You are on page 1of 17

T h i s i s You r T h y ro i d on

Drugs
Targetable Mutations and Fusions in Thyroid
Carcinoma
Ying-Hsia Chu, MD

KEYWORDS
 Kinase fusion  BRAF V600 E  NTRK  RET  Inhibitor  Resistance

Key points
 Activating alterations of receptor tyrosine kinases and downstream effectors in the mitogen-
activated protein kinase (MAPK) and PI3K pathways are the main drivers of thyroid carcinogenesis.
 BRAF V600 E mutation and rearrangements of RET and NTRK are the most common actionable targets
in thyroid cancer with the United States Food and Drug Administration (FDA)-approved inhibitor
therapy.
 Rearrangements of other kinases (eg, ALK, MET, ROS1, and BRAF), deregulation of the PI3K/Akt/
mTOR pathway, microsatellite instability status, and tumor mutation burden are emerging bio-
markers of therapeutic actionability under active research.

ABSTRACT past few decades.1,2 Rising incidences are seen

T
across different histologic subtypes, in microcarci-
his review aims to provide an overview of the nomas and large (>4 cm) tumors, in thyroid-
molecular pathogenesis thyroid carcinomas, confined and metastatic cases.1,2 Although most
emphasizing genetic alterations that are patients with thyroid cancer have a surgically
therapeutically actionable. The main pathways in manageable disease, postoperative persistent
thyroid carcinogenesis are the MAPK and PI3K and recurrent tumors cause significant morbidity
pathways. Point mutations and gene rearrange- and mortality in 10% to 30% of differentiated thy-
ments affecting the pathway effectors and recep- roid carcinoma (DTC) and the majority of medullary
tor tyrosine kinases are well-known drivers of and anaplastic carcinoma patients. To treat DTC,
thyroid cancer. Research over the past few de- radioactive iodine (RAI) has long been used to
cades has successfully introduced highly effective ablate remnant thyroid tissue and control residual
treatments for unresectable thyroid cancer, and recurrent disease.3 However, de novo and ac-
evolving from multi-kinase inhibitors to structurally quired RAI refractoriness, although controversially
selective agents, with constantly improving defined,3 is a well-recognized clinical conundrum
toxicity profiles and coverage of resistance mech- associated with adverse prognosis, with only
anisms. The pros and cons of major laboratory 10% of such patients reaching 10-year survival.4
techniques for therapeutic target identification Virtually all poorly differentiated and anaplastic
are discussed. carcinomas are RAI-refractory and respond poorly
to conventional chemoradiation, with a five-year
OVERVIEW disease-specific survival of 50% to 60% and
almost none, respectively.5,6 This underscores a
The overall incidence of thyroid cancer has been growing need for systemic therapy with
surgpath.theclinics.com

steadily increasing in the United States over the genomics-guided precision to improve patient

The author declares no conflict of interest.


Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street,
Guishan District, Taoyuan City 333, Taiwan
E-mail address: yinghsia.c@gmail.com

Surgical Pathology 16 (2023) 57–73


https://doi.org/10.1016/j.path.2022.09.007
1875-9181/23/Ó 2022 Elsevier Inc. All rights reserved.
58 Chu

outcomes. This review presents an overview of the the RL group consists mainly of highly differenti-
genetic underpinnings of thyroid cancer, focusing ated, follicular variant PTC driven by N/H/KRAS
on recent advances in targeted therapy. (11%), EIF1AX (1%), non-V600 E BRAF (1%) muta-
tions, and PAX8::PPARG fusion (1%).
MOLECULAR LANDSCAPE OF FOLLICULAR- Yoo and colleagues8 subsequently performed a
DERIVED THYROID CARCINOMA transcriptomic analysis of a large cohort of
follicular-patterned neoplasms including follicular
WELL-DIFFERENTIATED THYROID adenoma (FA), follicular thyroid carcinomas
CARCINOMAS (FTC), follicular variant PTC, and a comparison
group of classic PTC. In addition to affirming the
Activating alterations of receptor tyrosine kinases key genetic onco-drivers of the BVL and RL mo-
(RTKs) and the downstream mitogen-activated lecular subtypes observed by TCGA, the study
protein kinase (MAPK) pathway are the main identified a new non-BRAF-non-RAS (NBNR) sub-
drivers for well-differentiated thyroid carcinomas type associated with mutations in IDH1, DICER1,
(Fig. 1). The Cancer Genome Atlas (TCGA) pro- EIF1AX, PTEN, SPOP, SOS1, and PAX8::PPARG
gram analyzed 496 papillary thyroid carcinomas rearrangement. Similar to the RL samples, the
(PTC), composed mainly of classic (69%), follicular NBNR group was characterized by highly differen-
(21%), and tall cell (7.5%) types, and was able to tiated tumors with preserved expression of genes
divide the cohort into BRAFV600E-like (BVL) and involved in thyroid function and metabolism. All
RAS-like (RL) tumors using a BRAFV600E -RAS the FA and FTC samples clustered into the RL
score (BRS) derived from a 71-gene expression and NBNR categories, as did most encapsulated
signature.7 BRAF V600 E mutation (47% of all follicular variant PTC. Infiltrative follicular variant
cases), BRAF fusions (2%), and RET fusions PTCs, in contrast, had variable transcriptomic pro-
(5%) were the main drivers in the BVL group, which files that were distributed roughly equally to the
was characterized by decreased thyroid differenti- BVL and RL groups.
ation and a predominance of classic, tall cell, and Kinase fusions have a predilection for pediatric
infiltrative follicular variant histology. In contrast, and radiation-associated PTCs, at 56% and 41%

Fig. 1. Intracellular signaling


pathways in thyroid carcino-
genesis. Genetic alterations
of the MAPK and PI3K path-
ways are the most common
druggable drivers. Targeted
inhibitors are listed, with
investigational therapies that
have not received FDA
approval for thyroid cancer
placed in parentheses.
This is Your Thyroid on Drugs 59

prevalence, respectively,9,10 and have also been B). ALK fusion tumors are frequently follicular-
reported in up to 25% of adult PTC.11 In all the patterned (see Fig. 2C).14 An intriguing finding of
three patient populations, RET is the most glomeruloid papillary formation has been
commonly rearranged kinase gene (approximately described in NTRK fusion cases (see Fig. 2D).11
28%, 17%, 14% in pediatric, post-radiation, and In addition to PTC, primary thyroid secretory carci-
adult PTC, respectively), followed by NTRK nomas represent a rare phenotype of
(15%, 11%, and 8%).9–11 Other kinase fusions ETV6::NTRK3 fusion thyroid carcinoma.18 Histo-
involving BRAF, ALK, ROS1, and MET also rarely logically, these tumors may show microcystic,
occur.11 In RTK gene fusions, the partner genes tubular, and solid growth with production of eosin-
typically provide dimerization domains, such as ophilic secretions (see Fig. 2E–F). The nuclei are
the coiled coil domains in CCDC6, NCOA4, PPL, vacuolar with well-visible nucleoli (see Fig. 2F
TPR, TPM3, the WD domain in EML4, the PNT inset) and may show grooves and rare pseudoin-
domain in ETV6, the zinc finger domain in clusions. Most studies reported a tendency for
IRF2BP2, the PB1 domains in TFG and SQSTM1, aggressive behavior for fusion-positive tumors.
and the RNA recognition motif of RBPMS, which Over 50% of fusion-positive PTC presented as
enables the kinase domain to undergo ligand- T3 or T4 disease in pediatric, post-radiation, and
independent dimerization and activation. Fusions adult settings, and up to 81%, 39%, and 75%,
involving the RAF family serine/threonine kinase respectively, had lymph node metastases at the
genes such as BRAF, in contrast, cause aberrant time of diagnosis.9–11
activation by replacing the 50 autoinhibitory
domain-encoding sequence with a partner gene POORLY DIFFERENTIATED AND ANAPLASTIC
sequence that may not contain a dimerization CARCINOMAS
domain. In a recent analysis of 100 BRAF fusion-
positive melanocytic tumors, only 55% of the part- Progression of well-differentiated thyroid carci-
ner sequences were found to contain dimerization noma to poorly differentiated thyroid carcinoma
domains, in keeping the idea that the loss of auto- (PDTC) and anaplastic thyroid carcinoma (ATC) is
inhibition plays a major oncogenic role. In the pre- accompanied by increasing mutation burden.19,20
treatment setting, kinase fusions are mutually In addition to retaining the tumor-initiating BRAF/
exclusive with the BRAF V600 E and RAS muta- RAS mutations and kinase fusions, PDTC and
tions, but have been found concurrent with muta- ATC frequently harbor mutations in TP53, TERT
tions in TP53, SMARCA4, VHL, PIK3R1, GNAQ, promoter, mediators of the PI3K/Akt/mTOR
CDH1, AKT2, FBXW7, JAK2, GNA11, IDH1, pathway (PIK3CA, PTEN, AKT, TSC1/2, MTOR),
MEN1, SETD2, IGF2R, SDHA, CDKN2A, MSH6, SWI/SNF chromatin-remodeling complexes
and the TERT promoter.11,12 Interestingly, in the (ARID1A/1 B/2/5B, SMARCB1, PBRM1, ATRX),
TCGA study, NTRK (2%) and ALK (1%) fusion histone-modifying enzymes (KMT2A/2 C/2D and
cases tended to show intermediate BRS and a SETD2), and mismatch repair proteins (MSH2,
neutral transcriptional profile in the BVL-RL MSH6, MLH1) genes.19–22 Copy number alterations
spectrum.7 (CNAs) affecting cell cycle regulation (deletion of
Several recent studies have reported distinct CDKN2A/2B, amplification of CCNE1), RTKs
clinicopathologic features of kinase fusion-driven (KDR, KIT, PDGFRA), and immune evasion-related
PTC. In fine needle aspiration specimens, NTRK genes have also been associated with high-grade
and ALK fusion tumors have been noted to non-anaplastic and anaplastic carcinomas.20,22
frequently show indeterminate cytology with
most cases in The Bethesda System for Reporting ONCOCYTIC CARCINOMAS
Thyroid Cytopathology (TBS) categories III
and IV.13,14 Higher percentages of categories V Hurthle cell (oncocytic) carcinomas, as defined by
to VI have been reported by other studies,15,16 voluminous eosinophilic cytoplasm of the
reflecting possible confounding by inter-observer neoplastic cells as a result of mitochondrial hyper-
variability and a need for further research. plasia, characteristically harbor mitochondrial
Histology-wise, commonly observed features DNA (mtDNA) mutations and CNAs.23–26 Nonsilent
included multinodular growth, intratumoral mtDNA mutations, found in 71% of oncocytic car-
fibrosis, and lymphovascular spread cinomas, most commonly affect genes that
(Fig. 2).9,11,17 RET fusions have long been associ- encode subunits of the complex I (67%) of the
ated with the diffuse sclerosing type of PTC, char- electron transport chain, followed by complexes
acterized by extensive lymphovascular IV (9%), III (4%), and V (3%).24 Approximately
involvement, stromal fibrosis, lymphocytic inflam- 14% of mutations occur in mitochondrial transfer
mation, and squamous metaplasia (see Fig. 2A– RNA and ribosomal RNA.24 Pro-oncogenic
60 Chu

Fig. 2. Histologic features of kinase fusion-driven papillary carcinomas. Multinodular growth (A), intratumoral
fibrosis (A), and lymphovascular spread are frequently seen with various kinase fusions. RET fusions are associated
with the diffuse sclerosing variant with additional features of lymphocytic inflammation (B, white arrow) and squa-
mous metaplasia (B, black arrow). ALK fusion tumors are often follicular-patterned (C, magnified in inset). Glomer-
uloid papillary formation has been described in NTRK fusion tumors (D, white arrows). Primary thyroid secretory
carcinoma is a rare phenotype of NTRK-rearranged thyroid carcinoma, typically carrying ETV6::NTRK3 fusions. These
tumors are characterized by microcystic architecture (E) and intracellular and extracellular globules of eosinophilic
secretions (F, white arrows, also seen in inset). The nuclei are low grade-appearing with well-visible nucleoli (F, inset).

cytophysiologic deregulations in oncocytic carci- genesis.23 Another oncogenic force comes from
nomas have been shown in vitro, including a shift frequent whole chromosome duplications
from oxidative phosphorylation to aerobic glycol- (WCDs) of chromosomes 5, 7, and 12 in approxi-
ysis and increased reactive oxygen species mately 55% of oncocytic carcinomas.23–26 WCDs
This is Your Thyroid on Drugs 61

lead to amplification and overexpression of several Survival benefits were also observed for sorafenib
MAPK and PI3K pathway effectors, including RIC- in the DECISION trial, with a median PFS of
TOR and GOLPH3 on chromosome 5, MET, CDK6, 10.8 months versus 5.8 months in the placebo
and BRAF on chromosome 7, KRAS, CDK4, and group.31 Despite their inhibitory activities against
MDM2 on chromosome 12. Approximately 54% multiple additional kinases including RET, repur-
of oncocytic carcinomas show widespread chro- posing MKI for direct RET targeting has shown sig-
mosomal losses in a near-haploid tumor genome, nificant off-target toxicities and inferior
often with concurrent WCD of the remaining chro- pharmacokinetics compared with selective RET
mosomes 5, 7, and 12 leading to extensive loss of inhibitors.32 Most adverse effects of MKI are
heterozygosity (LOH), an adverse prognosticator related to impaired vascular repair and vasomotor
for oncocytic carcinoma.24,25 In addition, somatic regulation leading to hypertension and mucocuta-
mutations in the RAS/RAF/MAPK and PI3K/AKT/ neous inflammation. Toxicities frequently led to
mTOR pathways (55%), DNA damage and repair dose interruptions, reductions, and discontinua-
machineries (38%), epigenetic modifying enzymes tions in 66.2%, 64.3%, and 18.8% of subjects in
(59%), and the TERT promoter (22% to 32%) have the DECISION trial and 82.4%, 67.8%, and
been reported.23–26 14.2% in the SELECT trial,30,31 respectively. As
many patients with RAIR DTC can have slowly pro-
TARGETED THERAPY FOR FOLLICULAR- gressive disease, deciding when to initiate MKI
DERIVED CARCINOMAS therapy require careful clinical evaluation to bal-
ance between life quality and tumor control.
Currently, kinase fusions and the BRAF V600 E mu-
tation are the main druggable genetic alterations in MITOGEN-ACTIVATED PROTEIN KINASE
thyroid cancer. Although RAS mutations account PATHWAY BLOCKERS
for most FTC and many follicular-patterned PTC,
the development of RAS inhibitors has been hin- Direct BRAF inhibition has been evaluated in BRAF
dered by the lack of drug-binding pocket.27 V600E-mutated thyroid carcinoma in two open-
Although two covalent inhibitors of the KRAS label phase II trials.33–35 Brose and colleagues33
G12 C mutant, sotorasib and adagrasib, have investigated the use of vemurafenib in 51 patients
recently become available,28 KRAS G12 C is with RAIR PTC, including 26 patients without
extremely rare in thyroid cancer. Instead, most (cohort 1) and 25 with prior MKI treatment (cohort
RAS mutations in follicular-derived carcinomas 2). In cohort 1, over a median follow-up of
occur at the Q61 locus, which is currently undrug- 18$8 months, partial response was achieved in
gable. Nevertheless, thyroid cancer overall harbors 38.5%, and the median PFS was 18.2 months. In
the highest frequency of kinase fusions of all solid cohort 2, over a median follow-up of 12.0 months,
tumors.29 Selective kinase inhibitors have become partial response was achieved in 27.3%, and the
the first-choice treatment for clinically significant, median PFS was 8.9 months. It was noteworthy
radioactive iodine-refractory (RAIR) kinase fusion- that cutaneous squamous cell carcinoma (SCC)
positive tumors. For fusion-negative RAIR DTC, was the most common serious adverse effect in
multi-kinase inhibitors represent the current stan- the trial, developing in 27% of cohort 1 and 20%
dard of care based on well-established survival of cohort 2. BRAF inhibitor-associated SCC is
benefits from two pivotal phase III trials (Table 1). believed to result from paradoxical RAF activation
MAPK pathway blockade, through directly target- in BRAF-wildtype cells, and its occurrence can be
ing the BRAF V600 E mutant or inducing tumor effectively reduced with combination therapy of
redifferentiation, has also shown clinical efficacy. BRAF and MEK inhibitors.36,37 Indeed, in the
ROAR trial by Subbiah and colleagues34,35 on
MULTI-KINASE INHIBITORS treating 36 patients with BRAF V600E-mutated
ATC with dabrafenib and trametinib combination
Sorafenib and lenvatinib are the current first-line therapy, no secondary SCC was noted. The
kinase inhibitor therapy for clinically significant ROAR trial showed an objective response rate
RAIR DTC. These agents exert anti-tumoral effects (ORR) of 56% and a median PFS of 6.7 months,
mainly through targeting the vascular endothelial leading to the United States Food and Drug
growth factor receptors (VEGFRs) and suppress- Administration (FDA) approval of dabrafenib and
ing tumor-induced angiogenesis. In the SELECT trametinib for the treatment of ATC in 2018 fol-
trial, 392 patients were randomized to receive len- lowed by accelerated histology-agnostic approval
vatinib versus placebo.30 Median progression-free for BRAF V600E-positive solid tumors in 2022.34,35
survival (PFS) was significantly improved in the Despite the protective effect of co-administered
lenvatinib group (18.3 months vs 3.6 months).30 MEK inhibitor against MAPK pathway reactivation,
62
Table 1
FDA-approved targeted therapy for thyroid cancer

Chu
Approved Thyroid Registrational Tumor Histology
Agent Target(s) Indication(s) Trial Design (Case no.) ORR Median PFS
Lenvatinib VEGFR1/2/3, RAI-refractory SELECT30 Phase III PTC (200), FTC (75), 64.8% 18.3 months
FGFR1/2/3/4, thyroid cancer HCC (70), PDTC (47) (lenvatinib)
PDGFRɑ, vs 3.6 months
RET, KIT (placebo)
Sorafenib VEGFR1/2/3, RAI-refractory DECISION31 Phase III PTC (237), FTC (106), 12.2% 10.8 months
RET, RAF, thyroid cancer PDTC (40), WDC (3) (sorafenib) vs
PDGFRb, KIT 5.8 months
(placebo)
Cabozantinib VEGFR1/2/3, RET, DTC failing prior COSMIC-31176 Phase III PTC (102), FTC (90) 15% Not reached
MET, FLT3, KIT anti-VEGF therapy (cabozantinib)
vs 1.9 months
(placebo)
MTC EXAM75 Phase III MTC (330) 28% 11.2 months
(cabozantinib)
vs 4.0 months
(placebo)
Vandetanib VEGFR1/2/3, MTC ZETA74 Phase III MTC (331) 45% Not reached
RET, EGFR (vandetanib)
vs 19.3 months
(placebo)
Dabrafenib, BRAF BRAF V600E-mutant ROAR/ Phase II ATC (36) 56% 6.7 months
trametinib (dabrafeninb), solid tumorsb BRF11701935
MEK
(trametinib)
Selpercatinib RET RET fusion 1 solid LIBRETTO-00142 Phase I/II PTC (13), PDTC (3), 79% 20.1 months
tumorsb ATC (2), HCC (1)
RET-mutant MTC LIBRETTO-00142 Phase I/II MTC (55 with and 88 69%, 73%a Not reached
without prior MKI (previously treated
treatment) with MKI);
23.6 months (MKI-
naı̈ve)
Pralsetinib RET RET fusion 1 thyroid ARROW43 Phase I/II RET fusion-positive 89% Not reached
cancer thyroid cancer (11)
RET-mutant MTC ARROW43 Phase I/II 55 with and 21 60%, 71%a Not reached
without prior MKI
treatment
Larotrectinib NTRK1/2/3 NTRK fusion 1 solid NAVIGATE, SCOUT, Phase I/II PTC (20), ATC (7), FTC 71% 2.2 months for ATC;
tumorsb LOXO-TRK-1400146 (2) not reached for
other histologic
types
Entrectinib NTRK1/2/3, NTRK fusion 1 solid ALKA-372–001, Phase I/II NTRK fusion-positive 20% -
ROS1, ALK tumorsb STARTRK-1, thyroid cancer (5)
STARTRK-247

Abbreviations: ATC, anaplastic thyroid carcinoma; DTC, differentiated thyroid carcinoma; FTC, follicular thyroid carcinoma; HCC, Hurthle cell carcinoma; MKI, multikinase inhibitors
vandetanib and cabozantinib; MTC, medullary thyroid carcinoma; ORR, objective response rate; PDTC, poorly differentiated thyroid carcinoma; PFS, progression-free survival; PTC,
papillary thyroid carcinoma; RAI, radioactive iodine; WDC, well-differentiated carcinoma.
a
See text for details.
b
Histology-agnostic approval.

This is Your Thyroid on Drugs


63
64 Chu

resistance has since still emerged due to acquired carcinoma cases (20 PTC, 7 ATC, 2 FTC), with an
RAS mutations.38 BRAF and/or MEK inhibitors ORR of 71%.46 As for entrectinib, three phase 1/2
have also attracted significant research interest trials reported an ORR of 57% among all enrolled
in inducing tumor redifferentiation and restoring subjects and 20% among thyroid cancer cases.47
RAI sensitivity in RAIR DTC,39–41 which is outside Both larotrectinib and entrectinib were well toler-
the scope of this review. ated. Less than 5% of the trial subjects discontin-
ued treatment due to toxicities. Only 8%
SELECTIVE RET INHIBITORS (larotrectinib) and 30% (entrectinib) required dose
reductions due to anemia, elevated blood creati-
Pralsetinib and selpercatinib are selective RET in- nine, hepatic or pancreatic enzymes, fatigue, and
hibitors with significantly improved antitumoral ac- nausea.44,47 On-target resistance-mediating muta-
tivity and tolerability compared with MKI. tions have been reported in multiple tumor types
Selpercatinib received FDA approval in 2020 for (colonic, thyroid, pancreatic, pulmonary, salivary
treating RET fusion-positive advanced or metasta- gland carcinomas, cholangiocarcinoma, gastroin-
tic thyroid cancer followed by histology-agnostic testinal stromal tumor, sarcomas)48–50 at the sol-
approval in 2022 based on the efficacy data from vent front (NTRK1 G595 R, NTRK3 G623 R) and
the phase 1-2 trial LIBRETTO-001 (see Table 1).42 gatekeeper (NTRK1 F589 L, NTRK3 F617 L) posi-
The trial evaluated a total of 19 non-medullary thy- tions as well as in the xDFG motif (NTRK1
roid cancer patients, including 13 PTC, 1 oncocytic G667 C/S, NTRK3 G696 A).48–51 Fortunately,
carcinoma, 3 PDTC, and 2 ATC. The ORR was 79% next-generation NTRK inhibitors selitrectinib and
including 1 complete response and 14 partial re- repotrectinib have shown great promise in restoring
sponses, whereas the remaining patients all tumor response in resistant cases.52,53
showed stable disease.42 The median duration of
response was not reached, and the median PFS ALK inhibitors
was 20.1 months.42 Overall, toxicities led to dose
ALK fusions are rarely found in thyroid cancer, with
reductions in 30% of the patients and treatment
only scattered case reports in PTC,54 oncocytic
termination in 2% due to abnormal liver function
carcinoma,55 PDTC,11 ATC,56,57 and MTC.58 Cri-
and hypersensitivity.42 Pralsetinib was granted
zotinib is a first-generation ALK inhibitor with con-
accelerated FDA approval in 2020 for thyroid can-
current inhibitory activities against ROS1 and
cer based on the ARROW trial (see Table 1).43
MET. Originally approved by the FDA in 2011 for
This phase 1/2 trial evaluated 11 RET fusion-
treating ALK-rearranged lung cancer, crizotinib
positive thyroid cancer patients with an ORR of
has since shown therapeutic success in various
89% and a disease control rate of 100%.43 Ac-
organs including in thyroid cancer. However, resis-
quired resistance to selective RET inhibition has
tance inevitably develops 1 to 2 years into treat-
been emerging in RET fusion-positive thyroid can-
ment due to acquired kinase domain mutations59
cer, including one ATC that developed EGFR ampli-
and, less commonly, through activating mutations
fication as a bypassing mechanism that caused
or amplifications of genes involved in bypassing
selpercatinib resistance.11
pathways such as EGFR, KRAS, and MET.60
Among the five reported cases of ALK-rearranged
NTRK INHIBITORS thyroid carcinomas treated with crizotinib
(Table 2), initial crizotinib response was noted to
Larotrectinib and entrectinib are the first TRK inhib-
last for 6 to 36 months.11,54–56,58 One patient with
itors approved by the FDA for the treatment of adult
ATC was able to switch to second-generation in-
and pediatric NTRK-rearranged solid tumors.
hibitors ceritinib and brigatinib but eventually suc-
Oncogenic NTRK rearrangements are rare drivers
cumbed to progressive disease.56 None of the
of thyroid, breast, lung, pancreatic, and colonic car-
reported cases underwent post-treatment
cinomas, melanomas, gliomas, and sarcomas,
sequencing, and their resistance mechanisms
while being pathognomonic in several unusual
remained uncharacterized.
tumor types (eg, secretory carcinomas, infantile fi-
brosarcomas, and congenital mesoblastic nephro-
mas). The body-wide distribution of NTRK- PI3K/AKT/mTOR inhibition
rearranged tumors inspired the histology-agnostic The targeting of the PI3K/AKT/mTOR pathway has
design of clinical trials44–47 (see Table 1). thus far shown limited success in thyroid cancer.
Combining three phase 1/2 trials in 159 patients, Everolimus, an mTOR inhibitor, has been evaluated
larotrectinib showed an ORR of 79% across various by several recent phase II trials.61–63 Schneider and
solid tumors, with complete response in 16%.44 By colleagues63 studied 28 patients with RAIR DTC and
mid-2020, the trials included a total of 29 thyroid 7 ATC with daily treatment of 10 mg everolimus
This is Your Thyroid on Drugs 65

Table 2
Reported cases of thyroid carcinomas receiving ALK inhibitor therapy

Molecular Best Response,


Reference Histology Abnormalities ALK Inhibitor Duration Side Effects
54
PTC EML4::ALK; Crizotinib SD, over 6 months Fatigue, cough
mutations to the end of
in 55 genes study
55
OC ALK fusion with Crizotinib CR, 6 months Constipation
unknown
partner
11
PDTC STRN::ALK; TP53 Crizotinib SD, 11 months Fatigue
c.772 G > C
56
ATC STRN::ALK Crizotinib, CR, 36 months –
switched to (crizotinib);
ceritinib and PR, 16 months
then brigatinib (ceritinib); PR,
for resistance 8 months
(brigatinib)
58
MTC CCDC6::ALK Crizotinib, PR, over 280 days Myalgia
switched to to the end of
alectinib for the study
tolerance
Abbreviations: ATC, anaplastic thyroid carcinoma; CR, complete response; MTC, medullary thyroid carcinoma; OC, onco-
cytic carcinoma; PDTC, poorly differentiated thyroid carcinoma; PR, partial response; PTC, papillary thyroid carcinoma; SD,
stable disease.

10 mg orally. At a median follow-up of 38 months, Thyroid Association guidelines recommend the


65% achieved stable disease, whereas partial or use of external beam radiotherapy radiation and
complete response was not observed. Hanna and systemic therapy with MKI or selective RET inhib-
colleagues62 evaluated 33 RAIR DTC, 10 MTC, itors.66 MTC is known to have low response rate to
and 7 ATC patients with the same everolimus conventional cytotoxic chemotherapeutic agents,
regimen for a median follow-up of 31.8 months. which are therefore not recommended for the
The ORR was 3%, 10%, and 14% in DTC, MTC, first-line setting.
and ATC, whereas 82%, 80%, and 28% achieved
stable disease, respectively.62 Although both RET
studies performed genetic analysis and found
PI3K/AKT/mTOR pathway mutations in subsets of Activating alterations of RET serve as the key
patients, a solid correlation between tumor geno- genetic drivers for nearly all hereditary and
type and everolimus response has not been made approximately 40% of sporadic MTC. The RET
with the small cohort sizes. This remains to be proto-oncogene encodes a transmembrane RTK
resolved by several ongoing clinical trials at the composed of four extracellular cadherin-like do-
time of this writing. mains, a cysteine-rich region, a transmembrane
domain, and an intracellular part that contains
the kinase domain. Various point mutations can
MEDULLARY THYROID CARCINOMA cause aberrant kinase activation, and, in the he-
reditary setting, interesting genotype-phenotype
Medullary thyroid carcinomas (MTC) are neuroen- correlations have been characterized. Multiple
docrine tumors showing differentiation towards endocrine neoplasia (MEN) syndrome type 2A pre-
the parafollicular cells of thyroid gland. Despite sents with MTC, pheochromocytomas, hyperpara-
their uncommonness (approximately 5% of all thy- thyroidism, and cutaneous lichen amyloidosis
roid cancers), most patients present with (CLM). Over 90% of MEN-2A cases result from
advanced disease (50% to 70% with regional mutations in cysteine codons 611, 618, 620, and
and 105 to 15% with distant metastases) which 634. These mutations are thought to promote
is not amenable to RAI therapy.64,65 For patients intermolecular disulfide bone formation, leading
with advanced, recurrent, or metastatic disease to aberrant RET dimerization and activation. In
beyond surgical control, the current American contrast, the MEN type 2B syndrome arises from
66 Chu

a kinase domain mutation M918 T, which leads to (56%) and occasionally in KRAS (12%) and
increased ATP-binding affinity, loss of autoinhibi- NRAS (rare).80 Uncommonly, HRAS may be co-
tion, and ultimately ligand-independent mono- mutated along with RET in the same tumor.80
meric and dimeric autophosphorylation and Various mutations in codons 12, 13, and 61 have
activation.67 Unlike those affected by MEN-2A, pa- been reported in the Catalogue Of Somatic Muta-
tients with the MEN-2B syndrome tend not to tions In Cancer (COSMIC) database,81 such as
develop hyperparathyroidism and CLM, but G12 V/R, G13 V/R, and Q61 K/L/R. However,
instead present with mucosal and intestinal neu- KRAS G12 C, the primary target of sotorasib and
romas and a marfanoid appearance. Among spo- adagrasib, has been vanishingly rare. Other RAS-
radic MTC, the most common RET mutation is targeting strategies being developed include
M918 T (in 69%) followed by codon 634 mutations mutant-specific T cell receptor-engineered T cell
(11%).68 The M918 T mutation has been associ- therapy, cancer vaccines, and inhibition of post-
ated with more aggressive tumor behavior in transcriptional RAS processing.82 Multiple post-
both hereditary and sporadic settings.69,70 RET fu- transcriptional enzymatic steps are required for
sions are exceptionally rare in MTC.71 Fusions RAS proteins to become functional, including pre-
involving other kinases, such as ALK and BRAF, nylation by farnesyltransferase or geranylgeranyl-
have been reported in isolated cases with notable transferase. Unlike KRAS and NRAS, HRAS can
response to targeted therapy.58,72,73 only be prenylated by farnesyltransferase and is
Inhibition of RET signaling can be achieved therefore vulnerable to farnesyltransferase inhibi-
through MKI and selective RET inhibitors.74–76 tion. Tipifarnib is a farnesyltransferase inhibitor
Among MKI, vandetanib and cabozantinib have that has been studied in a combination therapy
both been approved by the FDA for treating MTC with sorafenib in a phase II trial that included 13
(see Table 1). In the registrational ZETA trial, van- MTC patients.83 The trial observed an ORR of
detanib led to a superior median PFS of 38%, whereas 31% had stable disease as the
30.5 months compared with 19.3 months in the best response.83 As the study did not analyze
placebo group.74 Patients receiving cabozantinib RAS mutation status in the MTC cases, the rela-
also showed a clear survival benefit, with a median tionship between tumor genotype and therapeutic
PFS of 11.2 months compared with 4.0 months in sensitivity was unclear.
those receiving placebo.75 To overcome MKI’s
frequent toxicities and to tackle MKI-resistant mu- LABORATORY IDENTIFICATION OF
tations such as V804 L/M, selective RET inhibitors
THERAPEUTIC TARGETS
have been developed with excellent efficacy and
tolerability (see Table 1). In the LIBRETTO-001 IMMUNOHISTOCHEMISTRY
trial, selpercatinib showed durable partial to com-
plete response in 69% of those with preceding Immunohistochemical assays (IHCs) have been
MKI treatment and 73% of MKI-naı̈ve patients.42 developed for detecting BRAF V600 E84,85 and
More recently, pralsetinib showed similarly RAS Q61 R86,87 mutations and for rearrangements
impressive clinical benefits, with an ORR of 60% of NTRK (pan-TRK),88,89 ALK,90,91 RET,92 and
and 71% among subjects with and without prior ROS1.93 IHC are advantageous for its short turn-
MKI exposure, respectively.43 Based on the trial around time, low tissue input and the ability to
results, both selpercatinib and pralsetinib have visualize biomarker expression within histologic
received the FDA approval for treating advanced context. Recent studies have reported reasonable
or metastatic RET-altered MTC. However, ac- performance for BRAF V600 E (clone VE1, sensi-
quired resistance to selpercatinib has since tivity 89–100%; specificity 62–100%85) and RAS
emerged due to solvent front mutations G810 C/ Q61 R86 (clone SP174, sensitivity 90.6%, speci-
S and hinge region mutations Y806 C/N.77,78 ficity 92.3%) mutant-directed IHC. The VE1 anti-
These mutations are also predicted to confer body does not label BRAF V600 K or K601 E
resistance to pralsetinib.78 Fortunately, next- mutations or BRAF fusions.85 The performance
generation RET inhibitors such as TPX-0046 of kinase-based IHC in detecting gene fusions
have shown potent in vitro and in vivo activity may be partner gene-dependent. For pan-TRK
against a broad range of RET alterations including IHC, Solomon and colleagues88 reported a sensi-
resistance-mediating solvent front mutations.79 tivity of 81.1% and a specificity of 99.9% in 87
NTRK fusion tumors. The sensitivity was higher in
RAS NTRK1 (96%) and NTRK2 (100%) fusions
compared with NTRK3-rearranged samples
Among RET-wildtype MTC, up to 68% harbor hot- (79%).88,94 It is noteworthy that tumors with neural
spot RAS mutations, most commonly in HRAS and smooth muscle differentiation can show
This is Your Thyroid on Drugs 67

positive TRK staining in the absence of NTRK fusions are overexpressed, the sequence incorpo-
rearrangement.89 Also partner-dependent was rated by the fusion transcript (eg, the 30 part of
RET-directed IHC, for which Yang and col- NTRK3) is detected at a much higher level
leagues92 reported a sensitivity of 100% for compared with the unincorporated part (eg, the
KIF5B-RET, 88.9% for CCDC6-RET cases, and 50 part of NTRK3).96 EI-based detection has been
50% for NCOA4-RET; the specificity was approx- used by several rapid testing platforms, including
imately 82%. Unlike ALK IHC which has nearly quantitative PCR,97 digital droplet PCR,98
100% sensitivity and specificity for ALK fusions, hybridization-based transcript enumeration,99,100
ROS1 IHC may show high background staining matrix-assisted laser desorption/ionization time-
that limits its clinical application.93 of-flight analysis100 as well as next-generation
sequencing (NGS) (discussed below) with variable
FLUORESCENCE IN SITU HYBRIDIZATION performance. EI-based detection can be signifi-
cantly hindered by endogenous expression of the
For a long time, fluorescence in situ hybridization wildtype gene. For example, ROS1 have a high
(FISH) has been the mainstay method for detecting physiologic expression in pneumocytes, which
gene rearrangements. Break-apart FISH probe has been shown to obscure EI in ROS1-rear-
design allows for fusion detection without knowl- ranged tumors and reduce fusion detection
edge about partner gene identity and breakpoint sensitivity.96,99
location. However, FISH has limited multiplexing
capability and cannot provide therapeutically rele- NEXT-GENERATION SEQUENCING
vant functional details such as the reading frame
and the preservation of drug-binding domain (eg, Over the past decade, NGS has significantly
the kinase domain in kinase fusions). As a result, improved the efficiency of therapeutic target identi-
biologically nonproductive fusions have led to fication through expanded throughput and drasti-
treatment failure in up to 40% of ALK FISH- cally reduced per-gene analytic time and cost.
positive lung cancer.95 Moreover, fusions second- Although detection of point mutations and small
ary to short-segment inversions, such as the indels has been well-validated in most platforms,
NCOA4-RET fusion commonly encountered in thy- several challenges persist for NGS-based fusion
roid carcinomas, may not produce visually detection. Fusion breakpoints are often intronic
discernible probe separation resulting in false and can be analyzed through hybrid capture or
negative results.92 amplicon-based enrichment (Fig. 3). However,
extensive intronic tiling can cause inefficient con-
CONVENTIONAL POLYMERASE CHAIN sumption of sequencing capacity. Furthermore,
REACTION-BASED ASSAYS intronic repetitive sequences and complex rear-
rangement events can render bioinformatic map-
Single-gene polymerase chain reaction (PCR)- ping challenging and error-prone with short-read
based assays are widely performed in clinical lab- sequencing. For this reason, instead of direct
oratories due to their technical versatility. Point DNA sequencing, fusion detection is often per-
mutations, such as BRAF V600 E, can be detected formed using tumor-derived complementary DNA
at the DNA level using Sanger sequencing, restric- (cDNA), in which introns have been spliced out,
tion enzyme processing, and allele-specific ampli- and the analysis can focus on the exons. One addi-
fication methods. Gene fusions are generally tional advantage of cDNA sequencing is that onco-
queried at the RNA level using revere- genic fusions are highly expressed, which aids in
transcription PCR as post-transcriptional splicing detection in low tumor purity samples. Comple-
circumvents several technical challenges caused mentary DNA sequencing can also characterize
by intronic breakpoints, as discussed in the next oncogenic splice variants such as the BRAF splice
section. Fusion-specific (FS) detection and variants in PTC and ATC101 as well as the NTRK1
expression imbalance (EI) quantification are the splice variants deltaTRKA and TRKAIII.102 The
two main assay design strategies used to detect main limitation of cDNA sequencing is RNA degra-
gene fusions.96 FS detection requires knowing dation. Molecular pathologists play an important
the identity of both partner genes and the break- role in ensuring the validity of sequencing results
point location for designing opposing primers by thoroughly evaluating quality metrics and veri-
(Fig. 3C), which precludes detection of novel fu- fying each fusion call based on the alignment qual-
sions. In contrast, EI-based detection only ana- ity, the predicted reading frame, and literature
lyzes one target gene and quantifies the support of biological relevance.
expression levels of the 30 and 50 regions using Very recently, the American Head and Neck So-
separate amplicons (Fig. 3E). As oncogenic ciety (AHNS) Endocrine Surgery Section and
68 Chu

Fig. 3. Common NGS-


based assay designs for
gene fusion detection.
Fusion breakpoints are
often intronic. Genes are
depicted with exons in
wide bands and introns
in thin lines. Detection at
the DNA level (A) requires
intronic bait tiling that
can be costly. Further-
more, repetitive se-
quences and complex
rearrangement events
can be difficult to resolve
with short-read
sequencing. RNA (cDNA)
sequencing detects tran-
scriptionally active fu-
sions. Target enrichment
can be achieved through
hybrid capture (B),
opposing primer ampli-
cons (C), anchored multi-
plex PCR (D), and 50 to 30
imbalance (E). Hybrid cap-
ture is highly scalable
without the limitation of
primer multiplexibility
such as primer dimer for-
mation. Amplicon-based
methods have been
believed to be more sensi-
tive in low-input and low-
purity specimens
compared with hybrid
capture. However, this
could be variable with constantly optimized and diversified assay designs in recent literature. The main downside
of conventional opposing primer amplicons is inability to cover novel fusions (C). Anchored multiplex PCR (AMP)
amplifies a fusion sequence using gene-specific primer on one side and adapter-based primer on the other side,
allowing detection without the identity of the partner gene (D). Expression imbalance has been used by several
non-NGS and NGS platforms such as the Oncomine panels. In fusion-negative tumors (E, upper part), there will be
approximately equal expressions of the 30 and 50 amplicons. In fusion-positive tumors (E, lower part), the 50 re-
gion, which is incorporated by the fusion, is overrepresented in tumor transcriptome compared with the 30 re-
gion. In tissues with high endogenous expression of the wildtype gene, expression imbalance may be
obscured leading to suboptimal sensitivity.

International Thyroid Oncology Group published a pretreatment setting. The advantage of upfront
consensus document on molecular testing in thy- use of NGS may include workflow acceleration
roid cancer.103 The expert panel recommends for rapidly progressive tumors such as ATC and
applying either upfront use of comprehensive tissue conservation for small biopsies and cyto-
NGS or a multi-step approach in identifying thera- logic samples. For characterizing acquired therapy
peutic targets in advanced DTC. The advantages resistance, a comprehensive panel analysis will be
of multi-step testing, starting with a screening the most ideal for exploring various on-target and
test for the BRAF V600 E mutation followed by bypassing mechanisms.
fusion testing using NGS or FISH in BRAF-nega-
tive cases, may include lower cost and faster turn- SUMMARY
around time compared with NGS, given that BRAF
V600 E accounts for the majority of PTC and is Despite the remarkable advances in targeted ther-
mutually exclusive with kinase fusions in the apy for thyroid cancer in recent years, several
This is Your Thyroid on Drugs 69

areas remain to be elucidated by future research. 5. Wong KS, Dong F, Telatar M, et al. Papillary thyroid
Firstly, although tumors driven by ALK and ROS1 carcinoma with high-grade features versus poorly
fusions have standard targeted therapy in the pul- differentiated thyroid carcinoma: an analysis of clin-
monary setting, those of thyroid origin are not icopathologic and molecular features and
currently FDA-approved indications of ALK and outcome. Thyroid 2021;31(6):933–40.
ROS1 inhibitors. MET fusions are even rarer and 6. Ibrahimpasic T, Ghossein R, Carlson DL, et al. Out-
also in need of evidence-based therapy. Secondly, comes in patients with poorly differentiated thyroid
microsatellite instability (MSI) status and tumor carcinoma. J Clin Endocrinol Metab 2014;99(4):
mutation burden (TMB) are increasingly recog- 1245–52.
nized predictors of immunotherapy response. 7. The Cancer Genome Atlas Research Network. Inte-
However, MSI have only been reported in a small grated genomic characterization of papillary thy-
number of thyroid carcinomas with limited data roid carcinoma. Cell 2014;159(3):676–90.
on their clinical behavior.19,41,104,105 TMB, when 8. Yoo SK, Lee S, Kim SJ, et al. Comprehensive Anal-
assessed using targeted panels instead of ysis of the Transcriptional and Mutational Land-
whole-exome sequencing, is well known to be scape of Follicular and Papillary Thyroid Cancers.
affected by sequencing panel design and preana- PLoS Genet 2016;12(8):e1006239.
lytical factors such as tumor purity and tissue qual- 9. Pekova B, Sykorova V, Dvorakova S, et al. RET,
ity. Standardization of TMB assessment is an NTRK, ALK, BRAF, and MET Fusions in a large
ongoing effort in the field to provide solid guidance cohort of pediatric papillary thyroid carcinomas.
for clinical decision-making. Lastly, several intra- Thyroid 2020;30(12):1771–80.
cellular pathways implicated in the high-grade 10. Morton LM, Karyadi DM, Stewart C, et al. Radia-
transformation of thyroid carcinoma, such as the tion-related genomic profile of papillary thyroid car-
PI3K/Akt/mTOR pathway, SWI/SNF chromatin- cinoma after the Chernobyl accident. Science
remodeling complexes, and histone-modifying 2021;372(6543):eabg2538–725.
enzymes, are the targets of a rapidly growing 11. Chu YH, Wirth LJ, Farahani AA, et al. Clinicopatho-
armamentarium of novel therapeutics, which logic features of kinase fusion-related thyroid carci-
stand great promises as new treatment options nomas: an integrative analysis with molecular
in the near future. characterization. Mod Pathol 2020;33(12):2458–72.
12. Vanden Borre P, Schrock AB, Anderson PM, et al.
pediatric, adolescent, and young adult thyroid car-
ACKNOWLEDGMENT cinoma harbors frequent and diverse targetable
Dr. Chu prepared Figure 2 with materials from genomic alterations, including kinase fusions.
Massachusetts General Hospital in Boston, Mas- Oncologist 2017;22(3):255–63.
sachusetts, United States from a study mentored 13. Lee YC, Hsu CY, Lai CR, et al. NTRK-rearranged
by Dr. Peter M. Sadow. papillary thyroid carcinoma demonstrates frequent
subtle nuclear features and indeterminate cytologic
REFERENCES diagnoses. Cancer Cytopathol 2022;130(2):136–43.
14. Panebianco F, Nikitski AV, Nikiforova MN, et al.
1. Megwalu UC, Moon PK. Thyroid cancer incidence Characterization of thyroid cancer driven by known
and mortality trends in the United States: 2000- and novel ALK fusions. Endocr Relat Cancer 2019;
2018. Thyroid 2022;32(5):560–70. 26(11):803–14.
2. Lim H, Devesa SS, Sosa JA, et al. Trends in Thyroid 15. Viswanathan K, Chu YH, Faquin WC, et al. Cyto-
Cancer Incidence and Mortality in the United morphologic features of NTRK-rearranged thyroid
States, 1974-2013. JAMA 2017;317(13):1338–48. carcinoma. Cancer Cytopathol 2020;128(11):
3. Tuttle RM, Ahuja S, Avram AM, et al. Controversies, 812–27.
consensus, and collaboration in the use of (131)i 16. Abi-Raad R, Prasad ML, Adeniran AJ, et al. Fine-
therapy in differentiated thyroid cancer: a joint needle aspiration cytomorphology of papillary thy-
statement from the american thyroid association, roid carcinoma with NTRK gene rearrangement
the european association of nuclear medicine, the from a case series with predominantly indetermi-
society of nuclear medicine and molecular imag- nate cytology. Cancer Cytopathol 2020;128(11):
ing, and the european thyroid association. Thyroid 803–11.
2019;29(4):461–70. 17. Prasad ML, Vyas M, Horne MJ, et al. NTRK fusion
4. Durante C, Haddy N, Baudin E, et al. Long-term oncogenes in pediatric papillary thyroid carcinoma
outcome of 444 patients with distant metastases in northeast United States. Cancer 2016;122(7):
from papillary and follicular thyroid carcinoma: ben- 1097–107.
efits and limits of radioiodine therapy. J Clin Endo- 18. Desai MA, Mehrad M, Ely KA, et al. secretory carci-
crinol Metab 2006;91(8):2892–9. noma of the thyroid gland: report of a highly
70 Chu

aggressive case clinically mimicking undifferenti- 32. Subbiah V, Cote GJ. Advances in targeting RET-
ated carcinoma and review of the literature. Head dependent cancers. Cancer Discov 2020;10(4):
Neck Pathol 2019;13(4):562–72. 498–505.
19. Landa I, Ibrahimpasic T, Boucai L, et al. Genomic 33. Brose MS, Cabanillas ME, Cohen EE, et al. Vemur-
and transcriptomic hallmarks of poorly differenti- afenib in patients with BRAF(V600E)-positive meta-
ated and anaplastic thyroid cancers. J Clin Invest static or unresectable papillary thyroid cancer
2016;126(3):1052–66. refractory to radioactive iodine: a non-
20. Pozdeyev N, Gay LM, Sokol ES, et al. Genetic anal- randomised, multicentre, open-label, phase 2 trial.
ysis of 779 advanced differentiated and anaplastic Lancet Oncol 2016;17(9):1272–82.
thyroid cancers. Clin Cancer Res 2018;24(13): 34. Subbiah V, Kreitman RJ, Wainberg ZA, et al. Dabra-
3059–68. fenib and trametinib treatment in patients with
21. Kunstman JW, Juhlin CC, Goh G, et al. Character- locally advanced or metastatic BRAF V600-mutant
ization of the mutational landscape of anaplastic anaplastic thyroid cancer. J Clin Oncol 2018;
thyroid cancer via whole-exome sequencing. Hum 36(1):7–13.
Mol Genet 2015;24(8):2318–29. 35. Subbiah V, Kreitman RJ, Wainberg ZA, et al. Dabra-
22. Xu B, David J, Dogan S, et al. Primary high-grade fenib plus trametinib in patients with BRAF V600E-
non-anaplastic thyroid carcinoma: a retrospective mutant anaplastic thyroid cancer: updated analysis
study of 364 cases. Histopathology 2022;80(2): from the phase II ROAR basket study. Ann Oncol
322–37. 2022;33(4):406–15.
23. Kumari S, Adewale R, Klubo-Gwiezdzinska J. The 36. Poulikakos PI, Zhang C, Bollag G, et al. RAF inhib-
molecular landscape of hürthle cell thyroid cancer itors transactivate RAF dimers and ERK signalling
is associated with altered mitochondrial function-a in cells with wild-type BRAF. Nature 2010;
comprehensive review. Cells 2020;9(7):1570. 464(7287):427–30.
24. Ganly I, Makarov V, Deraje S, et al. Integrated 37. Purdie KJ, Proby CM, Rizvi H, et al. The Role of Hu-
genomic analysis of hürthle cell cancer reveals man Papillomaviruses and Polyomaviruses in
oncogenic drivers, recurrent mitochondrial muta- BRAF-Inhibitor Induced Cutaneous Squamous
tions, and unique chromosomal landscapes. Can- Cell Carcinoma and Benign Squamoproliferative
cer Cell 2018;34(2):256–70.e255. Lesions. Front Microbiol 2018;9:1806.
25. Gopal RK, Kübler K, Calvo SE, et al. Widespread 38. Cabanillas ME, Dadu R, Iyer P, et al. Acquired sec-
chromosomal losses and mitochondrial DNA alter- ondary RAS mutation in BRAF(V600E)-mutated thy-
ations as genetic drivers in hürthle cell carcinoma. roid cancer patients treated with BRAF inhibitors.
Cancer Cell 2018;34(2):242–55.e245. Thyroid 2020;30(9):1288–96.
26. Gasparre G, Porcelli AM, Bonora E, et al. Disruptive 39. Ho AL, Grewal RK, Leboeuf R, et al. Selumetinib-
mitochondrial DNA mutations in complex I subunits enhanced radioiodine uptake in advanced thyroid
are markers of oncocytic phenotype in thyroid tu- cancer. N Engl J Med 2013;368(7):623–32.
mors. Proc Natl Acad Sci U S A 2007;104(21): 40. Tchekmedyian V, Dunn L, Sherman E, et al.
9001–6. Enhancing radioiodine incorporation in BRAF-
27. Cruz-Migoni A, Canning P, Quevedo CE, et al. mutant, radioiodine-refractory thyroid cancers with
Structure-based development of new RAS-effector vemurafenib and the anti-ErbB3 monoclonal anti-
inhibitors from a combination of active and inactive body CDX-3379: results of a pilot clinical trial. Thy-
RAS-binding compounds. Proc Natl Acad Sci U S roid 2022;32(3):273–82.
A 2019;116(7):2545–50. 41. Qiao PP, Tian KS, Han LT, et al. Correlation of
28. Liu J, Kang R, Tang D. The KRAS-G12C inhibitor: mismatch repair deficiency with clinicopathological
activity and resistance. Cancer Gene Ther 2022; features and programmed death-ligand 1 expression
29(7):875–8. in thyroid carcinoma. Endocrine 2022;76(3):660–70.
29. Stransky N, Cerami E, Schalm S, et al. The land- 42. Wirth LJ, Sherman E, Robinson B, et al. Efficacy of
scape of kinase fusions in cancer. Nat Commun selpercatinib in RET-altered thyroid cancers.
2014;5:4846. N Engl J Med 2020;383(9):825–35.
30. Schlumberger M, Tahara M, Wirth LJ, et al. Len- 43. Subbiah V, Hu MI, Wirth LJ, et al. Pralsetinib for pa-
vatinib versus placebo in radioiodine-refractory tients with advanced or metastatic RET-altered thy-
thyroid cancer. N Engl J Med 2015;372(7): roid cancer (ARROW): a multi-cohort, open-label,
621–30. registrational, phase 1/2 study. Lancet Diabetes
31. Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in Endocrinol 2021;9(8):491–501.
radioactive iodine-refractory, locally advanced or 44. Hong DS, DuBois SG, Kummar S, et al. Larotrecti-
metastatic differentiated thyroid cancer: a rando- nib in patients with TRK fusion-positive solid tu-
mised, double-blind, phase 3 trial. Lancet 2014; mours: a pooled analysis of three phase 1/2
384(9940):319–28. clinical trials. Lancet Oncol 2020;21(4):531–40.
This is Your Thyroid on Drugs 71

45. Laetsch TW, DuBois SG, Mascarenhas L, et al. Lar- anaplastic thyroid carcinoma. J Clin Oncol 2015;
otrectinib for paediatric solid tumours harbouring 33(20):e84–7.
NTRK gene fusions: phase 1 results from a multi- 58. Hillier K, Hughes A, Shamberger RC, et al. A novel
centre, open-label, phase 1/2 study. Lancet Oncol ALK fusion in pediatric medullary thyroid carci-
2018;19(5):705–14. noma. Thyroid 2019;29(11):1704–7.
46. Waguespack SG, Drilon A, Lin JJ, et al. Efficacy 59. Dagogo-Jack I, Rooney M, Lin JJ, et al. Treatment
and safety of larotrectinib in patients with TRK with next-generation ALK inhibitors fuels plasma
fusion-positive thyroid carcinoma. Eur J Endocrinol ALK mutation diversity. Clin Cancer Res 2019;
2022;186(6):631–43. 25(22):6662–70.
47. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib 60. Toyokawa G, Seto T. Updated evidence on the
in patients with advanced or metastatic NTRK mechanisms of resistance to ALK inhibitors and stra-
fusion-positive solid tumours: integrated analysis tegies to overcome such resistance: clinical and
of three phase 1-2 trials. Lancet Oncol 2020; preclinical data. Oncol Res Treat 2015;38(6):291–8.
21(2):271–82. 61. Lim SM, Chang H, Yoon MJ, et al. A multicenter,
48. Drilon A, Li G, Dogan S, et al. What hides behind phase II trial of everolimus in locally advanced or
the MASC: clinical response and acquired resis- metastatic thyroid cancer of all histologic subtypes.
tance to entrectinib after ETV6-NTRK3 identification Ann Oncol 2013;24(12):3089–94.
in a mammary analogue secretory carcinoma 62. Hanna GJ, Busaidy NL, Chau NG, et al. Genomic
(MASC). Ann Oncol 2016;27(5):920–6. correlates of response to everolimus in aggressive
49. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of radioiodine-refractory thyroid cancer: a phase II
larotrectinib in TRK fusion-positive cancers in study. Clin Cancer Res 2018;24(7):1546–53.
adults and children. N Engl J Med 2018;378(8): 63. Schneider TC, de Wit D, Links TP, et al. Everolimus
731–9. in patients with advanced follicular-derived thyroid
50. Russo M, Misale S, Wei G, et al. Acquired resis- cancer: results of a phase II clinical trial. J Clin En-
tance to the TRK inhibitor entrectinib in colorectal docrinol Metab 2017;102(2):698–707.
cancer. Cancer Discov 2016;6(1):36–44. 64. Roman S, Lin R, Sosa JA. Prognosis of medullary
51. Hong DS, Bauer TM, Lee JJ, et al. Larotrectinib in thyroid carcinoma: demographic, clinical, and
adult patients with solid tumours: a multi-centre, pathologic predictors of survival in 1252 cases.
open-label, phase I dose-escalation study. Ann On- Cancer 2006;107(9):2134–42.
col 2019;30(2):325–31. 65. Stamatakos M, Paraskeva P, Stefanaki C, et al.
52. Florou V, Nevala-Plagemann C, Whisenant J, et al. Medullary thyroid carcinoma: The third most com-
clinical activity of selitrectinib in a patient with mam- mon thyroid cancer reviewed. Oncol Lett 2011;
mary analogue secretory carcinoma of the parotid 2(1):49–53.
gland with secondary resistance to entrectinib. 66. Wells SA Jr, Asa SL, Dralle H, et al. Revised amer-
J Natl Compr Canc Netw 2021;19(5):478–82. ican thyroid association guidelines for the manage-
53. Murray BW, Rogers E, Zhai D, et al. Molecular char- ment of medullary thyroid carcinoma. Thyroid 2015;
acteristics of repotrectinib that enable potent inhibi- 25(6):567–610.
tion of TRK fusion proteins and resistant mutations. 67. Gujral TS, Singh VK, Jia Z, et al. Molecular mecha-
Mol Cancer Ther 2021;20(12):2446–56. nisms of RET receptor-mediated oncogenesis in
54. Demeure MJ, Aziz M, Rosenberg R, et al. Whole- multiple endocrine neoplasia 2B. Cancer Res
genome sequencing of an aggressive BRAF wild- 2006;66(22):10741–9.
type papillary thyroid cancer identified EML4-ALK 68. Chu YH, Lloyd RV. Medullary thyroid carcinoma:
translocation as a therapeutic target. World J recent advances including MicroRNA expression.
Surg 2014;38(6):1296–305. Endocr Pathol 2016;27(4):312–24.
55. de Salins V, Loganadane G, Joly C, et al. Complete 69. Leboulleux S, Travagli JP, Caillou B, et al. Medullary
response in anaplastic lymphoma kinase- thyroid carcinoma as part of a multiple endocrine
rearranged oncocytic thyroid cancer: a case report neoplasia type 2B syndrome: influence of the stage
and review of literature. World J Clin Oncol 2020; on the clinical course. Cancer 2002;94(1):44–50.
11(7):495–503. 70. Moura MM, Cavaco BM, Pinto AE, et al. Correlation
56. Leroy L, Bonhomme B, Le Moulec S, et al. Remark- of RET somatic mutations with clinicopathological
able response to ceritinib and brigatinib in an features in sporadic medullary thyroid carcinomas.
anaplastic lymphoma kinase-rearranged Br J Cancer 2009;100(11):1777–83.
anaplastic thyroid carcinoma previously treated 71. Grubbs EG, Ng PK, Bui J, et al. RET fusion as a
with crizotinib. Thyroid 2020;30(2):343–4. novel driver of medullary thyroid carcinoma.
57. Godbert Y, Henriques de Figueiredo B, Bonichon F, J Clin Endocrinol Metab 2015;100(3):788–93.
et al. Remarkable response to crizotinib in woman 72. Kasaian K, Wiseman SM, Walker BA, et al. Putative
with anaplastic lymphoma kinase-rearranged BRAF activating fusion in a medullary thyroid
72 Chu

cancer. Cold Spring Harb Mol Case Stud 2016; 86. Saliba M, Katabi N, Dogan S, et al. NRAS Q61R
2(2):a000729. immunohistochemical staining in thyroid pathology:
73. Ji JH, Oh YL, Hong M, et al. Identification of driving sensitivity, specificity and utility. Histopathology
ALK fusion genes and genomic landscape of med- 2021;79(4):650–60.
ullary thyroid cancer. PLoS Genet 2015;11(8): 87. Crescenzi A, Fulciniti F, Bongiovanni M, et al. De-
e1005467. tecting N-RAS Q61R Mutated Thyroid Neoplasias
74. Jr SAW, Robinson BG, Gagel RF, et al. Vandetanib by Immunohistochemistry. Endocr Pathol 2017;
in patients with locally advanced or metastatic 28(1):71–4.
medullary thyroid cancer: a randomized, double- 88. Solomon JP, Linkov I, Rosado A, et al. NTRK fusion
blind phase III trial. J Clin Oncol 2012;30(2): detection across multiple assays and 33,997
134–41. cases: diagnostic implications and pitfalls. Mod
75. Elisei R, Schlumberger MJ, Müller SP, et al. Cabo- Pathol 2020;33(1):38–46.
zantinib in progressive medullary thyroid cancer. 89. Solomon JP, Hechtman JF. Detection of NTRK fu-
J Clin Oncol 2013;31(29):3639–46. sions: merits and limitations of current diagnostic
76. Brose MS, Robinson B, Sherman SI, et al. Cabo- platforms. Cancer Res 2019;79(13):3163–8.
zantinib for radioiodine-refractory differentiated 90. Park G, Kim TH, Lee HO, et al. Standard immuno-
thyroid cancer (COSMIC-311): a randomised, histochemistry efficiently screens for anaplastic
double-blind, placebo-controlled, phase 3 trial. lymphoma kinase rearrangements in differentiated
Lancet Oncol 2021;22(8):1126–38. thyroid cancer. Endocr Relat Cancer 2015;22(1):
77. Solomon BJ, Tan L, Lin JJ, et al. RET Solvent Front 55–63.
Mutations Mediate Acquired Resistance to Selec- 91. Chou A, Fraser S, Toon CW, et al. A detailed clinico-
tive RET Inhibition in RET-Driven Malignancies. pathologic study of ALK-translocated papillary thy-
J Thorac Oncol 2020;15(4):541–9. roid carcinoma. Am J Surg Pathol 2015;39(5):
78. Subbiah V, Shen T, Terzyan SS, et al. Structural ba- 652–9.
sis of acquired resistance to selpercatinib and pral- 92. Yang SR, Aypar U, Rosen EY, et al. A performance
setinib mediated by non-gatekeeper RET comparison of commonly used assays to detect
mutations. Ann Oncol 2021;32(2):261–8. RET fusions. Clin Cancer Res 2021;27(5):1316–28.
79. Drilon AE, Zhai D, Rogers E, et al. The next- 93. Nozaki Y, Yamamoto H, Iwasaki T, et al. Clinico-
generation RET inhibitor TPX-0046 is active in pathological features and immunohistochemical
drug-resistant and naı̈ve RET-driven cancer utility of NTRK-, ALK-, and ROS1-rearranged papil-
models. J Clin Oncol 2020;38(15_suppl):3616. lary thyroid carcinomas and anaplastic thyroid car-
80. Moura MM, Cavaco BM, Pinto AE, et al. High prev- cinomas. Hum Pathol 2020;106:82–92.
alence of RAS mutations in RET-negative sporadic 94. Lee YC, Chen JY, Huang CJ, et al. Detection of
medullary thyroid carcinomas. J Clin Endocrinol NTRK1/3 rearrangements in papillary thyroid carci-
Metab 2011;96(5):E863–8. noma using immunohistochemistry, fluorescent in
81. Tate JG, Bamford S, Jubb HC, et al. COSMIC: the situ hybridization, and next-generation
catalogue of somatic mutations in cancer. Nucleic sequencing. Endocr Pathol 2020;31(4):348–58.
Acids Res 2019;47(D1):D941–7. 95. Rosenbaum JN, Bloom R, Forys JT, et al. Genomic
82. Moore AR, Rosenberg SC, McCormick F, et al. heterogeneity of ALK fusion breakpoints in non-
RAS-targeted therapies: is the undruggable small-cell lung cancer. Mod Pathol 2018;31(5):
drugged? Nat Rev Drug Discov 2020;19(8): 791–808.
533–52. 96. Chu YH, Barbee J, Yang SR, et al. Clinical utility
83. Hong DS, Cabanillas ME, Wheler J, et al. Inhibition and performance of an ultrarapid multiplex RNA-
of the Ras/Raf/MEK/ERK and RET kinase pathways based assay for detection of ALK, ROS1, RET,
with the combination of the multikinase inhibitor sor- and NTRK1/2/3 rearrangements and MET exon 14
afenib and the farnesyltransferase inhibitor tipifar- skipping alterations. J Mol Diagn 2022;24(6):
nib in medullary and differentiated thyroid 642–54.
malignancies. J Clin Endocrinol Metab 2011; 97. Tong Y, Zhao Z, Liu B, et al. 5’/3’ imbalance strat-
96(4):997–1005. egy to detect ALK fusion genes in circulating tumor
84. Loo E, Khalili P, Beuhler K, et al. BRAF V600E RNA from patients with non-small cell lung cancer.
mutation across multiple tumor types: correlation J Exp Clin Cancer Res 2018;37(1):68.
between DNA-based sequencing and mutation- 98. Liu Y, Wu S, Shi X, et al. Clinical evaluation of the
specific immunohistochemistry. Appl Immunohisto- effectiveness of fusion-induced asymmetric tran-
chem Mol Morphol 2018;26(10):709–13. scription assay-based reverse transcription droplet
85. Ritterhouse LL, Barletta JA. BRAF V600E mutation- digital PCR for ALK detection in formalin-fixed
specific antibody: a review. Semin Diagn Pathol paraffin-embedded samples from lung cancer.
2015;32(5):400–8. Thorac Cancer 2020;11(8):2252–61.
This is Your Thyroid on Drugs 73

99. Lira ME, Choi YL, Lim SM, et al. A single-tube multi- cancer: defining advanced thyroid cancer and its tar-
plexed assay for detecting ALK, ROS1, and RET fu- geted treatment. Head Neck 2022;44(6):1277–300.
sions in lung cancer. J Mol Diagn 2014;16(2): 104. Wong KS, Lorch JH, Alexander EK, et al. Clinico-
229–43. pathologic features of mismatch repair-deficient
100. Rogers TM, Arnau GM, Ryland GL, et al. Multi- anaplastic thyroid carcinomas. Thyroid 2019;
plexed transcriptome analysis to detect ALK, 29(5):666–73.
ROS1 and RET rearrangements in lung cancer. 105. Genutis LK, Tomsic J, Bundschuh RA, et al. Micro-
Sci Rep 2017;7:42259. satellite instability occurs in a subset of follicular
101. Baitei EY, Zou M, Al-Mohanna F, et al. Aberrant thyroid cancers. Thyroid 2019;29(4):523–9.
BRAF splicing as an alternative mechanism for
oncogenic B-Raf activation in thyroid carcinoma. FURTHER READING
J Pathol 2009;217(5):707–15.
102. Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive 1. Subbiah Vivek, Wolf Jürgen, Konda Bhavana, et al.
cancers and TRK inhibitor therapy. Nat Rev Clin Tumour-agnostic efficacy and safety of selpercatinib
Oncol 2018;15(12):731–47. in patients with RET fusion-positive solid tumours other
103. Shonka DC Jr, Ho A, Chintakuntlawar AV, et al. amer- than lung or thyroid tumours (LIBRETTO-001): a
ican head and neck society endocrine surgery sec- phase 1/2, open-label, basket trial. Lancet Oncology
tion and international thyroid oncology group 2022;23(10):1261–73. https://doi.org/10.1016/S1470-
consensus statement on mutational testing in thyroid 2045(22)00541-1. In this issue.

You might also like