielding a 70.6% SD rate; therefore, mTOR inhibition may prove a viable second-line option post-TKI progression. This was evaluated in a single-arm phase II study of DTC treated with everolimus plus sorafenib, following progression on sorafenib alone, where median PFS was 13.7 months (17), comparable with our cohort treated with mTOR inhibition alone.

In comparing our cohort with 399 thyroid cancer samples generated by The Cancer Genome Atlas (TCGA) Research Network (18), we observed several important differences: (i) our cohort represents a genetically enriched population with a higher mutational burden in that we selected for aggressive tumors and (ii) a significant proportion of our patients had mutations in the PI3K/mTOR/Akt (16/38, 42.1%) compared with TCGA (<5%). Our favorable outcomes may result from targeting the PI3K/mTOR/Akt pathway. Murine models with constitutive mTOR activation have demonstrated decreased in vitro tumor growth with the use of everolimus (19). We expected patients with alterations in the PI3K/mTOR/Akt to derive the greatest benefit from everolimus. That was, however, not the case with the exception of the PDTC/ATC subgroup where all patients with a significant response to treatment demonstrated mutations directly linked to mTOR activation.

Patients with DTC histology who were only BRAF-mutated experienced a PFS benefit greater than that of all other subgroups (43.4 months), fitting with their histology and low mutational burden. While hypothesis-generating, this observation could also have been due to chance alone or other confounders. Of interest, the presence of a coexisting TP53 mutation decreased the median PFS to just 6.8 months among PTC patients, in line with prior studies that indicate that TP53 mutations are an independent marker of poor outcomes in cancer (20). The three PI3K/mTOR/Akt–mutated patients among the FTC and HCTC subgroups had a median PFS of 18.6 months. The sequenced HCTC subgroup demonstrated a median PFS better than expected given that only two patients were TSC2-mutated. Although speculative, this may be explained by alternative mechanisms, such as a reduction in angiogenesis or immunomodulatory effects.

RET oncogene activation was observed in nearly all sequenced MTC patients (87.5%). Their median PFS approached 1 year with 2-year PFS comparable with DTC patients. RET-mutant cell lines demonstrate sustained growth suppression following mTOR inhibition (21), which could account for these findings despite the low number of mTOR alterations we identified. In our PDTC/ATC cohort, best response was SD for two patients and another achieved a near-CR and remained progression-free until approximately 18 months (23). This patient had an exceptional response to mTOR inhibition from a TSC2 nonsense mutation and subsequently developed acquired resistance with a new mTORF2108L mutation resulting in allosteric inhibition. The median PFS for the four ATC/PDTC patients who had PI3K/mTOR/Akt alterations (TSC2, FLCN, or NF1) was impressive (15.2 months). The coexistence of a TP53 mutation observed in two of these patients did not decrease survival. Although speculative, PI3K pathway alterations may represent a dominant oncogene in ATC.

Our genomic analysis has some important limitations, mainly related to sample size of each histologic subgroup. Our sequencing platform did not assess for translocations or epigenetic events. In addition, histologic subgroups analyzed were not always mutually exclusive of one another, given the overlap in genomic mutations and the possibility of mixed histologies.

The progression-free benefit of everolimus in this cohort is in keeping with previously studied TKIs and supports mTOR inhibition as a clinically meaningful target in thyroid cancer. Everolimus demonstrated a manageable toxicity profile and may represent a suitable alternative for those patients who cannot tolerate VEGFR inhibitor–specific side effects. This clinically aggressive cohort showed genomic heterogeneity, but enriched for PI3K/mTOR/Akt alterations. It may be that prior VEGFR TKI exposure promotes alternative pathway coactivation to enable tumor progression. PDTC/ATC appeared to benefit most from everolimus when a PI3K/mTOR/Akt alteration was present. While genomic profiling of tumors does not currently guide therapeutic selection in thyroid cancer, these data have important implications when selecting an mTOR inhibitor versus VEGFR TKI. This remains an active area of investigation as we continue to explore additional targeted therapies and the appropriate sequence of treatments in an era of personalized medicine.

N.L. Busaidy reports receiving speakers bureau honoraria from and is a consultant/advisory board member for Eisai, reports receiving commercial research grants from Novartis, and reports receiving commercial research support from Bayer. L.J. Wirth is a consultant/advisory board member for Eisai, Loxo and Novartis. R.I. Haddad is a consultant/advisory board member for Astra-Zeneca, Bristol-Myers Squibb, Eisai, Merck and Pfizer. G. Rabinowits is a consultant/advisory board member for EMD Serono and Pfizer. No potential conflicts of interest were disclosed by the other authors.

Conception and design: G.J. Hanna, N.L. Busaidy, L.J. Wirth, R.I. Haddad, K. Misiukiewicz, T. Thomas, J.H. Lorch

Development of methodology: G.J. Hanna, N.L. Busaidy, L.J. Wirth, K. Misiukiewicz, T. Thomas, J.H. Lorch

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G.J. Hanna, N.L. Busaidy, N.G. Chau, L.J. Wirth, J.A. Barletta, A. Calles, R.I. Haddad, S. Kraft, G. Rabinowits, E.K. Alexander, F.D. Moore, K. Misiukiewicz, T. Thomas, E. Marqusee, S.L. Lee, J.H. Lorch

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G.J. Hanna, N.L. Busaidy, N.G. Chau, L.J. Wirth, R.I. Haddad, A. O'Neill, E.K. Alexander, K. Misiukiewicz, T. Thomas, P.A. Janne, J.H. Lorch

Writing, review, and/or revision of the manuscript: G.J. Hanna, N.L. Busaidy, N.G. Chau, L.J. Wirth, J.A. Barletta, A. Calles, R.I. Haddad, M.E. Cabanillas, G. Rabinowits, A. O'Neill, S.A. Limaye, E.K. Alexander, F.D. Moore, T. Thomas, M.A. Nehs, E. Marqusee, S.L. Lee, P.A. Janne, J.H. Lorch

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G.J. Hanna, A. Calles, R.I. Haddad, A. O'Neill, J.H. Lorch

Study supervision: G.J. Hanna, R.I. Haddad, M.E. Cabanillas, J.H. Lorch

We would like to thank Tyler Haddad, Vicky Vergara, Julian Huang, Sarah Reilly, and Marge Suda for their assistance with data gathering. We thank the Center for Cancer Genome Discovery (CCGD) at the Dana-Farber Cancer Institute for their assistance in analyzing the sequencing data. We are grateful to our patients for their participation in this research. This investigator-initiated study was financially supported by Novartis and the National Institutes of Health/National Cancer Institute Cancer Center Support Grant (grant number P30-CA016672; to M.E. Cabanillas and N.L. Busaidy).

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.

1.
Brenner
H
. 
Long-term survival rates of cancer patients achieved by the end of the 20th century: a period analysis
.
Lancet
2002
;
360
:
1131
5
.
2.
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
.
3.
Wells
SA
 Jr
,
Asa
SL
,
Dralle
H
,
Elisei
R
,
Evans
DB
,
Gagel
RF
, et al
American Thyroid Association Guidelines Task Force on Medullary Thyroid Carcinoma. Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma
.
Thyroid
2015
;
25
:
567
610
.
4.
Nagaiah
G
,
Hossain
A
,
Mooney
CJ
,
Parmentier
J
,
Remick
SC
. 
Anaplastic thyroid cancer: a review of epidemiology, pathogenesis, and treatment
.
J Oncol
2011
;
2011
:
542358
.
5.
Brose
MS
,
Nutting
CM
,
Jarzab
B
,
Elisei
R
,
Siena
S
,
Bastholt
L
, et al
DECISION investigators. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial
.
Lancet
2014
;
384
:
319
28
.
6.
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
.
7.
Elisei
R
,
Schlumberger
MJ
,
Müller
SP
,
Schöffski
P
,
Brose
MS
,
Shah
MH
, et al
Cabozantinib in progressive medullary thyroid cancer
.
J Clin Oncol
2013
;
31
:
3639
46
.
8.
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
.
9.
Nikiforov
YE
,
Nikiforova
MN
. 
Molecular genetics and diagnosis of thyroid cancer
.
Nat Rev Endocrinol
2011
;
7
:
569
80
.
10.
Wullschleger
S
,
Loewith
R
,
Hall
MN
. 
TOR signaling in growth and metabolism
.
Cell
2006
;
124
:
471
84
.
11.
Agrawal
N
,
Akbani
R
,
Aksoy
BA
,
Ally
A
,
Arachchi
H
,
Asa
SL
, et al
Integrated genomic characterization of papillary thyroid carcinoma
.
Cell
2014
;
159
:
676
90
.
12.
Santoro
M
,
Dathan
NA
,
Berlingieri
MT
,
Bongarzone
I
,
Paulin
C
,
Grieco
M
, et al
Molecular characterization of RET/PTC3; a novel rearranged version of the RETproto-oncogene in a human thyroid papillary carcinoma
.
Oncogene
1994
;
9
:
509
16
.
13.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
,
Sivachenko
A
,
Jaffe
D
,
Sougnez
C
, et al
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
2013
;
31
:
213
9
.
14.
Wagle
N
,
Grabiner
BC
,
Van Allen
EM
,
Amin-Mansour
A
,
Taylor-Weiner
A
,
Rosenberg
M
, et al
Response and acquired resistance to everolimus in anaplastic thyroid cancer
.
N Engl J Med
2014
;
371
:
1426
33
.
15.
Lim
SM
,
Chang
H
,
Yoon
MJ
,
Hong
YK
,
Kim
H
,
Chung
WY
, et al
A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes
.
Ann Oncol
2013
;
24
:
3089
94
.
16.
Schneider
TC
,
de Wit
D
,
Links
TP
,
van Erp
NP
,
van der Hoeven
JJ
,
Gelderblom
H
, et al
Everolimus in patients with advanced follicular-derived thyroid cancer: results of a phase II clinical trial
.
J Clin Endocrinol Metab
2017
;
102
:
698
707
.
17.
Sherman
EJ
,
Ho
AL
,
Fury
MG
,
Baxi
SS
,
Haque
S
,
Lipson
BL
, et al
Phase II study of everolimus and sorafenib for the treatment of metastatic thyroid cancer
.
J Clin Oncol
31
:
15s
, 
2013
(
suppl; abstr 6024
).
18.
The Cancer Genomic Atlas Network
. 
Comprehensive genomic characterization of head and neck squamous cell carcinomas
.
Nature
2015
;
517
:
576
82
.
19.
Guigon
CJ
,
Fozzatti
L
,
Lu
C
,
Willingham
MC
,
Cheng
SY
. 
Inhibition of mTORC1 signaling reduces tumor growth but does not prevent cancer progression in a mouse model of thyroid cancer
.
Carcinogenesis
2010
;
31
:
1284
91
.
20.
Petitjean
A
,
Achatz
MI
,
Borresen-Dale
AL
,
Hainaut
P
,
Olivier
M
. 
TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes
.
Oncogene
2007
;
26
:
2157
65
.
21.
Gild
ML
,
Landa
I
,
Ryder
M
,
Ghossein
RA
,
Knauf
JA
,
Fagin
JA
. 
Targeting mTOR in RET mutant medullary and differentiated thyroid cancer cells
.
Endocr Relat Cancer
2013
;
20
:
659
67
.