You are on page 1of 10

Expert Review of Anticancer Therapy

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iery20

Evaluating new treatments for anaplastic thyroid


cancer

Andrés Coca-Pelaz, Juan P. Rodrigo, Fernando Lopez, Jatin P. Shah, Carl E.


Silver, Abir Al Ghuzlan, C.Willemien Menke-van der Houven van Oordt,
Robert C. Smallridge, Ashok R. Shaha, Peter Angelos, William M. Mendenhall,
Cesare Piazza, Kerry D. Olsen, June Corry, Ralph P. Tufano, Alvaro Sanabria,
Sandra Nuyts, Cherie-Ann Nathan, Vincent Vander Poorten, Fernando
Luiz Dias, Carlos Suarez, Nabil F. Saba, Pim de Graaf, Michelle D. Williams,
Alessandra Rinaldo & Alfio Ferlito

To cite this article: Andrés Coca-Pelaz, Juan P. Rodrigo, Fernando Lopez, Jatin P. Shah,
Carl E. Silver, Abir Al Ghuzlan, C.Willemien Menke-van der Houven van Oordt, Robert C.
Smallridge, Ashok R. Shaha, Peter Angelos, William M. Mendenhall, Cesare Piazza, Kerry
D. Olsen, June Corry, Ralph P. Tufano, Alvaro Sanabria, Sandra Nuyts, Cherie-Ann Nathan,
Vincent Vander Poorten, Fernando Luiz Dias, Carlos Suarez, Nabil F. Saba, Pim de Graaf,
Michelle D. Williams, Alessandra Rinaldo & Alfio Ferlito (2022) Evaluating new treatments
for anaplastic thyroid cancer, Expert Review of Anticancer Therapy, 22:11, 1239-1247, DOI:
10.1080/14737140.2022.2139680

To link to this article: https://doi.org/10.1080/14737140.2022.2139680

Published online: 09 Nov 2022. Submit your article to this journal

Article views: 403 View related articles

View Crossmark data Citing articles: 2 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iery20
EXPERT REVIEW OF ANTICANCER THERAPY
2022, VOL. 22, NO. 11, 1239–1247
https://doi.org/10.1080/14737140.2022.2139680

REVIEW

Evaluating new treatments for anaplastic thyroid cancer


Andrés Coca-Pelaza, Juan P. Rodrigoa, Fernando Lopeza, Jatin P. Shahb, Carl E. Silverc, Abir Al Ghuzland, C.
Willemien Menke-van der Houven van Oordte, Robert C. Smallridgef, Ashok R. Shahab, Peter Angelosg,
William M. Mendenhallh, Cesare Piazzai, Kerry D. Olsenj, June Corryk, Ralph P. Tufanol, Alvaro Sanabriam,
Sandra Nuytsn,o, Cherie-Ann Nathanp, Vincent Vander Poortenq,r, Fernando Luiz Diass, Carlos Suarezt, Nabil F. Sabau,
Pim de Graafv, Michelle D. Williamsw, Alessandra Rinaldox and Alfio Ferlitoy
a
Department of Otolaryngology, Hospital Universitario Central de Asturias, University of Oviedo, ISPA, IUOPA, CIBERONC, Oviedo, Spain; bHead and
Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; cDepartments of Surgery and Otolaryngology-
Head and Neck Surgery, Albert Einstein College of Medicine, Bronx, NY, USA; dDepartment of Medical Biology and Pathology, Institut Gustave
Roussy, Villejuif, France; eDepartment of Medical Oncology, Amsterdam Center for Endocrine and Neuro Endocrine Tumors (ACcENT), Amsterdam
UMC location Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, Netherlands; fDivision of Medicine,
Mayo Clinic School of Medicine, Jacksonville, FL, USA; gDepartment of Surgery and MacLean Center for Clinical Medical Ethics, The University of
Chicago, Chicago, IL, USA; hDepartment of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL, USA;
i
Otorhinolaryngology-Head and Neck Surgery Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health,
University of Brescia, ASST Spedali Civili, Brescia, Italy; jDepartment of Otorhinolaryngology, Mayo Clinic, Rochester, MN, USA; kDepartment
Radiation Oncology, GenesisCare St Vincent’s Hospital, Melbourne, Australia; lFPG Thyroid and Parathyroid Center, Division of Head and Neck
Endocrine Surgery, The Sarasota Memorial Health Care System, Sarasota, FL, USA; mDepartment of Surgery, Universidad de Antioquia, CEXCA
Centro de Excelencia en Enfermedades de Cabeza y cuello, Medellín, Colombia; nLaboratory of Experimental Radiotherapy, Department of
Oncology, University of Leuven, Belgium; oDepartment of Radiation Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven,
Belgium; pDepartment of Otolaryngology-Head and Neck Surgery, Louisiana State University-Health Shreveport, Shreveport, LA, USA; qDepartment
of Oncology, Section Head and Neck Oncology, KU Leuven, Leuven, Belgium; rOtorhinolaryngology, Head and Neck Surgery, University Hospitals
Leuven, Leuven Cancer Institute, Leuven, Belgium; sHead and Neck Surgery Section, Instituto Nacional do Câncer (INCA), Pontifícia Universidade
Católica do Rio de Janeiro, Rio de Janeiro, Brazil; tInstituto de Investigación Sanitaria del Principado de Asturias, IUOPA, CIBERONC, Oviedo, Spain;
u
Department of Hematology and Medical Oncology, The Winship Cancer Institute of Emory University, Atlanta, GA, USA; vCancer Center
Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
w
Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA; xUniversity of Udine School of Medicine,
Udine, Italy; yCoordinator of the International Head Neck Scientific Group, Padua, Italy

ABSTRACT ARTICLE HISTORY


Introduction: Anaplastic thyroid cancer (ATC) is one of the most lethal diseases known to humans with Received 20 June 2022
a median survival of 5 months. The American Thyroid Association (ATA) recently published guidelines Accepted 20 October 2022
for the treatment of this dreadful thyroid malignancy. KEYWORDS
Areas covered: This review presents the current therapeutic landscape of this challenging disease. We Anaplastic thyroid cancer;
also present the results from trials published over the last five years and summarize currently active immune checkpoint
clinical trials. inhibitors; immunotherapy;
Expert opinion: Recent attempts to improve the prognosis of these tumors are moving toward targeted therapy; treatment;
personalized medicine, basing the treatment decision on the specific genetic profile of the individual Tyrosine-kinase inhibitors
tumor. The positive results of dabrafenib and trametinib for ATC harboring the BRAF V600E mutation
have provided a useful treatment option. For the other genetic profiles, different drugs are available
and can be used to individualize the treatment, likely using drug combinations. Combinations of drugs
act on different molecular pathways and achieve inhibition at separate areas. With new targeted
therapies, average survival has improved considerably and death from local disease progression or
airway compromise is less likely with improvement in quality of life. Unfortunately, the results remain
poor in terms of survival.

1. Introduction related deaths [2,3]. Typically, it presents as a rapidly growing


thyroid mass, with local compression symptoms (neck pain,
Anaplastic thyroid cancer (ATC) is the most aggressive and dysphagia, and stridor), generally affecting patients in their
lethal disease of the thyroid gland, with a reported median sixth and seventh decades of life [4]. ATC is composed of
survival of only 5 months and the median 1-year survival of undifferentiated thyroid cells and typically presents at an
20% [1]. While accounting for less than 2% of thyroid malig­ advanced stage at the time of diagnosis [5]. ATC may evolve
nancies, ATC is responsible for 25% of all thyroid cancer- from a differentiated or poorly differentiated thyroid cancer,

CONTACT Andrés Coca-Pelaz acocapelaz@yahoo.es Department of Otolaryngology, Hospital Universitario Central de Asturias, University of Oviedo, ISPA,
IUOPA, CIBERONC, Av. Roma, s/n, 33011 Oviedo, Asturias, Spain
This article was written by members and invitees of the International Head and Neck Scientific Group (www.IHNSG.com).
© 2022 Informa UK Limited, trading as Taylor & Francis Group
1240 A. COCA-PELAZ ET AL.

In a report by de Ridder et al., all patients with ATC


Article highlights between 1989 and 2016 were identified from the
● Anaplastic thyroid cancer is one of the most lethal diseases known to
Netherlands Cancer Registry, concluding that the incidence
humans with a median survival of 5 months. has increased slightly in the last 30 years, but there is a sub­
● Personalized medicine is probably the key for improving the group of patients with longer survival, and they correspond to
outcome.
● Dabrafenib and trametinib for tumors harboring the BRAF V600E
cases with limited disease and who underwent a combination
mutation have provided a useful treatment option. of two or three treatment modalities [11].
● Combinations of drugs acting on different molecular pathways are In light of these disappointing results, it is the purpose of
the last attempt for fighting this cancer.
this review to present the current therapeutic landscape of
this challenging disease, focusing on the new therapeutic
options that are currently being used or investigated, with
the aim of synthesizing the latest advances, showing the
and while maintaining the genomic profile of the original effort that is being carried out to improve the prognosis of
tumor, however, ATC does not usually exhibit histologic char­ this disease. We also present the results from recent pub­
acteristics of follicular cells [6,7]. Poorly differentiated thyroid lished trials over the last five years and summarize the
cancer and the tall cell variant of papillary thyroid cancer were currently active clinical trials.
the most common subtypes found to be associated with these
mixed/transformed ATC sub-types in the Memorial Sloan
Kettering Cancer Center series [8].
2. Treatment options
In 2019, Janz et al. assessed the incidence of ATC using
the Surveillance Epidemiology and End Results (SEER) data­ In 2021, the American Thyroid Association (ATA) published
base from 1973 to 2014, identifying 1527 patients in the the most recent guidelines for management of patients with
database and a noted increased incidence from 0.2 per ATC, recommending total thyroidectomy for patients with
1,000,000 people in 1973 to 1.2 per 1,000,000 people in localized tumors confined to the thyroid gland in whom
2014 [9]. This finding has not been associated with a complete resection is feasible, followed by (chemo)radio­
a change in survival. Median disease-specific survival (DSS) therapy. For more advanced disease, the treatment must be
did not improve, being 4 months (95% CI: 2.26–5.74) from individualized according to the patient’s general medical
1995 to 1999 and remaining 4 months (95% CI: 3.26–4.74) in condition and his/her goals of care [3]. In relation to the
the period from 2010 to 2014. Patients who were not treated use of systemic therapy, in patients with unresectable or
surgically had a DSS of 2 months (95% CI: 1.65–2.35) com­ advanced disease wishing aggressive treatment, these
pared to 6 months (95% CI: 3.91–8.09) for patients treated by guidelines suggest (recommendation 19) early initiation of
subtotal or near-total thyroidectomy and 10 months (95% CI: cytotoxic chemotherapy as an initial and potentially brid­
7.70–12.30) for those who underwent a total thyroidectomy. ging approach until mutational interrogation results and/or
The authors point out that this increase in incidence could be mutationally specified therapies might be available [3].
explained by the expansion of the SEER database and Given the unchanged prognosis of these tumors in the
because the population of the United States is increasing. past few decades, a focus on the biology and the genetic
Furthermore, the increase could be due to improved diag­ profile of ATC is necessary to devise new therapeutic stra­
nostic guidelines for healthcare providers. Another significant tegies that impact key elements in the oncogenetic path­
finding of the article is that 47.5% of the patients did not way to improve patient outcomes. Along those lines, the
have any surgery and the percent of patients that received most commonly identified BRAF V600E mutation analysis
surgery remained stable during the study period. Conversely, should be performed within the context of next-
the percent of patients receiving no surgical management generation sequencing [7].
increased from 1995 to 1999, where it was 37.6% to 2000– Currently, targeting a number of pathways appears to
2004, where it was 55.5% (P < 0.001), showing the effort that offer some promise including multitarget tyrosine-kinase
is being made to find an effective treatment. inhibitors (TKI), epidermal growth factor receptor (EGFR),
Nevertheless, in a single-institution cohort study of 479 BRAF, mammalian target of rapamycin (mTOR) inhibitors,
patients with ATC spanning nearly 20 years, 1- and 2-year targeted peroxisome proliferator activated receptor-gamma
survival significantly increased from 35% and 18% in the (PPARγ) ligand molecules, and vascular disruptor molecules,
2000–2013 time period (n = 227) to 47% and 25% during such as vascular endothelial growth factor receptor (VEGFR)
2014–2016 (n = 100) and 59% and 42% in the 2017–2019 inhibitor and immune checkpoint inhibitor targeting pro­
time period (n = 152), respectively. The determining differen­ grammed cell-death 1 (PD-1) and its ligands (PD-L1). Each
tial fact was that more patients received targeted therapy in strategy has a distinct anti-tumor mechanism as well as
the 2017–2019 group vs the 2000–2013 and 2014–2016 specific side effects, which must be taken into account
groups (61%, 9%, and 43%, respectively). The median OS for when considering therapeutic options. In the following sec­
patients treated with targeted therapy, regardless of their tions, we will review these targeted agents relying on stu­
grouping, was 1.31 years (15.7 months) (95% CI, 1.07– dies published between 2016 and 2021. The Preferred
1.99 years) compared with 0.63 years (7.6 months) (95% CI, Reporting Items for Systematic Review and Meta-Analyses
0.52–0.72 years) in patients not having received any targeted (PRISMA) method was used to analyze the literature
therapy [10]. (Figure 1) [12].
EXPERT REVIEW OF ANTICANCER THERAPY 1241

Records identified through Records excluded


database search (n = 1319)
(n = 1345)

Full-text articles assessed Full-text articles


for eligibility excluded
(n = 26) (n = 10)

Articles included in the


study
(n = 16)

Figure 1. Flow chart showing the study selection process for our PRISMA systematic review.

2.1. Targeted therapy label, single-arm, multicenter, phase II trial of lenvatinib for
patients with ATC: Thirty-four patients were enrolled, all over
2.1.1. Multitargeted TKI
the age of 65 years, and 62% females and 71% had prior
2.1.1.1. Lenvatinib. Lenvatinib is an oral multitarget TKI that
anticancer therapy, 71% surgery and 65% radiation therapy
inhibits VEGF receptors 1–3, FGF receptors 1–4, PDGF recep­
[16]. More than half of the patients experienced tumor shrink­
tor-α, and RET and KIT proto-oncogenes [13].
age (partial response and stable disease), but the confirmed
We identified three clinical trials in the last 5 years using
overall response rate (ORR) was 0% as there were no patients
lenvatinib for advanced thyroid cancer. Tahara et al. per­
with a confirmed partial or complete response in the interim
formed a phase 2, single-arm, open-label study in patients
analysis set. Therefore, the study was halted based on the
with thyroid cancer that included 17 cases of ATC [14]. The
prespecified criteria for futility, as the minimum ORR threshold
most frequent TEAEs were decreased appetite (82%), hyper­
of 15% was not met. The median PFS was 2.6 months (95% CI,
tension (82%), fatigue (59%), nausea (59%), and proteinuria
1.4–2.8); while the median overall survival (OS) was 3.2 months
(59%). The median PFS was 7.4 months [95% CI: 1.7–12.9], the
(95% CI, 2.8–8.2). The most common treatment-related AEs
median OS was 10.6 months (95% CI: 3.8–19.8), and the objec­
were hypertension (56%), decreased appetite (29%), fatigue
tive response rate was 24%. Takahashi et al. published in 2019
(29%), and stomatitis (29%).
a phase II study including 51 patients who received lenvatinib
in advanced thyroid cancer [15]. Seventeen had ATC, with
a mean age of 65 years. Eleven of the patients were women. 2.1.1.2. Sorafenib. Sorafenib inhibits vascular endothelial
82% of the patients had previously undergone surgery, 41% growth factor receptors (VEGFR) 1, 2, and 3; platelet-derived
chemotherapy and 53% radiotherapy. All patients had treat­ growth factor receptor (PDGFR); mitogen-activated protein
ment-related adverse events (AEs), 88% of which were grade 3 kinase (MAPK) members BRAF and RET; chimeric protein RET/
or 4. Serious AEs were deemed related to lenvatinib in 13 PTC; and c-KIT [17].
patients, with 3 deaths, 1 life-threatening AE, and 13 patients In 2017, Ito et al. conducted an uncontrolled, open-label,
requiring inpatient hospitalization or prolongation of existing multicenter, single-arm, phase II clinical trial to evaluate sor­
hospitalization. The most common side effects of this drug afenib in Japanese patients with medullary thyroid cancer and
were hypertension and decreased appetite in 82% of the ATC [18]. Eighteen patients were included, of which 10 had
patients, as well as fatigue, proteinuria, and nausea in 59%. ATC. The most common drug-related AEs in patients with ATC
Additional adverse reactions included palmar-plantar erythro­ were palmar-plantar erythrodysesthesia (50%), rash (40%),
dysesthesia syndrome or stomatitis, which were seen in 47% hypertension (50%), and weight loss (50%). Median PFS was
of patients. The objective response rate was 24%, the disease 2.8 months (95% CI 0.7–5.6), and median OS was 5.0 months
control rate 94%, and the overall clinical benefit 71%, while (95% CI 0.7–5.7). Objective response (complete and partial
most of them were classified as having stable disease > response) and disease control rates (stable disease) were 0%
11 weeks. The median progression-free survival (PFS) for the and 40%, respectively. They concluded that sorafenib did not
ATC group was 7.4 months. Wirth et al. published an open- seem to be effective for ATC.
1242 A. COCA-PELAZ ET AL.

Sherman et al. evaluated the combination of sorafenib and 41–89%), with complete response in 1 patient and partial
temsirolimus in patients deemed to have radioactive iodine- response in 10 patients. The most common AEs were fatigue
refractory thyroid cancer in a phase II study [19]. Temsirolimus (38%), pyrexia (37%), and nausea (35%). With the combina­
is an inhibitor of mTOR, and it binds to an abundant intracel­ tion of BRAF and MEK inhibition resulting in a promising
lular protein, FKBP-12, forming a complex that inhibits mTOR clinical activity, this approach is now approved in the man­
complex 1 (mTORC1) signaling [20]. Interrupting mTOR signal­ agement of BRAF-positive ATC by the US Food and Drug
ing inhibits the protein synthesis regulating cell cycle progres­ Administration (FDA).
sion and angiogenesis [21,22]. mTORC1 activity in the thyroid In 2019, Wang et al. published a case series using this
cells is required for the proliferative effects of thyroid- treatment in 6 patients who were treated pre-operatively
stimulating hormone (TSH) in vitro and in vivo, and its inhibi­ with neoadjuvant dabrafenib plus trametinib, with initially
tion suppresses growth of thyroid cancer cell lines in vitro unresectable BRAFV600E-mutated ATC [32]. Thereafter, they
[23,24]. There were 36 patients treated. One of the 2 ATC underwent a surgical resection of the tumor and adjuvant
patients had a partial response and was on study for chemoradiation (3 patients also received pembrolizumab).
6.9 months. This patient had been previously treated with Remarkably, complete surgical resection was achieved in all
paclitaxel, pazopanib, and radiation therapy and was cases. The OS at 6 and 12 months was 100% and 83%, respec­
81 years of age. The most common grade 3 and 4 toxicities tively. The locoregional control rate was 100% and 2 patients
associated with these drugs included hyperglycemia, fatigue, died of distant metastases without evidence of locoregional
anemia, and oral mucositis. disease at 8 and 14 months from diagnosis. The remaining 4
patients had no evidence of disease at last follow-up (median:
2.1.1.3. Sunitinib. Sunitinib is an oral multitargeted TKI 16.5 months, range: 7.8–26.0 months). These results open the
against VEGFRs (1 and 2), PDGFRs (α and β), c KIT, FMS-like question of pre-operative (Induction) therapy as a possible
tyrosine kinase-3 (FLT3), glial cell-line derived neurotrophic valid option in the management of resectable disease.
factor receptor (RET), and the receptor of macrophage colony-
stimulating factor (CSF1R) [25]. Ravaud et al. published in 2017 2.1.3. Targeting NTRK, RET, and ALK fusions
the data from the THYSU study, which investigated the use of Fusions that involve NTRK, RET, and ALK genes are rare events
sunitinib in locally uncontrolled recurrent disease or advanced in thyroid cancers—including in papillary thyroid cancer,
metastases in thyroid cancer [26]. They included 71 patients poorly differentiated thyroid cancer, and ATC—generally
(45 differentiated follicular carcinomas or ATC and 26 medul­ mutually exclusive of other oncogenic driver mutations.
lary carcinomas). There were 4 ATC patients treated with Thus, in the absence of another candidate oncogenic driver,
sunitinib, and these patients died at 3.3, 3.5, 7.8, and fusions involving these genes should be investigated.
26.5 months. Asthenia was the most frequent side effect in Entrectinib inhibits TRK 1–3 but in addition also inhibits the
83.1% of patients including 25.4% of patients with grade 3 and ALK and ROS1 tyrosine kinases and is also approved for NTRK
1.4% with grade 4 fatigue. Of all patients, 14.1% had a cardiac fusion solid tumors. The clinical trial that led to the FDA
event. Nine unexpected side effects were reported, of which 5 approval included five thyroid cancer patients, but histologies
were fatal. The authors concluded that the treatment with were not specified [33]. One of five thyroid cancer patients
sunitinib is effective in medullary and differentiated thyroid achieved a PR to therapy. The 2021 ATA guidelines recom­
cancer but did not seem to improve the natural history of the mend the use of this drug in the context of a clinical trial [3].
disease in ATC patients. Selpercatinib is a selective RET inhibitor that has shown effi­
cacy in patients with medullary thyroid cancer harboring RET
2.1.2. Anti-BRAF molecules fusion and RET mutations [34]. However, the data about its
2.1.2.1. Dabrafenib/Trametinib. BRAF inhibitors (in combi­ efficacy on ATC are scarce as only two ATC patients with RET
nation with MEK inhibitors) can be effective in malignancies fusion were enrolled in that trial and one of these patients
with a BRAF V600E mutation. As 40–70% of ATC carry such responded for 18 months. Then, their use in ATC should be
mutation, BRAF/MEK inhibitory treatment is of interest recommended in the clinical trial setting. Although there are
[27,28]. Dabrafenib is a BRAFV600E kinase signaling inhibitor, case reports of ATC patients with ALK fusion who were suc­
whereas trametinib is a kinase inhibitor that blocks MEK, cessfully treated with ALK inhibitors [35], ALK fusions are very
which is downstream from BRAF in the MAPK pathway. rare in ATC and should be preferably used in a clinical trial
BRAF/MEK inhibition is given in combination to overcome setting.
early resistance to BRAF inhibitors [29,30]. This combination
treatment has been tested in a clinical trial with hopeful 2.1.4. Anti-mTOR molecules
results. In 2018, Subbiah et al. completed a study of 16 2.1.4.1. Everolimus. Constitutive activation of the phospha­
patients, 15 of which had BRAF V600E mutated tumors [31]. tidylinositol-3-kinase (PI3K)/AKT/mTOR pathway has been
The mutation had been tested before the start of the study observed in thyroid cancer pathogenesis [7]. Everolimus is
and confirmed again before treatment. The median age was a sirolimus-derived mTOR inhibitor. Administering everolimus
72 years, 63% were female, and 63% were of Asian heritage. to different ATC cell lines that previously had shown resistance
Prior treatments included surgery (88%), external beam radio­ to gefitinib showed a significant growth inhibition [36].
therapy (81%), and chemotherapy (38%). The median dura­ Schneider et al. included 28 patients (7 with ATC) in
tion of follow-up for the ATC cohort was 47 weeks (range 4– a nonrandomized, open-label, multicenter, single-arm phase
120). The confirmed overall response rate was 69% (95% CI, II trial, but the results were disappointing, with none of the
EXPERT REVIEW OF ANTICANCER THERAPY 1243

patients responding to the treatment [37]. The most frequent CI = 6.02, 14.83, range 5.4–40 months). The median OS and
AEs were anemia and cough, each occurring in 64% of the PFS from the addition of pembrolizumab were 6.93 months
patients, and time until death varied from 5 to 53 weeks. The (95% CI, 1.7–12.15; range 3–15.9 months) and 2.96 months
authors concluded that everolimus does not have a role in (95% CI, 2.2–3.7; range 0.57–13.14 months), respectively.
ATC treatment. The authors conclude that pembrolizumab may be an effec­
Hanna et al. conducted a single-arm, multi-institutional tive therapy added to TKI at the time of progression on
phase II study of patients with radioactive iodine-refractory these drugs.
thyroid cancer including 7 patients with ATC [38]. One patient
achieved a near-complete response, and one had stable dis­
3. Other therapies – current trials
ease for 26 months and no evidence of progression prior to
death (from congestive heart failure), while 4 progressed A search for the recent clinical trials available [45] resulted in
within 3 months of study entry. The last patient was unevalu­ 72 ongoing studies with ATC patients. We found nine clinical
able. All patients with a significant response to treatment had trials with preliminary unpublished results (Table 1) and 21
demonstrated mutations directly linked to mTOR activation. active trials with results pending (Table 2). When analyzing the
The median PFS of the ATC patients was 2.8 months (median clinical trials with available preliminary results, there are four
OS, 8.9 months). where a molecular compound is used in addition to che­
motherapy (i.e. Crolibulin+Cisplatin, Combretastatin
+Paclitaxel /Carboplatin). In general, no clear improvement
2.2. Immunotherapy
of the poor prognosis of ATC was observed. However,
2.2.1. Checkpoint inhibitor drugs targeting PD-1 or PD-L1 among the trials without results, we note a large number of
2.2.1.1. Spartalizumab. Macrophages and T-cells express drugs (21 studies with 25 drugs) that are being analyzed,
a glycoprotein called programmed cell-death 1 (PD-1), and indicating the great effort that is being undertaken to improve
when its ligands (PD-L1 or PD-L2) bind to it, they inhibit the treatment of ATC. When analyzing ongoing studies, there
cytotoxic T-cell immune response that leads to an immune is a clear trend toward the use of multiple therapies to target
escape of the cells that express these ligands. This glycopro­ different pathways simultaneously (TKI+anti PD1 or PD-L1, anti
tein is found in more than 65% of ATC cells [39–41]. BRAF+MEK inhibitor+anti PD1 or PD-L1).
Spartalizumab is a humanized immunoglobulin 4 monoclo­
nal antibody that binds PD-1 with sub-nanomolar activity,
4. Discussion
blocking interaction with PDL1 and PD-L2 [42]. In 2020,
Capdevila et al. published a phase II study on 42 patients ATC is the most aggressive and dreaded thyroid malignancy,
with ATC, 2 of them without tumor tissue available for central and when resectable, the preferred treatment is surgery fol­
pathology review [43]. The commonest AEs were diarrhea lowed by definitive radiation with or without chemotherapy
(12%), pruritus (12%), fatigue (7%), and pyrexia (7%). The over­ (taxane monotherapy or with platin or anthracycline) [3]. ATC
all response rate was 19%, including 3 patients with can arise as a result of terminal de-differentiation of well-
a complete response and 5 with a partial response. Twenty- differentiated thyroid cancer, and it can develop in a pre-
eight of the 40 patients with available biopsy material showed existing goiter (≥80% of ATC come from a longstanding his­
tumors that expressed PD-L1, and response rates were higher tory of multinodular thyroid disease) or may develop de novo
in PD-L1–positive (29%) versus PD-L1–negative (0%) patients. [46,47]. Some studies confirmed the presence of a well-
The highest rate of response (35%) was observed in patients differentiated thyroid cancer within ATC tumors, which
with PD-L1 ≥ 50%. Responses were seen in both BRAF- leads to the hypothesis of a post-malignant de-
nonmutant and BRAF-mutant patients and were durable, differentiation. This is controversial as only a very small num­
with a 1-year survival of 52.1% in the PD-L1–positive popula­ ber of differentiated carcinomas progress to ATC [48,49]. ATC
tion. This study demonstrated the responsiveness of ATC to tumorigenesis may be a multistep process with a biological
PD-1 blockade. transformation (synchronous or metachronous) from differen­
tiated thyroid cancer to ATC [50]. The recent understanding of
2.2.1.2. Pembrolizumab. Pembrolizumab is a selective the genomic profile of this disease has led to impressive
anti-PD-1 monoclonal antibody. It was used in combination improvement in patient outcomes using specific targeted
with chemoradiotherapy by Chintakuntlawar et al. in agents. Patients are currently selected for multimodal treat­
a phase II study on 3 ATC patients with unresectable disease ment at large academic centers. Management of this disease
[39]. After an initial tumor response, all patients died in less remains, however, very challenging in non-referral institutions
than 6 months, with a median OS of 2.76 months. Iyer et al. who see few ATC patients. In ATC, one can find mutations of
studied the use of pembrolizumab in combination with TKI BRAF and RAS, PI3K/Akt pathway, PIK3CA, p53 and B-Catenin
(lenvatinib, trametinib ± dabrafenib) at the time of progres­ (CTNNB1) [51–55] and investigations are ongoing trying to
sion on TKI alone [44]. Twelve patients were treated: 42% block these pathways in order to achieve a favorable tumor
had partial response, 33% had stable disease, and 25% had response.
progressive disease. Clinical benefit (partial response and In the most recent ATA guidelines for ATC, the authors
stable disease) was seen in 75% patients. Fatigue, anemia, suggested in their Recommendation 4 that assessment of
and hypertension were the most common AEs encountered. BRAF V600E mutation should be performed by immunohisto­
The median OS from the start of TKI was 10.43 months (95% chemistry and confirmed by molecular testing.
1244 A. COCA-PELAZ ET AL.

Table 1. Clinical trials on ATC with results available.


No. of patients
[NCT number] Response More common AE
Clinical Inmunotherapy PD-1 and PD- Pembrolizumab 6 (5♀-1♂) 3 PR Dyspnea (50%)
trials L1 inhibitors [NCT02688608] 1 SD Respiratory failure (33.3%)
(with 1 PD Rash (33.3%)
results) 1 Lost Mucositis (33.3%)
Vascular disruptors Combretastatin 55 CA4P+CT CA4P+CT: 5.2 months Bronchitis (9.8%)
(CA4P) (25♀-30♂) CT: 4 months Febrile Neutropenia (5.8%)
25 CT
(18♀-7♂)
[NCT00507429]
Crolibulin 27 Crolibulin NA Gastrointestinal and respiratory disorders
+Cisplatin (7♀- (16.67%)
20♂)
[NCT01240590]
Tyrosine-kinase inhibitors Lenvatinib 34 (21♀-13♂) PFS: 2.6 months Dyspnea (11.76%)
[NCT02657369] OS: 3.2 months Pulmonary embolism (8.82%)
27 deaths
Sorafenib 20 (7♀-13♂) 2 PR Dyspnea (10%)
[NCT00126568] 5 SD
11 PD
PFS: 1.9 months
OS: 3.9 months
Imatinib 11 (5♀-6♂) 2 PR Anemia (54.5%), hypertension (53.3%), nausea
[NCT00115739] 4 SD (45.45%), fatigue and edema (63.6%)
3 Lost
SR at 6 months: 46%
Pazopanib 15 (10♀-5♂) NA Fatigue (80%), anorexia (53.3%), diarrhea
[NCT00625846] (46.67%), nausea (40%)
PPARγ agonist CS7017 15 CS7017 + CT 1 PR Anaphylatic reaction, infections, dysphagia,
(10♀-5♂) 8 SD dyspnea
[NCT00603941] 4 PD
2 unknown
Inhibitor of HDAC and PI3K CUDC-907 7 Median survival after Gastrointestinal and general disorders
[4 ATC (2♀-2♂)] therapy: 127 days
[NCT03002623]
AE: Adverse events, CT: Chemotherapy, CRT: Chemoradiotherapy, DCC: Disease control rate, NA: Not available, NTC: ClinicalTrials.gov identifier, ORR: Overall
response rate, PD: Progressive disease, PR: Partial response, SD: Stable disease, SR: Survival rate.

Recommendation 5 proposes that molecular profiling should pathway, we can use inhibitors of RAS, RET, RAF, and MEK
be performed at diagnosis as well as at the time of progres­ kinases [56].
sion [3]. Initial treatment of stage IVB disease is chemoradia­ As stated above, anti-BRAF molecules are recommended by
tion. In stage IVB, if chemoradiation is not feasible for the the ATA for treatment of unresectable ATC with the
patient, and in the presence of BRAFV600E mutations, dabra­ BRAFV600E mutation that has been identified as the driver
fenib and trametinib, a combination treatment, approved by mutation in 45% of ATC cases [27]. BRAF inhibition resulted in
the U.S. Food and Drug Administration (FDA) for this indica­ diminished phosphorylation of ERK and MEK kinases in the
tion, may be used. In BRAFwt tumors, another genetic altera­ mitogen-activated protein kinase (MAPK) signaling cascade
tion such as ALK, NTRK, or RET fusions suggests that targeted [57,58]. Dabrafenib (BRAF inhibitor) plus trametinib (MEK inhi­
therapy should be considered, depending on the availability bitor) is a promising new combination targeted therapy for
of such treatment options. More clinical data are, however, patients with BRAF V600E–mutated ATC, demonstrating a high
needed to assess the role of these agents in unresectable overall response rate, prolonged duration of response, and
disease. In addition, a rapid genomic analysis remains survival with manageable toxicity [31]. Furthermore, in the
a major challenge for small non-referral centers and its use above cited study by Maniakas et al., surgical resection follow­
may be counter-productive by delaying therapy. In cases of ing neoadjuvant therapy was completed in 23 patients (15%)
excellent tumor response, subsequent surgery and definitive in the 2017–2019 group compared with none in the 2000–
radiotherapy or chemoradiotherapy should be undertaken. In 2013 and 2014–2016 groups [10]. In patients who received
cases of stage IVC disease, a similar strategy for targeted BRAF-directed therapy (most of them with dabrafenib/trame­
therapy can be followed. In addition, participation in immu­ tinib), those who underwent surgery following neoadjuvant
notherapy trials may be helpful for patients with tumors with therapy (n = 20) had statistically significant improvement in
high PD-L1 expression. median OS compared with those who did not. Nevertheless,
Different TKIs, such as lenvatinib, sorafenib, and others like similar to experience with other tumors, the emergence of RAS
imatinib and pazopanib have also been used for the treatment mutations appears to act as a mechanism of resistance to
of ATC. All of them have shown limited activity against ATC in BRAF inhibitors in thyroid cancers [59]. Other BRAFV600E
terms of PFS or OS. They are used because they target trans- kinase signaling inhibitors studied in prospective clinical trials
membrane tyrosine kinase receptors that initiate signaling include vemurafenib/cobimetinib, followed by atezolizumab.
through the MAP kinase pathway. In the same signaling From 10 patients with surgically unresectable disease enrolled
EXPERT REVIEW OF ANTICANCER THERAPY 1245

Table 2. Drugs used in active clinical trials without results available yet. regulation of the G1- to S-phase transition [62]. The results in
Drug terms of their effectiveness are heterogeneous.
[NTC] Checkpoint inhibitor drugs targeting PD-1 or PD-L1 repre­
Active clinical MEK inhibitors Trametinib sent the next frontier in treating ATC. T-cells express program
trials [NCT03085056]
(no results [NCT04739566] cell death-receptor 1 (PD-1) on their surface, which interact
available) [NCT04675710] with a ligand on normal tissues. Tumor cells express pro­
[NCT03975231] grammed cell death ligand 1 (PDL-1), and the interaction
Cobimetinib
[NCT03181100] between receptor and ligand suppresses T-cell mediated cyto­
Immunotherapy PD-1 and PD-L1 Durvalumab toxicity toward the thyroid tumor [63]. ATA guidelines for ATC
inhibitors [NCT03122496] suggested that in stage IVC patients with high PD-L1 expres­
Cemiplimab
[NCT04238624] sion, checkpoint (PD-L1, PD1) inhibitors can be considered as
Atezolizumab the first-line therapy in the absence of other targetable altera­
[NCT03181100] tions or as subsequent therapy, preferably in the context of
[NCT04400474]
Pembrolizumab a clinical trial [3].
[NCT04675710]
[NCT05119296]
[NCT05059470] 5. Conclusions
Tislelizumab
[NCT04579757] Treatment of ATC is based on a combination of surgery, if
Nivolumab possible, with (chemo)radiotherapy, but prognosis remains
[NCT03246958]
CTLA-4 inhibitors Tremelimumab poor. Recent attempts to improve the prognosis of these
[NCT03122496] tumors are moving toward personalized medicine, basing
Ipilimumab the treatment decision on the specific genetic profile of the
[NCT03246958]
mTOR inhibitor MLN0128 individual tumor. The positive results of dabrafenib and tra­
[NCT02244463] metinib for ATC harboring the BRAF V600E mutation have
PPARγ agonist Efatutazone provided a useful treatment option. For the other genetic
[NCT02152137]
Tyrosine-kinase Pazopanib profiles, different drugs are available and can be used to
inhibitors [NCT01236547] individualize the treatment, probably using some drugs
Surufatinib together. Combinations of drugs act on different molecular
[NCT04579757]
Selpercatinib pathways and achieve inhibition at separate areas. With new
[NCT04759911] targeted therapies, the average survival has improved consid­
Cabozantinib erably and death from local disease progression or airway
[NCT04400474]
[NCT02041260] compromise is less likely with improvement in quality of life.
CDK inhibitors Abemaciclib Unfortunately, the results are still poor in terms of survival. It is
[NCT04552769] necessary to continue exploring novel therapies for ATC.
CDK-002
[NCT04592484]
IL-2 Aldesleukin
[NCT03449108] 6. Expert opinion
Vascular disruptors Bevacizumab
[NCT03181100] The treatment of ATCs remains a challenge given their rapid
Anti-BRAF HLX208 clinical progression, which makes diagnosis by molecular
molecules [NCT05102292] studies and treatment difficult, as small delays greatly com­
Vemurafenib
[NCT03181100] promise patient symptomatology. The greatest efforts are
Dabrafenib currently being made in tumors considered unresectable,
[NCT04739566] trying to inhibit different molecular pathways in order to
[NCT04675710]
[NCT03975231] achieve a stable clinical response for as long as possible. In
this sense, the effectiveness of the combination of dabrafe­
nib with trametinib has been proven in tumors expressing
the BRAF V600E mutation. The problem arises in tumors with
in a cohort, six patients underwent complete resection of a different genetic profile, for which multiple drug combina­
primary or residual tumor [60]. tions are being tested in different ongoing clinical trials. It is
Anti-mTOR molecules have been used in an attempt to treat known that these tumors present several genetic alterations,
these tumors. Activation of the mTOR serine/threonine protein which stimulates the effort to inhibit or disrupt several path­
kinase has been reported in a variety of malignant tumors with ways, but while radiological responses are achieved, there
an estimated 70% of mTOR upregulation [61]. Everolimus are no clear increases in patient survival. It is true that with
(RAD001) is an orally active derivative of rapamycin, targeting all the advances that are being made, cause of death is
mTOR. This drug exerts its activity through high-affinity interac­ changing, going from extensive local tumors with rapid
tion with an intracellular receptor protein, the immunophilin respiratory and digestive involvement, to deaths due to dis­
FKBP12, and subsequently interacts with the mTOR protein tant disease, which represents an advance in terms of the
kinase, inhibiting downstream signaling events involved in quality of life of the patients. One of the major limitations
when trying to implement these treatments is the availability
1246 A. COCA-PELAZ ET AL.

of genetic analysis and next-generation sequencing in all 4. Wein RO, Weber RS. Anaplastic thyroid carcinoma: palliation or
centers, which makes it impossible to know the genetic treatment? Curr Opin Otolaryngol Head Neck Surg. 2011;19
(2):113–118.
profile of the tumor in a short time. If we want personalized
5. Lloyd R, Osamura R, Klöppel G, et al. WHO classification of tumours
medicine to become a reality, this is one of the great chal­ of endocrine organs. Lyon: International Agency for Research on
lenges to be addressed, given that genetic evaluation of Cancer; 2017.
both the primary tumor and its metastases is increasingly 6. Cabanillas ME, Ryder M, Jimenez C. Targeted therapy for advanced
essential. What does seem to have been demonstrated is thyroid cancer: kinase inhibitors and beyond. Endocr Rev. 2019;40
(6):1573–1604.
that monotherapy is not achieving its objectives, which
7. De Leo S, Trevisan M, Fugazzola L. Recent advances in the manage­
leads us to treatments with two or more drugs, thus improv­ ment of anaplastic thyroid cancer. Thyroid Res. 2020;13(1):17.
ing response and reducing toxicity, but the side effects of • Comprehensive review of the more recent advances in ana­
these treatments are a major concern. At this time, the plastic thyroid cancer treatment
objective of all these treatments must be to prolong survival 8. Xu B, Fuchs T, Dogan S, et al. Dissecting anaplastic thyroid carci­
noma: a comprehensive clinical, histologic, immunophenotypic,
and, if possible, to achieve a local tumor response that allows
and molecular study of 360 cases. Thyroid. 2020;30(10):1505–1517.
the tumor to be resectable and to be able to perform multi­ 9. Janz TA, Neskey DM, Nguyen SA, et al. Is the incidence of anaplastic
modality treatment. The future of ATC treatment will look at thyroid cancer increasing: a population based epidemiology study.
improving the genetic knowledge of these lesions, to achieve World J Otorhinolaryngol - head neck Surg. 2019;5(1):34–40
true personalized medicine, which allows the treatment of • Interesting article about the epidemiology of the anaplastic
thryoid cancer.
each patient to be individualized given the variations in the
10. Maniakas A, Dadu R, Busaidy NL, et al. Evaluation of overall survival
genetic profiles that these tumors present. Once this profile in patients with anaplastic thyroid carcinoma, 2000-2019. JAMA
is known, the development of new drugs will be aimed at Oncol. 2020;6(9):1397–1404.
inhibiting the largest possible number of pathways, as long •• Article about the overall survival of the anaplastic thyroid
as tolerance is acceptable and it manages to improve the cancer compared among different treatment eras
11. de Ridder M, Nieveen van Dijkum E, Engelsman A, et al. Anaplastic
condition of patients, which leads to being able to carry out
thyroid carcinoma: a nationwide cohort study on incidence, treat­
more aggressive treatments with proven efficacy. ment and survival in the Netherlands over 3 decades. Eur
Unfortunately, despite the great advances that have been J Endocrinol. 2020;183(2):203–209.
made, ATCs continue to cause devastating disease with 12. Shamseer L, Moher D, Clarke M, et al. Preferred reporting items for
a much poorer prognosis than desired. systematic review and meta-analysis protocols (PRISMA-P) 2015:
elaboration and explanation. BMJ. 2015;350(jan02 1):g7647.
13. Okamoto K, Kodama K, Takase K, et al. Antitumor activities of the
Funding targeted multi-tyrosine kinase inhibitor lenvatinib (E7080) against
RET gene fusion-driven tumor models. Cancer Lett. 2013;340
This paper received no funding. (1):97–103.
14. Tahara M, Kiyota N, Yamazaki T, et al. Lenvatinib for anaplastic
thyroid cancer. Front Oncol. 2017;7:25.
15. Takahashi S, Kiyota N, Yamazaki T, et al. A Phase II study of the
Declaration of interest safety and efficacy of lenvatinib in patients with advanced thyroid
cancer. Future Oncol. 2019;15(7):717–726.
MDW- Scientific Advisory and consulting for Bayer Pharmaceuticals. The
16. Wirth LJ, Brose MS, Sherman EJ, et al. Open-label, single-arm, multi­
authors have no relevant affiliations or financial involvement with any
center, phase II trial of lenvatinib for the treatment of patients with
organization or entity with a financial interest in or financial conflict with
anaplastic thyroid cancer. J Clin Oncol. 2021;39(21):2359–2366.
the subject matter or materials discussed in the manuscript. This includes
17. Brose MS, Nutting CM, Jarzab B, et al. Sorafenib in radioactive
employment, consultancies, honoraria, stock ownership or options, expert
iodine-refractory, locally advanced or metastatic differentiated
testimony, grants or patents received or pending, or royalties.
thyroid cancer: a randomised, double-blind, phase 3 trial. Lancet.
2014;384(9940):319–328.
18. Ito Y, Onoda N, Ito K-I, et al. Sorafenib in Japanese patients with
Reviewer disclosures locally advanced or metastatic medullary thyroid carcinoma and
anaplastic thyroid carcinoma. Thyroid. 2017;27(9):1142–1148.
Peer reviewers of this manuscript have no relevant financial or other 19. Sherman EJ, Dunn LA, Ho AL, et al. Phase 2 study evaluating the
relationships to disclose. combination of sorafenib and temsirolimus in the treatment of
radioactive iodine-refractory thyroid cancer. Cancer. 2017;123
(21):4114–4121.
20. Harding MW. Immunophilins, mTOR, and pharmacodynamic stra­
References tegies for a targeted cancer therapy. Clin Cancer Res. 2003;9
(8):2882–2886.
Papers of special note have been highlighted as either of interest (•)
21. Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes
or of considerable interest (••) to readers.
Dev. 2004;18(16):1926–1945.
1. Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: patho­
22. Del Bufalo D, Ciuffreda L, Trisciuoglio D, et al. Antiangiogenic
genesis and emerging therapies. Clin Oncol (R Coll Radiol). 2010;22
potential of the mammalian target of rapamycin inhibitor
(6):486–497.
temsirolimus. Cancer Res. 2006;66(11):5549–5554.
2. Perros P, Boelaert K, Colley S, et al. Guidelines for the management
23. Malaguarnera R, Chen K-Y, Kim T-Y, et al. Switch in signaling
of thyroid cancer. Clin Endocrinol (Oxf). 2014;81 Suppl 1:1–122.
control of mTORC1 activity after oncoprotein expression in thyroid
3. Bible KC, Kebebew E, Brierley J, et al. 2021 American Thyroid
Association guidelines for management of patients with anaplastic cancer cell lines. J Clin Endocrinol Metab. 2014;99(10):E1976–87.
thyroid cancer. Thyroid. 2021;31(3):337–386. 24. Guigon CJ, Fozzatti L, Lu C, et al. Inhibition of mTORC1 signaling reduces
• Essential article about the complete management of this tumor growth but does not prevent cancer progression in a mouse
pathology model of thyroid cancer. Carcinogenesis. 2010;31(7):1284–1291.
EXPERT REVIEW OF ANTICANCER THERAPY 1247

25. Papaetis GS, Syrigos KN. Sunitinib: a multitargeted receptor tyro­ 44. Iyer PC, Dadu R, Gule-Monroe M, et al. Salvage pembrolizumab
sine kinase inhibitor in the era of molecular cancer therapies. added to kinase inhibitor therapy for the treatment of anaplastic
BioDrugs. 2009;23(6):377–389. thyroid carcinoma. J Immunother Cancer. 2018;6(1):68.
26. Ravaud A, de la Fouchardière C, Caron P, et al. A multicenter phase 45. ClinicalTrials.gov [Internet]. Bethesda: National Library of Medicine;
II study of sunitinib in patients with locally advanced or metastatic [Cited 2022 Jan 4]. Available from: https://clinicaltrials.gov/.
differentiated, anaplastic or medullary thyroid carcinomas: mature 46. Aldinger KA, Samaan NA, Ibanez M, et al. Anaplastic carcinoma of
data from the THYSU study. Eur J Cancer. 2017;76:110–117. the thyroid: a review of 84 cases of spindle and giant cell carci­
27. Landa I, Ibrahimpasic T, Boucai L, et al. Genomic and transcriptomic noma of the thyroid. Cancer. 1978;41(6):2267–2275.
hallmarks of poorly differentiated and anaplastic thyroid cancers. 47. Mohebati A, Dilorenzo M, Palmer F, et al. Anaplastic thyroid carci­
J Clin Invest. 2016;126(3):1052–1066. noma: a 25-year single-institution experience. Ann Surg Oncol.
28. Deeken-Draisey A, Yang G-Y, Gao J, et al. Anaplastic thyroid carci­ 2014;21(5):1665–1670.
noma: an epidemiologic, histologic, immunohistochemical, and 48. Van der Laan BFAM, Freeman JL, Tsanq RW, et al. The association of
molecular single-institution study. Hum Pathol. 2018;82:140–148. well-differentiated thyroid carcinoma with insular or anaplastic
29. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and MEK thyroid carcinoma; evidence for dedifferentiation in tumor
inhibition in melanoma with BRAF V600 mutations. N Engl J Med. progression. Endocr Pathol. 1993;4(4):215–221.
2012;367(18):1694–1703. 49. Spires JR, Schwartz MR, Miller RH. Anaplastic thyroid carcinoma.
30. Kurata K, Onoda N, Noda S, et al. Growth arrest by activated BRAF Association with differentiated thyroid cancer. Arch Otolaryngol
and MEK inhibition in human anaplastic thyroid cancer cells. Head Neck Surg. 1988;114(1):40–44.
Int J Oncol. 2016;49(6):2303–2308. 50. Jannin A, Escande A, Al Ghuzlan A, et al. Anaplastic thyroid carci­
31. Subbiah V, Kreitman RJ, Wainberg ZA, et al. Dabrafenib and trame­ noma: an update. Cancers (Basel). 2022;14(4):1061.
tinib treatment in patients with locally advanced or metastatic 51. Xing M, Clark D, Guan H, et al. BRAF mutation testing of thyroid
BRAF V600-Mutant anaplastic thyroid cancer. J Clin Oncol. fine-needle aspiration biopsy specimens for preoperative risk stra­
2018;36(1):7–13. tification in papillary thyroid cancer. J Clin Oncol. 2009;27
32. Wang JR, Zafereo ME, Dadu R, et al. Complete surgical resection (18):2977–2982.
following neoadjuvant dabrafenib plus trametinib in 52. Hou P, Liu D, Shan Y, et al. Genetic alterations and their relation­
BRAFV600E-mutated anaplastic thyroid carcinoma. Thyroid. ship in the phosphatidylinositol 3-kinase/Akt pathway in thyroid
2019;29(8):1036–1043. cancer. Clin Cancer Res. 2007;13(4):1161–1170.
33. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with 53. García-Rostán G, Costa AM, Pereira-Castro I, et al. Mutation of the
advanced or metastatic NTRK fusion-positive solid tumours: inte­ PIK3CA gene in anaplastic thyroid cancer. Cancer Res. 2005;65
grated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21 (22):10199–10207.
(2):271–282. 54. Moretti F, Nanni S, Farsetti A, et al. Effects of exogenous p53
34. Wirth LJ, Sherman E, Robinson B, et al. Efficacy of selpercatinib in transduction in thyroid tumor cells with different p53 status.
RET-Altered thyroid cancers. N Engl J Med. 2020;383(9):825–835. J Clin Endocrinol Metab. 2000;85(1):302–308.
35. Leroy L, Bonhomme B, Le Moulec S, et al. Remarkable response to 55. Wiseman SM, Masoudi H, Niblock P, et al. Derangement of the
ceritinib and brigatinib in an anaplastic lymphoma E-cadherin/catenin complex is involved in transformation of differ­
kinase-rearranged anaplastic thyroid carcinoma previously treated entiated to anaplastic thyroid carcinoma. Am J Surg. 2006;191
with crizotinib. Thyroid. 2020;30(2):343–344. (5):581–587.
36. Onoda N, Nakamura M, Aomatsu N, et al. Significant cytostatic 56. O’Neill JP, Shaha AR. Anaplastic thyroid cancer. Oral Oncol. 2013;49
effect of everolimus on a gefitinib-resistant anaplastic thyroid can­ (7):702–706.
cer cell line harboring PI3KCA gene mutation. Mol Clin Oncol. 57. Salerno P, De Falco V, Tamburrino A, et al. Cytostatic activity of
2015;3(3):522–526. adenosine triphosphate-competitive kinase inhibitors in BRAF
37. Schneider TC, de Wit D, Links TP, et al. Everolimus in patients with mutant thyroid carcinoma cells. J Clin Endocrinol Metab. 2010;95
advanced follicular-derived thyroid cancer: results of a Phase II (1):450–455.
clinical trial. J Clin Endocrinol Metab. 2017;102(2):698–707. 58. Ouyang B, Knauf JA, Smith EP, et al. Inhibitors of Raf kinase activity
38. Hanna GJ, Busaidy NL, Chau NG, et al. genomic correlates of block growth of thyroid cancer cells with RET/PTC or BRAF muta­
response to everolimus in aggressive radioiodine-refractory thyr­ tions in vitro and in vivo. Clin Cancer Res. 2006;12(6):1785–1793.
oid cancer: a Phase II study. Clin Cancer Res. 2018;24 59. Cabanillas ME, Dadu R, Iyer P, et al. Acquired secondary RAS muta­
(7):1546–1553. tion in BRAFV600E-mutated thyroid cancer patients treated with
39. Chintakuntlawar AV, Rumilla KM, Smith CY, et al. Expression of PD-1 BRAF inhibitors. Thyroid. 2020;30(9):1288–1296.
and PD-L1 in anaplastic thyroid cancer patients treated with multi­ 60. Cabanillas M, Busaidy N, Dadu R, et al. OR27-6 combination vemur­
modal therapy: results from a retrospective study. J Clin Endocrinol afenib (BRAF Inhibitor)/Cobimetinib (MEK Inhibitor)/Atezolizumab
Metab. 2017;102(6):1943–1950. (Anti-PDL1 Inhibitor) in BRAF-V600E mutated anaplastic thyroid
40. Ahn S, Kim TH, Kim SW, et al. Comprehensive screening for PD-L1 cancer (ATC): initial safety and feasibility. J Endocr Soc. 2019;3
expression in thyroid cancer. Endocr Relat Cancer. 2017;24 (Supplement_1):OR27–6.
(2):97–106. 61. Hall MN. mTOR-what does it do? Transplant Proc. 2008;40(10
41. Cantara S, Bertelli E, Occhini R, et al. Blockade of the programmed Suppl):S5–8.
death ligand 1 (PD-L1) as potential therapy for anaplastic thyroid 62. Grozinsky-Glasberg S, Franchi G, Teng M, et al. Octreotide and
cancer. Endocrine. 2019;64(1):122–129. the mTOR inhibitor RAD001 (everolimus) block proliferation and
42. Naing A, Gainor JF, Gelderblom H, et al. A first-in-human phase 1 interact with the Akt-mTOR-p70S6K pathway in a
dose escalation study of spartalizumab (PDR001), an anti-PD-1 neuro-endocrine tumour cell line. Neuroendocrinology. 2008;87
antibody, in patients with advanced solid tumors. J Immunother (3):168–181.
Cancer. 2020;8(1):e000530. 63. Zhang G-Q, Wei W-J, Song H-J, et al. Programmed cell death-ligand
43. Capdevila J, Wirth LJ, Ernst T, et al. PD-1 blockade in anaplastic 1 overexpression in thyroid cancer. Endocr Pract. 2019;25
thyroid carcinoma. J Clin Oncol. 2020;38(23):2620–2627. (3):279–286.

You might also like