Next Article in Journal
Single-Cell Sequencing: An Emerging Tool for Biomarker Development in Nuclear Emergencies and Radiation Oncology
Previous Article in Journal
Identification of an Immune-Related Gene Signature for Prognostic Prediction in Glioblastoma: Insights from Integrated Bulk and Single-Cell RNA Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Systemic Therapeutic Options in Radioiodine-Refractory Differentiated Thyroid Cancer: Current Indications and Optimal Timing

by
Tamara Díaz Vico
1,2,*,
Brezo Martínez-Amores Martínez
1,2,
Luka Mihic Góngora
3,4,
Paula Jiménez-Fonseca
5,
Paloma Peinado Martín
3,
Irene Grao Torrente
1,
Alejandro García Muñoz-Nájar
1 and
Manuel Durán-Poveda
1,2
1
Department of General Surgery, Hospital Universitario Rey Juan Carlos, 28933 Madrid, Spain
2
Department of Health Sciences, Rey Juan Carlos University, 28933 Móstoles, Spain
3
HM CIOCC MADRID (Centro Integral Oncológico Clara Campal), Hospital Universitario HM Sanchinarro, 28050 Madrid, Spain
4
Instituto de Investigación Sanitaria HM Hospitales, Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, 28692 Cañada, Spain
5
Medical Oncology Department, Hospital Universitario Central de Asturias, ISPA, 33011 Oviedo, Spain
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(11), 1800; https://doi.org/10.3390/cancers17111800
Submission received: 3 May 2025 / Accepted: 24 May 2025 / Published: 28 May 2025
(This article belongs to the Section Cancer Therapy)

Simple Summary

Thyroid cancer (TC) is a common malignancy, with over 820,000 cases diagnosed globally in 2022. Differentiated thyroid carcinoma (DTC), including papillary and follicular types, generally responds well to treatment, but 5–15% of patients develop radioiodine-refractory (RAI-R) disease, leading to a poorer prognosis. In such cases, treatment decisions depend on disease progression. While some patients can be monitored through active surveillance, those with symptomatic or progressive disease require systemic therapy. Multikinase inhibitors (MKIs) like lenvatinib and sorafenib are the first-line treatment for RAI-R DTC, with cabozantinib recently approved for resistant cases. Additionally, targeted therapies, such as RET and NTRK inhibitors, and immune checkpoint inhibitors, offer new treatment possibilities. However, systemic therapies have significant side effects, impacting quality of life (QoL). This review summarizes current evidence on treatment options, optimal timing, and sequencing, highlighting the need for further research to improve personalized care for RAI-R TC patients.

Abstract

Thyroid cancer (TC) remains a prevalent malignancy, with over 820,000 global cases diagnosed in 2022. Differentiated thyroid carcinoma (DTC), primarily papillary and follicular types, accounts for most cases and has a favorable prognosis with total thyroidectomy and radioiodine (RAI) ablation. However, 5–15% of patients develop RAI-refractory (RAI-R) disease, leading to a significantly poorer outcome. For RAI-R patients, treatment decisions depend on disease progression. Active surveillance is suitable for indolent cases, while symptomatic or progressive disease requires systemic therapy. Multikinase inhibitors (MKIs) such as lenvatinib and sorafenib serve as first-line options, with cabozantinib recently approved for resistant cases. Additionally, novel targeted therapies, including RET and NTRK inhibitors, and immune checkpoint inhibitors, are under investigation, offering a personalized approach. A key challenge is determining the optimal timing for systemic therapy, balancing progression-free survival (PFS) benefits against MKI-related toxicities, which significantly impact quality of life (QoL). Molecular testing can identify actionable mutations, guiding therapy selection. Clinical guidelines (ATA, ESMO) recommend initiating treatment based on disease progression and patient condition, integrating strategies such as active surveillance, surgery, and radiotherapy when appropriate. Despite advances, systemic therapies carry significant adverse events (e.g., hypertension, fatigue, gastrointestinal toxicity), necessitating careful monitoring to prevent dose reductions or interruptions. A multidisciplinary approach is essential to optimize patient outcomes and maintain QoL. As targeted therapies continue to evolve, further research is needed to refine treatment sequencing and improve outcomes for RAI-R TC. This review synthesizes current evidence to guide clinical decision-making.

1. Introduction

Thyroid cancer (TC) is a relatively common malignancy, with an estimated 821,214 new cases worldwide in 2022, accounting for 4.1% of all cancer diagnoses [1]. In Spain, 5233 new cases and 314 annual deaths have been reported, which accounts for 0.27% of cancer-related mortality, according to the latest available data from the Global Cancer Observatory [1].
Differentiated thyroid carcinoma (DTC), encompassing predominantly papillary and follicular subtypes, comprises many TC cases [2,3]. Standard treatment typically includes total thyroidectomy followed by radioiodine (RAI) ablation to remove residual thyroid tissue and treat microscopic disease. This approach is generally effective, with a 10-year survival rate exceeding 90% in localized disease [4].
However, approximately 5–15% of patients with DTC develop RAI-refractory (RAI-R) disease [5,6]. In these patients, tumors no longer concentrate iodine, making RAI therapy ineffective and leading to a markedly poorer prognosis, with a median overall survival (OS) of 2.5 to 3.5 years after the development of distant metastases [5,6].
For patients with slow tumor growth and low burden, active surveillance may be appropriate, with local interventions considered when progression is limited. In contrast, systemic therapy is warranted for rapid and/or symptomatic progression involving multiple lesions or organs. Multikinase inhibitors (MKIs) such as lenvatinib and sorafenib have been approved as first-line treatments for advanced, progressive RAI-R DTC by both the European Medicines Agency (EMA) and the United States Food and Drug Administration (FDA), based on the SELECT and DECISION phase III trials [7,8]. More recently, cabozantinib has emerged as a second-line option for patients who have progress on initial MKI therapy targeting the vascular endothelial growth factor (VEGF) receptor (COSMIC-311 trial) [9,10]. These targeted therapies have demonstrated efficacy in delaying disease progression and, in some cases, improving OS.
Advances in next-generation sequencing (NGS) have enabled the identification of genetic alterations in TC, significantly impacting patient management. Novel selective targeted therapies—including rearranged during transfection (RET) inhibitors (selpercatinib and pralsetinib) and neurotrophic tyrosine receptor kinase (NTRK) inhibitors (larotrectinib and entrectinib)—have further expanded the therapeutic options for selected RAI-R TC cases [11,12,13,14]. Additionally, immune checkpoint inhibitors (ICIs) have shown promising results and are still under investigation [15,16,17].
The optimal timing for initiating MKIs in patients with RAI-R DTC remains controversial. Although MKIs extend progression-free survival (PFS), they are not curative and are associated with significant toxicities that can negatively impact health-related quality of life (HRQoL) [7,18,19]. Consequently, close monitoring of disease activity is essential to determine the appropriate moment for systemic intervention.
Given the lack of a robust consensus on the optimal timing for MKI initiation, particularly in asymptomatic progressive disease, and the emergence of novel targeted therapies, this narrative review aims to synthesize current evidence and best practices to guide clinical decision-making in the management of RAI-R DTC.

2. Pathophysiology of RAI-R TC

Several genetic mutations, chromosomal rearrangements, and molecular alterations in various signaling pathways reduce iodine uptake in tumor cells, contributing to refractoriness in DTC. The sodium/iodide symporter (NIS), located on the basolateral membrane of thyroid follicular cells, is essential for actively transporting iodine necessary for synthesizing thyroxine (T4) and triiodothyronine (T3). This process also involves crucial molecules, including the pendrin transporter and thyroid peroxidase (TPO). TPO catalyzes the iodination of thyroglobulin (TG) to form monoiodotyrosine (MIT) and diiodotyrosine (DIT) and subsequently mediates the coupling of these iodinated tyrosines to produce T3 and T4 [20]. Alterations in the expression or subcellular localization of these proteins, particularly NIS, are considered the primary mechanisms underlying radioiodine refractoriness in DTC [21].

2.1. Alteration of Signaling Pathways

The Mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase/protein kinase Bα/mammalian target of rapamycin (PI3K/AKT/mTOR) pathways are central to TC development and progression, with mutations in genes such as BRAF, RAS, and PTEN playing key roles in RAI-R disease.
-
MAPK pathway:
This pathway plays a central role in thyroid cell differentiation and proliferation.
Its dysregulation triggers dedifferentiation in DTC, characterized by reduced expression of genes essential for thyroid hormone biosynthesis, including NIS, TPO, and TG. A pivotal event is decreased histone acetylation at the NIS gene promoter, which diminishes iodine uptake [22].
Chronic activation of both the MAPK and PI3K pathways leads to uncontrolled cell proliferation, impaired apoptosis, enhanced dedifferentiation, increased migration, and angiogenesis [23].
Abnormal MAPK activation in RAI-R DTC is most commonly driven by the BRAFV600E mutation; however, other BRAF mutations, rearrangements, RAS mutations, and MEK gene alterations also contribute [24,25].
-
PI3K/AKT pathway:
The PI3K pathway consists of three key molecules: PI3K, AKT, and mTOR. Its activation, alongside cAMP-independent signaling, suppresses the cAMP-dependent pathway responsible for thyroid-specific protein expression, including NIS [26]. Mutations in PIK3CA and alterations in PTEN further drive TC progression and RAI resistance [26]. Overactivation of this pathway is believed to enhance tumor cell survival and proliferation.
-
Transforming growth factor-β (TGF-β)/Smad pathway:
Abnormal TGF-β signaling, acting as a tumor promoter, is implicated in TC. In papillary TC (PTC), the BRAFV600E mutation upregulates functional TGF-β1, establishing an autocrine loop that increases Smad3 phosphorylation and levels. This, in turn, enhances NADPH oxidase 4 (NOX4) expression via a complex with p22phox, producing reactive oxygen species (ROS) that serve as second messengers to inhibit differentiation and promote proliferation and metastasis [27]. Thus, the TGF-β/Smad pathway is integrated to BRAFV600E-induced RAI-R DTC [28].
-
Wnt/β-catetin pathway:
The Wnt family of secreted glycoproteins activates β-catenin, a transcription factor critical for cell growth and differentiation. In TC, particularly those harboring BRAFV600E mutations, β-catenin is markedly elevated in cancer stem cells, supporting their self-renewal and contributing to chemotherapy resistance [29]. Abnormal β-catenin activation can also alter NIS localization, further contributing to RAI resistance [30].
-
Notch pathway:
The Notch receptor is a versatile transmembrane protein that influences cell differentiation, proliferation, and survival. In dedifferentiated TC, upregulation of Notch receptor expression helps restore differentiation by enhancing thyroid-specific gene expression like NIS and TPO and reducing tumor growth [31].
-
Thyroid-stimulating hormone receptor (TSHR) pathway:
Thyroid-stimulating hormone (TSH) binds to its receptor (TSHR), a member of the G protein-coupled receptor (GPCR), and activates signaling cascades that prove vital for thyroid cell growth and hormone synthesis [32]. In addition, the TSH-TSHR axis contributes to immune evasion. TSHα and TSHβ2, abundantly expressed in monocyte-derived dendritic cells (moDCs), upregulate immune checkpoint protein programmed death-ligand 1 (PD-L1) via the TSHR/AC/PKA/JNK/c-JUN pathway, a process that results in the inhibition of T cell-mediated immune responses against TC cells [33].

2.2. Tumor Genetic Profiling

-
BRAF mutation and rearrangement:
The most prevalent BRAF mutation in TC is BRAFV600E, which substitutes valine with glutamic acid at codon 60, leading to constitutive activation of the MAPK pathway [34]. This mutation is present in approximately 40–60% of PTCs and is associated with reduced NIS expression, increased tumor aggressiveness, higher recurrence rates, lymph node metastasis, and RAI refractoriness [34].
BRAF rearrangements, such as the BRAF-KIAA1549 fusion, are less common but can indicate a more aggressive disease course. Efanov et al. [35] studied the PTC tissues of 65 Ukrainian Americans affected by Chernobyl radiation using targeted NGS and RNA sequencing. Their findings revealed rearrangements in MACF-BRAF, MBP-BRAF, and POR-BRAF, which were identified as key mechanisms driving TC associated with radiation exposure from the Chernobyl disaster.
-
RAS mutation:
The RAS gene family comprises three key genes: HRAS, KRAS, and NRAS. These genes encode proteins that regulate cell signaling pathways essential for growth and differentiation. Activating mutations in these genes occur in approximately 20–30% of follicular TC (FTC) and 10-15% of PTC, and they appear less frequently in anaplastic TC (ATC). These mutations often disrupt critical downstream signaling pathways, including the MAPK/ERK and PI3K/AKT pathways, which play vital roles in cell proliferation and survival. Moreover, RAS mutations may interact with other genetic changes, such as BRAF mutations in PTC, thereby affecting tumor behavior and treatment responses [36]. Overall, RAS mutations associate with a more aggressive tumor phenotype.
-
RET rearrangement:
RET is a proto-oncogene that encodes a receptor tyrosine kinase. Its rearrangement, commonly seen in PTC, especially in RET/PTC1 and RET/PTC3 fusions, results in constitutive activation of RET signaling, promoting tumor growth and survival [37]. RET rearrangements are often associated with childhood radiation exposure, while germline RET mutations are a hallmark of medullary TC (MTC) and familial syndromes like Multiple Endocrine Neoplasia type 2 (MEN2).
-
Telomerase reverse transcriptase (TERT) promoter mutation:
Notably, the coexistence of BRAFV600E and TERT promoter mutations increases the likelihood of RAI refractoriness due to diminished iodine uptake [38].
The TERT gene is essential for telomere maintenance. Mutations in its promoter region create binding sites for ETS transcription factors, leading to telomerase reactivation and enabling limitless cell division [38]. TERT promoter mutations frequently co-occur with BRAFV600E, RAS, or TP53 mutations. Notably, the coexistence of BRAFV600E and TERT promoter mutations increases the likelihood of RAI refractoriness due to diminished iodine uptake [39].
-
NTRK fusion:
Fusions involving NTRK1, NTRK2, and NTRK3 result in chimeric proteins with constitutive kinase activity activating downstream signaling pathways such as MAPK and PI3K/AKT [14]. Patients harboring NTRK fusion exhibit more aggressive tumor behavior, with a higher risk of distant metastases and RAI refractoriness compared to those with RAS or BRAF mutations [40].
-
Anaplastic lymphoma kinase (ALK) mutation and rearrangement:
Although more commonly associated with lung cancer, ALK gene fusions or rearrangements have been identified in TC, particularly in more aggressive forms such as ATC and occasionally in PTC [41].
-
SWI/SNF (SWItch/Sucrose Non-Fermentable) complex mutation:
The SWI/SNF complex, which includes subunits like SMARCA4, SMARCB1, ARID1A, ARID1B, SMARCE1, and PBRM1, regulates chromatin remodeling and gene transcription. Mutations or deletions in these subunits, especially in ATC and poorly DTC, can promote BRAFV600E-driven tumorigenesis and can impair the efficacy of MAPK inhibitors and redifferentiation therapies [42].

2.3. Tumor Microenvironment

The tumor microenvironment in RAI-R TC is marked by increased angiogenesis and mechanisms of immune evasion. Tumor angiogenesis, primarily driven by VEGF signaling, not only promotes tumor growth but also metastasis, making VEGF a key target in the development of systemic treatment strategies.
Additionally, the combined impact of reduced iodine uptake and genetic alterations underscores the need for systemic therapies that can target these pathways and address both the tumor cells and their supportive microenvironment to effectively halt disease progression.

3. Current Systemic Therapeutic Options

Several systemic therapies have been developed for the treatment of RAI-R TC. Among these, tyrosine kinase inhibitors (TKIs) have emerged as the most widely used approach, demonstrating notable improvements in PFS and, in some studies, OS, albeit with significant treatment-related toxicity.

3.1. TKIs

TKIs are a class of targeted cancer therapies that competitively inhibit adenosine triphosphate (ATP) binding at the catalytic site of tyrosine kinases and serve as the cornerstone of systemic treatment for RAI-R TC. Landmark phase III clinical trials have led to the approved of lenvatinib, sorafenib, and cabozantinib for differentiated TC, resulting in significant improvements in PFS. Although these agents provide meaningful clinical benefits, their impact on OS remains modest, and their use carries substantial adverse effects.
-
Lenvatinib:
Lenvatinib is an oral TKI targeting VEGFR1-3, FGFR1-4, PDGFRβ, RET, and KIT. The SELECT phase III trial demonstrated that lenvatinib significantly prolonged PFS compared to placebo (18.3 months vs. 3.6 months, hazard ratio (HR) for progression or death 0.21, 99% CI 0.14–0.31, p < 0.00) in 392 patients with RAI-R DTC, treated in first or second line (with up to one prior VEGF- or VEGFR-targeted therapy) [7]. The objective response rate (ORR) was 65% and the median OS had not been reached. Furthermore, mature data revealed that lenvatinib responders experienced a markedly prolonged OS compared to non-responders (52.2 vs. 19.0 months, HR 0.32, 95% CI 0.23–0.46), and patients with a low (≤40 mm) baseline tumor burden had significantly longer OS than those with a high (>40 mm) tumor burden (median OS not reached vs. 29.1 months, HR 0.42, 95% CI 0.28–0.63) [43]. The most common any-grade treatment-related adverse events (AEs) (all grades) were hypertension (68%), diarrhea (59%), decreased appetite (50%), weight loss (46%), and nausea (41%). Of these common events, a subset were severe (grade 3–4): hypertension (41.8%), diarrhea (8%), decreased appetite (5.4%), weight loss (9.6%), and nausea (2.3%).
Dose reductions occurred in 78.5% of patients, and treatment discontinuation due to AEs was required in 14.2% of patients [44].
-
Sorafenib:
Sorafenib is an oral inhibitor of VEGFR-1, PDGFRβ, Raf kinases, BRAF, c-KIT, FLT-3, and CRAF. The DECISION phase III trial demonstrated that sorafenib significantly improved PFS compared to placebo (10.8 months vs. 5.8 months, HR 0.58, 95% CI 0.45–0.75, p < 0.0001) in 417 patients with RAI-R DTC [8]. The ORR was 12%, and stable disease for at least 6 months was achieved in 42%. The median OS did not differ significantly between groups. The most common any-grade treatment-related AEs included palmar–plantar erythrodysesthesia (76%), diarrhea (69%), alopecia (67%), rash/desquamation (50%), fatigue (50%), weight loss (47%), and hypertension (41%). Among these common events, only a subset were grade 3–4: palmar–plantar erythrodysesthesia (20%), diarrhea (6%), rash/desquamation (5%), fatigue (6%), weight loss (6%), and hypertension (10%). Dose reductions occurred in 66.2% of patients and treatment discontinuation due to AEs was required in 18.8% of patients [43].
-
Cabozantinib:
Cabozantinib is an oral TKI that primarily targets MET, VEGFR2, RET, c-KIT, FLT-3, and AXL [45,46]. In the phase III COSMIC-311 trial, cabozantinib demonstrated an improvement in PFS compared to placebo (median not reached vs. 1.9 months; p < 0.0001) in 258 patients with RAI-R DTC who had received prior treatment [9]. The ORR was 11%. In the mature data, despite 40 patients in the placebo group crossing over to cabozantinib, a trend toward improved OS was observed in the cabozantinib arm, with an HR of 0.76 (95% CI, 0.45–1.31) [47]. The most common treatment-related AEs of any grade were diarrhea (62%), palmar–plantar erythrodysesthesia (47%), and hypertension (32%). Of these, grade 3-4 were as follows: diarrhea (8%), palmar–plantar erythrodysesthesia (10%), and hypertension (11%) [46,48].
In addition, phase III trials in MTC demonstrated a significantly PFS benefit compared to placebo (11.2 vs. 4.0 months) [45,49].
-
Vandetanib
Vandetanib is a TKI that targets RET, VEGFR, and EGFR, approved by the FDA in 2011 and EMA in 2012 for treating advanced MTC. Clinical trials have shown that vandetanib significantly improves PFS in MTC patients [50,51]. However, its impact on OS remains inconclusive [52]. Vandetanib is generally well tolerated, but can cause AEs such as fatigue, hypertension, QTc prolongation, and diarrhea [53,54]. The drug’s efficacy may be limited by the development of resistance over time [55]. Ongoing research is exploring vandetanib’s long-term efficacy, potential combinations with other agents, and strategies to overcome resistance [52,55]. Despite its limitations, vandetanib represents a significant advancement in the treatment of advanced MTC.
-
TKIs with evidence from phase II clinical trials:
Entrectinib is an oral TKI targeting NTRK, ROS1, and ALK fusions, approved by the FDA and EMA for ROS1-positive non-small cell lung cancer and NTRK fusion-positive solid tumors, including TC, in patients ≥ 12 years [56,57,58]. It has shown efficacy in NTRK-rearranged TC, which typically exhibits aggressive clinical behavior and a high metastatic risk [59]. In a case report, entrectinib demonstrated a response in a patient with metastatic PTC harboring an EZR-ROS1 fusion [60]. Clinical trials have reported durable responses and intracranial activity [61], with a favorable safety profile marked primarily by low-grade AEs [60]. Entrectinib offers a promising option for NTRK fusion-positive TC, for which no approved drugs were previously available [62,63].
Larotrectinib is a selective inhibitor of tropomyosin receptor kinases (TRKs), specifically TRKA, TRKB, and TRKC, encoded by NTRK1-3. Its mechanism of action involves blocking downstream signaling pathways such as RAS/MAPK, PI3K/AKT, and PLCγ, thereby inhibiting tumor proliferation, survival, and migration.
Approved by both the FDA and EMA, larotrectinib has reported high ORRs (71% to 90%) in DTC patients [64], with rapid and durable responses [65], and a favorable safety profile, with mostly grade 1–2 AEs [66]. The treatment has shown potential for improved long-term outcomes compared to standard care in metastatic DTC [67]. Interestingly, larotrectinib may exert a redifferentiating effect in some RAI-R TC, potentially restoring iodine uptake [68]. These findings support routine testing for NTRK gene fusions in advanced non-MTC patients before initiating systemic therapy [64].
Axitinib, a selective inhibitor of VEGFRs, demonstrated antitumor activity across all histologic TC subtypes in phase II trials, with ORRs of 30–38% and median PFS of 15–18.1 months [69]. A compassionate use program in Spain reported superior outcomes with first-line axitinib compared to its use in second-line settings [70].
Pharmacokinetic/pharmacodynamic analyses further suggest that higher axitinib plasma levels correlate with greater tumor size reduction [69].
The efficacy of sunitinib is attributed to its multi-targeted inhibition of VEGFR, PDGF-R, RET, c-KIT, FLT3, and receptor of macrophage colony-stimulating factor (CSF1R) [71]. Clinical studies have reported partial response rates ranging from 13% to 55.5% in DTC and 0% to 55% in MTC [71,72]. Notably, sunitinib is associated with a high incidence of hypothyroidism, possibly due to its inhibition effect on TPO activity [73].
Pazopanib, an MKI targeting VEGFR and PDGFR, has shown promising activity in DTC, with a 49% ORR [74].
Mechanisms of action for these drugs are summarized in Table 1.

3.2. Immune Checkpoint Inhibitors (ICIs)

The role of immunotherapy in RAI-R TC is still evolving, with several ongoing clinical trials investigating its potential. ICIs, such as pembrolizumab (anti-PD-1), may offer benefit in advanced TC, particularly when combined with TKIs.
-
Pembrolizumab:
In advanced DTC, pembrolizumab monotherapy has shown limited efficacy, with an ORR of 9% [82]. In ATC, pembrolizumab combined with chemoradiotherapy as initial treatment yielded disappointing survival outcomes [83]. However, adding pembrolizumab to kinase inhibitors at disease progression has shown potential benefit in some ATC patients [84,85]. Notably, the combination of lenvatinib and pembrolizumab demonstrated promising results in ATC and poorly DTC, with a median PFS of 17.75 months and some long-term remissions [17]. In patients with RAI-R DTC progressing on lenvatinib, the addition of pembrolizumab resulted in a 15% partial response rate and a median PFS of 12.6 months [86]. These findings suggest that pembrolizumab, particularly in combination regimens, may have a role in advanced TC treatment.

3.3. Chemotherapy

Traditional chemotherapy has limited efficacy in TC and is generally reserved for patients who have exhausted other treatment options. Agents such as doxorubicin were used historically, but their role in current treatment protocols is minimal due to the advent of more effective targeted therapies.

3.4. Other Novel Therapies

-
RET Inhibitors:
Targeted therapies against RET fusions, such as selpercatinib (LOXO-292) and pralsetinib (BLU-667), have shown remarkable efficacy in RET-altered TCs.
The LIBRETTO-001 trial demonstrated durable responses in RET fusion-positive TC, with ORRs of 95.8% in treatment-naïve patients and 85.4% in pre-treated patients [87]. In RET-mutant MTC, ORRs were 81.0% in patients naïve to cabozantinib/vandetanib and 73.5% in pre-treated patients [88]. Selpercatinib exhibited a consistent safety profile, with common AEs including hypertension and increased liver enzymes [89]. Based on these results, selpercatinib has received FDA and EMA approval for treating RET-altered TC and lung cancers [90,91].
Similarly, pralsetinib has shown high ORRs (73.1–90.9%) and durable responses, with median PFS of 25.4–25.8 months across various cohorts in the ARROW study [81,92]. Consequently, the FDA granted accelerated approval for pralsetinib for TC harboring RET mutations or fusions, although its label included warnings for AEs such as pneumonitis, hypertension, and hepatotoxicity [93].
-
BRAF Inhibitors:
In patients with BRAFV600E-mutant RAI-R TC, targeted therapy with the BRAF inhibitor dabrafenib in combination with the MEK inhibitor trametinib has demonstrated promising efficacy, especially in ATC. Phase II trials have reported high ORR ranging from 56% to 73.1% in both ATC and PTC [94,95,96]. This combination has been associated with improved long-term survival in ATC patients, with 12-month OS rates reported between 51.7% and 80% [94,95]. Real-world data support these findings, showing median OS of 6.9 months in ATC patients [97]. Additionally, the combination therapy may induce redifferentiation in poorly DTC [98]. However, dose reductions due to AEs are common, and some studies suggest that the combination may not offer significant advantages over dabrafenib monotherapy in RAI-R TC [96,99].
Neoadjuvant BRAF-directed therapy followed by surgery has also been explored in stage IVb ATC, showing improved survival (12-month OS of 93.6% and PFS of 84.4%), reduced tumor size, and lower surgical morbidity, though outcomes were poorer in patients with residual disease post-surgery; these promising findings require validation in prospective trials [100].
Preclinical studies have further highlighted the potential of combining targeted agents. Trametinib, alone or with pazopanib, effectively inhibited TC growth in cell lines and xenograft models [101]. A phase I trial of pazopanib and trametinib in DTC showed a 33% ORR and median PFS of 10.7 months [102]. Intermittent dosing of dabrafenib and trametinib in BRAFV600E-mutated PTC also produced encouraging responses in case reports [103]. Although pazopanib monotherapy showed a 14.3% partial response rate and a 9.4-month median PFS in DTC [104], its single-agent activity in ATC remains limited despite preclinical promise [74]. Adding pembrolizumab to these regimens may further enhance survival outcomes [105].
Together, these studies underscore the potential of novel targeted therapies in various TC subtypes, warranting further investigation in prospective clinical trials.

4. Clinical Indications for Systemic Therapy

Systemic therapy should be initiated in patients with progressive or symptomatic RAI-R TC, as well as those with high-risk metastatic disease. Current guidelines from the American Thyroid Association (ATA) and European Society for Medical Oncology (ESMO) offer clear criteria for patient selection.

4.1. Criteria for Determining Refractoriness (RAI-R) [106]

-
Tumors that show no RAI uptake in all local or distant lesions on a post-treatment whole-body scan, despite being DTC.
-
Partial loss of RAI uptake, with some lesions failing to concentrate RAI.
-
Progressive disease within 6–12 months, despite receiving adequate doses of RAI, as defined by RECIST criteria.
-
Patients who have received a cumulative RAI dose (typically >600 mCi) without a significant response.
-
Locally advanced thyroid tumors in which surgical resection is not feasible, precluding proper assessment of RAI uptake.

4.2. Criteria for Determining Advanced/Aggressive Forms

According to the framework developed by the American Head and Neck Society (AHNS) and the International Thyroid Oncology Group, advanced TC is defined by several dimensions [107]:
  • Structural/surgical category:
    -
    Invasive or inoperable locoregional disease.
    -
    Recurrence of the disease.
    -
    Presence of distant metastases.
    -
    Rapid progression detectable on imaging studies.
  • Biochemical category:
    -
    Tumors resistant to RAI treatment.
    -
    Tumors unresponsive to TSH suppression therapy.
    -
    Rapid doubling time of specific biomarkers (e.g., TG).
  • Histologic/molecular category:
    -
    Aggressive histologic variants (e.g., poorly DTC or ATC).
    -
    High Ki67 proliferation index.
    -
    Elevated mitotic count.
  • Clinical judgment:
Physicians may also designate cases as aggressive based on observed clinical behavior, even if all criteria are not met.
It is important to note that a recent consortium of experts from the ATA, ETA, the European Association of Nuclear Medicine, and the Society of Nuclear Medicine and Molecular Imaging has highlighted that no single definition, classification, criterion, or clinical scenario serves as an absolute indicator of RAI-R DTC [108].

4.3. Patient Selection

In RAI-R TC, the decision to initiate systemic therapy should be made by specialized endocrinologists or oncologists, considering several factors, such as disease progression, tumor burden, symptoms, and patient-specific considerations [109].
Molecular testing has become an invaluable tool in the diagnosis and management of TC. Advanced techniques, like targeted NGS panels (e.g., ThyroSeq), can accurately detect mutations in multiple cancer-related genes with high accuracy and sensitivity, while requiring minimal DNA input [110]. This is particularly useful in cases with indeterminate cytology, where the presence of mutations correlates with a higher risk of malignancy [110]. By complementing fine-needle aspiration cytology, molecular testing enhances diagnostic accuracy and guides clinical management [111,112]. Additionally, molecular profiling is essential for characterizing advanced TC and developing targeted therapies [107].
Genomic testing should be considered for patients with advanced or metastatic RAI-R DTC under specific conditions: (a) the patient has good performance status, (b) systemic treatment is planned, (c) first-line MKI therapy has failed, and (d) access to targeted therapies is feasible [107]. Molecular analysis may be performed at various stages of RAI-R disease progression, at diagnosis, if possible, upon structural progression (as defined by RECIST criteria), or after progression following one or two lines of MKI treatment [109].
Further research is needed to optimize treatment sequencing and address potential drug resistance [113].

5. Timing of Systemic Therapy

Various guidelines recommend systemic therapies for patients with progressive, symptomatic, or metastatic RAI-R TC, and address the timing for starting MKIs. For instance, NCCN guidelines recommend considering MKIs for patients with rapidly growing or symptomatic lesions [114]. The ATA guidelines suggest MKI administration for patients with life-threatening lesions, diffuse disease progression, or symptomatic disease [115], while ESMO guidelines advise their use in patients with multiple symptomatic lesions or asymptomatic patients with progressive multifocal disease [116]. The ETA emphasizes the importance of patient-related factors (age, health status, comorbidities, and contraindications) and patient preferences [117]. Similarly, the Japan Association of Endocrine Surgeons’ guidelines indicate that MKIs should not be started unconditionally in all patients with rapid tumor growth or symptoms; instead, the timing should balance potential benefits against risks in the context of the patient’s overall condition [118]. Indeed, patients exhibiting asymptomatic, stable disease can be closely monitored without immediate intervention, especially in cases of low tumor burden [118]. The optimal sequence of MKIs remains undetermined due to limited evidence from subgroup analyses of phase III trials.
The decision to initiate systemic therapy is critical. While early intervention may prevent disease complications, it must be balanced against the risks of long-term toxicity, particularly in patients with slow-growing tumors (Figure 1, Table 2).

5.1. Early vs. Late Intervention

-
Early intervention:
Some experts advocate for the early initiation of systemic therapy in metastatic disease to prevent complications such as bone fractures or organ failure. However, this approach may expose patients to treatment-related side effects for an extended period [7,8].
-
Late intervention:
Delaying systemic therapy until symptomatic progression is another approach, particularly for patients with indolent disease. This allows patients to avoid toxicities until the disease becomes more aggressive [115,119].
In a global non-interventional study by Brose et al. [120], the median time to symptomatic progression in asymptomatic RAI-R DTC patients treated with MKIs was 55.4 months overall, 55.4 months in the cohort that initiated MKI treatment at study entry, and 51.4 months in the cohort that did not initiate MKI treatment at study entry. Median PFS from the start of MKI therapy was 19.2 months and from the start of sorafenib therapy 16.7 months, while median OS from RAI-R classification was 167 months. For sorafenib-treated patients, 70% required dose modifications, and 89% experienced treatment-emergent AEs. The study provides valuable real-world insights into outcomes for patients with asymptomatic, progressive RAI-R DTC under observation or MKI treatment in the current era.

5.2. Active Surveillance

Although some patients are refractory to RAI therapy, they may maintain stable metastatic disease for several years without additional treatment [121]. The therapeutic approach is primarily guided by tumor burden and the rate of disease progression. For lesions < 1–2 cm, active surveillance is generally recommended [122]. Studies have shown that small metastatic lymph nodes (<1 cm) and thyroid bed nodules can be safely monitored over long periods using ultrasound [123,124,125]. Similarly, small soft tissue metastases can be followed with cross-sectional imaging. Depending on their location, stable or very slowly progressing lesions (i.e., without significant growth over 12–16 months) may also be managed conservatively, except when they are near critical structures, where even minimal growth could cause symptoms [126].

5.3. Integration with Local Treatments

-
Surgery
For patients with oligometastatic disease (≤5 metastatic lesions), surgical resection may be effectively combined with systemic therapy. In DTC patients with extracervical metastases, a multifaceted approach is essential for accurate diagnosis, treatment, and monitoring. While the initial goal is cure, especially for small, radioiodine-avid pulmonary metastases, the focus shifts to improving survival and managing symptoms in persistent disease. Asymptomatic, stable RAI-R metastases may be managed with active surveillance, whereas large or painful lesions often require directed therapies such as external beam radiotherapy (EBRT), surgery, or embolization [127]. Multimodal imaging (CT, MRI, bone scans, and 18FDG-PET) plays a key role in guiding treatment decisions.
-
Radiation therapy
EBRT is an important adjunct in the management of TC, particularly for high-risk patients with bone or brain metastases, when used in conjunction with systemic therapies. In DTC, EBRT can improve local control in patients over 45 with microscopic residual disease or extensive extrathyroid invasion [128]. It is also recommended for gross residual or unresectable disease [129]. In MTC, EBRT may benefit patients with surgically inaccessible or residual tumors [130]. For ATC, a combination of surgery, EBRT, and chemotherapy can yield higher local control rates and prolonged survival [131]. Advances in EBRT techniques, such as intensity-modulated radiation therapy, can improve treatment accuracy and reduce toxicity [132]. However, the impact of EBRT on OS remains uncertain, and its routine use is still debated in the absence of randomized controlled studies [133,134].

6. AEs and Toxicity Management

Systemic therapies, especially TKIs, have improved outcomes in advanced TC, but are associated with numerous AEs, particularly during the initial phases of therapy. Common AEs affect the cardiovascular, gastrointestinal, dermatological, and metabolic systems, with over 90% of patients experiencing at least one AE [135]. Hypothyroidism is the most frequently observed endocrine toxicity [136]. Furthermore, TKIs significantly increase the risk of arterial thromboembolic events, although venous events are not similarly affected [137]. Early assessment and management of AEs are crucial to mitigate their impact on QoL and to avoid treatment discontinuation [138].
In a prospective real-world study involving 20 patients treated with lenvatinib, 100% experienced at least one AE, and 80% had grade ≥ 3 toxicity. The most commonly self-reported symptoms included fatigue, anorexia, abdominal pain, xerostomia, and gastrointestinal disturbances, all of which significantly interfered with daily activities. HRQoL, assessed using the EQ-5D-3L and EQ-VAS questionnaires, showed a statistically significant decline at 3 and 6 months of treatment (p < 0.05), followed by partial recovery at 12 months, possibly due to dose adjustments and proactive symptom management [139]. These findings were complemented by the phase II randomized, double-blind Study 211, which compared HRQoL in patients with RAI-R DTC initiating lenvatinib at either 18 mg/day or the approved 24 mg/day starting dose. No statistically significant differences were found between the two arms in longitudinal changes in EQ-5D-3L or FACT-G scores. Similarly, time to deterioration of HRQoL was comparable (16–28 weeks), and no clinical benefit was observed with the lower starting dose [140]. Both studies identified a trend toward better HRQoL preservation in patients who achieved objective tumor responses, suggesting that disease control may positively influence overall well-being.
Despite the high frequency of AEs, QoL can be maintained—or even restored—through appropriate toxicity management and close monitoring. The integration of patient-reported outcome measures allows clinicians to detect symptoms that might be overlooked during routine assessments and supports a more patient-centered approach to care. A multidisciplinary approach, including dedicated endocrinologists, is recommended for optimal management of TKI-related AEs [141,142]. Regular monitoring of thyroid function is essential throughout treatment [143]. In addition, prehabilitation measures prior to initiating TKIs—particularly lenvatinib—may enhance patient resilience and reduce toxicity. Such measures include optimizing fluid and electrolyte balance, adjusting dietary intake, and implementing appropriate skin care protocols. For patients with pre-existing hypertension, intensifying antihypertensive treatment and optimizing blood pressure control are also recommended [144]. These strategies aim to minimize the severity of AEs and support better treatment tolerance and overall outcomes.

6.1. Common AEs

Over 90% of patients receiving TKIs experience at least one AE, with a higher incidence in the first 6–8 weeks of treatment. Frequently affected systems include dermatological, gastrointestinal, cardiovascular, and metabolic, potentially compromising patient HRQoL. For lenvatinib, the most common AEs are hypertension (67%), fatigue (66%), diarrhea (59%), and decreased appetite (50%) [7]. Serious events, such as proteinuria and thromboembolic events, occur in a minority of patients. Common AEs from sorafenib include hand–foot syndrome (76%), rash (50%), and alopecia (67%) [44].
In a study of 29 patients on MKIs, 17.2% experienced rare AEs [145]. These included heart failure, thrombocytopenia, and squamous cell carcinoma during sorafenib treatment, as well as squamous cell carcinoma and oophoritis with intestinal perforation during vandetanib therapy. The study followed patients for a mean of 13.7 months and included individuals with DTC and MTC. While most MKI-related adverse effects are manageable, the authors emphasize the importance of being aware of these rare complications.

6.2. Toxicity Management

Dose reductions or temporary treatment discontinuation may be necessary in cases of severe toxicities, particularly in older or frail patients.
Management strategies for common AEs include the following:
-
Fatigue/asthenia
Thyroid function should be assessed before starting lenvatinib and monitored monthly, with adjustments to thyroid-replacement therapy as needed. If no other cause is found, supportive care, such as proper nutrition, exercise, and stress reduction techniques, is recommended. For moderate to severe fatigue, consider temporary dose interruptions of MKIs.
-
Hypertension
MKI-induced hypertension is thought to result from decreased nitric oxide production resulting from VEGF inhibition. Hypertension caused by antiangiogenic therapies is believed to reflect the agent’s intended pharmacologic activity. This AE, which may correlate with prolonged PFS, requires proactive management with antihypertensive medications to mitigate risks [146].
-
Proteinuria
Before initiating MKIs, baseline urinalysis and protein-to-creatinine ratio assessments are recommended. Decisions regarding dose modification or treatment interruption should be individualized, as proteinuria is a class effect, and switching agents may not be beneficial.
-
Gastrointestinal AEs
Management includes dietary modifications (avoiding foods that worsen diarrhea and promoting stool consistency), dehydration control, use of loperamide, and dose adjustments for severe AEs (grades 3 or 4). Once symptoms subside, optimal dosing may be resumed.
-
Stomatitis/mucositis
Patients should avoid irritants such as mint-flavored toothpaste, alcohol-based mouthwashes, and spicy or acidic foods. Ongoing clinical trials are evaluating steroid mouthwashes, which may reduce the incidence and severity of stomatitis in patients on lenvatinib. Topical treatment (lidocaine or steroid ointment) can provide relief. For severe stomatitis (grade ≥ 3), dose modifications are recommended [147].
-
Palmar–plantar erythrodysesthesia syndrome (PPES):
Patients should be advised to minimize exposure of hands and feet to hot water, avoid tight footwear, and limit vigorous exercise to prevent PPES. This condition typically develops within the first 2–4 weeks of TKI therapy, so avoiding trauma and ensuring adequate rest during this period is crucial. Dose reduction or interruption may be required until symptoms return to grade ≤ 1 [148].
-
Hemorrhagic events
Mechanisms may include blood vessel destabilization, vascular integrity loss, and thrombocytopenia. MKIs should be withheld in patients with grade 3 hemorrhage until resolution to grade 0 or 1. Treatment can resume at a reduced dose, depending on the event’s severity. For grade 4 hemorrhages, MKI should be permanently discontinued [146].
-
Hepatotoxicity
TKIs can induce liver injury through mitochondrial toxicity, inhibition of glycolysis, and formation of reactive metabolites, ranging from mild elevation of liver enzymes to severe liver damage [149,150]. Management strategies include close monitoring of liver function tests, dose adjustments, and, in some cases, corticosteroid use or discontinuation of the drug are essential.
Early risk assessment and prompt AE identification are crucial for optimal patient care. A multidisciplinary team, including nursing support, plays a vital role in preventing and managing these side effects to maximize treatment benefits and minimize risks. Effective AE management helps avoid drug discontinuation or interruption that could potentially accelerate disease progression. Based on the tumor’s molecular characteristics and local regulatory framework, a clinician may consider switching to second-line treatment, such as cabozantinib or newer targeted therapies, when necessary.
By supporting patients throughout their clinical-therapeutic journey, healthcare professionals can improve treatment outcomes and enhance patients’ QoL.

7. Challenges, Practical Implications, and Future Directions

7.1. Challenges and Limitations of Emerging Therapies

Despite the promising results of ICIs and novel targeted therapies in RAI-R TC, several challenges remain. ICIs, particularly pembrolizumab, have shown limited monotherapy efficacy in DTC, and their clinical benefit often requires combination regimens, which can increase toxicity and complicate management [17,82,83,84,85,86]. Moreover, access to ICIs and next-generation targeted therapies is uneven globally, largely due to high costs and the need for specialized molecular testing to identify actionable mutations (e.g., RET and NTRK fusions). Even in high-income countries, financial toxicity can be substantial, impacting both health care systems and individual patients.
Furthermore, many emerging agents (e.g., RET inhibitors, NTRK inhibitors, and novel BRAF/MEK combinations) have primarily been evaluated in small, selected patient populations or in early-phase trials, limiting the generalizability of the results. Long-term data on survival benefit, QoL, and resistance patterns are still lacking. These limitations underscore the need for continued research and real-world studies to better define the optimal use and sequencing of these therapies.

7.2. Practical Recommendations for Clinicians and Researchers

To maximize the clinical impact of current evidence, we suggest several actionable steps:
-
Routine molecular profiling: All patients with progressive RAI-R TC should undergo comprehensive molecular testing early in the disease course to identify potential targets for therapy.
-
Multidisciplinary evaluation: Treatment decisions should involve tumor boards including endocrinologists, oncologists, nuclear medicine specialists, and molecular pathologists to ensure individualized care.
-
Cost-effectiveness analysis: Institutions should prioritize the implementation of cost-effectiveness assessments when introducing new targeted therapies, especially in resource-constrained settings.
-
Proactive toxicity management: Early recognition and intervention for treatment-related AEs can prolong therapy duration and maintain QoL.
-
Participation in clinical trials: Clinicians should encourage eligible patients to enroll in ongoing studies evaluating novel agents or treatment sequences to help build robust evidence.

7.3. Integration of Artificial Intelligence (AI) in Clinical Practice

The integration of AI and machine learning technologies offers exciting opportunities to enhance clinical decision-making in RAI-R TC. AI-based algorithms can assist with the following:
-
Molecular profiling: AI tools can rapidly analyze NGS data, identifying actionable mutations and predicting potential resistance mechanisms.
-
Treatment selection: Predictive models based on real-world data could support clinicians in selecting the most appropriate systemic therapy based on patient and tumor characteristics.
-
Monitoring and toxicity prediction: AI-driven digital platforms, including mobile health apps and wearable devices, can enable real-time monitoring of symptoms and early detection of AEs.
-
Clinical trial matching: Automated systems can help match patients to appropriate clinical trials based on their molecular profiles and clinical parameters, facilitating access to emerging therapies.
The implementation of AI must be accompanied by rigorous validation, transparency, and careful integration into clinical workflows to ensure its effectiveness and acceptance among healthcare providers.

8. Conclusions

The management of RAI-R DTC remains a complex clinical challenge that requires balancing tumor progression, treatment-related toxicity, and HRQoL. The introduction of MKIs such as lenvatinib and sorafenib has significantly improved outcomes in advanced cases, although their AEs necessitate careful monitoring and dose adjustments. At the same time, emerging targeted therapies, including RET and NTRK inhibitors, are expanding treatment options, particularly for patients with actionable genetic alterations, highlighting the potential of precision medicine.
To optimize patient outcomes, clinicians should prioritize molecular profiling at diagnosis or upon disease progression to identify actionable mutations. Early and proactive management of AEs, particularly hypertension, fatigue, and gastrointestinal toxicity, is essential to minimize treatment discontinuation. In selected cases, particularly those with low tumor burden and indolent progression, active surveillance may be considered to delay systemic therapy while maintaining disease control.
Recent advances and challenges in the field—including the accessibility and cost implications of novel therapies, the need for practical guidance for clinicians, and the integration of AI into molecular profiling and treatment selection—offer new opportunities for improving patient care. Addressing these aspects is essential to further personalize therapy, enhance decision-making, and overcome current limitations.
From a research perspective, ongoing clinical trials should focus on defining the optimal sequencing of systemic therapies, addressing drug resistance mechanisms, and improving the risk–benefit ratio of treatment strategies. Future efforts should also explore how digital innovations can support clinical practice, ultimately enhancing QoL and outcomes for patients with RAI-R DTC.

Author Contributions

Conceptualization, T.D.V. and M.D.-P.; methodology, T.D.V. and B.M.-A.M.; software, I.G.T., L.M.G. and P.P.M.; validation, P.J.-F., P.P.M., A.G.M.-N. and M.D.-P.; formal analysis, T.D.V., B.M.-A.M. and P.J.-F.; investigation, T.D.V., L.M.G. and P.J.-F.; resources, T.D.V.; data curation, T.D.V. and P.J.-F.; writing—original draft preparation, T.D.V.; writing—review and editing, T.D.V., B.M.-A.M. and P.J.-F.; visualization, P.J.-F., P.P.M., A.G.M.-N. and M.D.-P.; supervision, P.J.-F., P.P.M., A.G.M.-N. and M.D.-P.; project administration, M.D.-P.; funding acquisition, M.D.-P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization International Agency for Research on Cancer (IARC). GLOBOCAN: Estimated Cancer Incidence, Mortality and Prevalence Worlwide. Available online: http://gco.iarc.fr/en (accessed on 8 February 2024).
  2. Cabanillas, M.E.; McFadden, D.G.; Durante, C. Thyroid cancer. Lancet 2016, 388, 2783–2795. [Google Scholar] [CrossRef] [PubMed]
  3. La Vecchia, C.; Negri, E. Thyroid cancer: The thyroid cancer epidemic—Overdiagnosis or a real increase? Nat. Rev. Endocrinol. 2017, 13, 318–319. [Google Scholar] [CrossRef] [PubMed]
  4. Sciuto, R.; Romano, L.; Rea, S.; Marandino, F.; Sperduti, I.; Maini, C.L. Natural history and clinical outcome of differentiated thyroid carcinoma: A retrospective analysis of 1503 patients treated at a single institution. Ann. Oncol. 2009, 20, 1728–1735. [Google Scholar] [CrossRef] [PubMed]
  5. Durante, C.; Haddy, N.; Baudin, E.; Leboulleux, S.; Hartl, D.; Travagli, J.P.; Caillou, B.; Ricard, M.; Lumbroso, J.D.; De Vathaire, F.; 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, 2892–2899. [Google Scholar] [CrossRef]
  6. Lirov, R.; Worden, F.P.; Cohen, M.S. The Treatment of Advanced Thyroid Cancer in the Age of Novel Targeted Therapies. Drugs 2017, 77, 733–745. [Google Scholar] [CrossRef]
  7. Schlumberger, M.; Tahara, M.; Wirth, L.J.; Robinson, B.; Brose, M.S.; Elisei, R.; Habra, M.A.; Newbold, K.; Shah, M.H.; Hoff, A.O.; et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N. Engl. J. Med. 2015, 372, 621–630. [Google Scholar] [CrossRef]
  8. Brose, M.S.; Nutting, C.M.; Jarzab, B.; Elisei, R.; Siena, S.; Bastholt, L.; de la Fouchardiere, C.; Pacini, F.; Paschke, R.; Shong, Y.K.; et al. Sorafenib in radioactive iodine-refractory, locally advanced or metastatic differentiated thyroid cancer: A randomised, double-blind, phase 3 trial. Lancet 2014, 384, 319–328. [Google Scholar] [CrossRef]
  9. Brose, M.S.; Robinson, B.; Sherman, S.I.; Krajewska, J.; Lin, C.C.; Vaisman, F.; Hoff, A.O.; Hitre, E.; Bowles, D.W.; Hernando, J.; et al. Cabozantinib for radioiodine-refractory differentiated thyroid cancer (COSMIC-311): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2021, 22, 1126–1138. [Google Scholar] [CrossRef]
  10. Gild, M.L.; Tsang, V.H.M.; Clifton-Bligh, R.J.; Robinson, B.G. Multikinase inhibitors in thyroid cancer: Timing of targeted therapy. Nat. Rev. Endocrinol. 2021, 17, 225–234. [Google Scholar] [CrossRef]
  11. Wirth, L.J.; Sherman, E.; Robinson, B.; Solomon, B.; Kang, H.; Lorch, J.; Worden, F.; Brose, M.; Patel, J.; Leboulleux, S.; et al. Efficacy of Selpercatinib in RET-Altered Thyroid Cancers. N. Engl. J. Med. 2020, 383, 825–835. [Google Scholar] [CrossRef]
  12. Subbiah, V.; Hu, M.I.; Wirth, L.J.; Schuler, M.; Mansfield, A.S.; Curigliano, G.; Brose, M.S.; Zhu, V.W.; Leboulleux, S.; Bowles, D.W.; et al. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): A multi-cohort, open-label, registrational, phase 1/2 study. Lancet Diabetes Endocrinol. 2021, 9, 491–501. [Google Scholar] [CrossRef] [PubMed]
  13. Huang, F.W.; Feng, F.Y. A Tumor-Agnostic NTRK (TRK) Inhibitor. Cell 2019, 177, 8. [Google Scholar] [CrossRef] [PubMed]
  14. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747. [Google Scholar] [CrossRef] [PubMed]
  15. Cunha, L.L.; Ward, L.S. Translating the immune microenvironment of thyroid cancer into clinical practice. Endocr. Relat. Cancer 2022, 29, R67–R83. [Google Scholar] [CrossRef]
  16. Oh, D.Y.; Algazi, A.; Capdevila, J.; Longo, F.; Miller WJr Chun Bing, J.T.; Bonilla, C.E.; Chung, H.C.; Guren, T.K.; Lin, C.C.; Motola-Kuba, D.; et al. Efficacy and safety of pembrolizumab monotherapy in patients with advanced thyroid cancer in the phase 2 KEYNOTE-158 study. Cancer 2023, 129, 1195–1204. [Google Scholar] [CrossRef]
  17. Dierks, C.; Seufert, J.; Aumann, K.; Ruf, J.; Klein, C.; Kiefer, S.; Rassner, M.; Boerries, M.; Zielke, A.; la Rosee, P.; et al. Combination of Lenvatinib and Pembrolizumab Is an Effective Treatment Option for Anaplastic and Poorly Differentiated Thyroid Carcinoma. Thyroid 2021, 31, 1076–1085. [Google Scholar] [CrossRef]
  18. Reed, N.; Glen, H.; Gerrard, G.; Good, J.; Lei, M.; Lyon, A.R.; Strachan, M.; Wadsley, J.; Newbold, K. Expert Consensus on the Management of Adverse Events During Treatment with Lenvatinib for Thyroid Cancer. Clin. Oncol. (R. Coll. Radiol.) 2020, 32, e145–e153. [Google Scholar] [CrossRef]
  19. Haddad, R.I.; Schlumberger, M.; Wirth, L.J.; Sherman, E.J.; Shah, M.H.; Robinson, B.; Dutcus, C.E.; Teng, A.; Gianoukakis, A.G.; Sherman, S.I. Incidence and timing of common adverse events in Lenvatinib-treated patients from the SELECT trial and their association with survival outcomes. Endocrine 2017, 56, 121–128. [Google Scholar] [CrossRef]
  20. Xing, M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat. Rev. Cancer 2013, 13, 184–199. [Google Scholar] [CrossRef]
  21. Castro, M.R.; Bergert, E.R.; Goellner, J.R.; Hay, I.D.; Morris, J.C. Immunohistochemical analysis of sodium iodide symporter expression in metastatic differentiated thyroid cancer: Correlation with radioiodine uptake. J. Clin. Endocrinol. Metab. 2001, 86, 5627–5632. [Google Scholar] [CrossRef]
  22. Zhang, Z.; Liu, D.; Murugan, A.K.; Liu, Z.; Xing, M. Histone deacetylation of NIS promoter underlies BRAF V600E-promoted NIS silencing in thyroid cancer. Endocr. Relat. Cancer 2014, 21, 161–173. [Google Scholar] [CrossRef] [PubMed]
  23. Romitti, M.; Ceolin, L.; Siqueira, D.R.; Ferreira, C.V.; Wajner, S.M.; Maia, A.L. Signaling pathways in follicular cell-derived thyroid carcinomas (review). Int. J. Oncol. 2013, 42, 19–28. [Google Scholar] [CrossRef] [PubMed]
  24. Nagarajah, J.; Le, M.; Knauf, J.A.; Ferrandino, G.; Montero-Conde, C.; Pillarsetty, N.; Bolaender, A.; Irwin, C.; Krishnamoorthy, G.P.; Saqcena, M.; et al. Sustained ERK inhibition maximizes responses of BrafV600E thyroid cancers to radioiodine. J. Clin. Investig. 2016, 126, 4119–4124. [Google Scholar] [CrossRef] [PubMed]
  25. Borrelli, N.; Panebianco, F.; Condello, V.; Barletta, J.A.; Kaya, C.; Yip, L.; Nikiforova, M.N.; Nikiforov, Y.E. Characterization of Activating Mutations of the MEK1 Gene in Papillary Thyroid Carcinomas. Thyroid 2019, 29, 1279–1285. [Google Scholar] [CrossRef] [PubMed]
  26. Shen, H.; Zhu, R.; Liu, Y.; Hong, Y.; Ge, J.; Xuan, J.; Niu, W.; Yu, X.; Qin, J.J.; Li, Q. Radioiodine-refractory differentiated thyroid cancer: Molecular mechanisms and therapeutic strategies for radioiodine resistance. Drug Resist. Updat. 2024, 72, 101013. [Google Scholar] [CrossRef]
  27. Zhu, S.; Guo, Y.; Zhang, X.; Liu, H.; Yin, M.; Chen, X.; Peng, C. Pyruvate kinase M2 (PKM2) in cancer and cancer therapeutics. Cancer Lett. 2021, 503, 240–248. [Google Scholar] [CrossRef]
  28. Azouzi, N.; Cailloux, J.; Cazarin, J.M.; Knauf, J.A.; Cracchiolo, J.; Al Ghuzlan, A.; Hartl, D.; Polak, M.; Carré, A.; El Mzibri, M.; et al. NADPH Oxidase NOX4 Is a Critical Mediator of BRAFV600E-Induced Downregulation of the Sodium/Iodide Symporter in Papillary Thyroid Carcinomas. Antioxid. Redox Signal 2017, 26, 864–877. [Google Scholar] [CrossRef]
  29. Zhang, W.; Ruan, X.; Li, Y.; Zhi, J.; Hu, L.; Hou, X.; Shi, X.; Wang, X.; Wang, J.; Ma, W.; et al. KDM1A promotes thyroid cancer progression and maintains stemness through the Wnt/β-catenin signaling pathway. Theranostics 2022, 12, 1500–1517. [Google Scholar] [CrossRef]
  30. Lan, L.; Basourakos, S.; Cui, D.; Zuo, X.; Deng, W.; Huo, L.; Chen, L.; Zhang, G.; Deng, L.; Shi, B.; et al. Inhibiting β-catenin expression promotes efficiency of radioiodine treatment in aggressive follicular thyroid cancer cells probably through mediating NIS localization. Oncol. Rep. 2017, 37, 426–434. [Google Scholar] [CrossRef]
  31. Somnay, Y.R.; Yu, X.M.; Lloyd, R.V.; Leverson, G.; Aburjania, Z.; Jang, S.; Jaskula-Sztul, R.; Chen, H. Notch3 expression correlates with thyroid cancer differentiation, induces apoptosis, and predicts disease prognosis. Cancer 2017, 123, 769–782. [Google Scholar] [CrossRef]
  32. Szkudlinski, M.W.; Fremont, V.; Ronin, C.; Weintraub, B.D. Thyroid-stimulating hormone and thyroid-stimulating hormone receptor structure-function relationships. Physiol. Rev. 2002, 82, 473–502. [Google Scholar] [CrossRef] [PubMed]
  33. Wu, Z.; Xi, Z.; Xiao, Y.; Zhao, X.; Li, J.; Feng, N.; Hu, L.; Zheng, R.; Zhang, N.; Wang, S.; et al. TSH-TSHR axis promotes tumor immune evasion. J. Immunother. Cancer 2022, 10, e004049. [Google Scholar] [CrossRef] [PubMed]
  34. Fiskus, W.; Mitsiades, N. B-Raf Inhibition in the Clinic: Present and Future. Annu. Rev. Med. 2016, 67, 29–43. [Google Scholar] [CrossRef] [PubMed]
  35. Efanov, A.A.; Brenner, A.V.; Bogdanova, T.I.; Kelly, L.M.; Liu, P.; Little, M.P.; Wald, A.I.; Hatch, M.; Zurnadzy, L.Y.; Nikiforova, M.N.; et al. Investigation of the Relationship Between Radiation Dose and Gene Mutations and Fusions in Post-Chernobyl Thyroid Cancer. J. Natl. Cancer Inst. 2018, 110, 371–378. [Google Scholar] [CrossRef]
  36. Zou, M.; Baitei, E.Y.; Alzahrani, A.S.; BinHumaid, F.S.; Alkhafaji, D.; Al-Rijjal, R.A.; Meyer, B.F.; Shi, Y. Concomitant RAS, RET/PTC, or BRAF mutations in advanced stage of papillary thyroid carcinoma. Thyroid 2014, 24, 1256–1266. [Google Scholar] [CrossRef]
  37. Romei, C.; Ciampi, R.; Elisei, R. A comprehensive overview of the role of the RET proto-oncogene in thyroid carcinoma. Nat. Rev. Endocrinol. 2016, 12, 192–202. [Google Scholar] [CrossRef]
  38. Greider, C.W. Telomere length regulation. Annu. Rev. Biochem. 1996, 65, 337–365. [Google Scholar] [CrossRef]
  39. Cao, J.; Zhu, X.; Sun, Y.; Li, X.; Yun, C.; Zhang, W. The genetic duet of BRAF V600E and TERT promoter mutations predicts the poor curative effect of radioiodine therapy in papillary thyroid cancer. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 3470–3481. [Google Scholar] [CrossRef]
  40. Prasad, M.L.; Vyas, M.; Horne, M.J.; Virk, R.K.; Morotti, R.; Liu, Z.; Tallini, G.; Nikiforova, M.N.; Christison-Lagay, E.R.; Udelsman, R.; et al. NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States. Cancer 2016, 122, 1097–1107. [Google Scholar] [CrossRef]
  41. Kelly, L.M.; Barila, G.; Liu, P.; Evdokimova, V.N.; Trivedi, S.; Panebianco, F.; Gandhi, M.; Carty, S.E.; Hodak, S.P.; Luo, J.; et al. Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc. Natl. Acad. Sci. USA 2014, 111, 4233–4238. [Google Scholar] [CrossRef]
  42. Saqcena, M.; Leandro-Garcia, L.J.; Maag, J.L.V.; Tchekmedyian, V.; Krishnamoorthy, G.P.; Tamarapu, P.P.; Tiedje, V.; Reuter, V.; Knauf, J.A.; de Stanchina, E.; et al. SWI/SNF Complex Mutations Promote Thyroid Tumor Progression and Insensitivity to Redifferentiation Therapies. Cancer Discov. 2021, 11, 1158–1175. [Google Scholar] [CrossRef] [PubMed]
  43. Kiyota, N.; Tahara, M.; Robinson, B.; Schlumberger, M.; Sherman, S.I.; Leboulleux, S.; Lee, E.K.; Suzuki, T.; Ren, M.; Fushimi, K.; et al. Impact 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 2022, 128, 2281–2287. [Google Scholar] [CrossRef] [PubMed]
  44. Tahara, M.; Brose, M.S.; Wirth, L.J.; Suzuki, T.; Miyagishi, H.; Fujino, K.; Dutcus, C.E.; Gianoukakis, A. Impact of dose interruption on the efficacy of lenvatinib in a phase 3 study in patients with radioiodine-refractory differentiated thyroid cancer. Eur. J. Cancer 2019, 106, 61–68. [Google Scholar] [CrossRef] [PubMed]
  45. Viola, D.; Cappagli, V.; Elisei, R. Cabozantinib (XL184) for the treatment of locally advanced or metastatic progressive medullary thyroid cancer. Future Oncol. 2013, 9, 1083–1092. [Google Scholar] [CrossRef]
  46. Grüllich, C. Cabozantinib: Multi-kinase Inhibitor of MET, AXL, RET, and VEGFR2. Recent Results Cancer Res. 2018, 211, 67–75. [Google Scholar]
  47. Brose, M.S.; Robinson, B.G.; Sherman, S.I.; Jarzab, B.; Lin, C.C.; Vaisman, F.; Hoff, A.O.; Hitre, E.; Bowles, D.W.; Sen, S.; et al. Cabozantinib for previously treated radioiodine-refractory differentiated thyroid cancer: Updated results from the phase 3 COSMIC-311 trial. Cancer 2022, 128, 4203–4212. [Google Scholar] [CrossRef]
  48. Cochin, V.; Gross-Goupil, M.; Ravaud, A.; Godbert, Y.; Le Moulec, S. Cabozantinib: Modalités d’action, efficacité et indications [Cabozantinib: Mechanism of action, efficacy and indications]. Bull Cancer 2017, 104, 393–401. [Google Scholar] [CrossRef]
  49. Krajewska, J.; Olczyk, T.; Jarzab, B. Cabozantinib for the treatment of progressive metastatic medullary thyroid cancer. Expert Rev. Clin. Pharmacol. 2016, 9, 69–79. [Google Scholar] [CrossRef]
  50. Chau, N.G.; Haddad, R.I. Vandetanib for the treatment of medullary thyroid cancer. Clin. Cancer Res. 2013, 19, 524–529. [Google Scholar] [CrossRef]
  51. Deshpande, H.; Roman, S.; Thumar, J.; Sosa, J.A. Vandetanib (ZD6474) in the Treatment of Medullary Thyroid Cancer. Clin. Med. Insights Oncol. 2011, 5, 213–221. [Google Scholar] [CrossRef]
  52. Sim, M.W.; Cohen, M.S. The discovery and development of vandetanib for the treatment of thyroid cancer. Expert Opin. Drug Discov. 2014, 9, 105–114. [Google Scholar] [CrossRef] [PubMed]
  53. Campbell, M.J.; Seib, C.D.; Gosnell, J. Vandetanib and the management of advanced medullary thyroid cancer. Curr. Opin. Oncol. 2013, 25, 39–43. [Google Scholar] [CrossRef] [PubMed]
  54. Karras, S.; Anagnostis, P.; Krassas, G.E. Vandetanib for the treatment of thyroid cancer: An update. Expert Opin. Drug Metab. Toxicol. 2014, 10, 469–481. [Google Scholar] [CrossRef]
  55. Fallahi, P.; Di Bari, F.; Ferrari, S.M.; Spisni, R.; Materazzi, G.; Miccoli, P.; Benvenga, S.; Antonelli, A. Selective use of vandetanib in the treatment of thyroid cancer. Drug Des. Devel Ther. 2015, 9, 3459–3470. [Google Scholar]
  56. Liu, D.; Offin, M.; Harnicar, S.; Li, B.T.; Drilon, A. Entrectinib: An orally available, selective tyrosine kinase inhibitor for the treatment of NTRK, ROS1, and ALK fusion-positive solid tumors. Ther. Clin. Risk Manag. 2018, 14, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  57. Delgado, J.; Pean, E.; Melchiorri, D.; Migali, C.; Josephson, F.; Enzmann, H.; Pignatti, F. The European Medicines Agency review of entrectinib for the treatment of adult or paediatric patients with solid tumours who have a neurotrophic tyrosine receptor kinase gene fusions and adult patients with non-small-cell lung cancer harbouring ROS1 rearrangements. ESMO Open 2021, 6, 100087. [Google Scholar]
  58. Marcus, L.; Donoghue, M.; Aungst, S.; Myers, C.E.; Helms, W.S.; Shen, G.; Zhao, H.; Stephens, O.; Keegan, P.; Pazdur, R. FDA Approval Summary: Entrectinib for the Treatment of NTRK gene Fusion Solid Tumors. Clin. Cancer Res. 2021, 27, 928–932. [Google Scholar] [CrossRef]
  59. Chu, Y.H.; Dias-Santagata, D.; Farahani, A.A.; Boyraz, B.; Faquin, W.C.; Nosé, V.; Sadow, P.M. Clinicopathologic and molecular characterization of NTRK-rearranged thyroid carcinoma (NRTC). Mod Pathol 2020, 33, 2186–2197. [Google Scholar] [CrossRef]
  60. Liu, S.V.; Macke, L.A.; Colton, B.S.; Imran, S.S.; Christiansen, J.; Chow-Maneval, E.; Hornby, Z.; Multani, P.S. Response to Entrectinib in Differentiated Thyroid Cancer With a ROS1 Fusion. JCO Precis. Oncol. 2017, 1, PO.17.00105. [Google Scholar] [CrossRef]
  61. Drilon, A.; Siena, S.; Ou, S.I.; Patel, M.; Ahn, M.J.; Lee, J.; Bauer, T.M.; Farago, A.F.; Wheler, J.J.; Liu, S.V.; et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017, 7, 400–409. [Google Scholar] [CrossRef]
  62. Ardini, E.; Menichincheri, M.; Banfi, P.; Bosotti, R.; De Ponti, C.; Pulci, R.; Ballinari, D.; Ciomei, M.; Texido, G.; Degrassi, A.; et al. Entrectinib, a Pan-TRK, ROS1, and ALK Inhibitor with Activity in Multiple Molecularly Defined Cancer Indications. Mol. Cancer Ther. 2016, 15, 628–639. [Google Scholar] [CrossRef] [PubMed]
  63. Cabanillas, M.E.; Ryder, M.; Jimenez, C. Targeted Therapy for Advanced Thyroid Cancer: Kinase Inhibitors and Beyond. Endocr. Rev. 2019, 40, 1573–1604. [Google Scholar] [CrossRef] [PubMed]
  64. Waguespack, S.G.; Drilon, A.; Lin, J.J.; Brose, M.S.; McDermott, R.; Almubarak, M.; Bauman, J.; Casanova, M.; Krishnamurthy, A.; Kummar, S.; et al. Efficacy and safety of larotrectinib in patients with TRK fusion-positive thyroid carcinoma. Eur. J. Endocrinol. 2022, 186, 631–643. [Google Scholar] [CrossRef]
  65. Pitoia, F. Complete response to larotrectinib treatment in a patient with papillary thyroid cancer harboring an ETV6-NTRK3 gene fusion. Clin. Case Rep. 2021, 9, 1905–1912. [Google Scholar] [CrossRef]
  66. Hong, D.S.; Bauer, T.M.; Lee, J.J.; Dowlati, A.; Brose, M.S.; Farago, A.F.; Taylor, M.; Shaw, A.T.; Montez, S.; Meric-Bernstam, F.; et al. Larotrectinib in adult patients with solid tumours: A multi-centre, open-label, phase I dose-escalation study. Ann. Oncol. 2019, 30, 325–331. [Google Scholar] [CrossRef] [PubMed]
  67. Suh, K.; Carlson, J.J.; Xia, F.; Williamson, T.; Sullivan, S.D. The potential long-term comparative effectiveness of larotrectinib vs standard of care for treatment of metastatic TRK fusion thyroid cancer, colorectal cancer, and soft tissue sarcoma. J. Manag. Care Spec. Pharm. 2022, 28, 622–630. [Google Scholar] [CrossRef]
  68. Groussin, L.; Theodon, H.; Bessiene, L.; Bricaire, L.; Bonnet-Serrano, F.; Cochand-Priollet, B.; Leroy, K.; Garinet, S.; Pasmant, E.; Zerbit, J.; et al. Redifferentiating Effect of Larotrectinib in NTRK-Rearranged Advanced Radioactive-Iodine Refractory Thyroid Cancer. Thyroid 2022, 32, 594–598. [Google Scholar] [CrossRef]
  69. Cohen, E.E.; Tortorici, M.; Kim, S.; Ingrosso, A.; Pithavala, Y.K.; Bycott, P. A Phase II trial of axitinib in patients with various histologic subtypes of advanced thyroid cancer: Long-term outcomes and pharmacokinetic/pharmacodynamic analyses. Cancer Chemother. Pharmacol. 2014, 74, 1261–1270. [Google Scholar] [CrossRef]
  70. Capdevila, J.; Trigo, J.M.; Aller, J.; Manzano, J.L.; Adrián, S.G.; Llopis, C.Z.; Reig, Ò.; Bohn, U.; Cajal, T.R.Y.; Duran-Poveda, M.; et al. Axitinib treatment in advanced RAI-resistant differentiated thyroid cancer (DTC) and refractory medullary thyroid cancer (MTC). Eur. J. Endocrinol. 2017, 177, 309–317. [Google Scholar] [CrossRef]
  71. Ferrari, S.M.; Centanni, M.; Virili, C.; Miccoli, M.; Ferrari, P.; Ruffilli, I.; Ragusa, F.; Antonelli, A.; Fallahi, P. Sunitinib in the Treatment of Thyroid Cancer. Curr. Med. Chem. 2019, 26, 963–972. [Google Scholar] [CrossRef]
  72. Gómez-Sáez, J.M. Sunitinib for the treatment of thyroid cancer. Expert Opin. Investig. Drugs 2016, 25, 1345–1352. [Google Scholar] [CrossRef] [PubMed]
  73. Wong, E.; Rosen, L.S.; Mulay, M.; Vanvugt, A.; Dinolfo, M.; Tomoda, C.; Sugawara, M.; Hershman, J.M. Sunitinib induces hypothyroidism in advanced cancer patients and may inhibit thyroid peroxidase activity. Thyroid 2007, 17, 351–355. [Google Scholar] [CrossRef] [PubMed]
  74. Bible, K.C.; Suman, V.J.; Menefee, M.E.; Smallridge, R.C.; Molina, J.R.; Maples, W.J.; Karlin, N.J.; Traynor, A.M.; Kumar, P.; Goh, B.C.; et al. A multiinstitutional phase 2 trial of pazopanib monotherapy in advanced anaplastic thyroid cancer. J. Clin. Endocrinol. Metab. 2012, 97, 3179–3184. [Google Scholar] [CrossRef] [PubMed]
  75. Wells SAJr Robinson, B.G.; Gagel, R.F.; Dralle, H.; Fagin, J.A.; Santoro, M.; Baudin, E.; Elisei, R.; Jarzab, B.; Vasselli, J.R.; Read, J.; 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, 134–141. [Google Scholar] [CrossRef]
  76. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  77. Hong, D.S.; DuBois, S.G.; Kummar, S.; Farago, A.F.; Albert, C.M.; Rohrberg, K.S.; van Tilburg, C.M.; Nagasubramanian, R.; Berlin, J.D.; Federman, N.; et al. Larotrectinib in patients with TRK fusion-positive solid tumours: A pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020, 21, 531–540. [Google Scholar] [CrossRef]
  78. Carr, L.L.; Mankoff, D.A.; Goulart, B.H.; Eaton, K.D.; Capell, P.T.; Kell, E.M.; Bauman, J.E.; Martins, R.G. Phase II study of daily sunitinib in FDG-PET-positive, iodine-refractory differentiated thyroid cancer and metastatic medullary carcinoma of the thyroid with functional imaging correlation. Clin. Cancer Res. 2010, 16, 5260–5268. [Google Scholar] [CrossRef]
  79. Bible, K.C.; Menefee, M.E.; Lin, C.J.; Millward, M.J.; Maples, W.J.; Goh, B.C.; Karlin, N.J.; Kane, M.A.; Adkins, D.R.; Molina, J.R.; et al. An International Phase 2 Study of Pazopanib in Progressive and Metastatic Thyroglobulin Antibody Negative Radioactive Iodine Refractory Differentiated Thyroid Cancer. Thyroid 2020, 30, 1254–1262. [Google Scholar] [CrossRef]
  80. Hadoux, J.; Elisei, R.; Brose, M.S.; Hoff, A.O.; Robinson, B.G.; Gao, M.; Jarzab, B.; Isaev, P.; Kopeckova, K.; Wadsley, J.; et al. Phase 3 Trial of Selpercatinib in Advanced RET-Mutant Medullary Thyroid Cancer. N. Engl. J. Med. 2023, 389, 1851–1861. [Google Scholar] [CrossRef]
  81. Subbiah, V.; Hu, M.I.; Mansfield, A.S.; Taylor, M.H.; Schuler, M.; Zhu, V.W.; Hadoux, J.; Curigliano, G.; Wirth, L.; Gainor, J.F.; et al. Pralsetinib in Patients with Advanced/Metastatic Rearranged During Transfection (RET)-Altered Thyroid Cancer: Updated Efficacy and Safety Data from the ARROW Study. Thyroid 2024, 34, 26–40. [Google Scholar] [CrossRef]
  82. Mehnert, J.M.; Varga, A.; Brose, M.S.; Aggarwal, R.R.; Lin, C.C.; Prawira, A.; de Braud, F.; Tamura, K.; Doi, T.; Piha-Paul, S.A.; et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced, PD-L1-positive papillary or follicular thyroid cancer. BMC Cancer 2019, 19, 196. [Google Scholar] [CrossRef] [PubMed]
  83. Chintakuntlawar, A.V.; Yin, J.; Foote, R.L.; Kasperbauer, J.L.; Rivera, M.; Asmus, E.; Garces, N.I.; Janus, J.R.; Liu, M.; Ma, D.J.; et al. A Phase 2 Study of Pembrolizumab Combined with Chemoradiotherapy as Initial Treatment for Anaplastic Thyroid Cancer. Thyroid 2019, 29, 1615–1622. [Google Scholar] [CrossRef] [PubMed]
  84. Iyer, P.C.; Dadu, R.; Gule-Monroe, M.; Busaidy, N.L.; Ferrarotto, R.; Habra, M.A.; Zafereo, M.; Williams, M.D.; Gunn, G.B.; Grosu, H.; et al. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J. Immunother. Cancer 2018, 6, 68. [Google Scholar] [CrossRef]
  85. Boudin, L.; Morvan, J.B.; Thariat, J.; Métivier, D.; Marcy, P.Y.; Delarbre, D. Rationale Efficacy and Safety Evidence of Lenvatinib and Pembrolizumab Association in Anaplastic Thyroid Carcinoma. Curr. Oncol. 2022, 29, 7718–7731. [Google Scholar] [CrossRef]
  86. Haugen, B.; French, J.; Worden, F.; Konda, B.; Sherman, E.; Dadu, R.; Gianoukakis, A.; Mccue, S.; Foster, N.; Bowles, D.; et al. 1917P Pembrolizumab salvage add-on therapy in patients with radioiodine-refractory (RAIR), progressive differentiated thyroid cancer (DTC) progressing on lenvatinib: Results of a multicenter phase II International Thyroid Oncology Group Trial. Ann. Oncol. 2020, 31, S1086–S1087. [Google Scholar] [CrossRef]
  87. Wirth, L.; Subbiah, V.; Worden, F.; Solomon, B.; Robinson, B.; Hadoux, J.; Tomasini, P.; Weiler, D.; Deschler-Baier, B.; Tan, D.; et al. 2229P Updated safety and efficacy of selpercatinib in patients (pts) with RET-activated thyroid cancer: Data from LIBRETTO-001. Ann. Oncol. 2023, 34, S1147–S1148. [Google Scholar] [CrossRef]
  88. Kroiss, M.; Sherman, E.; Wirth, L.; Cabanillas, M.; Robinson, B.; Subbiah, V.; Drilon, A.; Godbert, Y.; Fasnacht, N.; Soldatenkova, V.; et al. 1656P Durable efficacy of selpercatinib in patients (pts) with medullary thyroid cancer (MTC): Update of the LIBRETTO-001 trial. Ann. Oncol. 2022, 33, S1299–S1300. [Google Scholar] [CrossRef]
  89. Subbiah, V.; Wolf, J.; Konda, B.; Kang, H.; Spira, A.; Weiss, J.; Takeda, M.; Ohe, Y.; Khan, S.; Ohashi, K.; et al. Tumour-agnostic efficacy and safety of selpercatinib in patients with RET fusion-positive solid tumours other than lung or thyroid tumours (LIBRETTO-001): A phase 1/2, open-label, basket trial. Lancet Oncol. 2022, 23, 1261–1273. [Google Scholar] [CrossRef]
  90. Markham, A. Selpercatinib: First Approval. Drugs 2020, 80, 1119–1124. [Google Scholar] [CrossRef]
  91. Della Corte, C.M.; Morgillo, F. Rethinking treatment for RET-altered lung and thyroid cancers: Selpercatinib approval by the EMA. ESMO Open 2021, 6, 100041. [Google Scholar] [CrossRef]
  92. Zheng, X.; Fang, M.; Fan, Y.; Sun, Y.; Sun, M.; Yang, A.; Zhang, B.; Liu, Q.; Liu, H.; Zhou, X.; et al. Efficacy and safety of pralsetinib in Chinese advanced RET-mutant medullary thyroid cancer patients. Endocr. Relat. Cancer 2024, 31, e230134. [Google Scholar] [CrossRef] [PubMed]
  93. Kim, J.; Bradford, D.; Larkins, E.; Pai-Scherf, L.H.; Chatterjee, S.; Mishra-Kalyani, P.S.; Wearne, E.; Helms, W.S.; Ayyoub, A.; Bi, Y.; et al. FDA Approval Summary: Pralsetinib for the Treatment of Lung and Thyroid Cancers With RET Gene Mutations or Fusions. Clin. Cancer Res. 2021, 27, 5452–5456. [Google Scholar] [CrossRef] [PubMed]
  94. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.; Cabanillas, M.E.; Urbanowitz, G.; et al. Dabrafenib and Trametinib Treatment in Patients With Locally Advanced or Metastatic BRAF V600-Mutant Anaplastic Thyroid Cancer. J. Clin. Oncol. 2018, 36, 7–13. [Google Scholar] [CrossRef] [PubMed]
  95. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Cho, J.Y.; Schellens, J.H.M.; Soria, J.C.; Wen, P.Y.; Zielinski, C.C.; Cabanillas, M.E.; Boran, A.; et al. Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: Updated analysis from the phase II ROAR basket study. Ann. Oncol. 2022, 33, 406–415. [Google Scholar] [CrossRef]
  96. Jeon, Y.; Park, S.; Lee, S.H.; Kim, T.H.; Kim, S.W.; Ahn, M.J.; Jung, H.A.; Chung, J.H. Combination of Dabrafenib and Trametinib in Patients with Metastatic BRAFV600E-Mutated Thyroid Cancer. Cancer Res. Treat. 2024, 56, 1270–1276. [Google Scholar] [CrossRef]
  97. Lorimer, C.; Cheng, L.; Chandler, R.; Garcez, K.; Gill, V.; Graham, K.; Grant, W.; Sardo Infirri, S.; Wadsley, J.; Wall, L.; et al. Dabrafenib and Trametinib Therapy for Advanced Anaplastic Thyroid Cancer—Real-World Outcomes From UK Centres. Clin. Oncol. (R. Coll. Radiol.) 2023, 35, e60–e66. [Google Scholar] [CrossRef]
  98. Leboulleux, S.; Dupuy, C.; Lacroix, L.; Attard, M.; Grimaldi, S.; Corre, R.; Ricard, M.; Nasr, S.; Berdelou, A.; Hadoux, J.; et al. Redifferentiation of a BRAFK601E-Mutated Poorly Differentiated Thyroid Cancer Patient with Dabrafenib and Trametinib Treatment. Thyroid 2019, 29, 735–742. [Google Scholar] [CrossRef]
  99. Busaidy, N.L.; Konda, B.; Wei, L.; Wirth, L.J.; Devine, C.; Daniels, G.A.; DeSouza, J.A.; Poi, M.; Seligson, N.D.; Cabanillas, M.E.; et al. Dabrafenib Versus Dabrafenib + Trametinib in BRAF-Mutated Radioactive Iodine Refractory Differentiated Thyroid Cancer: Results of a Randomized, Phase 2, Open-Label Multicenter Trial. Thyroid 2022, 32, 1184–1192. [Google Scholar] [CrossRef]
  100. Zhao, X.; Wang, J.R.; Dadu, R.; Busaidy, N.L.; Xu, L.; Learned, K.O.; Chasen, N.N.; Vu, T.; Maniakas, A.; Eguia, A.A.; et al. Surgery After BRAF-Directed Therapy Is Associated with Improved Survival in BRAFV600E Mutant Anaplastic Thyroid Cancer: A Single-Center Retrospective Cohort Study. Thyroid 2023, 33, 484–491. [Google Scholar] [CrossRef]
  101. Ball, D.W.; Jin, N.; Xue, P.; Bhan, S.; Ahmed, S.R.; Rosen, D.M.; Schayowitz, A.; Clark, D.P.; Nelkin, B.D. Trametinib with and without pazopanib has potent preclinical activity in thyroid cancer. Oncol. Rep. 2015, 34, 2319–2324. [Google Scholar] [CrossRef]
  102. Kurzrock, R.; Ball, D.W.; Zahurak, M.L.; Nelkin, B.D.; Subbiah, V.; Ahmed, S.; O’Connor, A.; Karunsena, E.; Parkinson, R.M.; Bishop, J.A.; et al. A Phase I Trial of the VEGF Receptor Tyrosine Kinase Inhibitor Pazopanib in Combination with the MEK Inhibitor Trametinib in Advanced Solid Tumors and Differentiated Thyroid Cancers. Clin. Cancer Res. 2019, 25, 5475–5484. [Google Scholar] [CrossRef] [PubMed]
  103. White, P.S.; Pudusseri, A.; Lee, S.L.; Eton, O. Intermittent Dosing of Dabrafenib and Trametinib in Metastatic BRAFV600E Mutated Papillary Thyroid Cancer: Two Case Reports. Thyroid 2017, 27, 1201–1205. [Google Scholar] [CrossRef]
  104. Bible, K.C.; Suman, V.J.; Molina, J.R.; Smallridge, R.C.; Maples, W.J.; Menefee, M.E.; Rubin, J.; Karlin, N.; Sideras, K.; Morris JC3rd McIver, B.; et al. A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: MC057H. J. Clin. Endocrinol. Metab. 2014, 99, 1687–1693. [Google Scholar] [CrossRef]
  105. Hamidi, S.; Iyer, P.C.; Dadu, R.; Gule-Monroe, M.K.; Maniakas, A.; Zafereo, M.E.; Wang, J.R.; Busaidy, N.L.; Cabanillas, M.E. Checkpoint Inhibition in Addition to Dabrafenib/Trametinib for BRAFV600E-Mutated Anaplastic Thyroid Carcinoma. Thyroid 2024, 34, 336–346. [Google Scholar] [CrossRef] [PubMed]
  106. Schlumberger, M.; Brose, M.; Elisei, R.; Leboulleux, S.; Luster, M.; Pitoia, F.; Pacini, F. Definition and management of radioactive iodine-refractory differentiated thyroid cancer. Lancet Diabetes Endocrinol. 2014, 2, 356–358. [Google Scholar] [CrossRef]
  107. Shonka DCJr Ho, A.; Chintakuntlawar, A.V.; Geiger, J.L.; Park, J.C.; Seetharamu, N.; Jasim, S.; Abdelhamid Ahmed, A.H.; Bible, K.C.; Brose, M.S.; Cabanillas, M.E.; et al. American Head and Neck Society Endocrine Surgery Section and International Thyroid Oncology Group consensus statement on mutational testing in thyroid cancer: Defining advanced thyroid cancer and its targeted treatment. Head Neck 2022, 44, 1277–1300. [Google Scholar] [CrossRef]
  108. Tuttle, R.M.; Ahuja, S.; Avram, A.M.; Bernet, V.J.; Bourguet, P.; Daniels, G.H.; Dillehay, G.; Draganescu, C.; Flux, G.; Führer, D.; et al. Controversies, Consensus, and Collaboration in the Use of 131I Therapy in Differentiated Thyroid Cancer: A Joint Statement from the American Thyroid Association, the European Association of Nuclear Medicine, the Society of Nuclear Medicine and Molecular Imaging, and the European Thyroid Association. Thyroid 2019, 29, 461–470. [Google Scholar] [PubMed]
  109. Pitoia, F.; Jerkovich, F.; Trimboli, P.; Smulever, A. New approaches for patients with advanced radioiodine-refractory thyroid cancer. World J. Clin. Oncol. 2022, 13, 9–27. [Google Scholar] [CrossRef]
  110. Nikiforova, M.N.; Wald, A.I.; Roy, S.; Durso, M.B.; Nikiforov, Y.E. Targeted next-generation sequencing panel (ThyroSeq) for detection of mutations in thyroid cancer. J. Clin. Endocrinol. Metab. 2013, 98, E1852–E1860. [Google Scholar] [CrossRef]
  111. Giordano, T.J.; Beaudenon-Huibregtse, S.; Shinde, R.; Langfield, L.; Vinco, M.; Laosinchai-Wolf, W.; Labourier, E. Molecular testing for oncogenic gene mutations in thyroid lesions: A case-control validation study in 413 postsurgical specimens. Hum. Pathol. 2014, 45, 1339–1347. [Google Scholar] [CrossRef]
  112. Vitale, M. Mutational testing and its utility in thyroid cancer management: The need for something more. Biomark. Med. 2013, 7, 571–574. [Google Scholar] [CrossRef] [PubMed]
  113. Babu, G.; Kainickal, C.T. Update on the systemic management of radioactive iodine refractory differentiated thyroid cancer (Review). Mol. Clin. Oncol. 2021, 14, 35. [Google Scholar] [CrossRef] [PubMed]
  114. Haddad, R.I.; Bischoff, L.; Ball, D.; Bernet, V.; Blomain, E.; Busaidy, N.L.; Campbell, M.; Dickson, P.; Duh, Q.; Ehya, H.; et al. Thyroid Carcinoma, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 925–951. [Google Scholar] [CrossRef] [PubMed]
  115. Haugen, B.R.; Alexander, E.K.; Bible, K.C.; Doherty, G.M.; Mandel, S.J.; Nikiforov, Y.E.; Pacini, F.; Randolph, G.W.; Sawka, A.M.; Schlumberger, M.; et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 2016, 26, 1–133. [Google Scholar] [CrossRef]
  116. Filetti, S.; Durante, C.; Hartl, D.; Leboulleux, S.; Locati, L.D.; Newbold, K.; Papotti, M.G.; Berruti, A.; ESMO Guidelines Committee. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 1856–1883. [Google Scholar] [CrossRef]
  117. Fugazzola, L.; Elisei, R.; Fuhrer, D.; Jarzab, B.; Leboulleux, S.; Newbold, K.; Smit, J. 2019 European Thyroid Association Guidelines for the Treatment and Follow-Up of Advanced Radioiodine-Refractory Thyroid Cancer. Eur. Thyroid J. 2019, 8, 227–245. [Google Scholar] [CrossRef]
  118. Ito, Y.; Onoda, N.; Okamoto, T. The revised clinical practice guidelines on the management of thyroid tumors by the Japan Associations of Endocrine Surgeons: Core questions and recommendations for treatments of thyroid cancer. Endocr. J. 2020, 67, 669–717. [Google Scholar] [CrossRef]
  119. Bible, K.C.; Ryder, M. Evolving molecularly targeted therapies for advanced-stage thyroid cancers. Nat. Rev. Clin. Oncol. 2016, 13, 403–416. [Google Scholar] [CrossRef]
  120. Brose, M.S.; Smit, J.W.A.; Lin, C.C.; Tori, M.; Bowles, D.W.; Worden, F.; Shen, D.H.; Huang, S.M.; Tsai, H.J.; Alevizaki, M.; et al. Multikinase Inhibitors for the Treatment of Asymptomatic Radioactive Iodine-Refractory Differentiated Thyroid Cancer: Global Noninterventional Study (RIFTOS MKI). Thyroid 2022, 32, 1059–1068. [Google Scholar] [CrossRef]
  121. Vaisman, F.; Tala, H.; Grewal, R.; Tuttle, R.M. In differentiated thyroid cancer, an incomplete structural response to therapy is associated with significantly worse clinical outcomes than only an incomplete thyroglobulin response. Thyroid 2011, 21, 1317–1322. [Google Scholar] [CrossRef]
  122. American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer; Cooper, D.S.; Doherty, G.M.; Haugen, B.R.; Kloos, R.T.; Lee, S.L.; Mandel, S.J.; Mazzaferri, E.L.; McIver, B.; Pacini, F.; et al. Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 2009, 19, 1167–1214. [Google Scholar]
  123. Tufano, R.P.; Clayman, G.; Heller, K.S.; Inabnet, W.B.; Kebebew, E.; Shaha, A.; Steward, D.L.; Tuttle, R.M.; American Thyroid Association Surgical Affairs Committee Writing Task Force. Management of recurrent/persistent nodal disease in patients with differentiated thyroid cancer: A critical review of the risks and benefits of surgical intervention versus active surveillance. Thyroid 2015, 25, 15–27. [Google Scholar] [CrossRef] [PubMed]
  124. Urken, M.L.; Milas, M.; Randolph, G.W.; Tufano, R.; Bergman, D.; Bernet, V.; Brett, E.M.; Brierley, J.D.; Cobin, R.; Doherty, G.; et al. Management of recurrent and persistent metastatic lymph nodes in well-differentiated thyroid cancer: A multifactorial decision-making guide for the Thyroid Cancer Care Collaborative. Head Neck 2015, 37, 605–614. [Google Scholar] [CrossRef]
  125. Guy, A.; Hirsch, D.; Shohat, T.; Bachar, G.; Tirosh, A.; Robenshtok, E.; Shimon, I.; Benbassat, C.A. Papillary thyroid cancer: Factors involved in restaging N1 disease after total thyroidectomy and radioactive iodine treatment. J. Clin. Endocrinol. Metab. 2014, 99, 4167–4173. [Google Scholar] [CrossRef]
  126. Tuttle, R.M.; Haddad, R.I.; Ball, D.W.; Byrd, D.; Dickson, P.; Duh, Q.Y.; Ehya, H.; Haymart, M.; Hoh, C.; Hunt, J.P.; et al. Thyroid carcinoma, version 2.2014. J. Natl. Compr. Cancer Netw. 2014, 12, 1671–1680; quiz 1680. [Google Scholar] [CrossRef] [PubMed]
  127. Haugen, B.R.; Kane, M.A. Approach to the thyroid cancer patient with extracervical metastases. J. Clin. Endocrinol. Metab. 2010, 95, 987–993. [Google Scholar] [CrossRef]
  128. Brierley, J.D.; Tsang, R.W. External-beam radiation therapy in the treatment of differentiated thyroid cancer. Semin. Surg. Oncol. 1999, 16, 42–49. [Google Scholar] [CrossRef]
  129. Brierley, J.D.; Tsang, R.W. External beam radiation therapy for thyroid cancer. Endocrinol. Metab. Clin. N. Am. 2008, 37, 497–509. [Google Scholar] [CrossRef]
  130. Pöllinger, B.; Dühmke, E. External radiotherapy of thyroid cancer. Onkologie 2001, 24, 134–138. [Google Scholar] [CrossRef]
  131. Mangoni, M.; Gobitti, C.; Autorino, R.; Cerizza, L.; Furlan, C.; Mazzarotto, R.; Monari, F.; Simontacchi, G.; Vianello, F.; Basso, M.; et al. External beam radiotherapy in thyroid carcinoma: Clinical review and recommendations of the AIRO “Radioterapia Metabolica” Group. Tumori 2017, 103, 114–123. [Google Scholar] [CrossRef]
  132. Brierley, J.D. Update on external beam radiation therapy in thyroid cancer. J. Clin. Endocrinol. Metab. 2011, 96, 2289–2295. [Google Scholar] [CrossRef] [PubMed]
  133. Roukoz, C.; Gregoire, V. Indications of external beams radiation for thyroid cancer. Curr. Opin. Otolaryngol. Head Neck Surg. 2022, 30, 137–144. [Google Scholar] [CrossRef]
  134. Giuliani, M.; Brierley, J. Indications for the use of external beam radiation in thyroid cancer. Curr. Opin. Oncol. 2014, 26, 45–50. [Google Scholar] [CrossRef] [PubMed]
  135. De Leo, A.; Di Simone, E.; Spano, A.; Puliani, G.; Petrone, F. Nursing Management and Adverse Events in Thyroid Cancer Treatments with Tyrosine Kinase Inhibitors: A Narrative Review. Cancers 2021, 13, 5961. [Google Scholar] [CrossRef] [PubMed]
  136. De Leo, S.; Trevisan, M.; Moneta, C.; Colombo, C. Endocrine-related adverse conditions induced by tyrosine kinase inhibitors. Ann. Endocrinol. 2023, 84, 374–381. [Google Scholar] [CrossRef]
  137. Bai, Y.; Li, J.Y.; Li, J.; Zhang, B.; Liu, Y.H.; Zhang, B.Y.; Jing, J. Risk of venous and arterial thromboembolic events associated with tyrosine kinase inhibitors in advanced thyroid cancer: A meta-analysis and systematic review. Oncotarget 2018, 10, 4205–4212. [Google Scholar] [CrossRef]
  138. Yu, S.T.; Ge, J.N.; Luo, J.Y.; Wei, Z.G.; Sun, B.H.; Lei, S.T. Treatment-related adverse effects with TKIs in patients with advanced or radioiodine refractory differentiated thyroid carcinoma: A systematic review and meta-analysis. Cancer Manag. Res. 2019, 11, 1525–1532. [Google Scholar] [CrossRef]
  139. Nervo, A.; Ragni, A.; Piovesan, A.; Marica, V.; Migliore, E.; Gallo, M.; Arvat, E. Quality of Life during Treatment with Lenvatinib for Thyroid Cancer: The Patients’ Perspective beyond the Medical Evaluation. Eur. Thyroid J. 2021, 10, 65–71. [Google Scholar] [CrossRef]
  140. Taylor, M.H.; Leboulleux, S.; Panaseykin, Y.; Konda, B.; de La Fouchardiere, C.; Hughes, B.G.M.; Gianoukakis, A.G.; Park, Y.J.; Romanov, I.; Krzyzanowska, M.K.; et al. Health-related quality-of-life analyses from a multicenter, randomized, double-blind phase 2 study of patients with differentiated thyroid cancer treated with lenvatinib 18 or 24 mg/day. Cancer Med. 2023, 12, 4332–4342. [Google Scholar] [CrossRef]
  141. Enokida, T.; Tahara, M. Management of VEGFR-Targeted TKI for Thyroid Cancer. Cancers 2021, 13, 5536. [Google Scholar] [CrossRef]
  142. Krajewska, J.; Paliczka-Cieslik, E.; Jarzab, B. Managing tyrosine kinase inhibitors side effects in thyroid cancer. Expert Rev. Endocrinol. Metab. 2017, 12, 117–127. [Google Scholar] [CrossRef] [PubMed]
  143. Gabora, K.; Piciu, A.; Bădulescu, I.C.; Larg, M.I.; Stoian, I.A.; Piciu, D. Current evidence on thyroid related adverse events in patients treated with protein tyrosine kinase inhibitors. Drug Metab. Rev. 2019, 51, 562–569. [Google Scholar] [CrossRef]
  144. Jimenez-Fonseca, P. Use of multikinase inhibitors/lenvatinib in patients with high cardiovascular risk/vasculopathy and radioiodine refractory-differentiated thyroid cancer. Cancer Med. 2022, 11 (Suppl. S1), 17–25. [Google Scholar] [CrossRef] [PubMed]
  145. Pitoia, F.; Schmidt, A.; Bueno, F.; Abelleira, E.; Jerkovich, F. Rare complications of multikinase inhibitor treatment. Arch. Endocrinol. Metab. 2018, 62, 636–640. [Google Scholar] [CrossRef] [PubMed]
  146. Cabanillas, M.E.; Takahashi, S. Managing the adverse events associated with lenvatinib therapy in radioiodine-refractory differentiated thyroid cancer. Semin. Oncol. 2019, 46, 57–64. [Google Scholar] [CrossRef]
  147. Cohen, R.B.; Oudard, S. Antiangiogenic therapy for advanced renal cell carcinoma: Management of treatment-related toxicities. Investig. New Drugs 2012, 30, 2066–2079. [Google Scholar] [CrossRef]
  148. Lacouture, M.E.; Ciccolini, K.; Kloos, R.T.; Agulnik, M. Overview and management of dermatologic events associated with targeted therapies for medullary thyroid cancer. Thyroid 2014, 24, 1329–1340. [Google Scholar] [CrossRef]
  149. Teo, Y.L.; Ho, H.K.; Chan, A. Risk of tyrosine kinase inhibitors-induced hepatotoxicity in cancer patients: A meta-analysis. Cancer Treat. Rev. 2013, 39, 199–206. [Google Scholar] [CrossRef]
  150. Béchade, D.; Chakiba, C.; Desjardin, M.; Bécouarn, Y.; Fonck, M. Toxicité hépatique des inhibiteurs des tyrosines kinases: Mécanismes en cause et conséquences pratiques [Hepatotoxicity of tyrosine kinase inhibitors: Mechanisms involved and practical implications]. Bull. Cancer 2018, 105, 290–298. [Google Scholar] [CrossRef]
Figure 1. Therapeutic sequencing and decision-making pathway for RAI-R DTC.
Figure 1. Therapeutic sequencing and decision-making pathway for RAI-R DTC.
Cancers 17 01800 g001
Table 1. Available agents in the treatment of radioiodine-refractory differentiated thyroid carcinoma (RAI-R DTC), medullary TC (MTC), and anaplastic TC (ATC).
Table 1. Available agents in the treatment of radioiodine-refractory differentiated thyroid carcinoma (RAI-R DTC), medullary TC (MTC), and anaplastic TC (ATC).
DrugClinical TrialPhaseIndicationMain Molecular TargetsDosagePFS Compared to Placebo (Months)
Lenvatinib [7]NCT01321554 IIIFirst/second-line DTC VEGFR1-3, FGFR1-4, PDGFRα, RET, c-KIT24 mg orally per day18.3 vs. 3.6, HR 0.21 (99% CI 0.14–0.31, p < 0.001)
Sorafenib [8]NCT00984282 IIIFirst * line DTCVEGFR1-3, PDGFRβ, Raf, c-KIT, FLT-3. BRAF, CRAF 400 mg orally twice daily 10.8 vs. 5.8, HR 0.58 (95% CI 0.45–0.75, p < 0.0001)
Cabozantinib [9,45]NCT01811212/NCT00704730III (DTC)/
III (MTC)
Second-line DTC
First-line non-RETm MTC
Second-line RETm MTC
MET, VEGFR2, RET, c-KIT, FLT-3, AXL60 mg orally per dayDTC: NE vs. 1.9, HR 0.22 (96% CI 0.13–0.36; p < 0.0001)
MTC: 11.2 vs. 4.0; HR 0.28 (95% CI 0.19–0.40; p < 0.001)
Vandetanib [75]NCT00537095/NCT00410761II/IIIFirst-line non-RETm MTC
Second-line RETm MTC
Subsequent in DTC
VEGFR2/3, EGFR, RET300 mg orally per dayDTC: 11.1 (95% CI, 7.7–14.0)
MTC: 30.5 (model prediction; NE) vs. 19.3, HR 0.46 (95% CI 0.31–0.69; p < 0.001)
Entrectinib [76] NCT02568267IIAny line NTRK, ROS1, and ALK fusions600 mg orally per day19.9 (95% CI, 6.5–33.8)
Larotrectinib [77]NCT02122913/
NCT02637687/NCT02576431
I/IIAny line NTRK fusions100 mg orally twice daily 44.0 (95% CI, 16.6–NE) at a median follow-up of 38.7 months
Axitinib [69]NCT00094055 IISubsequent line if no driver mutationVEGFR1-35 mg orally twice daily 15–18.1 (95% CI)
Sunitinib [78]NCT00381641 IISubsequent line if no driver mutationVEGFR1/2, PDGFR, RET, KIT, FLT3, CSF1R37.5 mg orally per day 12.8 (95% CI, 8.9–NE)
Pazopanib [79]NCT00625846 IISubsequent line if no driver mutationVEGFR, PDGFR800 mg orally per day 11.4 (95% CI)
Selpercatinib [80]NCT04211337IIIFirst/second-line DTC (targeted therapy)
First-line RETm MTC
First-line RETm ATC
RET160 mg orally twice daily NE vs. 16.8, HR 0.28 (95% CI 0.16–0.48; p < 0.001)
Pralsetinib [81]NCT03037385IITargeted therapy RET400 mg orally per day 25.9, NR, 25.4 (95% CI) **
PFS: progression-free survival; RETm: RET mutation; NE: not estimable. * Can be considered as a subsequent line. ** 25.9 months in patients with RETm MTC who had received prior cabozantinib/vandetanib; NR in patients with treatment-naïve RETm MTC; 25.4 months in patients with previously treated RET fusion-positive TC.
Table 2. Therapeutic sequencing pathway for RAI-R DTC.
Table 2. Therapeutic sequencing pathway for RAI-R DTC.
Clinical SituationRecommended Approach
1Diagnosis of RAI-R DTCAssess tumor burden and disease progression
2Low tumor burden, indolent diseaseActive surveillance with periodic monitoring
3Progressive or symptomatic diseaseSystemic therapy initiation (TKIs or targeted agents)
4First-line systemic therapyLenvatinib or Sorafenib
5Disease progression after first-line therapyCabozantinib or alternative systemic agents
6Molecularly targeted therapyRET/NTRK inhibitors, immune checkpoint inhibitors
7Consideration of local treatmentsSurgery, radiotherapy, or ablative technique
8Quality of life assessmentMultidisciplinary team review and supportive care
9Palliative care if necessaryEnd-of-life care planning and symptom management
RAI-R: radioiodine-refractory; DTC: differentiated thyroid carcinoma; TKIs: tyrosine kinase inhibitors; RET: rearranged during transfection; NTKR: neurotrophic tyrosine receptor kinase.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Díaz Vico, T.; Martínez-Amores Martínez, B.; Mihic Góngora, L.; Jiménez-Fonseca, P.; Peinado Martín, P.; Grao Torrente, I.; García Muñoz-Nájar, A.; Durán-Poveda, M. Systemic Therapeutic Options in Radioiodine-Refractory Differentiated Thyroid Cancer: Current Indications and Optimal Timing. Cancers 2025, 17, 1800. https://doi.org/10.3390/cancers17111800

AMA Style

Díaz Vico T, Martínez-Amores Martínez B, Mihic Góngora L, Jiménez-Fonseca P, Peinado Martín P, Grao Torrente I, García Muñoz-Nájar A, Durán-Poveda M. Systemic Therapeutic Options in Radioiodine-Refractory Differentiated Thyroid Cancer: Current Indications and Optimal Timing. Cancers. 2025; 17(11):1800. https://doi.org/10.3390/cancers17111800

Chicago/Turabian Style

Díaz Vico, Tamara, Brezo Martínez-Amores Martínez, Luka Mihic Góngora, Paula Jiménez-Fonseca, Paloma Peinado Martín, Irene Grao Torrente, Alejandro García Muñoz-Nájar, and Manuel Durán-Poveda. 2025. "Systemic Therapeutic Options in Radioiodine-Refractory Differentiated Thyroid Cancer: Current Indications and Optimal Timing" Cancers 17, no. 11: 1800. https://doi.org/10.3390/cancers17111800

APA Style

Díaz Vico, T., Martínez-Amores Martínez, B., Mihic Góngora, L., Jiménez-Fonseca, P., Peinado Martín, P., Grao Torrente, I., García Muñoz-Nájar, A., & Durán-Poveda, M. (2025). Systemic Therapeutic Options in Radioiodine-Refractory Differentiated Thyroid Cancer: Current Indications and Optimal Timing. Cancers, 17(11), 1800. https://doi.org/10.3390/cancers17111800

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop