Skip to main content
Endocrine Oncology logoLink to Endocrine Oncology
. 2023 Jan 16;3(1):e220062. doi: 10.1530/EO-22-0062

Kinase inhibitors in thyroid cancers

Vineeth Sukrithan 1,*, Prachi Jain 1,*, Manisha H Shah 1, Bhavana Konda 1,
PMCID: PMC10305552  PMID: 37434642

Abstract

Objective

The treatment landscape for thyroid cancers has changed rapidly with the availability of kinase inhibitors against VEGFR, BRAF, MEK, NTRK, and RET. We provide an up-to-date review of the role of kinase inhibitors in thyroid cancer and discuss upcoming trials.

Design & Methods

A comprehensive review of the available literature describing kinase inhibitors in thyroid cancer was performed.

Results and Conclusions

Kinase inhibitors have become the standard of care for patients with metastatic radioactive iodine-refractory thyroid cancer. Short-term treatment can re-sensitize differentiated thyroid cancer to radioactive iodine, thereby potentially improving outcomes and sparing toxicities associated with the long-term use of kinase inhibitors. The approval of cabozantinib as salvage therapy for progressive radioactive iodine-refractory differentiated thyroid cancer following failure with sorafenib or lenvatinib adds to the available armamentarium of active agents. Vandetanib and cabozantinib have become mainstay treatments for metastatic medullary thyroid cancer regardless of RET mutation status. Selpercatinib and pralsetinib, potent and selective receptor kinase inhibitors with activity against RET, have revolutionized the treatment paradigm for medullary thyroid cancers and other cancers with driver mutations in RET. Dabrafenib plus trametinib for BRAF mutated anaplastic thyroid cancer provides an effective treatment option for this aggressive cancer with a dismal prognosis. In order to design the next generation of agents for thyroid cancer, future efforts will need to focus on developing a better understanding of the mechanisms of resistance to kinase inhibition including bypass signaling and escape mutations.

Keywords: sorafenib, lenvatinib, selpercatinib, dabrafenib, cabozantinib, thyroid, medullary thyroid cancer, anaplastic thyroid cancer, differentiated thyroid cancer

Introduction

Differentiated thyroid cancers (DTCs) represent the most common type of thyroid cancers (85%), followed by medullary thyroid cancers (MTCs) and anaplastic thyroid cancers (ATCs), representing 2 to 8% and 1%, respectively (Porter & Wong 2020). Localized DTCs, comprising papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), and Hurthle cell thyroid cancer, have an excellent 10-year prognosis of about 95% (Ganly et al. 2015). Patients with unresectable, advanced, or refractory DTC and MTC have median 10-year survival rates of around 40–42% (Durante et al. 2006, Tuttle et al. 2010, Giunti et al. 2013). Poorly DTCs (PDTCs) represent a subset with features that are in between DTC and ATC. The incidence is around 6% of all cases; in patients with distant metastases, 10-year survival is around 31% (Ibrahimpasic et al. 2014, Fagin & Wells 2016). ATCs are the most aggressive type of thyroid cancers and are historically associated with a median survival of only 3–6 months (Lim et al. 2012). DTCs are mostly diagnosed in the isolated or loco-regionally advanced stage and rarely with metastasis (10%). The standard of care for either localized or metastatic DTC consists of thyroidectomy with or without lymph node dissection, long-term thyroid-stimulating hormone-suppressive levothyroxine, and adjuvant radioactive iodine (RAI) for high-risk or residual disease as well for distant metastases. Between 25 and 50% of patients with advanced or metastatic DTC develop resistance to RAI (RAI-R). These cases are associated with a median 10-year survival of 10–20% due to their aggressiveness and limited therapeutic options (Durante et al. 2006, Anderson et al. 2013). Kinase inhibitors (KIs) have become the standard of care for progressive, symptomatic metastatic DTC or RAI-R thyroid cancers and have been shown to improve progression-free survival (PFS), but at the cost of some treatment-related toxicity.

While localized MTCs are surgically curable, with a 10-year survival rate of 96%, a considerable number of patients (13%) have distant metastasis at the time of diagnosis (Roman et al. 2006, Wells et al. 2015). This is associated with a poor median survival of around 3 years, for which multitargeted receptor KIs are the mainstay of therapy. However, a statistically significant overall survival (OS) advantage has yet to be demonstrated with these therapies (Boostrom et al. 2009, Wells et al. 2012, Elisei et al. 2013).

ATC is an extremely aggressive cancer, associated with a median OS of 4 months, a 6-month OS of 35%, and disease-specific mortality of 98–99% (Maniakas et al. 2020). ATCs account for up to 50% of thyroid cancer-related deaths (Are & Shaha 2006, Smallridge et al. 2009, Bible et al. 2021). ATCs are often diagnosed at locoregionally advanced stages, invading adjacent structures or with distant metastasis, making them surgically inoperable. However, the advent of novel targeted therapies that act on mitogen-activated protein kinase (MAPK) signaling has led to improved survival in patients with ATC (Maniakas et al. 2020). The resources available for the treatment of advanced or metastatic thyroid cancers continue to expand with the availability of novel therapeutics (Porter & Wong 2020). The role of immune checkpoint blockade is emerging with robust evidence of efficacy in ATC. Here, we review the role of KIs as therapeutic options in advanced or metastatic thyroid cancers, either as single agents or in combination with other therapies.

Molecular targets in thyroid cancer

Differentiated thyroid cancer

Alterations in BRAF, RET, RAS, PIK3CA, and PTEN activate the MAPK and phosphoinositide 3-kinase (PI3K) pathways leading to the pathogenesis of many DTCs. PTC is the most common DTC (80%), wherein the common driver mutations are the BRAF V600E mutation (50%), followed by RAS mutation (15%) and RET fusions. FTC is characterized by the presence of either RAS (40–50%) or PAX8/PPAR fusion oncogenes (Nikiforova et al. 2003, Elisei et al. 2008, Howell et al. 2013).

Poorly differentiated thyroid cancer

Compared to DTC, there is an increased prevalence of TERT promotor mutations (33–40%), EIF1AX mutations (10%), and SWI/SNF (6%) mutations in cases of PDTC, representing pathways of clonal evolution from slow-growing DTCs to a more aggressive phenotype closer in behavior to ATCs.

Anaplastic thyroid cancer

The most common mutations found in ATC are TP53 (up to 70%), BRAF (20–45%), and RAS (20–40%) (Bonhomme et al. 2017, Bowles et al. 2018). Mutations in the SWI/SNF components are seen in 18–36% of cases. TERT promoter mutations are seen in 43–73% of cases (Kunstman et al. 2015, Landa et al. 2016, Pozdeyev et al. 2018).

Medullary thyroid cancer

MTC can be either familial (25%) or sporadic (75%). Virtually, all familial cases (>98%) have germline RET mutations (Elisei et al. 2019). A large study of the mutational landscape of 208 cases of sporadic MTCs showed a predilection for RET mutations in 56%, followed by RAS mutations in 24.3% (Ciampi et al. 2019).

Kinase inhibitors for re-differentiation of radioactive iodine refractory differentiated thyroid cancer

Re-sensitization to RAI therapy through BRAF and/or MEK inhibition

RAI is the cornerstone of treatment for metastatic DTCs. RAI refractory tumors are defined as tumors that show no uptake of RAI in the post-RAI-therapy scan. There are other ways to define RAI-R tumors, including tumors that show RAI uptake on initial scans, but progress within a relatively short time interval (12–14 months). Finally, it remains controversial whether patients subjected to more than 600 mCi of RAI should be considered as having RAI-R (Haugen et al. 2016). RAI-R tumors, when treated with conventional methods, have a poor outcome with 10-year survival as low as 10% (Gild et al. 2018).

KIs have been approved for progressive or advanced thyroid cancers, but the eventual emergence of resistance and treatment-related toxicities are some important limiting factors. Hence, re-sensitizing RAI-R thyroid tumors to RAI is an area of active clinical and research interest (Fullmer et al. 2021).

Activating mutations in driver pathways, such as BRAF and NRAS mutations in the MAPK pathway, lead to hyperactive signaling, which decreases the expression of sodium/iodide symporter and reduces iodine uptake (Liu et al. 2007). Pre-clinical evidence of restored RAI uptake with BRAF inhibition in vivo led to a clinical study of MEK inhibition with selumetinib for the purpose of re-sensitization. Four weeks of treatment resulted in a clinically meaningful increase in RAI uptake in 40% patients (8 of 20) with metastatic RAI-R DTC including 100% of NRAS-mutated subjects (5 of 5) and 44% of BRAF-mutated patients (44%, 4 of 9). Of the eight patients treated with RAI, 63% (5 of 8) had confirmed partial responses (PRs) (Chakravarty et al. 2011, Ho et al. 2013). A report by Rothenberg et al. demonstrated the clinical efficacy of the selective BRAF inhibitor, dabrafenib, in inducing RAI uptake in BRAF V600E-mutated unresectable or metastatic RAI-R PTC. In this cohort, 60% (6 of 10) had increased RAI uptake post-treatment with dabrafenib. Two of these six patients (33%) had a PR (Rothenberg et al. 2015). A pilot trial of vemurafenib, a BRAF inhibitor, was conducted in 12 patients with BRAF-mutated RAI-R DTC. Four of the 10 evaluable patients had uptake meeting thresholds for treatment; PRs were seen in 50% (2 of 4 patients), with the other 2 patients having the stable disease (SD) at the 6-month mark (Dunn et al. 2019).

A retrospective review at MD Anderson of 13 patients on long-term BRAF or a MEK inhibitor with RAI uptake reported results from 9 patients (3 RAS, 5 BRAF,1 wild type) who subsequently underwent RAI treatment (Jaber et al. 2018). Responses were seen in 40% (2 of 5) BRAF-positive patients and 33% (1/3) RAS-mutant patients with the rest having stable disease. Another retrospective study reported six patients (three with NRAS mutation and three with BRAF V600E mutation) who were treated with either a MEK inhibitor or the combination of a BRAF and MEK inhibitor for RAS- and BRAF-mutated patients, respectively (Iravani et al. 2019). One NRAS- and all BRAF V600E-mutated cases demonstrated restoration of RAI uptake and proceeded to RAI therapy. The NRAS-mutated case (1 of 1) and 66% (2 of 3) of patients with a BRAF V600E mutation demonstrated PR beyond a 3-month follow-up.

These studies have led to the use of KIs to re-sensitize metastatic unresectable RAI-R thyroid tumors to further doses of RAI, thereby salvaging this valuable treatment option.

MERAIODE is a prospective multicentric phase 2 trial of the combination of dabrafenib and trametinib for re-differentiation in patients with BRAF V600E mutations and trametinib alone in RAS-mutated RAI-R metastatic DTC with RECIST progression within 18 months (Dunn et al. 2019). Among 21 evaluable patients with a BRAF mutation, at 6 months, the rate of PR was 38%, 52% had SD, and 10% had progressive disease (PD). In a separate cohort of 10 evaluable RAS-mutated patients, the rate of PR was 20% with SD in 70% and PD in 10% (Leboulleux et al. 2021).

ASTRA was a phase 3 trial of adjuvant RAI vs RAI with a MEK inhibitor (selumetinib) in patients with high-risk DTC after resection (Ho et al. 2022). Patients with high-risk disease were randomized in a 2:1 ratio to either selumetinib or placebo for 5 weeks. A single dose of adjuvant RAI therapy was administered on treatment days 29 to 31. The trial failed to show a statistically significant difference in the complete response rate at 18 months (OR = 1.07; P = 0.82). Treatment-related grade ≥3 adverse events (AEs) were reported in 25 of 154 patients (16%) with selumetinib while there were none with placebo. An acneiform rash was seen in 7% of patients.

Unlike agents that target the MAPK pathway, various other therapies including retinoids and multitargeted KIs such as sorafenib and lenvatinib have failed to show clinically meaningful response in re-sensitizing RAI-R thyroid cancers to iodine (Hoftijzer et al. 2009).

Re-sensitization to RAI therapy through RET and NTRK inhibition

There is emerging evidence that DTCs with fusions in RET and NTRK are amenable to re-differentiation through therapies that target the driver alterations. A case series reported re-differentiation in two out of three consecutive patients with NTRK-rearranged RAI-resistant thyroid cancers treated with the NTRK inhibitor larotrectinib (Groussin et al. 2020, 2022). A pediatric case series of two patients with NTRK and RET fusion treated with larotrectinib and selpercatinib reported decreased tumor sizes and restored RAI uptake (Lee et al. 2021). Another case series reported two pediatric cases and documented a case of re-differentiation with selpercatinib targeting a RET fusion gene in metastatic RAI-resistant PTC (Groussin et al. 2021). Interestingly, in one of the pediatric case reports of metastatic papillary thyroid carcinoma, larotrectinib was shown to induce redifferentiation when given in the neoadjuvant setting prior to RAI, thereby achieving an improved outcome (Waguespack et al. 2022b ). Prospective studies using such targeted agents are needed to confirm these findings and further expand the options to restore radioiodine uptake in RAI-resistant thyroid cancers.

Kinase inhibitors for advanced radioactive iodine refractory differentiated thyroid cancer

VEGFR-targeted kinase inhibitors

While sorafenib is approved as a frontline treatment, lenvatinib has efficacy in the first- or second-line setting. Cabozantinib (brand name: Cabometyx) has been approved in the second-line setting for patients with RAI-R DTC who have progressed following up to two prior vascular endothelial growth factor receptor (VEGFR)-targeted therapies.

Initial phase 2 trials of sorafenib in DTC showed median PFS ranging from 58 weeks to 84 weeks with a manageable toxicity profile, paving the way for larger phase 3 trials (Gupta-Abramson et al. 2008, Ahmed et al. 2008, Brose et al. 2009, Hoftijzer et al. 2009, Kloos et al. 2009). In 2013, the US Food and Drug Administration (FDA) approved sorafenib in the first line for RAI-R advanced or metastatic DTC based on data from the phase 3 decision trial, which showed significantly improved median PFS of 10.8 months vs 5.8 months in the placebo group (HR, 0·59; P < 0.0001) (Brose et al. 2014). This benefit was observed regardless of BRAF or RAS mutation. While improved PFS was supportive of the use of sorafenib as a treatment option in RAI-R DTC, responses occurred in only 12% of subjects. The most common AEs in the sorafenib arm were hand–foot skin reaction, diarrhea, alopecia, rash/desquamation, fatigue, weight loss, and hypertension.

Lenvatinib is a multitargeted KI, which inhibits VEGFR 1–3 (VEGFR1–3), fibroblast growth factor receptors 1–4 (FGFR1–4), RET, c-KIT, and platelet-derived growth factor receptor α (PDGFRα). In 2015, the FDA approved lenvatinib for RAI-refractory DTC with disease progression, including those with prior exposure to one tyrosine kinase inhibitor (TKI), based on the phase 3 trial (SELECT). This study showed a significantly improved median PFS of 18.3 vs 3.6 months (HR: 0.21; P < 0.001) in addition to an increased response rate of 64.8% vs 1.5% (OR 28.87; P < 0.001) (Schlumberger et al. 2015). After adjusting for crossover effects, there was a trend toward improved OS (HR, 0.62; 95% CI, 0.40–1.00; P = 0.05). The response was maintained regardless of the patient’s BRAF or RAS mutation status. Commonly reported AEs were hypertension, diarrhea, fatigue/asthenia, and decreased appetite.

Cabozantinib, a multikinase inhibitor targeting VEGFR2, MET, AXL, and RET, was approved by the FDA in 2021 for RAI-R DTC patients who progressed after up to two prior VEGFR-targeted therapies, based on data from the phase 3 COSMIC-311 study (Brose et al. 2021b ). The trial demonstrated significantly improved median PFS for the cabozantinib group over the placebo group of 11 vs 1.9 months (HR, 0.22; 95% CI, 0.15–0.31) (Brose et al. 2021a,b , Capdevila et al. 2021). Although it did not meet the co-primary endpoint of ORR – 15% for cabozantinib vs 0% for placebo (P = 0.0281) – a favorable OS trend was observed (HR 0.54, 95% CI 0.27–1.11). The benefit in PFS was seen across all subgroups, either with prior sorafenib or lenvatinib treatment alone or after progression on both. Cabozantinib is therefore a promising agent for salvage in patients who have progressed on the prior-approved VEGFR-targeted therapies. Common AEs were diarrhea, palmar-plantar erythrodysesthesia, fatigue, hypertension, and stomatitis.

Sunitinib, a multi-targeted inhibitor of VEGFR, RET, and PDGFR, was studied in a single-center phase 2 study among 23 patients who had received at least one dose of RAI. Six (26%) patients achieved a PR, and 13 (57%) had SD with a median PFS of 8 months (Bikas et al. 2016). Another phase 2 study that enrolled 71 patients showed sunitinib to be active in metastatic DTC and MTC, with ORR of 22 and 38.5%, respectively, and OS of 26.4 and 29.4 months, respectively (Ravaud et al. 2017). The study did not support the proposed dose of 50 mg/day due to the occurrence of pronounced side effects. Overall, there is limited evidence of the clinical activity of sunitinib in advanced thyroid cancer (Cohen et al. 2008, Carr et al. 2009, Gómez-Sáez 2016).

Pazopanib is another multikinase inhibitor targeting VEGFR, PDGFR, and KIT, which showed promising activity and manageable toxicity in a phase 2 trial of a pre-treated group of 60 patients with progressive RAI-R DTC (Bible et al. 2010). The trial demonstrated an ORR of 37%, a median PFS of 11.4 months, and a median OS of 2.6 years. Hypertension, fatigue, and neutropenia were commonly observed grade 3–5 toxicities.

Axitinib, a potent selective inhibitor of VEGFR, was shown to have activity in advanced thyroid cancer, across all histologic subtypes including RAI-R DTC in two phase 2 trials. The two trials reported an ORR of 38 and 35%, PFS of 15 and 16.1 months, and duration of response of 21 and 17.2 months, respectively. The median OS was 35 and 27.2 months, respectively (Cohen et al. 2014, Locati et al. 2014). Commonly reported AEs were fatigue, diarrhea, nausea, anorexia, hypertension, and weight loss.

mTOR-targeted kinase inhibitors

Everolimus, an mTOR inhibitor, was shown to have limited efficacy in the second-line setting post-progression on a prior TKI in a trial of 50 subjects. ORRs of 3, 10, and 14% were reported for DTC, MTC, and ATC, respectively (Hanna et al. 2018). Another multi-institutional phase 2 study showed improved PFS benefit with the addition of everolimus to sorafenib in a cohort with RAI-R Hurthle cell cancer (24.7 vs 10.9 months), but similar OS (Sherman et al. 2021).

ALK-targeted kinase inhibitors

Anaplastic lymphoma kinase (ALK)-targeted therapies, such as crizotinib, ceritinib, brigatinib, loratinib, and alectinib, are well-established FDA-approved treatment options in ALK-translocated non-small cell lung cancer (NSCLC). Genomic profiling of thyroid cancers has led to the discovery of ALK rearrangements in around 2% of PTCs, including a novel ALK fusion product,STRN-ALK, that is sensitive to available ALK inhibitors (Kelly et al. 2014). A case study showed the clinical efficacy of lorlatinib in a refractory DTC harboring EML4-ALK fusion. The patient had a PR that was ongoing at 7 months, with no treatment-related AEs (Aydemirli et al. 2021).

BRAF/MEK-targeted kinase inhibitors

The role of BRAF inhibition with driver mutations at the BRAF V600 locus is a topic of active study. A clinically meaningful response to vemurafenib in BRAF V600E-mutated PTC was shown in a nonrandomized phase 2 trial, with PR rates of 38.5 vs 27.3% in VEGFR-TKI naïve and VEGFR TKI experienced cohorts, respectively. The median PFS was 18.2 and 8.9 months, respectively (Brose et al. 2016).

A randomized phase 2 trial of dabrafenib alone or dabrafenib and trametinib was conducted in 53 cases of BRAF-mutated papillary thyroid carcinoma (Busaidy et al. 2022). Patients with refractory DTC with PD within 13 months were eligible. The primary outcome of the response rate by per-protocol modified RECIST was not statistically significantly different (42 vs 48%, P = 0.67) in the single and combination arms, respectively. The objective response rate by RECIST 1.1 was 35% (9 of 26) with dabrafenib and 30% (8 of 27) with dabrafenib and trametinib. The median PFS was numerically prolonged in the combination arm but did not reach statistical significance (10.7 vs 15.1 months, P = 0.65). In a small subset of 11 patients who were pre-treated with multikinase inhibitors, the median PFS with dabrafenib was 3.7 and 18.8 months with combination therapy (P = 0.01)

RET-targeted kinase inhibitors

Large-scale genomics studies indicate that RET rearrangements occur in around 7% of PTCs (Cancer Genome Atlas Research Network 2014). In a trial of the novel selective RET inhibitor selpercatinib that reported data from 19 patients with RET fusion-positive, previously treated thyroid cancer, objective responses were seen in 79% (Wirth et al. 2020). The activity was observed across the board in thyroid cancers including anaplastic carcinomas, and across different fusion partners. Pralsetinib, another potent, selective RET inhibitor, was also shown to have clinically meaningful activity in RET fusion-positive thyroid cancer with an ORR of 89% (8 of 9 patients) (Subbiah et al. 2021a, Mansfield et al. 2022).

NTRK-targeted kinase inhibitors

Chromosomal rearrangements of NTRK genes are oncogenic driver mutations seen in around 3% of patients with thyroid cancer (Solomon et al. 2020b). NTRK fusions occur across a range of pediatric cancers including infantile fibrosarcoma and secretory breast cancer in which such fusions are nearly pathognomonic (Blauel & Laetsch 2022). Pooled data of thyroid cancers treated with the NTRK inhibitor larotrectinib from three trials (NCT02576431, NCT02122913 and NCT02637687) showed that the objective response rates were 86% for DTC and 29% among those with ATC (Waguespack et al. 2022a). In an integrated analysis of three phase 1/2 trials of patients with driver NTRK fusions treated with entrectinib, among 13 patients with thyroid cancer, responses were seen in 54% (7 of 13) and a median PFS of 19.9 months was reported (Demetri et al. 2022).

Select clinical trials of KIs in DTC with associated clinical results are summarized in Table 1.

Table 1.

Select clinical trials of kinase inhibitors (KIs) in differentiated thyroid cancer.

Agent Molecular targets Trial phase Subject population Outcome
Median PFS (months) and/or DoR ORR (%) Reference
RAI + BRAF/MEK inhibitors (use of KIs as redifferentiating agents)
Selumetinib + RAI MEK Prospective RAIR DTC (follicular thyroid cancer) NA 62% (5/8) Ho et al. 2013
Dabrafenib + RAI BRAF Prospective BRAF V600E RAIR DTC NA 33% (2/6) Rothenberg et al. 2015
Selumetinib + RAI vs RAI MEK Phase 3 Adjuvant treatment in high-risk DTC NA 40% (62/155) Ho et al. 2022
VEGFR KIs in DTC
Sorafenib VEGFR 1–3, PDGFR, RET, cKIT, BRAF Phase 3 Advanced RAIR DTC 10.8 vs 5.8 Placebo
12.2% (24/196) Brose et al. 2014
Lenvatinib VEGFR 1–3, FGFR 1–4, PDGFR, RET, c-KIT Phase 3 Advanced RAIR DTC 18.3 vs 3.6 Placebo
64.8% (169/261) Schlumberger et al. 2015
Cabozantinib VEGFR, MET, RET, AX Phase 3 Advanced RAIR DTC 11 vs 1.9 Placebo 15% (10/67) Brose et al. 2021a ;
Brose et al. 2021b
BRAF/MAPK inhibitors in DTC
Dabrafenib + 
trametinib vs dabrafenib
BRAF/MEK Phase 2 RAIR PTC D + T: 15.1 (11.7–NR)
D: 11.4 (3.8–NR)
D + T: 35% (9/26)
D: 45% (10/22)
Busaidy et al. 2022
Vemurafenib BRAF V600E Phase 2 Advanced (metastatic/unresectable) RAIR PTC (KI Naïve) 18·2 38.5% (10/26) Brose et al. 2016
Advanced (metastatic/unresectable) RAIR PTC (prior exposure to KI) 8.9 27.3% (6/22)
NTRK inhibitors in NTRK fusion positive DTC/ATC
Larotrectinib NTRK1, NTRK2, NTRK3 fusion inhibitor Phase 1/2 Advanced DTC DoR of 24 months = 84% PFS at 24 months = 84% 86% (18/21) Waguespack et al. 2022a
Anaplastic thyroid cancer DoR of 12 months = 50%
Median PFS = 2.2 months
29% (2/7)
Entrectenib NTRK1,NTRK2 ,NTRK3 fusion inhibitor Phase 1/2 Unspecified thyroid cancer mPFS = 19.9 months
mDoR = 13.2 months
54% (7/13) Demetri et al. 2022
RET inhibitors in RET-fusion positive DTC/ATC
Selpercatinib RET inhibitor Phase 1/2 Multiple histologic types (papillary thyroid cancer, Hurthle cell, poorly differentiated, ATC) PFS at 12 months = 64% 79% (15/19) Wirth et al. 2020
Pralsetinib RET inhibitor Phase 1/2 RET fusion positive thyroid cancer previously treated mDoR = 17.5 months
mPFS = 19.4 months
86% (18/21) Mansfield et al. 2022

ATC, anaplastic thyroid carcinoma; DoR, duration of response; DTC, differentiated thyroid carcinoma; MTC, medullary thyroid carcinoma; NR, not reached; ORR, overall response rate; PFS, progression-free survival; RAI-R, radioactive iodine-refractory.

Kinase inhibitors in anaplastic thyroid cancer

ATCs are one of the most aggressive malignancies with a historical median OS of around 4 months (Lin et al. 2019). Recent advances in understanding the molecular biology of these cancers have begun to translate to better survival. Unique characteristics such as a prevalence of BRAF mutations, high PD-L1 staining, and increased neo-angiogenesis make ATC susceptible to novel molecular targeted therapies such as BRAF inhibition and VEGFR inhibitors in combination with immunotherapy (Bastman et al. 2016, Chintakuntlawar et al. 2017, Zwaenepoel et al. 2017, Bowles et al. 2018, Rosenbaum et al. 2018, Cantara et al. 2019, Zhang et al. 2019). A single-center retrospective study of OS by treatment era in stage 4-C ATC patients (n = 253) from 2000 to 2019 showed an improvement in the rate of survival to 1 year: 35% in the pre-2013 group, 47% in the 2014–2016 cohort, and 59% in the 2017–2019 cohort (hazard ratio: 0.5) (Maniakas et al. 2020). The importance of rapid molecular genotyping for the immediate initiation of targeted therapies against BRAF, NTRK, and other KIs cannot be overstated.

Dabrafenib plus trametinib was approved by the FDA in 2018 for patients with locally advanced or metastatic BRAF V600E-mutated ATC with no satisfactory locoregional treatment options, based on a clinical trial that showed a high overall response rate (69%), prolonged duration of response, and prolonged survival with manageable toxicity (Subbiah et al. 2018). Further updated analysis showed continued clinically meaningful benefits with a median PFS and OS of 6.7 and 14.5 months, respectively (Subbiah et al. 2022). Neoadjuvant treatment of unresectable BRAF-mutated ATCs with BRAF/MEK inhibitors is associated with significant, rapid therapeutic responses, rendering them surgically operable in many cases (Cabanillas et al. 2018, Wang et al. 2019, Maniakas et al. 2020).

Multi-receptor KIs do not have a role in ATC as single agents. Sorafenib and pazopanib failed to show any treatment efficacy in ATC (Garon et al. 2015, Villaruz & Socinski 2016, Villaruz et al. 2019). Lenvatinib as a single agent was studied in a multicenter single-arm phase 2 trial. Three percent (1 of 34) had a PR and 50% (17 of 34) of patients achieved SD (≥5 weeks). Even though more than half of the evaluable patients experienced some degree of tumor shrinkage and three patients experienced a >30% tumor reduction, the response duration was short-lived: the median PFS and OS were 2.6 and 3.2 months, respectively (Wirth et al. 2021). There is promising evidence of added efficacy with the combination of lenvatinib with immune-checkpoint inhibitors in ATC, which is summarized in the section ‘combination of kinase inhibitors with immune checkpoint inhibitors’.

In a phase 1/2 study, a deep and durable PR was reported in one patient with widely metastatic ATC, harboring a CCDC6-RET gene fusion, treated with the RET inhibitor LOXO-292 following progression on conventional treatment (Dias-Santagata et al et al. 2020). Further studies are needed to establish the benefit of RET inhibitors in this cohort of RET-harboring, progressive ATC.

Kinase inhibitors in medullary thyroid cancer

KIs have become the mainstay treatment for advanced, progressive, symptomatic, and metastatic MTCs. Vandetanib (RET, VEGFR2/3, EGFR) and cabozantinib (brand name: Cometriq) are multitargeted KIs, approved in 2011 and 2012, respectively, based on data from randomized, placebo-controlled phase 3 trials for unresectable or metastatic MTC. In the ZETA trial, an ORR of 44% and PFS of 30.5 months were reported with vandetanib when compared to 19.3 months for placebo. The EXAM trial was another phase 3 trial that showed an ORR of 28% and PFS of 11.2 months with cabozantinib when compared to 4 months with placebo (Wells et al. 2012, Elisei et al. 2013). These trials did not pre-specify analysis of clinical outcomes by RET mutation status, and it is not currently known if clinical efficacy depends on the presence of RET mutations. There are numerous differences in the patient demographics of these studies that preclude inter-trial comparisons. There are no trials of direct head-to-head comparisons of vandetanib with cabozantinib and KI selection depends on individual patient characteristics. Common side effects with vandetanib include diarrhea, rash, acne, fatigue, nausea, hypertension, headache, vomiting, and photosensitivity. Vandetanib carries a black box warning of QT interval prolongation, torsade de pointes, and sudden death. Rare but serious AEs were reported, including reversible posterior leukoencephalopathy and Stevens–Johnson syndrome. The most commonly reported adverse drug reactions with cabozanitinib are diarrhea, stomatitis, palmar-plantar erythrodysesthesia syndrome, decreased weight, decreased appetite, nausea, fatigue, oral pain, hair color changes, dysgeusia, hypertension, abdominal pain, and constipation. It comes with a black box warning of hemorrhage, perforations, and fistulas. Other serious AEs include thromboembolic events, poor wound healing, osteonecrosis of the jaw, and reversible posterior leukoencephalopathy syndrome.

A meta-analysis studying KIs in MTC that included 1 phase 4 study, 2 phase 3 trials, 20 phase 1 or 2 studies, and 10 retrospective-observational studies showed that grade 3 AEs led to treatment discontinuation in 39.6 and 66.7% of patients treated with vandetanib and cabozantinib, respectively. A risk–benefit discussion with patients is important for decision-making to ascertain the best timing to start KIs (Efstathiadou et al. 2021). Case reports have shown a possibility of a cessation trial in patients with documented SD, but further prospective clinical trials would be needed before a discussion on the cessation of KIs (Brandenburg et al. 2021).

Selpercatinib is a selective RET KI with activity against diverse RET alterations, including the acquired gatekeeper resistance mutation at residue V804. Similar high response rates were seen in TKI-naive (73%) and pre-treated patients with RET-mutated MTC (69%) (Wirth et al. 2020). Pralsetinib is another RET-specific TKI with a treatment efficacy (ORR of 74% in TKI-naïve patients and 60% in those with prior TKI exposure) and side-effect profile similar to selpercatinib (Subbiah et al. 2021a). These agents are well tolerated with only 2% of patients on the trial of selpercatinib discontinuing medication due to toxicity. The most common grade 3/4 AEs were hypertension (21%), increased alanine aminotransferase level (11%), increased aspartate aminotransferase level (9%), hyponatremia (8%), and diarrhea (6%). There were no treatment-related deaths. The emergence of solvent-front mutations in RET at the G810 locus is associated with resistance to selective RET inhibitors; next-generation agents with putative activity against escape mutations are under active investigation (NCT05241834). Select clinical trials of KIs with associated clinical results in MTC are summarized in Table 2.

Table 2.

Select trials of kinase inhibitors in medullary thyroid cancer.

Agent Molecular targets Trial phase Subject population (# of prior lines) Outcome
Median PFS (months) ORR (%) Reference
Cabozantinib VEGFR, MET, RET, AX Phase 3 MTC 11.2 vs 4 28% (58/208) Elisei et al. 2013
Vandetanib VEGFR, EGFR, c-KIT, RET Phase 3 MTC 30.5 vs 19.5 44% (101/231) Wells et al. 2012
Selpercatinib RET Phase 1/2 MTC: treatment naive NR 73% (64/88)
69% (38/55)
Wirth et al. 2020
MTC: previously treated
Pralsetinib RET Phase 1/2 MTC: treatment naive NR 71% (15/21)
60% (33/55)
Subbiah et al. 2021a
MTC: previously treated

MTC, medullary thyroid carcinoma; NR, not reached; ORR, overall response rate; PFS, progression-free survival.

For patients with RET-mutant MTC, the advent of potent and selective RET inhibitors has led to a paradigm shift in treatment due to the highly improved risk–benefit profile with these novel agents when compared to older non-selective KIs, such as vandetanib and cabozantinib. For patients with vascular or bowel tumor involvement or elevated risk of thrombosis or hemorrhage, selective RET inhibitors open up treatment avenues that were otherwise closed due to unacceptably elevated risks with earlier agents. The efficacy of these agents in patients who have progressed on novel RET-selective agents is currently unknown and is an area of active research.

Combination of kinase inhibitors with immune checkpoint inhibitors

There is emerging evidence that angiogenic signaling in the tumor and microenvironment contributes to immune exclusion and primary resistance to checkpoint blockade. In a thyroid cancer tissue banking study, PD-1 and PD-L1 expression were noted in 20 and 64% of DTC samples, respectively (Bastman et al. 2016). FoxP3, PD-1 CD8, and PD-1 CD4 T cells were all noted to be abundant in DTC. Thus, immune evasion plays a vital role in the pathogenesis of DTC; subsequently, low response rates are encountered in single-agent immune checkpoint inhibitor treatment (ORR: 9%) (Mehnert et al. 2019). Currently, checkpoint inhibition is not approved for thyroid cancers. Pembrolizumab may still be used in advanced thyroid cancers with a high mutational burden or with mutations in DNA mismatch repair genes due to its approval in a tumor-agnostic fashion in this subset of patients.

The synergistic effect of the combination of checkpoint blockade and KIs in metastatic refractory thyroid cancer was reported in a single-institution case series (Porter et al. 2020). A single-arm phase 2 trial of the combination of lenvatinib (20 mg) and pembrolizumab in patients with RAI-R DTC with PD within 14 months was reported (Haugen et al. 2020). Patients with prior exposure to VEGFR-targeted therapy were excluded. Of 29 evaluable patients, 18 (62%) had a PR and 10 (35%) had SD. The PFS at 12 months was 74%.

A phase 2 study exploring the combination of cabozantinib with nivolumab and ipilimumab in patients with RAI-R DTC and disease progression with up to two prior VEGFR-targeted therapies is currently underway (NCT03914300). In ATC, immunotherapy with spartalizumab, an anti-PD-1 agent, is associated with an ORR of 19% (Capdevila et al. 2020). Results from a phase 2 trial of lenvatinib and pembrolizumab in patients with ATC and poorly differentiated thyroid cancer (ATLEP) showed that the combination may have significant synergy when compared to each drug alone. Among the first 26 patients with ATC treated with the regimen, ORR after 3 months of treatment was 38.5% (10 of 26, all PR). Additionally, 57.6% (15 of 26) patients achieved SD. One patient had PD. At the 2-year survival landmark, among 16 evaluable patients, the best-observed response rate was 68.5% (11 of 16), indicating that responses tended to improve with time. The median PFS was 8.3 months and OS was 10 months (Dierks 2021).

Mechanisms of resistance to kinase inhibition in thyroid cancer

Resistance to BRAF/MEK-targeted KIs

The eventual development of resistance to KIs remains a major limitation in the treatment of advanced thyroid cancers. A better understanding of the biologic underpinnings of resistance is needed to develop the next generation of thyroid cancer treatments. There is emerging evidence that resistance to BRAF inhibition can occur by bypass activation of the MAPK pathway through de novoRAS mutations (Owen et al. 2019, Cabanillas et al. 2020). In BRAF V600E-mutated cells, epigenetic up-regulation of HER3 expression through an autocrine NRG1 loop has been described (Montero-Conde et al. 2013). Also, mutations altering cell cycle or DNA damage recognition pathways are alternate mechanisms of resistance (Duquette et al. 2015, Antonello et al. 2017). Morphological de-differentiation through activating RAC1 (P34R) mutations and gene amplification has also been described. Epithelial to mesenchymal transformation through the amplification of the TWIST1 transcription factor and MET gene amplification are other potential mechanisms. Excess RAC1 activity leads to an increase in phosphorylated PAK1, which promotes the abnormal organization of mitotic spindles (Bagheri-Yarmand et al. 2021). Emerging evidence suggests that mutations in SWI/SNF chromatin remodeling complexes may predict resistance to re-differentiation with BRAF/MEK inhibitors; novel strategies that target this will need to be elucidated (Saqcena et al. 2021). Current ongoing clinical trials of KIs in DTC are summarized in Table 3.

Table 3.

Select ongoing clinical trials of KIs for thyroid cancers (ClinicalTrials.gov).

Agent Phase Population NCT
BRAF/MAP kinase pathway inhibitors +/− RAI in DTC
BRAF: dabrafenib and trametinib
RAS: trametinib + RAI
Phase 2 BRAF- or RAS-positive DTC with progression within 18 months NCT03244956
Selumetinib plus RAI vs RAI alone Phase 2 DTC with RAI avidity NCT02393690
Selumetinib and RAI Phase 2 DTC with progression within 12 months SEL-I-METRY
Encorafenib and binimetinib with or without nivolumab Phase 2 Metastatic RAI-R BRAF V600-mutant DTC NCT04061980
Vemurafenib plus copanlisib Phase 1 RAI-R DTC NCT04462471
Spartalizumab in combination with MAPK pathway inhibitors Phase 2 RAI-R DTC NCT04544111
Dabrafenib and lapatinib Phase 1 RAI-R/unresectable DTC NCT01947023
Cabozantinib in combination with atezolizumab Phase 1/2 Locally advanced or metastatic solid tumors (DTC) NCT03170960
Cabozantinib, nivolumab, and ipilimumab Phase 2 Advanced RAI-R DTC with progression on up to 2 VEGFR-targeted therapies NCT03914300
Lenvatinib and pembrolizumab Phase 2 DTC NCT02973997
Lenvatinib and everolimus Phase 2 Metastatic DTC with progression on lenvatinib alone NCT03139747
Anlotinib Phase 2 Locally advanced thyroid cancer NCT04309136
Apatinib Phase 3 RAI-R DTC NCT03048877
VEGFR inhibitors and immunotherapy combinations in ATC
Lenvatinib and pembrolizumab Phase 2 Unresectable stage IVB and IVC anaplastic thyroid cancers NCT04171622
Lenvatinib and pembrolizumab Phase 2 Anaplastic thyroid cancers and poorly differentiated thyroid cancers ATLEP
RET inhibitors
TPX-0046 Phase 1/2 Advanced or metastatic solid tumors harboring RET mutations or alterations NCT04161391
TAS0953 Phase 1/2 RET-altered solid tumors NCT04683250
Selpercatinib Phase 2 Neoadjuvant therapy in RET-altered thyroid cancer NCT04759911
BOS172738 Phase 1 Advanced RET altered tumors NCT03780517
LOXO-260 Phase 1 RET fusion-positive solid tumors, medullary thyroid cancer, and other tumors with RET activation refractory to selective RET inhibitors NCT05241834
NTRK inhibitors
Larotrectinib Phase 2 Locally advanced thyroid cancers harboring NTRK fusions NCT02576431
Larotrectinib Phase 1 Solid tumors harboring NTRK fusion NCT02122913
Entrectinib Phase 2 Locally advanced papillary thyroid cancer with RET fusions NCT02568267
Selitrectinib Phase 1 Solid tumors harboring NTRK fusion NCT03215511
Repotrectinib Phase 1/2 Locally advanced/metastatic solid tumors NCT03093116

DTC, differentiated thyroid carcinoma; RAI, radioactive iodine; RAI-R, RAI-refractory.

Resistance to RET-targeted KIs

Treatment with novel RET-targeted KIs, selpercatinib and pralsetinib, can overcome resistance from gatekeeper mutations but, in turn, can lead to the emergence of resistance mediated by novel non-gatekeeper mutations.

Solvent-front mutations at the G810 site were first described after two patients with MTC and RET fusion-positive NSCLC developed resistance to selpercatinib (Solomon et al. 2020a ). In-depth studies using X-ray crystallography confirmed that mutations at the C-lobe solvent-front (G810C/S/R), hinge site (Y806C/N), and the β2 strand (V738A) all led to a steric clash at the drug-binding site (Subbiah et al. 2021b ).

A study analyzing post-treatment tissue and/or liquid biopsies obtained from 18 patients with resistance to a RET-selective inhibitor showed acquired RET mutations at the RET G810 residue (11%), MET amplification (17%), or KRAS amplification (6%) as the most frequently occurring molecular mechanisms of resistance (Lin et al. 2020). Novel agents that can target solvent-front mutations at the G810 site (LOXO-260, TPX-0046, BOS172738, TAS0953) are currently being investigated (Table 3).

Resistance to NTRK-targeted KIs

A study of larotrectinib in TRK fusion-positive cancers in adults and children reported patterns of resistance in a subset of patients who experienced disease progression during treatment after a documented objective response or SD for at least 6 months (n = 10 patients) (Drilon et al. 2018). Multiple plasma samples obtained upon progression in nine patients showed kinase domain mutations in the NTRK gene, including substitutions in the solvent-front position (NTRK1 G595R or NTRK3 G623R, 78%), gatekeeper mutations (NTRK1 F589L, 22%), or xDFG activation-loop mutations (NTRK1 G667S or NTRK3 G696A, 22%).

A retrospective study of 18 patients with progression on first-generation TRK inhibitors (larotrectinib = 13 and entrectinib = 5) showed that the majority had on-target resistance (83%, n = 15 of 18) compared to off-target resistance (11%, n = 2 of 18), and no identifiable mechanism (6%, n = 1 of 18) (Harada et al. 2022). Among patients with on-target resistance, the most common mutation involved the solvent front (87%, n = 13 of 15: n = 7 NTRK3 G623R, n = 4 NTRK1 G595R, n = 1 NTRK2 G639L, n = 1 NTRK3 G623E) followed by the gatekeeper region (13%, n = 2 of 15: n = 1 NTRK1 F589L, n = 1 NTRK3 F617I). Two patients developed off-target alterations including a BRAF V600E mutation and a MET amplification.

Second-generation NTRK inhibitors with clinical activity against resistance mutations, such as selitrectinib (NCT03215511) and repotrectinib (NCT03093116), are currently being explored in separate phase 1/2 trials.

Clinical considerations for treatment with KIs in thyroid cancers

KIs have revolutionized the treatment of thyroid cancers. However, these treatments are not without significant issues including a challenging side-effect profile and the emergence of drug-resistant clones with treatment. In an open-label phase 2 trial of patients with BRAF-driven solid tumors that included 36 patients with ATC, the combination of BRAF and MEK inhibition with dabrafenib and trametinib was associated with treatment-related serious AEs in 19% (7 of 36). Grade 3/4 AEs were seen in 58% (21 of 36) with anemia (19%) and pneumonia (19%) being the two most common (Subbiah et al. 2022). Pyrexia occurred in 47% of patients, requiring the skillful use of steroids and treatment delays and/or dose reductions. Caution has to be used in patients with heart failure with reduced ejection fraction as there is a potential for cardiomyopathy with MEK inhibitors. A baseline echocardiogram and repeat echocardiograms every 3 months are warranted. Other rare but noteworthy adverse effects include uveitis and hemorrhage.

KIs that target VEGFR have unique adverse effects such as hypertension, hemorrhage, cardiovascular thrombosis, proteinuria, hand-foot syndrome, and fistulas. Patients with a predisposition to these conditions need to be screened out appropriately. A meta-analysis of trials of KIs in thyroid cancers revealed that the discontinuation rate was similar for sorafenib (16%), lenvatinib (16%), and vandetanib (19%). Sorafenib had increased rates of grade 3/4 hand–foot skin-related toxicities (21%) and rash (6%). Lenvatinib was associated with grade 3/4 hypertension in 28% of patients, and 3% had grade 3/4 proteinuria. The incidence of deaths caused by fatal AEs associated with each KI was 0.7, 1.9, and 1.7% for sorafenib, lenvatinib, and vandetanib, respectively (Oba et al. 2020). The newer-generation selective inhibitors of RET – selpercatinib and pralsetinib – have relatively lower rates of treatment-related grade 3 or higher AEs: 28 and 54%, respectively (Subbiah et al. 2021a ). A baseline EKG and close monitoring of QTc is imperative for patients on vandetanib and selpercatinib due to the risk of QTc prolongation. Treatment with vandetanib also requires educating patients on strategies to limit UV exposure to limit photosensitivity reactions.

The risks and attendant toxicities associated with VEGFR KIs pose difficulties in deciding when to start KI therapy in patients with RAI-R DTCs. Some patients have relatively SD for prolonged periods and starting a KI in these patients may have an unacceptable risk/benefit profile. A post-hoc analysis was performed to assess if there was a differential benefit in OS based on baseline tumor burden in patients treated with lenvatinib from the SELECT trial (Kiyota et al. 2022). In an unadjusted analysis in the subgroup of patients with baseline sums > 4 cm (n = 182), there was seemingly no improvement in survival with lenvatinib when compared to placebo (29.1 vs 31.6 months, HR = 1.07; 95% CI, 0.78–1.48). However, to account for the high rate of early crossover from placebo to lenvatinib in the high tumor burden group, an adjusted rank-preserving structural failure time model analysis was performed; patients with high baseline tumor burden who received lenvatinib were also confirmed to have increased survival vs those who received placebo (29.1 vs 14.3 months, HR = 0.49; 95% CI, 0.30–0.74).

Patients with slow-growing diseases may end up needing to take these medications for prolonged periods of time, thereby exposing them to a higher risk of side effects and financial toxicity. Therefore, VEGFR KIs are currently recommended in cases with an increased rate of tumor burden growth or cases in which a high tumor burden portends a high risk of poor overall prognosis. The availability of multiple systemic options in patients with molecular drivers of disease such as RET, BRAF, and NTRK alterations poses a question of how best to sequence these modalities. This remains an open question of active investigation, which can be best answered with randomized clinical trials. A general approach for agent selection should aim to individualize decision-making after considering the risks and benefits in each situation.

Conclusions and future perspectives

The advent of KIs targeting angiogenesis and MAPK signaling has changed the treatment paradigm for thyroid cancers. Sorafenib, a KI with activity against MAPK and VEGFR signaling, was the first KI approved for RAI-R DTC, which was followed by the validation and approval of lenvatinib and cabozantinib. Vandetanib and cabozantinib are multikinase inhibitors that are approved for advanced MTC. In PTCs and MTCs with driver RET alterations, the advent of potent and selective RET inhibitors, such as selpercatinib and pralsetinib, has led to substantial increases in efficacy and quality of life with limited toxicity. NTRK-driven thyroid cancers are highly sensitive to KIs such as larotrectinib and entrectinib. There is evidence that selective KIs that target BRAF, MEK, RET, NTRK, and ALK can re-sensitize refractory DTCs to take up RAI. The outcomes of ATC patients with BRAF V600E/K mutations have improved significantly with combination strategies utilizing BRAF and MEK inhibition. Emerging evidence also indicates that the combination of lenvatinib with the immune-checkpoint inhibitor pembrolizumab shows significant synergy in ATC.

Outstanding questions in the field include the optimal strategy of sequencing different therapies, especially in patients with driver BRAF-mutated tumors who have the added option of agents targeting the MAPK pathway in addition to the anti-angiogenic agents. New targeted agents that can inhibit bypass signaling through RAS mutations and RAC1 activation may help combat resistance to BRAF/MEK inhibition. Novel next-generation RET and NTRK targeted agents that have sensitivity to solvent-front resistance mutations and gatekeeper mutations are in early-phase trials and hold promise for patients with resistance that is driven by these alterations. Ongoing trials of note are summarized in Table 3.

Declaration of interest

MS declares research funding from Merck and Eli-Lilly. BK declares research funding (to institution) from Eli Lilly & Co, Merck, Eisai, and Xencor. VS declares research funding from Eli Lilly & Co.

Funding

This review was not funded by any entities.

Author contribution statement

PJ and VS contributed to manuscript writing and editing. BK and MS contributed to manuscript content and editing. All authors contributed to the article and approved the submitted version.

References

  1. Ahmed M Barbachano Y Riddell A Whittaker S Newbold K Harrington K Marais R & Nutting C. 2008An open labelled phase 2 study evaluating the safety and efficacy of sorafenib in metastatic advanced thyroid cancer. Annals of Oncology 26(Supplement 15); 6060–6060. (https://doi.org10.1200/jco.2008.26.15_suppl.6060) [Google Scholar]
  2. Anderson RT, Linnehan JE, Tongbram V, Keating K, Wirth LJ.2013Clinical, safety, and economic evidence in radioactive iodine–refractory differentiated thyroid cancer: a systematic literature review. Thyroid 23392–407. ( 10.1089/thy.2012.0520) [DOI] [PubMed] [Google Scholar]
  3. Antonello ZA, Hsu N, Bhasin M, Roti G, Joshi M, Van Hummelen P, Ye E, Lo AS, Karumanchi SA, Bryke CRet al. 2017Vemurafenib-resistance via de novo RBM genes mutations and chromosome 5 aberrations is overcome by combined therapy with palbociclib in thyroid carcinoma with BRAFV600E. Oncotarget 884743–84760. ( 10.18632/oncotarget.21262) [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Are C &, Shaha AR.2006Anaplastic thyroid carcinoma: biology, pathogenesis, prognostic factors, and treatment approaches. Annals of Surgical Oncology 13453–464. ( 10.1245/ASO.2006.05.042) [DOI] [PubMed] [Google Scholar]
  5. Aydemirli MD, Van Eendenburg JDH, Van Wezel T, Oosting J, Corver WE, Kapiteijn E, Morreau H.2021Targeting EML4-ALK gene fusion variant 3 in thyroid cancer. Endocrine-Related Cancer 28377–389. ( 10.1530/ERC-20-0436) [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bagheri-Yarmand R, Busaidy NL, Mcbeath E, Danysh BP, Evans KW, Moss TJ, Akcakanat A, Ng PKS, Knippler CM, Golden JAet al. 2021RAC1 alterations induce acquired dabrafenib resistance in association with anaplastic transformation in a papillary thyroid cancer patient. Cancers (Basel) 134950. ( 10.3390/cancers13194950) [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bastman JJ, Serracino HS, Zhu Y, Koenig MR, Mateescu V, Sams SB, Davies KD, Raeburn CD, Mcintyre RC, Haugen BRet al. 2016Tumor-infiltrating T cells and the PD-1 checkpoint pathway in advanced differentiated and anaplastic thyroid cancer. Journal of Clinical Endocrinology and Metabolism 1012863–2873. ( 10.1210/jc.2015-4227) [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, Rubin J, Sideras K, Morris JC, Mciver Bet al. 2010Efficacy of pazopanib in progressive, radioiodine-refractory, metastatic differentiated thyroid cancers: results of a phase 2 consortium study. The Lancet Oncology 11962–972. ( 10.1016/S1470-2045(1070203-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bible KC, Kebebew E, Brierley J, Brito JP, Cabanillas ME, Clark TJ, Di Cristofano A, Foote R, Giordano T, Kasperbauer Jet al. 2021American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid 31337–386. ( 10.1089/thy.2020.0944) [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bikas A, Kundra P, Desale S, Mete M, O'Keefe K, Clark BG, Wray L, Gandhi R, Barett C, Jelinek JSet al. 2016Phase 2 clinical trial of sunitinib as adjunctive treatment in patients with advanced differentiated thyroid cancer. European Journal of Endocrinology 174373–380. ( 10.1530/EJE-15-0930) [DOI] [PubMed] [Google Scholar]
  11. Blauel ER, Laetsch TW.2022The promise of TRK inhibitors in pediatric cancers with NTRK fusions. Cancer Genetics 262–26371–79. ( 10.1016/j.cancergen.2022.01.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bonhomme B, Godbert Y, Perot G, Al Ghuzlan A, Bardet S, Belleannée G, Crinière L, Do Cao C, Fouilloux G, Guyetant Set al. 2017Molecular pathology of anaplastic thyroid carcinomas: a retrospective study of 144 cases. Thyroid 27682–692. ( 10.1089/thy.2016.0254) [DOI] [PubMed] [Google Scholar]
  13. Boostrom SY, Grant CS, Thompson GB, Farley DR, Richards ML, Hoskin TL, Hay ID.2009. Need for a revised staging consensus in medullary thyroid carcinoma. Archives of Surgery 144 663–669. ( 10.1001/archsurg.2009.122) [DOI] [PubMed] [Google Scholar]
  14. Bowles DW Ross JS Gay LM Agrawal V Ali SM & Khan SA. 2018Comprehensive genomic profiling of anaplastic thyroid carcinoma. Journal of Clinical Oncology 366089–6089. ( 10.1200/JCO.2018.36.15_suppl.6089) [DOI] [Google Scholar]
  15. Brandenburg T, Tiedje V, Muchalla P, Theurer S, Weber F, Schmid Kw, Dralle H, Führer D.2021Continued discontinuation of TKI treatment in medullary thyroid carcinoma – lessons from individual cases with long-term follow-up. Frontiers in Endocrinology 12718418. ( 10.3389/fendo.2021.718418) [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Brose MS, Troxel AB, Harlacker K, Redlinger M, Chalian AA, Loevner LA, Mandel SJ, O'Dwyer Pj, Keefe SM.200951LBA Completion of a Phase II study of sorafenib for advanced thyroid cancer. European Journal of Cancer Supplements 7 22. ( 10.1016/S1359-6349(0972086-5) [DOI] [Google Scholar]
  17. Brose MS, Nutting CM, Jarzab B, Elisei R, Siena S, Bastholt L, De La Fouchardiere C, Pacini F, Paschke R, Shong YKet al. 2014Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet 384319–328. ( 10.1016/S0140-6736(1460421-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Brose MS, Cabanillas ME, Cohen EE, Wirth LJ, Riehl T, Yue H, Sherman SI, Sherman EJ.2016Vemurafenib 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. Oncology 171272–1282. ( 10.1016/S1470-2045(1630166-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Brose MS, Robinson B, Sherman SI, Jarzab B, Lin C-C, Vaisman F, Hoff A, Hitre E, Bowles DW, Faoro Let al. 2021aCabozantinib versus placebo in patients with radioiodine-refractory differentiated thyroid cancer who have progressed after prior VEGFR-targeted therapy: results from the phase 3 COSMIC-311 trial. Journal of Clinical Oncology 396001–6001. ( 10.1200/JCO.2021.39.15_suppl.6001) [DOI] [Google Scholar]
  20. Brose MS, Robinson B, Sherman SI, Krajewska J, Lin CC, Vaisman F, Hoff AO, Hitre E, Bowles DW, Hernando Jet al. 2021bCabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncology 221126–1138. ( 10.1016/S1470-2045(2100332-6) [DOI] [PubMed] [Google Scholar]
  21. Busaidy NL, Konda B, Wei L, Wirth LJ, Devine C, Daniels GA, Desouza JA, Poi M, Seligson ND, Cabanillas MEet al. 2022Dabrafenib versus Dabrafenib + trametinib in BRAF-mutated radioactive iodine refractory differentiated thyroid cancer: results of a randomized, Phase 2, open-label multicenter trial. Thyroid 321184–1192. ( 10.1089/thy.2022.0115) [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cabanillas ME, Ferrarotto R, Garden AS, Ahmed S, Busaidy NL, Dadu R, Williams MD, Skinner H, Gunn GB, Grosu Het al. 2018Neoadjuvant BRAF- and immune-directed therapy for anaplastic thyroid carcinoma. Thyroid 28945–951. ( 10.1089/thy.2018.0060) [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Cabanillas ME, Dadu R, Iyer P, Wanland KB, Busaidy NL, Ying A, Gule-Monroe M, Wang JR, Zafereo M, Hofmann MC.2020Acquired secondary RAS mutation in BRAF(V600E)-mutated thyroid cancer patients treated with BRAF inhibitors. Thyroid 301288–1296. ( 10.1089/thy.2019.0514) [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cancer Genome Atlas Research Network 2014Integrated genomic characterization of papillary thyroid carcinoma. Cell 159676–690. ( 10.1016/j.cell.2014.09.050) [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cantara S, Bertelli E, Occhini R, Regoli M, Brilli L, Pacini F, Castagna MG, Toti P.2019Blockade of the programmed death ligand 1 (PD-L1) as potential therapy for anaplastic thyroid cancer. Endocrine 64122–129. ( 10.1007/s12020-019-01865-5) [DOI] [PubMed] [Google Scholar]
  26. Capdevila J, Wirth LJ, Ernst T, Ponce Aix S, Lin CC, Ramlau R, Butler MO, Delord JP, Gelderblom H, Ascierto PAet al. 2020PD-1 blockade in anaplastic thyroid carcinoma. Journal of Clinical Oncology 382620–2627. ( 10.1200/JCO.19.02727) [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Capdevila J, Robinson B, Sherman SI, Jarzab B, Lin CC, Vaisman F, Hoff AO, Hitre E, Bowles DW, Sen Set al. 2021LBA67 cabozantinib versus placebo in patients with radioiodine-refractory differentiated thyroid cancer who have progressed after prior VEGFR-targeted therapy: updated results from the phase III COSMIC-311 trial and prespecified subgroup analyses by prior therapy. Annals of Oncology 32 S1343. ( 10.1016/j.annonc.2021.08.2148) [DOI] [Google Scholar]
  28. Carr L, Goulart B, Martins R, Keith E, Kell E, Wallace S, Capell P, Mankoff D.2009Phase II trial of continuous dosing of sunitinib in advanced, FDG-PET avid, medullary thyroid carcinoma (MTC) and well-differentiated thyroid cancer (WDTC). Journal of Clinical Oncology 276056–6056. ( 10.1200/jco.2009.27.15_suppl.6056) [DOI] [Google Scholar]
  29. Chakravarty D, Santos E, Ryder M, Knauf JA, Liao XH, West BL, Bollag G, Kolesnick R, Thin TH, Rosen Net al. 2011Small-molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. Journal of Clinical Investigation 1214700–4711. ( 10.1172/JCI46382) [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chintakuntlawar AV, Rumilla KM, Smith CY, Jenkins SM, Foote RL, Kasperbauer JL, Morris JC, Ryder M, Alsidawi S, Hilger Cet al. 2017. Expression of PD-1 and PD-L1 in Anaplastic Thyroid Cancer Patients Treated with Multimodal Therapy: results from a retrospective study. Journal of Clinical Endocrinology and Metabolism 1021943–1950. ( 10.1210/jc.2016-3756) [DOI] [PubMed] [Google Scholar]
  31. Ciampi R, Romei C, Ramone T, Prete A, Tacito A, Cappagli V, Bottici V, Viola D, Torregrossa L, Ugolini Cet al. 2019Genetic landscape of somatic mutations in a large cohort of sporadic medullary thyroid carcinomas studied by next-generation targeted sequencing. iScience 20324–336. ( 10.1016/j.isci.2019.09.030) [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Cohen EE, Needles BM, Cullen KJ, Wong SJ, Wade JL, Ivy SP, Villaflor VM, Seiwert TY, Nichols K, Vokes EE.2008Phase 2 study of sunitinib in refractory thyroid cancer. Journal of Clinical Oncology 266025–6025. ( 10.1200/jco.2008.26.15_suppl.6025) [DOI] [Google Scholar]
  33. Cohen EEW, Tortorici M, Kim S, Ingrosso A, Pithavala YK, Bycott P.2014A Phase II trial of axitinib in patients with various histologic subtypes of advanced thyroid cancer: long-term outcomes and pharmacokinetic/pharmacodynamic analyses. Cancer Chemotherapy and Pharmacology 741261–1270. ( 10.1007/s00280-014-2604-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Demetri GD, De Braud F, Drilon A, Siena S, Patel MR, Cho BC, Liu SV, Ahn MJ, Chiu CH, Lin JJet al. 2022Correction: updated integrated analysis of the efficacy and safety of entrectinib in patients with NTRK fusion-positive solid tumors. Clinical Cancer Research 28 2196. ( 10.1158/1078-0432.CCR-22-1108) [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Dias-Santagata D, Lennerz JK, Sadow PM, Frazier RP, Govinda Raju S, Henry D, Chung T, Kherani J, Rothenberg SM, Wirth LJ.2020Response to RET-specific therapy in RET fusion-positive anaplastic thyroid carcinoma. Thyroid 301384–1389. ( 10.1089/thy.2019.0477) [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Dierks C, Seufert J, Aumann K, Ruf J, Klein C, Kiefer S, Rassner M, Boerries M, Zielke A, la Rosee P, et al. 2021Combination of lenvatinib and pembrolizumab is an effective treatment option for anaplastic and poorly differentiated thyroid carcinoma. Thyroid 311076–1085. ( 10.1089/thy.2020.0322) [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Drilon A, Laetsch TW, Kummar S, Dubois SG, Lassen UN, Demetri GD, Nathenson M, Doebele RC, Farago AF, Pappo ASet al. 2018Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. New England Journal of Medicine 378731–739. ( 10.1056/NEJMoa1714448) [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Dunn LA, Sherman EJ, Baxi SS, Tchekmedyian V, Grewal RK, Larson SM, Pentlow KS, Haque S, Tuttle RM, Sabra MMet al. 2019Vemurafenib redifferentiation of BRAF mutant, RAI-refractory thyroid cancers. Journal of Clinical Endocrinology and Metabolism 1041417–1428. ( 10.1210/jc.2018-01478) [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Duquette M, Sadow PM, Husain A, Sims JN, Antonello ZA, Fischer AH, Song C, Castellanos-Rizaldos E, Makrigiorgos GM, Kurebayashi Jet al. 2015Metastasis-associated MCL1 and P16 copy number alterations dictate resistance to vemurafenib in a BRAFV600E patient-derived papillary thyroid carcinoma preclinical model. Oncotarget 642445–42467. ( 10.18632/oncotarget.6442) [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Durante C, Haddy N, Baudin E, Leboulleux S, Hartl D, Travagli JP, Caillou B, Ricard M, Lumbroso JD, De Vathaire Fet al. 2006Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy. Journal of Clinical Endocrinology and Metabolism 912892–2899. ( 10.1210/jc.2005-2838) [DOI] [PubMed] [Google Scholar]
  41. Efstathiadou ZA, Tsentidis C, Bargiota A, Daraki V, Kotsa K, Ntali G, Papanastasiou L, Tigas S, Toulis K, Pazaitou-Panayiotou Ket al. 2021Benefits and limitations of TKIs in patients with medullary thyroid cancer: a systematic review and meta-analysis. European Thyroid Journal 10125–139. ( 10.1159/000509457) [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Elisei R, Ugolini C, Viola D, Lupi C, Biagini A, Giannini R, Romei C, Miccoli P, Pinchera A, Basolo F.2008BRAF(V600E) mutation and outcome of patients with papillary thyroid carcinoma: a 15-year median follow-up study. Journal of Clinical Endocrinology and Metabolism 933943–3949. ( 10.1210/jc.2008-0607) [DOI] [PubMed] [Google Scholar]
  43. Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, Licitra L, Jarzab B, Medvedev V, Kreissl MCet al. 2013Cabozantinib in Progressive Medullary Thyroid Cancer. Journal of Clinical Oncology 313639–3646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Elisei R, Tacito A, Ramone T, Ciampi R, Bottici V, Cappagli V, Viola D, Matrone A, Lorusso L, Valerio Let al. 2019Twenty-five years experience on RET genetic screening on hereditary MTC: an update on the prevalence of germline RET mutations. Genes (Basel) 10698. ( 10.3390/genes10090698) [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fagin JA, Wells SA.2016Biologic and clinical perspectives on thyroid cancer. New England Journal of Medicine 3751054–1067. ( 10.1056/NEJMra1501993) [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fullmer T, Cabanillas ME, Zafereo M.2021Novel therapeutics in radioactive iodine-resistant thyroid cancer. Frontiers in Endocrinology 12720723. ( 10.3389/fendo.2021.720723) [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Ganly I, Nixon IJ, Wang LY, Palmer FL, Migliacci JC, Aniss A, Sywak M, Eskander AE, Freeman JL, Campbell MJet al. 2015Survival from differentiated thyroid cancer: what has age got to do with it? Thyroid 251106–1114. ( 10.1089/thy.2015.0104) [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn Let al. 2015Pembrolizumab for the Treatment of Non–Small-Cell Lung Cancer. New England Journal of Medicine 3722018–2028. ( 10.1056/NEJMoa1501824) [DOI] [PubMed] [Google Scholar]
  49. Gild ML, Topliss DJ, Learoyd D, Parnis F, Tie J, Hughes B, Walsh JP, Mcleod DSA, Clifton-Bligh RJ, Robinson BG.2018Clinical guidance for radioiodine refractory differentiated thyroid cancer. Clinical Endocrinology 88529–537. ( 10.1111/cen.13508) [DOI] [PubMed] [Google Scholar]
  50. Giunti S, Antonelli A, Amorosi A, Santarpia L.2013Cellular signaling pathway alterations and potential targeted therapies for medullary thyroid carcinoma. International Journal of Endocrinology 2013 803171. ( 10.1155/2013/803171) [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gómez-Sáez JM.2016Sunitinib for the treatment of thyroid cancer. Expert Opinion on Investigational Drugs 251345–1352. ( 10.1080/13543784.2016.1242577) [DOI] [PubMed] [Google Scholar]
  52. Groussin L, Clerc J, Huillard O.2020Larotrectinib-enhanced radioactive iodine uptake in advanced thyroid cancer. New England Journal of Medicine 3831686–1687. ( 10.1056/NEJMc2023094) [DOI] [PubMed] [Google Scholar]
  53. Groussin L, Bessiene L, Arrondeau J, Garinet S, Cochand-Priollet B, Lupo A, Zerbit J, Clerc J, Huillard O.2021Letter to the editor: selpercatinib-enhanced radioiodine uptake in RET-rearranged thyroid cancer. Thyroid 311603–1604. ( 10.1089/thy.2021.0144) [DOI] [PubMed] [Google Scholar]
  54. Groussin L, Theodon H, Bessiene L, Bricaire L, Bonnet-Serrano F, Cochand-Priollet B, Leroy K, Garinet S, Pasmant E, Zerbit Jet al. 2022Redifferentiating effect of larotrectinib in NTRK-rearranged advanced radioactive-iodine refractory thyroid cancer. Thyroid 32594–598. ( 10.1089/thy.2021.0524) [DOI] [PubMed] [Google Scholar]
  55. Gupta-Abramson V, Troxel AB, Nellore A, Puttaswamy K, Redlinger M, Ransone K, Mandel SJ, Flaherty KT, Loevner LA, O'dwyer PJet al. 2008Phase II trial of sorafenib in advanced thyroid cancer. Journal of Clinical Oncology 264714–4719. ( 10.1200/JCO.2008.16.3279) [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hanna GJ, Busaidy NL, Chau NG, Wirth LJ, Barletta JA, Calles A, Haddad RI, Kraft S, Cabanillas ME, Rabinowits Get al. 2018Genomic correlates of response to everolimus in aggressive radioiodine-refractory thyroid cancer: a Phase II study. Clinical Cancer Research 241546–1553. ( 10.1158/1078-0432.CCR-17-2297) [DOI] [PubMed] [Google Scholar]
  57. Harada G Choudhary NJ Schram AM Rosen E Murciano-Goroff YR Falcon CJ Wilhelm C Kaplanis LA Liu D Chang JC et al. 2022Mechanisms of acquired resistance to TRK inhibitors. Journal of Clinical Oncology 40(Supplement 16) 3104. ( 10.1200/JCO.2022.40.16_suppl.3104) [DOI] [Google Scholar]
  58. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, Pacini F, Randolph GW, Sawka AM, Schlumberger Met al. 2016American 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 261–133. ( 10.1089/thy.2015.0020) [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Haugen B, French J, Worden FP, Konda B, Sherman EJ, Dadu R, Gianoukakis AG, Wolfe EG, Foster NR, Bowles DWet al. 2020Lenvatinib plus pembrolizumab combination therapy in patients with radioiodine-refractory (RAIR), progressive differentiated thyroid cancer (DTC): results of a multicenter phase II international thyroid oncology group trial. Journal of Clinical Oncology 386512–6512. ( 10.1200/JCO.2020.38.15_suppl.6512) [DOI] [Google Scholar]
  60. Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, Pentlow KS, Zanzonico PB, Haque S, Gavane Set al. 2013Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. New England Journal of Medicine 368623–632. ( 10.1056/NEJMoa1209288) [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Ho AL, Dedecjus M, Wirth LJ, Tuttle RM, Inabnet WB, Tennvall J, Vaisman F, Bastholt L, Gianoukakis AG, Rodien Pet al. 2022Selumetinib plus adjuvant radioactive iodine in patients with high-risk differentiated thyroid cancer: a Phase III, randomized, placebo-controlled trial (Astra). Journal of Clinical Oncology 401, 870–1. ( 10.1200/JCO.21.00714) [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hoftijzer H, Heemstra KA, Morreau H, Stokkel MP, Corssmit EP, Gelderblom H, Weijers K, Pereira AM, Huijberts M, Kapiteijn Eet al. 2009Beneficial effects of sorafenib on tumor progression, but not on radioiodine uptake, in patients with differentiated thyroid carcinoma. European Journal of Endocrinology 161923–931. ( 10.1530/EJE-09-0702) [DOI] [PubMed] [Google Scholar]
  63. Howell GM, Hodak SP, Yip L.2013RAS mutations in thyroid cancer. Oncologist 18926–932. ( 10.1634/theoncologist.2013-0072) [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Ibrahimpasic T, Ghossein R, Carlson DL, Nixon I, Palmer FL, Shaha AR, Patel SG, Tuttle RM, Shah JP, Ganly I.2014Outcomes in patients with poorly differentiated thyroid carcinoma. Journal of Clinical Endocrinology and Metabolism 991245–1252. ( 10.1210/jc.2013-3842) [DOI] [PubMed] [Google Scholar]
  65. Iravani A, Solomon B, Pattison DA, Jackson P, Ravi Kumar A, Kong G, Hofman Ms, Akhurst T, Hicks RJ.2019Mitogen-activated protein kinase pathway inhibition for redifferentiation of radioiodine refractory differentiated thyroid cancer: an evolving protocol. Thyroid 291634–1645. ( 10.1089/thy.2019.0143) [DOI] [PubMed] [Google Scholar]
  66. Jaber T, Waguespack SG, Cabanillas ME, Elbanan M, Vu T, Dadu R, Sherman SI, Amit M, Santos EB, Zafereo Met al. 2018Targeted therapy in advanced thyroid cancer to resensitize tumors to radioactive iodine. Journal of Clinical Endocrinology and Metabolism 1033698–3705. ( 10.1210/jc.2018-00612) [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Kelly LM, Barila G, Liu P, Evdokimova VN, Trivedi S, Panebianco F, Gandhi M, Carty SE, Hodak SP, Luo Jet al. 2014Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. PNAS 1114233–4238. ( 10.1073/pnas.1321937111) [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kiyota N, Tahara M, Robinson B, Schlumberger M, Sherman SI, Leboulleux S, Lee EK, Suzuki T, Ren M, Fushimi Ket al. 2022Impact of baseline tumor burden on overall survival in patients with radioiodine-refractory differentiated thyroid cancer treated with lenvatinib in the SELECT global phase 3 trial. Cancer 1282281–2287. ( 10.1002/cncr.34181) [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kloos RT, Ringel MD, Knopp MV, Hall NC, King M, Stevens R, Liang J, Wakely PE, Vasko VV, Saji Met al. 2009Phase II trial of sorafenib in metastatic thyroid cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 271675–1684. ( 10.1200/JCO.2008.18.2717) [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Kunstman JW, Juhlin CC, Goh G, Brown TC, Stenman A, Healy JM, Rubinstein JC, Choi M, Kiss N, Nelson-Williams Cet al. 2015Characterization of the mutational landscape of anaplastic thyroid cancer via whole-exome sequencing. Human Molecular Genetics 242318–2329. ( 10.1093/hmg/ddu749) [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah RH, Dogan S, Ricarte-Filho JC, Krishnamoorthy GP, Xu Bet al. 2016Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. Journal of Clinical Investigation 1261052–1066. ( 10.1172/JCI85271) [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Leboulleux S, Benisvy D, Taieb D, Attard M, Bournaud C, Terroir M, Al Ghuzlan A, Lamartina L, Schlumberger MJ, Godbert Yet al. 20211743MO MERAIODE: a redifferentiation phase II trial with trametinib followed by radioactive iodine for metastatic radioactive iodine refractory differentiated thyroid cancer patients with a RAS mutation. Annals of Oncology 32 S1204. ( 10.1016/j.annonc.2021.08.889) [DOI] [Google Scholar]
  73. Lee YA, Lee H, Im SW, Song YS, Oh DY, Kang HJ, Won JK, Jung KC, Kwon D, Chung EJet al. 2021NTRK and RET fusion-directed therapy in pediatric thyroid cancer yields a tumor response and radioiodine uptake. Journal of Clinical Investigation 131e14484. ( 10.1172/JCI144847) [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Lim SM Shin S-J Chung WY Park CS Nam K-H Kang S-W Keum KC Kim JH Cho JY & Hong YK. 2012. Treatment outcome of patients with anaplastic thyroid cancer: a single center experience. Yonsei Medical Journal 53 352. ( 10.3349/ymj.2012.53.2.352) [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Lin B, Ma H, Ma M, Zhang Z, Sun Z, Hsieh IY, Okenwa O, Guan H, Li J, Lv W.2019The incidence and survival analysis for anaplastic thyroid cancer: a SEER database analysis. American Journal of Translational Research 115888–5896. [PMC free article] [PubMed] [Google Scholar]
  76. Lin JJ, Liu SV, Mccoach CE, Zhu VW, Tan AC, Yoda S, Peterson J, Do A, Prutisto-Chang K, Dagogo-Jack Iet al. 2020Mechanisms of resistance to selective RET tyrosine kinase inhibitors in RET fusion-positive non-small-cell lung cancer. Annals of Oncology 311725–1733. ( 10.1016/j.annonc.2020.09.015) [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Liu D, Hu S, Hou P, Jiang D, Condouris S, Xing M.2007Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant. Clinical Cancer Research 131341–1349. ( 10.1158/1078-0432.CCR-06-1753) [DOI] [PubMed] [Google Scholar]
  78. Locati LD, Licitra L, Agate L, Ou SH, Boucher A, Jarzab B, Qin S, Kane MA, Wirth LJ, Chen Cet al. 2014Treatment of advanced thyroid cancer with axitinib: Phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments. Cancer 1202694–2703. ( 10.1002/cncr.28766) [DOI] [PubMed] [Google Scholar]
  79. Maniakas A, Dadu R, Busaidy NL, Wang JR, Ferrarotto R, Lu C, Williams MD, Gunn GB, Hofmann MC, Cote Get al. 2020Evaluation of overall survival in patients with anaplastic thyroid carcinoma, 2000–2019. JAMA Oncology 61397–1404. ( 10.1001/jamaoncol.2020.3362) [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Mansfield AS, Subbiah V, Schuler MH, Zhu VW, Hadoux J, Brose MS, Curigliano G, Wirth LJ, Garralda E, Adkins Det al. 2022Pralsetinib in patients (pts) with advanced or metastatic RET-altered thyroid cancer (TC): updated data from the ARROW trial. Journal of Clinical Oncology 406080–6080. ( 10.1200/JCO.2022.40.16_suppl.6080) [DOI] [Google Scholar]
  81. Mehnert JM, Varga A, Brose MS, Aggarwal RR, Lin C-C, Prawira A, De Braud F, Tamura K, Doi T, Piha-Paul SAet al. 2019Safety and antitumor activity of the anti–PD-1 antibody pembrolizumab in patients with advanced, PD-L1–positive papillary or follicular thyroid cance. BMC Cancer 19196. ( 10.1186/s12885-019-5380-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Montero-Conde C, Ruiz-Llorente S, Dominguez JM, Knauf JA, Viale A, Sherman EJ, Ryder M, Ghossein RA, Rosen N, Fagin JA.2013Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF-mutant thyroid carcinomas. Cancer Discovery 3520–533. ( 10.1158/2159-8290.CD-12-0531) [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Nikiforova MN, Lynch RA, Biddinger PW, Alexander EK, Dorn GW, Tallini G, Kroll TG, Nikiforov YE.2003RAS point mutations and PAX8-PPARγ rearrangement in thyroid tumors: evidence for distinct molecular pathways in thyroid follicular carcinoma. Journal of Clinical Endocrinology and Metabolism 882318–2326. ( 10.1210/jc.2002-021907) [DOI] [PubMed] [Google Scholar]
  84. Oba T, Chino T, Soma A, Shimizu T, Ono M, Ito T, Kanai T, Maeno K, Ito KI.2020Comparative efficacy and safety of tyrosine kinase inhibitors for thyroid cancer: a systematic review and meta-analysis. Endocrine Journal 671215–1226. ( 10.1507/endocrj.EJ20-0171) [DOI] [PubMed] [Google Scholar]
  85. Owen DH, Konda B, Sipos J, Liu T, Webb A, Ringel MD, Timmers CD, Shah MH.2019KRAS G12V mutation in acquired resistance to combined BRAF and MEK inhibition in papillary thyroid cancer. Journal of the National Comprehensive Cancer Network 17409–413. ( 10.6004/jnccn.2019.7292) [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Porter A, Wong DJ.2020Perspectives on the treatment of advanced thyroid cancer: approved therapies, resistance mechanisms, and future directions. Frontiers in Oncology 10 592202. ( 10.3389/fonc.2020.592202) [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Porter A, Zeh A, Cohen E, Chai-Ho W, Wong DJ.2020Combination immunotherapy and TKI in metastatic refractory thyroid cancer, a case series. International Journal of Radiation Oncology*Biology*Physics 106 1197. ( 10.1016/j.ijrobp.2019.11.112) [DOI] [Google Scholar]
  88. Pozdeyev N, Gay LM, Sokol ES, Hartmaier R, Deaver KE, Davis S, French JD, Borre PV, Labarbera DV, Tan ACet al. 2018Genetic analysis of 779 advanced differentiated and anaplastic thyroid cancers. Clinical Cancer Research 243059–3068. ( 10.1158/1078-0432.CCR-18-0373) [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Ravaud A, De La Fouchardière C, Caron P, Doussau A, Do Cao C, Asselineau J, Rodien P, Pouessel D, Nicolli-Sire P, Klein Met al. 2017A multicenter phase II study of sunitinib in patients with locally advanced or metastatic differentiated, anaplastic or medullary thyroid carcinomas: mature data from the THYSU study. European Journal of Cancer 76110–117. ( 10.1016/j.ejca.2017.01.029) [DOI] [PubMed] [Google Scholar]
  90. Roman S, Lin R, Sosa JA.2006Prognosis of medullary thyroid carcinoma: demographic, clinical, and pathologic predictors of survival in 1252 cases. Cancer 1072134–2142. ( 10.1002/cncr.22244) [DOI] [PubMed] [Google Scholar]
  91. Rosenbaum MW Gigliotti BJ Pai SI Parangi S Wachtel H Mino-Kenudson M Gunda V & Faquin WC. 2018PD-L1 and IDO1 are expressed in poorly differentiated thyroid carcinoma. Endocrine Pathology 2959–67. ( 10.1007/s12022-018-9514-y) [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Rothenberg SM, Mcfadden DG, Palmer EL, Daniels GH, Wirth LJ.2015Redifferentiation of iodine-refractory BRAF V600E-mutant metastatic papillary thyroid cancer with dabrafenib. Clinical Cancer Research 211028–1035. ( 10.1158/1078-0432.CCR-14-2915) [DOI] [PubMed] [Google Scholar]
  93. Saqcena M, Leandro-Garcia LJ, Maag JLV, Tchekmedyian V, Krishnamoorthy GP, Tamarapu PP, Tiedje V, Reuter V, Knauf JA, De Stanchina Eet al. 2021SWI/SNF complex mutations promote thyroid tumor progression and insensitivity to redifferentiation therapies. Cancer Discovery 111, 15, 8–1. ( 10.1158/2159-8290.CD-20-0735) [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, Habra MA, Newbold K, Shah MH, Hoff AOet al. 2015Lenvatinib versus Placebo in Radioiodine-Refractory thyroid Cancer. New England Journal of Medicine 372621–630. ( 10.1056/NEJMoa1406470) [DOI] [PubMed] [Google Scholar]
  95. Sherman EJ, Foster NR, Su YB, Shergill A, Ho AL, Konda B, Ghossein RA, Ganly I, Schwartz GK.2021Randomized phase II study of sorafenib with or without everolimus in patients with radioactive iodine refractory Hürthle cell thyroid cancer (HCC) (Alliance A091302/ ITOG 1706). Journal of Clinical Oncology 396076–6076. ( 10.1200/JCO.2021.39.15_suppl.6076) [DOI] [Google Scholar]
  96. Smallridge RC Marlow LA & Copland JA. 2009Anaplastic thyroid cancer: molecular pathogenesis and emerging therapies. Endocrine-Related Cancer 1617–44. ( 10.1677/ERC-08-0154) [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Solomon BJ, Tan L, Lin JJ, Wong SQ, Hollizeck S, Ebata K, Tuch BB, Yoda S, Gainor JF, Sequist LVet al. 2020aRET solvent front mutations mediate acquired resistance to selective RET inhibition in RET-driven malignancies. Journal of Thoracic Oncology 15541–549. ( 10.1016/j.jtho.2020.01.006) [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Solomon JP, Linkov I, Rosado A, Mullaney K, Rosen EY, Frosina D, Jungbluth AA, Zehir A, Benayed R, Drilon Aet al. 2020bNTRK fusion detection across multiple assays and 33,997 cases: diagnostic implications and pitfalls. Modern Pathology 3338–46. ( 10.1038/s41379-019-0324-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski C, Cabanillas ME, Urbanowitz Get al. 2018Dabrafenib and Trametinib Treatment in patients with locally advanced or metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer. Journal of Clinical Oncology 367–13. ( 10.1200/JCO.2017.73.6785) [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Subbiah V, Hu MI, Wirth LJ, Schuler M, Mansfield AS, Curigliano G, Brose MS, Zhu VW, Leboulleux S, Bowles DWet al. 2021aPralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): a multi-cohort, open-label, registrational, phase 1/2 study. Lancet. Diabetes and Endocrinology 9491–501. ( 10.1016/S2213-8587(2100120-0) [DOI] [PubMed] [Google Scholar]
  101. Subbiah V, Shen T, Terzyan SS, Liu X, Hu X, Patel KP, Hu M, Cabanillas M, Behrang A, Meric-Bernstam Fet al. 2021bStructural basis of acquired resistance to selpercatinib and pralsetinib mediated by non-gatekeeper RET mutations. Annals of Oncology 32261–268. ( 10.1016/j.annonc.2020.10.599) [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, Wen PY, Zielinski CC, Cabanillas ME, Boran Aet al. 2022Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: updated analysis from the phase II ROAR basket study. Annals of Oncology 33406–415. ( 10.1016/j.annonc.2021.12.014) [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Tuttle RM, Ball DW, Byrd D, Dilawari RA, Doherty GM, Duh QY, Ehya H, Farrar WB, Haddad RI, Kandeel Fet al. 2010Thyroid carcinoma. Journal of the National Comprehensive Cancer Network 81228–1274. ( 10.6004/jnccn.2010.0093) [DOI] [PubMed] [Google Scholar]
  104. Villaruz LC, Socinski MA.2016The clinical utility of PD-L1 testing in selecting non-small cell lung cancer patients for PD1/PD-L1-directed therapy. Clinical Pharmacology and Therapeutics 100212–214. ( 10.1002/cpt.385) [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Villaruz LC, Ancevski Hunter K, Kurland BF, Abberbock S, Herbst C, Dacic S.2019Comparison of PD-L1 immunohistochemistry assays and response to PD-1/L1 inhibitors in advanced non-small-cell lung cancer in clinical practice. Histopathology 74269–275. ( 10.1111/his.13729) [DOI] [PubMed] [Google Scholar]
  106. Waguespack SG, Drilon A, Lin JJ, Brose MS, Mcdermott R, Almubarak M, Bauman J, Casanova M, Krishnamurthy A, Kummar Set al. 2022aEfficacy and safety of larotrectinib in patients with TRK fusion-positive thyroid carcinoma. European Journal of Endocrinology 186631–643. ( 10.1530/EJE-21-1259) [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Waguespack SG, Tewari SO, Busaidy NL, Zafereo ME.2022bLarotrectinib before initial radioactive iodine therapy in pediatric TRK fusion-positive papillary thyroid carcinoma: time to reconsider the treatment paradigm for distantly metastatic disease? JCO Precision Oncology 6 e2100467. ( 10.1200/PO.21.00467) [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Wang JR, Zafereo ME, Dadu R, Ferrarotto R, Busaidy NL, Lu C, Ahmed S, Gule-Monroe MK, Williams MD, Sturgis EMet al. 2019Complete Surgical Resection Following Neoadjuvant Dabrafenib Plus Trametinib in BRAFV600E-Mutated Anaplastic Thyroid Carcinoma. Thyroid 291036–1043. ( 10.1089/thy.2019.0133) [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Wells SA, Robinson BG, Gagel RF, Dralle H, Fagin JA, Santoro M, Baudin E, Elisei R, Jarzab B, Vasselli JRet al. 2012Vandetanib in patients with locally advanced or metastatic medullary thyroid cancer: a randomized, double-blind phase III trial. Journal of Clinical Oncology 30134–141. ( 10.1200/JCO.2011.35.5040) [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Wells SA, Asa SL, Dralle H, Elisei R, Evans DB, Gagel RF, Lee N, Machens A, Moley JF, Pacini Fet al. 2015Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma. Thyroid 25567–610. ( 10.1089/thy.2014.0335) [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, Worden F, Brose M, Patel J, Leboulleux Set al. 2020Efficacy of selpercatinib in RET-altered thyroid cancers. New England Journal of Medicine 383825–835. ( 10.1056/NEJMoa2005651) [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Wirth LJ, Brose MS, Sherman EJ, Licitra L, Schlumberger M, Sherman SI, Bible KC, Robinson B, Rodien P, Godbert Yet al. 2021Open-label, single-arm, multicenter, Phase II trial of lenvatinib for the treatment of patients with anaplastic thyroid cancer. Journal of Clinical Oncology 392359–2366. ( 10.1200/JCO.20.03093) [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Zhang G-Q Wei W-J Song H-J Sun Z-K Shen C-T Zhang X-Y Chen X-Y & Qiu Z-L. 2019. Programmed Cell Death–Ligand 1 Overexpression in Thyroid Cancer. Endocrine Practice 25279–286. ( 10.4158/EP-2018-0342) [DOI] [PubMed] [Google Scholar]
  114. Zwaenepoel K, Jacobs J, De Meulenaere A, Silence K, Smits E, Siozopoulou V, Hauben E, Rolfo C, Rottey S, Pauwels P.2017CD70 and PD-L1 in anaplastic thyroid cancer - promising targets for immunotherapy. Histopathology 71357–365. ( 10.1111/his.13230) [DOI] [PubMed] [Google Scholar]

Articles from Endocrine Oncology are provided here courtesy of Bioscientifica Ltd.

RESOURCES