You are on page 1of 15

Medical Oncology (2020) 37:19

https://doi.org/10.1007/s12032-020-1345-2

REVIEW ARTICLE

Preclinical and clinical combination therapies in the treatment


of anaplastic thyroid cancer
Daniela Gentile1 · Paola Orlandi1 · Marta Banchi1 · Guido Bocci1 

Received: 28 December 2019 / Accepted: 12 February 2020


© Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive form of thyroid cancer, and novel therapies are urgently needed to
prolong patient survival and improve clinical outcomes. Very few scientific reviews have examined the literature on com-
bination therapies with the goal of describing the available preclinical and clinical data and suggesting future clinical com-
bination treatment schedules. The present review focuses on preclinical and clinical studies of drug combination therapies
in ATC. The relevant literature from PubMed and Scopus was reviewed in this article; the ClinicalTrials.gov database was
searched for clinical trials not yet published. Recent data from preclinical models strongly support the idea that combination
treatments that utilize drugs from different antineoplastic classes have synergistic antitumour activity in ATC. However,
rapid translation of these therapies into the clinic is impeded by the difficulty in recruiting enough patients for randomized
clinical trials. Although promising results have been obtained in preclinical studies, additional clinical research is required
to elucidate the efficacy of combination treatments for clinical practice.

Keywords  Anaplastic thyroid cancer · Combination treatment · Synergism · Clinical trials · Mouse models

Introduction neck. However, early diagnosis of ATC is very rare, and


usually is discovered incidentally during a routine physical
Thyroid tumours are typically classified into benign thyroid examination. As ATC grows, it compresses the trachea and
adenomas, papillary thyroid cancer (PTC), follicular thyroid causes dyspnoea, dysphagia, neck pain, and hoarseness [4].
cancer (FTC), medullary thyroid cancer (MTC) and ana- Indeed, ATC patients have an aggressive course, and present
plastic thyroid cancer (ATC). Differentiated thyroid cancer with locally advanced disease or metastasis [5]. Although
(DTC) usually indicates PTC and FTC, which originate from the causes of ATC are unknown, thyroid cancer may arise
follicular cells, whereas MTC originates from parafollicular de novo, or from one of the differentiated cancers, as sug-
cells [1]. PTC, FTC and MTC are collectively called DTC, gested by the simultaneous occurrence of areas, in the same
as they show differentiated features of their origin cells, and tumour mass, of differentiated or poorly differentiated thy-
generally have a good prognosis with > 98% 5-years survival roid carcinoma [6].
[1]. Conversely, ATC is undifferentiated, and is the most ATC responds poorly to conventional therapies and
aggressive form of thyroid cancer [2, 3]. Although it repre- requires a multidisciplinary approach for its management.
sents only 1–2% of all thyroid tumours, it accounts for up to According to the American Thyroid Association guidelines,
50% of all thyroid cancer-related mortalities. ATC is most the options for improving patient prognosis are surgery and
common in people over age 60, and has a mean survival of radiotherapy, followed by chemotherapy [7]. However, it is
only six months after diagnosis [4]. ATC can be diagnosed important to emphasize that any treatment used alone usu-
by fine needle aspiration biopsy of either a rapidly growing ally fails to control local disease, which is often the cause of
thyroid mass, or an abnormal lymph node in the patient’s death [8]. Previous studies have suggested that surgery with
adjuvant radiotherapy improves patient survival, while other
* Guido Bocci studies recommend adding both radiotherapy and chemo-
guido.bocci@med.unipi.it therapy to the routine management of ATC [9]. Complete
surgical resection remains the cornerstone of therapy in
1
Dipartimento di Medicina Clinica e Sperimentale, Università resectable tumours. Radiotherapy is often administered
di Pisa, Scuola Medica – Via Roma 55, 56126 Pisa, Italy

13
Vol.:(0123456789)
19   Page 2 of 15 Medical Oncology (2020) 37:19

preoperatively to reduce or destroy the tumour as well as issues associated with monotherapy (single-agent admin-
post-operatively, to ablate the remaining thyroid tissue, istration) include drug resistance at cellular and tumour
eliminate any suspected micrometastases, or remove recur- levels, a harsh tumour microenvironment that hinders drug
rent disease [9]. Chemotherapy is usually administered penetration and efficacy, tumour heterogeneity, and dose-
for advanced ATC treatment, and includes drugs such as limiting toxicity [16]. These issues are applicable to ATC
doxorubicin, paclitaxel, cisplatin, and carboplatin. The as well. The combination of chemotherapy regimens that
clinical activity of these commonly used chemotherapeu- contain two or more anticancer drugs is a practice that has
tics (i.e. doxorubicin) is low, with a response rate of 22%. been applied clinically for decades, in a variety of cancers
Thus, more active compounds need to be employed in the [17]. Similarly, a combination of different drugs is often
therapeutic strategy against ATC [10]. Due to the failure used in preclinical studies, and sometime in clinical trials,
of single-agent chemotherapy, a combination of two or to improve therapeutic efficacy in ATC [8]. However, it is
more drugs such as paclitaxel, cisplatin, doxorubicin, peg- important to determine which drug combinations are syn-
filgrastim and docetaxel has been suggested as therapeutic ergistic, and which have data to suggest that similar drug
for ATC patients [10]. Moreover, a second-line treatment combinations will fail clinically. To date, very few reviews
could use targeted therapies including tyrosine kinase inhibi- have been conducted on combination treatments in ATC,
tors (TKIs), anti-angiogenic drugs, and multikinase inhibi- with the goal of describing preclinical data and identifying
tors [8]. In addition, RAF/MAPK signalling is an integral future combination therapy schedules to be used clinically.
part of ATC progression, and some molecular therapeutics Therefore, the present review will discuss the preclinical and
such as the BRAF inhibitor (i.e. vemurafenib) and the MEK clinical studies that have used drug combination therapies
inhibitor (i.e. selumetinib) target this signalling cascade for ATC. The relevant literature from PubMed and Scopus
[8]. In general, these single-targeted inhibitors have failed were reviewed in this article; the ClinicalTrials.gov database
to show significant therapeutic responses in ATC patients, was searched for clinical trials not yet published.
which has led to the use of multikinase inhibitors that act on
two or more targets [8]. Recently, molecular characteriza- Methodology
tion of ATC tumours has facilitated an investigation into a
variety of molecular-targeted agents that inhibit TK recep- Data sources and searches
tors, as these receptors are responsible for tumour growth
and angiogenesis [8]. Moreover, some anecdotal accounts The PubMed and Scopus databases were searched for rel-
of sunitinib’s effects on ATC patients have been reported evant research articles published between January 2000 and
[11, 12]. Several TK inhibitors are currently under clinical December 2019 using the search terms “anaplastic thyroid
investigation. Indeed, sorafenib, imatinib, and axitinib have cancer” AND “combination treatment”; “anaplastic thyroid
all been tested in small clinical trials, and have shown prom- cancer” AND “synergism”; “anaplastic thyroid cancer”
ising disease control rates that range from 35 to 75% [13]. AND “combination” AND “clinical trials”; “anaplastic
In the last few years, several TKIs have been tested for the thyroid cancer” AND “mouse models” AND “combina-
treatment of advanced, progressive and radioactive iodine tion”. Research articles published in English that reported
(RAI)-refractory thyroid cancers. Of these, lenvatinib and preclinical and clinical combination of drugs in anaplastic
sorafenib have been approved by the Food and Drug Admin- thyroid cancer were included in this review. No restrictions
istration (FDA) and the European Medical Agency for the were adopted for the geographical origin of the articles: data
treatment of radioactive iodine-refractory and progressive from Asia, Africa, Europe, North and South America and
DTC, but not in poorly DTC [14]. Sorafenib is a multikinase Oceania were allowed.
inhibitor that inhibits RAF, VEGFRs, the PDGF receptor,
and RET, lenvatinib is a multikinase inhibitor that acts on Inclusion/exclusion criteria
VEGFRs [15].
Research studies were included in the review if they met the
following inclusion criteria: (1) the study looked at the pre-
Combination treatments in ATC​ clinical combination therapies in vitro and in vivo for ATC;
(2) the study examined the clinical combination studies in
Considering that ATC is very aggressive and that no chemo- ATC; (3) the study reported data from abstracts on ATC
therapeutic or targeted-therapy drug has demonstrated sat- combined therapies presented at international meetings but
isfactory activity acting alone (with the exception of dab- not yet published in extenso on scientific journals. Publica-
rafenib and trametinib combination on BRAF V600E-mutant tions were excluded if they were reviews, commentaries, edi-
ATC), recent research efforts have focused on novel drugs torials or letters to the editor. However, additional articles,
and, above all, on new drug combinations. The common such as reviews and editorials which dealt with the general

13
Medical Oncology (2020) 37:19 Page 3 of 15  19

concepts in the treatment of ATC and the mechanisms of tumour growth arrest and blocked ATC progression in an
cited drugs were included into some sections of the review. orthotopic mouse model [19]. The combination of irinotecan
Following removal of duplicate studies, the authors indepen- and the VEGFR-2 TKI PTK787/ZK222584 (vatalanib) also
dently reviewed each article for potential eligibility based on caused a decrease in ATC growth in an orthotopic mouse
the title and abstract. The full text was obtained for articles model, as well as a decrease in both the tumour endothelium
considered to be eligible by each author. and tumour microvessel density [20]. Recently, Di Desidero
and colleagues have investigated the in vitro activity in ATC
Data synthesis and analysis of the camptothecin analogue topotecan, a drug belonging to
the same therapeutic class of irinotecan, in combination with
The authors independently reviewed data from the identi- the tyrosine kinase inhibitor pazopanib. Interestingly, the
fied studies. Each study was analysed and a summary of the concomitant schedule caused a strong synergism with a sig-
findings was elaborated. The results of this review process nificant increase of intracellular concentrations of topotecan
were compared, and any discrepancies were resolved by con- lactone, decreasing the mRNA expression of ATP-binding
sensus following discussion. Using the search items “ana- cassette transporter G2, hypoxia-inducible factor-1α and
plastic thyroid cancer” AND “combination treatment” we vascular endothelial growth factor [21].
identified 450, and 205 articles in the PubMed and Scopus The taxane paclitaxel (a microtubule inhibitor) plus the
databases, respectively; using the search items “anaplastic TKI lenvatinib showed a significant antitumour effect in
thyroid cancer” AND “synergism” we identified 49, and 24 ATC cells through an increased percentage of ATC cells
articles in these two databases, respectively; whereas the in G2/M phase [22]. In addition, tumour size was reduced
search items “anaplastic thyroid cancer” AND “combina- in an ATC xenograft treated with the same drug combina-
tion” AND “clinical trials” identified 41, and 61 articles in tion, where tumour reduction was greater compared with
PubMed, and Scopus, respectively. Finally, adopting the lenvatinib or paclitaxel monotherapy [22]. Manumycin,
terms anaplastic thyroid cancer” AND “mouse models” a potent and selective farnesyl-transferase inhibitor [23],
AND “combination”, we found 81 and 13 articles in Pub- enhanced the cytotoxic effect of the chemotherapeutic drug
med and Scopus, respectively. After the removal of duplicate paclitaxel, and the combination of these drugs increased
studies, we identified and reviewed a total of 163 articles, of apoptotic cell death in ATC cells compared with either drug
which 39 met the inclusion criteria and were included in the alone [24]. This combination was also effective in vivo, with
study (Tables 1 and 2). no significant toxicity observed [24]. In a subsequent study,
paclitaxel plus manumycin was reported to be an effec-
Preclinical studies tive drug combination against ATC xenograft growth and
was able to inhibit tumour angiogenesis [25]. Moreover,
Several studies have been performed to investigate the syn- in an ATC xenograft model, the triple-drug combination
ergistic relationship between two or three drugs in ATC. of paclitaxel plus manumycin and minocycline (an anti-
Several of these have shown compelling antitumour effects biotic with potent inhibitory effects on matrix metallo-
in in vitro and in vivo models. In the following sections, we proteinases) resulted in the lowest average tumour growth
review the most recent preclinical studies on the manage- rate and the longest survival, compared with manumycin
ment of ATC (Table 1). alone, paclitaxel alone, or manumycin plus paclitaxel [26].
Other triple-drug combinations (i.e. combrestatin A4 phos-
Chemotherapy‑based combination studies phate + paclitaxel + manumycin and combretastatin A4
phosphate + paclitaxel + carboplatin) demonstrated excel-
The simultaneous combination of the topoisomerase I inhib- lent antineoplastic activity and marked vascular disruption
itor camptothecin analogue irinotecan and the TK inhibi- activity in ATC xenograft models [27]. Paclitaxel in combi-
tor sunitinib showed synergistic ATC antitumour activity nation with the novel high-affinity peroxisome proliferator-
in both in vitro and in vivo experiments [18]. In particular, activated receptor-γ (PPARγ) agonist RS5444 demonstrated
when these drugs were given simultaneously, they showed an additive anti-proliferative activity in cell culture, but also
a high degree of synergism in ATC cells. Specifically, they inhibited ATC tumour growth in vivo [28]. While RS5444
increased the intracellular concentration of SN-38, the active did not induce apoptosis by itself, it doubled the apoptotic
metabolite of irinotecan. Moreover, this synergistic combi- index of paclitaxel alone, when given in combination with
nation greatly decreased the gene expression and protein lev- the chemotherapeutic [28]. In contrast, some studies have
els of VEGF, colony stimulating factor-1, and ATP-binding described instances in which drug combinations exhibit
cassette transporter G2 in ATC cells [18]. In another pre- antagonistic or non-synergistic relationships. For example,
clinical study, a combination therapy of irinotecan and the paclitaxel and the cholesterol-lowering drug lovastatin were
monoclonal antibody cetuximab (an EGFR inhibitor) caused cytotoxic in two ATC cell lines, and increased apoptosis

13
Table 1  Combination treatments on ATC preclinical models
19  

Combination studies Preclinical models Results References

13
Chemotherapy-based combination studies
Irinotecan (0.01–100 μM)  + sunitinib (0.01– ATC cell lines: 8305C and FB3 Significant synergistic ATC antitumour activity Di Desidero et al. [18]
100 μM) Mice: 6-week-old CD nu/nu male in vitro and in vivo; increased intracellular
Page 4 of 15

Irinotecan (100 mg/kg/week i.p.) + sunitinib SN-38 concentrations; decreased expression of


(25 mg/kg/every 2 days i.p.) VEGF, colony stimulating factor-1 and ATP-
binding cassette transporter G2
Cetuximab (0.5 g/mL) + irinotecan ATC cell line: ARO Significant synergistic ATC antitumour growth Kim et al. [19]
Irinotecan (50 mg/kg once per week i.p.) + cetux- Mice: Male athymic nude mice, age 8 to and progression; decreased incidence of lymph
imab (1 mg/injection twice per week i.p.) 12 weeks node metastasis, laryngeal invasion, and
tumour microvessel density
Irinotecan (50 mg/kg once/week i.p.) + PTK787/ Mice: Male athymic nude mice, age 8 to ATC growth decreased; decline of tumour Kim et al. [20]
ZK222584 (vatalanib) 12 weeks endothelium and microvessel density
50 mg/kg once/day by oral gavage
Topotecan (0.01–100 μM) + pazopanib (0.01– ATC cell lines: 8305C and FB3 Significant synergistic ATC antitumour activ- Di Desidero et al. [21]
100 μM) ity in vitro; increased intracellular topotecan
lactone concentrations; decreased expression of
VEGF, colony stimulating factor-1, hypoxia-
inducible factor-1α and ATP-binding cassette
transporter G2
Paclitaxel (500 nM) + lenvatinib (5 µM) ATC cell lines: C643, 8305C and 8505C Significant antitumour effects in ATC cells; Jing et al. [22]
Paclitaxel (5 mg/kg twice per week i.p.) + len- Mice: Female athymic nude mice increased percentage of ATC cells in G2/M
vatinib (5 mg/kg/day oral route) phase; reduced tumour size in vivo
Manunycin (0–100 µM) + paclitaxel (0–10 µM) ATC cell lines: ARO, C643, DRO, Hth-74, KAT- Manumycin enhanced the cytotoxic effect of Yeung et al. [24]
Manumycin (7.5 mg/kg i.p.) + paclitaxel (20 mg/ 4, and KAT-18 paclitaxel in all six of the cell lines
kg i.p.) Mice: 7-week-old nude mouse The combination had significant inhibitory
effects on tumour growth
Manumycin (15 mg/kg·week oral) + paclitaxel Mice: 7-week-old nude mouse (nu/nu BALB-c Inhibited ATC xenograft growth and tumour Xu et al. [25]
(40 mg/kg·week i.p.) mice bred) angiogenesis
Paclitaxel (50 mg/kg/day i.p.) + manumycin Mice: 5-week-old nude mouse (nu/nu BALB-c) Lowest average tumour growth rate compared to She et al. [26]
(2.5 mg/kg/day i.p.) + minocycline (subcuta- monotherapies; longer survival than manumy-
neous implantation of a slow-release tablet cin alone, paclitaxel alone, or manumycin plus
(10 mg released over 60 days) paclitaxel in vivo
Manumycin (3 mg/kg days on days 1–5 Mice: 5-week-old nude (nu/nu BALB-c) Antineoplastic activity and a marked vascular Yeung et al. [27]
i.p.) + paclitaxel (50 mg/kg days on days 1 of disruption in vivo
Medical Oncology

each 7-days cycle i.p.)


Combrestatin A4 phosphate 50 mg/kg/daily, days
1–5, 2 h after the manumycin or paclitaxel
injection
Paclitaxel (0.01–1000 nM) + RS5444 (0.1– ATC cell line: DRO Additive anti-proliferative activity in vitro; inhi- Copland et al. [28]
(2020) 37:19

1000 nM) Mice: 3–4 week athymic female nude mice bition of ATC tumour growth in vivo; RS5444
Paclitaxel (0.2–0.3 mg/0.1 ml twice weekly (Harlan) doubled the apoptotic index of paclitaxel alone
i.p.) + 0.025% RS5444
Table 1  (continued)
Combination studies Preclinical models Results References

Paclitaxel (5,10,20 nM) + lovastatin (5,10,20 µM) ATC cell lines: 8505C and BHT-101 Cytotoxic in both ATC cell lines; caused Chung et al. [29]
Paclitaxel (2.5, 5, 15, 10, 20 nM) + lovastatin (5, antagonism that affected the cytotoxicity and
Medical Oncology

10, 20, 30, 40 µM) pro-apoptotic effects on 8505C cells


Baicalein (0, 10, 20, 50, and 100 µM) + docetaxel ATC cell line: 8505C Induced apoptosis and inhibited the metastatic Park et al. [30]
(10 nM) process of ATC cells by downregulating the
expression of apoptotic and angiogenic pro-
teins, and by blocking the ERK and Akt/mTOR
pathways
(2020) 37:19

Selinexor (25, 50, 100, 200, 400 nM) + doxoru- ATC cell lines: SW1736 and ATC351 Synergistic decrease in cell proliferation of both Garg et al. [31]
bicin (25, 50,100,200, 400 nM) ATC cell lines
Tyrosine kinase inhibitors-based combination studies
Sunitinib (10 µM) + SL327 (10 µM) ATC cell lines: CAL62 and KAT4 Reduced viability, increased apoptosis and sup- Wang et al. [32]
Sunitinib (25 mg/kg/day for 2 weeks CAL62 and KAT4 (doxorubicin-resistant ATC pressed migration of cells; induced significant
i.p.) + SL327 (25 mg/kg/day for 2 weeks i.p.) cells) additive suppression of tumour growth in vivo
Mice: nud/nud mice
Sorafenib (0.89, 1.79, 3.58 µmol/L) + quinacrine ATC cell lines: THJ-16 T ATC cells responded in an additive/synergistic Abdulghani et al. [33]
(0.42, 0.85, 1.7, 3.4,6.8 µmol/L) Mice: manner; improved survival of immunodeficient
Sorafenib (30 mg/kg daily for 5 C57BL6/J mice mice injected orthotopically with ATC cells
day0.42 s) + quinacrine (100 mg/kg every other compared to mice administered with either
day via oral gavage) compound alone or doxorubicin; inhibited
expression of the pro-survival MCL-1 factor,
signal transducer and activator of transcription
3 (pSTAT3), and dampened NFκB signalling
Sorafenib ­IC50 6.3 µmol/L + withaferin ­IC50 ATC cell lines: BCPAP Synergistic efficacy in anaplastic thyroid cancers Cohen et al. [34]
0.155 µmol/L SW1736 in vitro with significant pro-apoptotic effects
Sorafenib ­IC50 7.6 µmol/L + withaferin A ­IC50
2.5 µmol/L
HNHA (0.5, 1, 5, 10, 20, 40, 80 µM) + sorafenib ATC cell lines:8505C, SNU-80, and GSA1 Synergistically decreased cell viability and Park et [35]
(0.5, 1, 5, 10, 20, 40, 80 µM) Mice: female BALB/c nude mice significantly increased apoptotic cell death
HNHA (6.5 mg/kg i.p.) + sorafenib (25 mg/kg in ATC cells, as proved by the cleavage of
once every 2 days orally) caspase-3 and DNA fragmentation; signifi-
cantly decreased the vessel density and tumour
volume in ATC xenograft model, increasing
survival
Pazopanib (800 nM) + trametinib (10 nM) ATC cell lines: BCPAP, 8505C and CAL62 cells Significant tumour volume reduction of 50% or Ball et al. [36]
Pazopanib (100 mg/kg daily by oral gav- Mice: 4- to 6-week-old female athymic nude (nu/ more in tumours harbouring K ­ RASG12R and
age) + trametinib (1 mg/kg daily by oral gavage) nu) mice ­BRAFV600E mutations by modulation of the
ERK pathway
Apatinib (20 μM) + chloroquine (10 µM) ATC cell lines: KHM-5 M and C643 Induced protective autophagy and apoptosis Feng et al. [37]
Apatinib (50 mg/kg daily via oral gavage) + chlo- Mice: 4-week-old male BALB/c nude mice through the downregulation of p-Akt and
Page 5 of 15 

roquine (60 mg/kg daily via oral gavage) p-mTOR signals via the Akt/mTOR pathway
in vitro and in vivo
19

13
Table 1  (continued)
19  

Combination studies Preclinical models Results References

13
MK-2206 (1 μM) + tyrphostin AG 1296 (1 μM) ATC cell lines: AL62 and KAT4 Suppressed ATC cell viability, migration and Che et al. [38]
MK-2206 (100 mg/kg) + tyrphostin AG1296 Mice: nud/nud mice invasion, and tumour growth; induced apop-
(100 mg/kg for 2 weeks i.p.) tosis
Page 6 of 15

Combination between different antineoplastic drugs


Anti-PD-L1 antibody (10F.9G2) (200 µg/mouse/ Mice: 10-week-old female SCID mutant mice PD-L1 potentiated the effect of PLX4720 on Brauner et al. and Gunda et al. [39, 40]
every other day i.p.) + PLX4720 (PLX4720- tumour regression and intensified the antitu-
impregnated chow 4 total doses) mour response; increased CD8+ T-cell infiltra-
tion and improved T-cell cytotoxicity profile;
reduced ATC tumour volumes, prolonged mice
survival and improved antitumour immune
profile; maximal tumour reduction was associ-
ated with increased cytotoxicity and number
of CD8+ T cells, NK cells, and M1-polarized
tumour-associated macrophages; decrease in
myeloid-derived suppressor-like cells
PLX4720 (10 μM) + dasatinib (50 nM) ATC cell lines: 8505c, BCPAP, TBP-3868, TBP- Reduced the tumour size in an orthotopic ATC Vanden Borre et al. [41]
PLX4720-impregnated chow ad libitum and 3743, TBPt-3403 and TBPt-3610R mouse model, increased immune cell infiltra-
orally gavaged + dasatinib (50 mg/kg 5 days) Mice: B6129SF1/J syngeneic mice tion and induced apoptosis
PLX4720 (1 μM) + U0126 (10 μM) ATC cell lines: BCPAP and SW1736 No enhanced inhibition of the invasive potential Ingeson-Carlsson et al. [42]
of ATC cells
Troglitazone (5 μM) + lovastatin (1 μM) ATC cell lines: 8305C and SW1736 Significant inhibition of the EGF-induced migra- Chin et al. [43]
tion of ATC cells by increasing E-cadherin
expression and decreasing the vimentin expres-
sion. cAMP response element-binding protein
(CREB) and ERK phosphorylation level was
decreased in ATC cells
Lovastatin (0.5, 1, and 2 μM) + troglitazone (5 ATC cell line: SW1736 Anti-proliferative activity in ATC cell culture Zhong et al. [45]
and 10 μM) Mice: Bagg’s albino (BALB)/c nude mice systems and a tumour regression in the mouse
Lovastatin (2 mg/kg/day) + troglitazone (5 mg/ xenograft model, through a cell cycle arrest at
kg/day) G0/G1 phase
Omipalisib (0–100 nM) + palbociclib (0–100 nM) ATC cell line: Mouse (T4888M, T3533, T1860, Omipalisib potentiated and extended the activity Wong et al. [46]
Omipalisib 0.3 mg/kg/day + palbociclib 30 mg/ T1903, A274, A275, T7152, T7189) of palbociclib in all the tested ATC cell lines;
kg/day, via oral gavage Human (THJ16T, CAL62, C643, OCUT2, low-dose combination inhibited tumour growth
T238, 8505C, SW1736, THJ21T, T243, T235, in vivo
Medical Oncology

HTH104, ACT1)
Mice: NOD-scid IL2rγnull (NSG) mice, aged 6- to
8-weeks old
BP-14 (5, 10, 25 and 50 nM) and everolimus (25, ATC cell lines: FRO, SW1736, 8505C Strong synergistic effect; significant loss of cell Allegri et al. [47]
50, 100 and 150 nM) viability and downregulation of epithelial–mes-
enchymal transition-related genes
(2020) 37:19
Table 1  (continued)
Combination studies Preclinical models Results References

BI6727 (600, 1000, 1750 nM) + BKM120 (600, ATC cell lines: T4888M, A275, THJ16T, Significant synergistic effect on ATC cells; De Martino et al. [48]
1000, 1750 nM) OCUT2 enhanced growth suppression at doses for
Medical Oncology

BI6727 (8 mg/kg/day via oral gavage) + BKM120 Mice: 6–8 week-old 129S6/SvE mice which single drugs showed no effect, and led to
(24 mg/kg/day via oral gavage) massive reduction of tetraploid cell popula-
tion; combined inhibition of PI3K and PLK1
was effective in vivo in the immunocompetent
allograft ATC model
SAHA (1–4 μM) + PJ34 (5–30 μM) ATC cell line: SW1736 Synergistic effect against SW1736 cell growth Baldan et al. [49]
(2020) 37:19

in vitro
PXD101 (1, 3, and 5 μM) + AUY922 (1, 3, and ATC cell lines: 8505C and CAL62 AUY922 had synergistic activity with PXD101 Kim et al. [50]
5 μM) in induction of cytotoxicity in conjunction with
the inactivation of PI3K/Akt signalling and
activation of DNA damage response
SNX5422 (5 μM) + HDAC inhibitors (PXD101, ATC cell lines: 8505C and CAL62 Synergistic effects in induction of ATC cell Kim et al. [51]
SAHA, trichostatin A) (5 μM) death; SNX5422 synergizes with HDAC
inhibitors to induce cytotoxicity, suppression
of PI3K/Akt/mTOR signalling and survival,
and the overexpression of DNA damage-related
proteins in ATC cells
Carfilzomib (0, 4, 6, 8, 10, 12 nm) + CUDC-101 ATC cell lines: 8505C, C-643, SW-1736, Inhibited cellular proliferation and caused cell Zhang et al. [52]
(0, 0.4, 0.8, 1.2, 1.6, 2 μM) THJ16T and THJ-29T death in multiple ATC cell lines; increased
anti-ATC effect was associated with a syner-
gistically enhanced G2/M cell cycle arrest and
increased caspase 3/7 activity; increased p21
expression and poly (ADP-ribose) polymerase
protein cleavage
Page 7 of 15 
19

13
19  

13
Page 8 of 15

Table 2  Clinical trials regarding combination schedules in ATC patients


Combination treatments Patients enrolled Phase Results Adverse drug reactions References

Paclitaxel (80 mg/m2/weekly) + valproic acid 25 II/III Not impaired paclitaxel pharmacokinetics; No severe adverse effect was observed. In Catalano et al. [53]
(1000 mg/day) vs. paclitaxel alone median overall survival and the median one patient, paclitaxel was reduced to 75%,
time to progression were not found statisti- as a consequence of allergic reaction
cally different in the two arms
Sorafenib (200 mg twice daily) + temsiroli- 36 II Active combination, with a progression-free Five patients suffered of bowel perforation Sherman et al. [55]
mus (25 mg weekly) survival rate at 1 year of 30.5%, in ATC (2), excessive fatigue (2), and pneumonitis
patients (1), leaving the trial; the most common
grade 3 and 4 toxicities included: hypergly-
cemia, fatigue, anaemia, and oral mucositis
Dabrafenib (150 mg twice daily) + trametinib 16 II An overall response rate of 69% and a Common adverse events were fatigue (38%), Subbiah et al. [56]
(2 mg once daily) complete response; durable responses, pyrexia (37%), and nausea (35%); grade
with a 12-month Kaplan–Meier estimate of 3–4 diarrhoea, anaemia, and hyperglycae-
response duration around 90% mia were also described
Efatutazone (0.15, 0.3, or 0.5 mg doses twice 15 I Seven patients achieved a stable disease, and Grade 3–4 septic shock, dysphagia, dyspnea, Smallridge et al. [58]
daily) + paclitaxel (every three weeks) 1 patient a partial response anaemia, pneumonia, loss of conscious-
ness, and anaphylactic reaction were
described
Triple association of combretastatin (60 mg/ 80 II/III No significant differences of overall survival Combination schedule usually well toler- Sosa et al. [59]
m2 for days 1, 8, 15 for 6 cycles), paclitaxel between the two arms ated but with described grade 3 and 4
(200 mg/m2 on day 1), and carboplatin hematologic adverse events (neutropenia,
(AUC 6 on day 1 following paclitaxel) vs. leukopenia and anaemia)
paclitaxel + carboplatin
Crolibulin (8–20 mg/m2) + cisplatin 27 (16 ATC patients) I/II One complete response and a stable disease The most common grade 3 toxicities were: Gramza et al. [60]
(75–100 mg/m2) lymphopenia, hyponatremia, anae-
mia, hypertension during infusion, and
hypophosphatemia. A laryngeal haem-
orrhage related to tumour erosion was
Medical Oncology

described
(2020) 37:19
Medical Oncology (2020) 37:19 Page 9 of 15  19

in 8505C cells. However, in these cells, the drug combina- in vitro and in vivo by downregulating p-Akt and p-mTOR
tion had antagonistic effects on both cytotoxicity and pro- signals via the Akt/mTOR pathway [37]. The combined
apoptotic events [29]. Therefore, rationally selecting proven administration of the Akt inhibitor MK-2206 and tyrphostin
combinations of drugs is crucial for improving therapeutic AG1296, a novel PDGFR inhibitor, suppressed ATC cell
effects on this type of anaplastic tumour. viability, migration, and invasion. This combination also
Park and colleagues reported that the simultaneous induced apoptosis and suppressed ATC tumour growth [38].
administration of the flavonoid baicalein and the microtube
stabilizer docetaxel, induced apoptosis and inhibited the Combination between different antineoplastic drugs
metastasis of ATC cells by downregulating antiapoptotic and
angiogenic proteins and by blocking the ERK and Akt/mam- Eran Brauner and collaborators showed that anti-PD-L1
malian target of rapamycin (mTOR) pathways [30]. Finally, immunotherapy potentiated the effect of the BRAFV600E
the combination of the exportin-1 inhibitor selinexor, with inhibitor PLX4720 on tumour regression and intensified the
the topoisomerase II inhibitor doxorubicin, caused a syner- antitumour immune response in an immunocompetent model
gistic decrease in the cellular proliferation of several ATC of ATC. This was achieved through increased CD8+ T-cell
cell lines [31]. infiltration, and an improved cytotoxic T-cell profile [39].
Another study that combined PLX4720 with antibodies to
Tyrosine kinase inhibitor‑based combination studies PD-L1 or PD-1 dramatically reduced murine ATC volumes,
prolonged mice survival time, and improved the antitumour
The combination of the multi-targeted TK inhibitor sunitinib immune profile [40]. Indeed, in this study maximal tumour
and the MEK1/2 inhibitor SL327 caused reduced viability, reduction was associated with increased cytotoxicity, and
increased apoptosis, and suppressed migration of doxoru- increased ­CD8+ T-cell and NK cell number. M1-polar-
bicin-resistant ATC cells. Moreover, this combination had ized tumour-associated macrophages were also increased,
a significantly additive effect on repression of doxorubicin- whereas a decrease in myeloid-derived suppressor-like cells
resistant ATC tumour growth in vivo [32]. Abdulghani and was described [40]. Moreover, the combination of PLX4720
colleagues also reported that ATC cells responded in an and dasatinib, a Src tyrosine receptor/Bcr-Abl family inhibi-
additive/synergistic manner to the combination of sorafenib tor, reduced tumour size, increased immune cell infiltration,
and the nuclear factor kappa B (NFκB) inhibitor quina- and induced apoptosis in an orthotopic ATC mouse model
crine [33]. Notably, this drug combination also improved [41]. Conversely, PLX4720 and the MEK inhibitor U0126
the survival of immunodeficient mice injected orthotopi- did not enhance the inhibition of ATC cells’ invasive poten-
cally with ATC cells, as compared with mice administered tial, suggesting that migration and invasion in ATC cells are
either doxorubicin or one compound alone [33]. Moreover, mediated by other non-MEK mechanisms [42].
at the molecular level, quinacrine and sorafenib dampened Co-administration of the PPARγ ligand troglitazone
NFκB signalling, and inhibited the expression of both signal and lovastatin exhibited significant inhibition of the EGF-
transducer and activator of transcription 3 (STAT3) and the induced migration of ATC cells, by increasing E-cadherin
pro-survival factor myeloid cell leukaemia-1 (MCL-1) [33]. expression and decreasing vimentin expression [43]. Moreo-
Sorafenib plus withaferin A (WA), a natural withanolide, ver, in ATC cells co-treated with troglitazone and lovastatin,
showed synergistic efficacy in papillary and anaplastic thy- both cAMP response element-binding protein (CREB) and
roid cancers in vitro, with significant pro-apoptotic effects ERK phosphorylation levels were decreased [43]. Indeed,
[34]. The histone deacetylase inhibitor N-hydroxy-7-(2- CREB and p-ERK are two crucial signalling molecules for
naphthylthio) heptanomide (HNHA) and sorafenib syner- EGF-induced cysteine-rich 61 expression, which is a secre-
gistically decreased cell viability, and significantly increased tory protein involved in the regulation of cell adhesion,
apoptotic cell death, as demonstrated by caspase-3 cleav- DNA synthesis, angiogenesis, cell survival, and cell migra-
age and DNA fragmentation in ATC cells. Moreover, the tion [44]. Lovastatin and troglitazone co-administration also
combination therapy of HNHA and sorafenib significantly cause cell cycle arrest at G0/G1, leading to anti-proliferative
decreased vessel density and tumour volume in an ATC xen- activity in ATC cell culture systems, and tumour regression
ograft model, thereby increasing the survival of the mice in a mouse xenograft model [45].
[35]. The combination of the TK inhibitor pazopanib and Wong et al. established that omipalisib, a PI3K/mTOR
the MEK inhibitor trametinib led to a 50% or greater reduc- inhibitor, potentiated and extended the activity of palbo-
tion in tumour volume, for tumours harbouring ­KRASG12R ciclib, a cyclin-dependent kinase (CDK) inhibitor, in all
and ­BRAFV600E mutations. This significant reduction was tested ATC cell lines [46]. More importantly, their low-dose
mediated through modulation of the ERK pathway [36]. combination was extremely effective in inhibiting tumour
The combination of a VEGFR-2 inhibitor such as apatinib growth in vivo [46]. Allegri and colleagues also reported
and the autophagy inhibitor chloroquine, induced apoptosis a strong synergistic effect of the new CDK inhibitor BP-14

13
19   Page 10 of 15 Medical Oncology (2020) 37:19

in combination with everolimus, a mTOR inhibitor. This across 5 centres in northwest Italy. The experimental arm
drug pairing demonstrated a significant loss in cell viabil- received a combination of paclitaxel (80 mg/m2/weekly)
ity in three human ATC cell lines, and downregulation of and valproic acid (1000 mg/day), whereas the control arm
epithelial–mesenchymal transition-related genes in two of received paclitaxel alone. Valproic acid has emerged as a
them [47]. promising antineoplastic agent due to its activity as a his-
Another example of combined treatment using two novel tone deacetylase inhibitor [53, 54]. The co-administration
antineoplastic drugs was a treatment with BI6727, a polo- of valproic acid did not influence the pharmacokinetics of
like kinase 1 (PLK1) inhibitor, and BKM120, a PI3K inhibi- paclitaxel, but the median overall survival and the median
tor. This combination resulted in a significant synergistic time to progression were not statistically different between
effect on ATC cells [48]. The two drugs together enhanced the two arms [53].
growth suppression at doses for which each single drug In a phase II, single-arm study conducted in 36 patients
showed no effect and led to a massive reduction in the tetra- with radioactive iodine-refractory thyroid carcinoma,
ploid cell population. Finally, the combined inhibition of sorafenib (200 mg twice daily) and temsirolimus (25 mg
PI3K and PLK1 was extremely effective in vivo, in an immu- weekly), appeared to be an active combination in compari-
nocompetent allograft model of ATC [48]. son to historical data using sorafenib only. This combination
The combination of suberoylanilide hydroxamic acid showed a progression-free survival rate at 1 year of 30.5%
(SAHA), a HDAC inhibitor, and PJ34, a poly ADP ribose for patients who had received no prior treatment [55].
polymerase (PARP) inhibitor, exhibited a synergistic effect In another phase II, open-label trial, the dabrafenib plus
against SW1736 cell growth in vitro [49]. Similar synergis- reatment schedule had important clinical activity in 16
tic effects were observed with another HDAC inhibitor, the patients with predefined BRAFV600E mutations, and a good
compound PXD101, and the heat shock protein 90 (hsp90) toxicity profile [56]. Indeed, dabrafenib (150  mg twice
inhibitor NVP-AUY922 (AUY922). Indeed, AUY922 daily) plus trametinib (2 mg once daily) combination ther-
showed synergistic activity with PXD101 in inducing cyto- apy resulted in a confirmed overall response rate of 69%. In
toxicity in conjunction with the inactivation of PI3K/Akt addition, one patient showed a complete response, achiev-
signalling, and the activation of a DNA damage response in ing resolution of multiple pulmonary metastases within the
ATC cells [50]. Moreover, the combination of another hsp90 first 8 weeks of therapy. Responses were also durable, with
inhibitor, SNX5422, and various HDAC inhibitors such as a 12-month Kaplan–Meier estimate of response duration
PXD101, SAHA, and trichostatin A (TSA), revealed syner- around 90% [56].
gistic effects in inducing ATC cell death. Indeed, SNX5422 A multicenter phase I/II dose-finding study has been
synergizes with HDAC inhibitors, inducing the suppres- conducted in 15 ATC patients treated with efatutazone and
sion of PI3K/Akt/mTOR signalling and survivin [51]. The paclitaxel. Efatutazone is an oral PPARγ agonist that was
second-generation proteasome inhibitor carfilzomib and the previously tested as a monotherapy in patients with solid
histone deacetylase and multikinase inhibitor CUDC-101 tumours [57]. In this study, it was taken orally twice daily at
synergistically inhibited cellular proliferation, and caused 0.15, 0.3, or 0.5 mg doses, and paclitaxel was administered
cell death in multiple ATC cell lines [52]. This increased every three weeks by intravenous infusion. This combination
anti-ATC effect was attributed to the drug combination’s was safe and tolerated, with 7 patients achieving a stable
effects on enhanced G2/M cell cycle arrest and increased disease state, and 1 patient showing a partial response [58].
caspase 3/7 activity. Interestingly, the treatment with carfil- A multicentre, open-label, randomized controlled trial
zomib and CUDC-101 also increased p21 expression and was designed to compare the overall survival of ATC
poly (ADP-ribose) polymerase protein cleavage [52]. patients treated with the triple combination of combretasta-
tin (also named fosbretabulin; 60 mg/m2 on day 1, 8, 15 for
Clinical studies on combined treatments 6 cycles), paclitaxel (200 mg/m2 on day 1), and carboplatin
area under curve (AUC) 6 on day 1 following paclitaxel,
Newer strategies involving systemic treatments are desper- compared to the double treatment of paclitaxel and car-
ately needed for ATC patients. It is important to note that boplatin. This study enrolled 80 patients out of a planned
clinical trials of potentially effective treatments for ATC are 180, and did not show statistical significance in improving
hampered by its low incidence and aggressiveness, which overall survival with the addition of combretastatin (5.2 vs.
limits patient enrolment, limits the treatment time-frame, 4.0 months). However, the combination schedule was well
and leads to poor statistical power [8]. However, some ran- tolerated, and no significant cardiovascular adverse drug
domized or single-arm clinical trials using combined treat- reactions were registered [59].
ments have been performed (Table 2). A Phase I/II randomized trial was performed to investi-
Catalano and colleagues [53] carried out a randomized gate the effect of crolibulin (dose range from 8 to 20 mg/
controlled phase II/III trial on a total of 25 ATC patients m2), a microtubule inhibitor, plus cisplatin (dose range

13
Medical Oncology (2020) 37:19 Page 11 of 15  19

from 75 to 100 mg/m2) in 27 adults with solid tumours, study will also evaluate the combination’s effect on tumour
including 16 ATC patients. Of these ATC patients, one had shrinkage and growth.
a complete response, and another achieved a stable disease. A multicentre phase II Study (NCT02152137) is currently
However, grade 3 and 4 toxicities were registered in at least active, but not recruiting, to evaluate the effects of efatuta-
one third of patients [60]. A phase I trial (NCT00004074) zone in combination with paclitaxel, in patients affected by
to investigate the effectiveness of interleukin-12 and tras- advanced ATC. Patients receive paclitaxel intravenously
tuzumab in patients who have ATC with high levels of over 3 h on day 1, and efatutazone dihydrochloride orally
HER2/neu and have not responded to previous therapy has twice daily on days 1 through 21.
been completed, but results are not yet available. Finally, An active but not recruiting pilot prognostic study
a phase I, pharmacokinetic and biologic correlative study (NCT02862470) has been set up to analyse exosomal bio-
(NCT00005842) using the farnesyl-transferase inhibitor logical markers in thyroid cancer patients treated with lov-
R115777 (NSC702818) and trastuzumab in combination astatin and vildagliptin, an oral anti-hyperglycaemic agent
has been performed in patients with advanced or metastatic of the new a dipeptidyl peptidase-4 inhibitor class of drugs
ATC cancer. Patients received trastuzumab IV over 90 min [61]. The investigators expect to enrol 30 patients with pap-
on days 1, 8, 15, and 22, plus oral R115777 twice daily for illary, follicular, or ATC tumours, and collect their urine
3 weeks. Treatment continued every 28 days in the absence samples before surgery, immediately after surgery and post-
of disease progression or unacceptable toxicity. Cohorts of operatively at 3, 6 and 12 months.
3 to 6 patients received escalating doses of R115777 until Another phase II study (NCT03246958) plans to inves-
the maximum tolerated dose (MTD) was determined. The tigate the effects of nivolumab, a monoclonal antibody
MTD is defined as the dose at which no more than 1 of 6 directed against PD-1 [62], plus ipilimumab, an antibody
patients experience dose-limiting toxicities. This trial has against cytotoxic T-lymphocyte antigen 4 [63], in RAI-
been completed, but results have not been published yet. refractory, aggressive thyroid cancers. This study also will
include exploratory cohorts in medullary cancer and ATC,
Ongoing clinical trials on combination schedules and is in the recruiting phase. The combination of nivolumab
and ipilimumab is currently approved by FDA as treatment
Ongoing ATC clinical trials were retrieved from www.clini​ for patients with metastatic melanoma [64]. Ipilimumab will
caltri​ als.gov using the search phrase “anaplastic thyroid can- be administered via IV infusion, starting two weeks after
cer” (last accessed November 2019). The search was filtered nivolumab.
to “recruiting”, and “active” trials. Five ongoing clinical tri- Finally, a phase II, non-randomized, open-label clinical
als that use combined treatments were identified with this trial is currently recruiting patients affected by anaplastic and
search, and their characteristic are reported in Table 3. poorly differentiated thyroid carcinomas (NCT03181100).
A pilot study indicated to be early phase I The treatment is centred on the combinations of atezoli-
(NCT03085056) is currently recruiting ATC patients to test zumab, a humanized anti-programmed cell death ligand-1
the safety and tolerability of the therapeutic combination (PD-L1) monoclonal antibody [65], with vemurafenib, cobi-
of paclitaxel (80 mg/m2 weekly for 3 out of 4 weeks) plus metinib, bevacizumab and nab-paclitaxel, depending on the
trametinib (2 mg p.o. daily during each 4 week cycle). This mutational status of the tumours. The primary aim of the

Table 3  Ongoing clinical Combination treatments Phase Status ClinicalTrials.


trials regarding combination gov Identifier
schedules in ATC patients
Paclitaxel + trametinib Early phase I Recruiting NCT03085056
Cohort I (B-raf mutation): vemu- Phase II Recruiting NCT03181100
rafenib + cobimetinib + atezoli-
zumab
Cohort II (RAS, NF1, or NF2 muta-
tion): cobimetinib + atezolizumab
Cohort III (non-B-raf/non-RAS
mutation): bevacizumab + atezoli-
zumab
Cohort IV: nab-paclitaxel + atezoli-
zumab
Paclitaxel + efatutazone Phase II Active but not recruiting NCT02152137
Lovastatin + vildagliptin Pilot prognostic study Active but not recruiting NCT02862470
Nivolumab + ipilimumab Phase II Recruiting NCT03246958

13
19   Page 12 of 15 Medical Oncology (2020) 37:19

study is to determine if targeted therapy plus atezolizumab in highly specialized centres with no budget limitations. On
will lead to improved overall survival in ATC patients. In the other hand, a pragmatic approach could also be possible
particular, the first cohort of B-RAF mutated ATC patients and sustainable, using off-label drugs already registered in
receives vemurafenib p.o. twice daily on days 1–21, cobi- other neoplastic or non-neoplastic diseases.
metinib p.o. once daily on days 1–21, and atezolizumab Indeed, preclinical settings should emphasize giving
i.v. over 30–60 min on days 1 and 15, whereas the second already in-use antineoplastic drugs, such as chemothera-
cohort including patients with RAS, NF1, or NF2 mutations peutics (e.g. campthotecins or microtubule inhibitors) and
is treated with cobimetinib and atezolizumab. TKIs (e.g. sorafenib, levatinib, pazopanib or sunitinib) in
A third and a fourth cohort of patients not harbouring combination. This could achieve new therapeutic successes
any mutation in B-RAF and RAS genes will be adminis- in a clinical setting, using drugs with already known toxicity
tered with atezolizumab and bevacizumab or nab-paclitaxel. profiles, at lower costs, and with easier patient management.
Cycles will be repeated every 21 days in the absence of dis- However, it is also possible that well-known drugs combined
ease progression and unacceptable toxicity. in new schedules may lead to unexpected toxicities.
In conclusion, although continuous efforts and promising
results have been obtained in the preclinical research area,
Concluding remarks and future perspective additional clinical studies are still required to better eluci-
date the activity of combination treatments in the context of
ATC is a complex disease with a bad prognosis [66]. Con- clinical practice.
ventional treatments based on surgery, radioiodine and
chemotherapy always fail to cure ATC [66]. To date, new
insights from preclinical research on the genetic alterations Author contributions  GB had the idea for the article. DG, PO, MB
and GB performed the literature search and data analysis. DG and GB
and dysfunction of different signalling pathways that affect drafted and/or critically revised the work.
ATC development and biology offer the unprecedented pos-
sibility of having therapies for these new molecular targets. Funding  This study was supported by a grant from Associazione Itali-
Currently, enormous effort is being made to understand ATC ana per la Ricerca sul Cancro (AIRC; IG-17672) to GB.
molecular pathogenesis, as exemplified by the elucidation
of the fundamental role of several major signalling path- Compliance with ethical standards 
ways, and related molecular derangements. Indeed, many
of these molecular alterations represent novel diagnostic Conflict of interest  All authors declare that they have no conflicts of
interest.
and prognostic molecular markers, and therapeutic targets
for thyroid cancer. This then provides new opportunities for Ethical approval  Human and animal rights: no animal or human was
further research, and clinical development of novel treat- involved in this study.
ment strategies. To date, results obtained with experimental
models support the idea that combination treatments seem to
have synergistic antitumour activity in ATC, although their
rapid translation into the clinics is impeded by the difficulty References
in recruiting enough patients for randomized clinical stud-
ies. Moreover, embracing a personalized approach based on 1. Sherman SI. Thyroid carcinoma. Lancet. 2003;361(9356):501–11.
advanced genomics and proteomics will be crucial for devel- 2. Are C, Shaha AR. Anaplastic thyroid carcinoma: biology, patho-
oping tailored treatment plans that improve the chances of genesis, prognostic factors, and treatment approaches. Ann Surg
Oncol. 2006;13(4):453–64.
clinical success. For some authors, new affordable genomic 3. Voigt W, Kegel T, Weiss M, Mueller T, Simon H, Schmoll HJ.
analyses and the possibility to test these new drugs in pri- Potential activity of paclitaxel, vinorelbine and gemcitabine
mary cell cultures propagated in vitro from each ATC patient in anaplastic thyroid carcinoma. J Cancer Res Clin Oncol.
could ameliorate the personalization of therapy, increasing 2005;131(9):585–90. https:​ //doi.org/10.1007/s00432​ -005-0673-0.
4. Chintakuntlawar AV, Foote RL, Kasperbauer JL, Bible KC.
therapeutic effectiveness and avoiding the use of ineffec- Diagnosis and management of anaplastic thyroid cancer. Endo-
tive treatments [67]. However, the preclinical and clinical crinol Metab Clin North Am. 2019;48(1):269–84. https​://doi.
research on combination treatments in ATC may face a great org/10.1016/j.ecl.2018.10.010.
challenge in the near future. On one hand, efforts could be 5. Chen J, Tward JD, Shrieve DC, Hitchcock YJ. Surgery and radio-
therapy improves survival in patients with anaplastic thyroid
focused on the molecular characterization of tumours from carcinoma: analysis of the surveillance, epidemiology, and end
individual patients, and identifying the best corresponding results 1983–2002. Am J Clin Oncol. 2008;31(5):460–4. https​://
drug treatment, using novel and still experimental com- doi.org/10.1097/COC.0b013​e3181​6a61f​3.
pounds. This choice is of course highly desirable, because of 6. Molinaro E, Romei C, Biagini A, Sabini E, Agate L, Mazzeo
S, et al. Anaplastic thyroid carcinoma: from clinicopathology
its high probability of success, but it could be achieved only

13
Medical Oncology (2020) 37:19 Page 13 of 15  19

to genetics and advanced therapies. Nat Rev Endocrinol. inhibitors. J Med Chem. 2004;47(27):6812–20. https​: //doi.
2017;13(11):644–60. https​://doi.org/10.1038/nrend​o.2017.76. org/10.1021/jm030​502y.
7. American Thyroid Association. ATA Guidelines. 2018. https​ 24. Yeung SC, Xu G, Pan J, Christgen M, Bamiagis A. Manumycin
://www.thyro​id.org/profe​ssion​als/ata-profe​ssion​al-guide​lines​/. enhances the cytotoxic effect of paclitaxel on anaplastic thyroid
Accessed 14 June 2019. carcinoma cells. Cancer Res. 2000;60(3):650–6.
8. Saini S, Tulla K, Maker AV, Burman KD, Prabhakar BS. Thera- 25. Xu G, Pan J, Martin C, Yeung SC. Angiogenesis inhibition in
peutic advances in anaplastic thyroid cancer: a current perspec- the in vivo antineoplastic effect of manumycin and paclitaxel
tive. Mol Cancer. 2018;17(1):018–0903. against anaplastic thyroid carcinoma. J Clin Endocrinol Metab.
9. Tennvall J, Lundell G, Wahlberg P, Bergenfelz A, Grimelius L, 2001;86(4):1769–77. https​://doi.org/10.1210/jcem.86.4.7374.
Akerman M, et al. Anaplastic thyroid carcinoma: three protocols 26. She M, Jim Yeung SC. Combining a matrix metalloprotein-
combining doxorubicin, hyperfractionated radiotherapy and sur- ase inhibitor, a farnesyltransferase inhibitor, and a taxane
gery. Br J Cancer. 2002;86(12):1848–53. https​://doi.org/10.1038/ improves survival in an anaplastic thyroid cancer model. Can-
sj.bjc.66003​61. cer Lett. 2006;238(2):197–201. https​://doi.org/10.1016/j.canle​
10. Smallridge RC, Ain KB, Asa SL, Bible KC, Brierley JD, Bur- t.2005.07.012.
man KD, et al. American Thyroid Association guidelines for 27. Yeung SC, She M, Yang H, Pan J, Sun L, Chaplin D. Combi-
management of patients with anaplastic thyroid cancer. Thyroid. nation chemotherapy including combretastatin A4 phosphate
2012;22(11):1104–39. https​://doi.org/10.1089/thy.2012.0302. and paclitaxel is effective against anaplastic thyroid cancer
11. Pasqualetti G, Ricci S, Boni G, Tognini S, Polini A, Mariani G, in a nude mouse xenograft model. J Clin Endocrinol Metab.
et al. Off-label use of sunitinib in patients with advanced, epithe- 2007;92(8):2902–9. https​://doi.org/10.1210/jc.2007-0027.
lial thyroid cancer: a retrospective analysis. Recent Pat Endocr 28. Copland JA, Marlow LA, Kurakata S, Fujiwara K, Wong AK,
Metab Immune Drug Discov. 2012;6(2):171–6. Kreinest PA, et al. Novel high-affinity PPARgamma agonist
12. Schoenfeld JD, Odejide OO, Wirth LJ, Chan AW. Survival of a alone and in combination with paclitaxel inhibits human ana-
patient with anaplastic thyroid cancer following intensity-modu- plastic thyroid carcinoma tumor growth via p21WAF1/CIP1.
lated radiotherapy and sunitinib—a case report. Anticancer Res. Oncogene. 2006;25(16):2304–17. https​: //doi.org/10.1038/
2012;32(5):1743–6. sj.onc.12092​67.
13. Denaro N, Nigro CL, Russi EG, Merlano MC. The role of 29. Chung YS, Cho S, Ryou HJ, Jee HG, Choi JY, Yoon K, et al. Is
chemotherapy and latest emerging target therapies in anaplastic there a treatment advantage when paclitaxel and lovastatin are
thyroid cancer. Onco Targets Ther. 2013;9:1231–41. https​://doi. combined to dose anaplastic thyroid carcinoma cell lines? Thy-
org/10.2147/ott.s4654​5. roid. 2011;21(7):735–44. https​://doi.org/10.1089/thy.2010.0304.
14. Worden F. Treatment strategies for radioactive iodine-refrac- 30. Park CH, Han SE, Nam-Goong IS, Kim YI, Kim ES. Combined
tory differentiated thyroid cancer. Ther Adv Med Oncol. effects of baicalein and docetaxel on apoptosis in 8505c anaplas-
2014;6(6):267–79. https​://doi.org/10.1177/17588​34014​54818​8. tic thyroid cancer cells via downregulation of the ERK and Akt/
15. Cabanillas ME, Habra MA. Lenvatinib: Role in thyroid cancer and mTOR pathways. Endocrinol Metab (Seoul). 8505c;33(1):121–32.
other solid tumors. Cancer Treat Rev. 2016;42:47–55. https​://doi. https​://doi.org/10.3803/EnM.2018.33.1.121.
org/10.1016/j.ctrv.2015.11.003. 31. Garg M, Kanojia D, Mayakonda A, Ganesan TS, Sadhanandhan B,
16. Tran S, DeGiovanni PJ, Piel B, Rai P. Cancer nanomedicine: Suresh S, et al. Selinexor (KPT-330) has antitumor activity against
a review of recent success in drug delivery. Clin Transl Med. anaplastic thyroid carcinoma in vitro and in vivo and enhances
2017;6(1):44. https​://doi.org/10.1186/s4016​9-017-0175-0. sensitivity to doxorubicin. Sci Rep. 2017;7(1):9749. https​://doi.
17. Zhang RX, Wong HL, Xue HY, Eoh JY, Wu XY. Nanomedicine org/10.1038/s4159​8-017-10325​-x.
of synergistic drug combinations for cancer therapy - Strategies 32. Wang W, Zhou J, Zhao L, Chen S. Combination of SL327 and
and perspectives. J Control Release. 2016;240:489–503. https​:// Sunitinib Malate leads to an additive anti-cancer effect in doxo-
doi.org/10.1016/j.jconr​el.2016.06.012. rubicin resistant thyroid carcinoma cells. Biomed Pharmacother.
18. Di Desidero T, Antonelli A, Orlandi P, Ferrari SM, Fioravanti 2017;88:985–90. https​://doi.org/10.1016/j.bioph​a.2017.01.135.
A, Ali G, et al. Synergistic efficacy of irinotecan and sunitinib 33. Abdulghani J, Gokare P, Gallant JN, Dicker D, Whitcomb T,
combination in preclinical models of anaplastic thyroid cancer. Cooper T, et al. Sorafenib and quinacrine target anti-apoptotic
Cancer Lett. 2017;411:35–433. https​://doi.org/10.1016/j.canle​ protein MCL1: a poor prognostic marker in anaplastic thyroid
t.2017.09.032. cancer (ATC). Clin Cancer Res. 2016;22(24):6192–203. https​://
19. Kim S, Prichard CN, Younes MN, Yazici YD, Jasser SA, Bekele doi.org/10.1158/1078-0432.ccr-15-2792.
BN, et al. Cetuximab and irinotecan interact synergistically to 34. Cohen SM, Mukerji R, Timmermann BN, Samadi AK, Cohen MS.
inhibit the growth of orthotopic anaplastic thyroid carcinoma A novel combination of withaferin A and sorafenib shows syner-
xenografts in nude mice. Clin Cancer Res. 2006;12(2):600–7. gistic efficacy against both papillary and anaplastic thyroid can-
https​://doi.org/10.1158/1078-0432.ccr-05-1325. cers. Am J Surg. 2012;204(6):895–900. https​://doi.org/10.1016/j.
20. Kim S, Yazici YD, Barber SE, Jasser SA, Mandal M, Bekele amjsu​rg.2012.07.027.
BN, et al. Growth inhibition of orthotopic anaplastic thyroid 35. Park KC, Kim SM, Jeon JY, Kim BW, Kim HK, Chang HJ, et al.
carcinoma xenografts in nude mice by PTK787/ZK222584 and Synergistic activity of N-hydroxy-7-(2-naphthylthio) heptano-
CPT-11. Head Neck. 2006;28(5):389–99. https:​ //doi.org/10.1002/ mide and sorafenib against cancer stem cells. Anaplastic Thyroid
hed.20369​. Cancer Neoplasia. 2017;19(3):145–53. https​://doi.org/10.1016/j.
21. di Desidero T, Orlandi P, Gentile D, Bocci G. Effects of pazo- neo.2016.12.005.
panib monotherapy vs pazopanib and topotecan combination on 36. Ball DW, Jin N, Xue P, Bhan S, Ahmed SR, Rosen DM, et al.
anaplastic thyroid cancer cells. Front Oncol. 2019;9:1202. https​ Trametinib with and without pazopanib has potent preclinical
://doi.org/10.3389/fonc.2019.01202​. activity in thyroid cancer. Oncol Rep. 2015;34(5):2319–24. https​
22. Jing C, Gao Z, Wang R, Yang Z, Shi B, Hou P. Lenvatinib ://doi.org/10.3892/or.2015.4225.
enhances the antitumor effects of paclitaxel in anaplastic thyroid 37. Feng H, Cheng X, Kuang J, Chen L, Yuen S, Shi M, et al. Apat-
cancer. Am J Cancer Res. 2017;7(4):903–12. inib-induced protective autophagy and apoptosis through the
23. Millet R, Domarkas J, Houssin R, Gilleron P, Goossens JF, AKT-mTOR pathway in anaplastic thyroid cancer. Cell Death Dis.
Chavatte P, et  al. Potent and selective farnesyl transferase 2018;9(10):1030. https​://doi.org/10.1038/s4141​9-018-1054-3.

13
19   Page 14 of 15 Medical Oncology (2020) 37:19

38. Che HY, Guo HY, Si XW, You QY, Lou WY. Additive effect by Metab. 2015;100(2):E253–E261261. https​://doi.org/10.1210/
combination of Akt inhibitor, MK-2206, and PDGFR inhibitor, jc.2014-3101.
tyrphostin AG 1296, in suppressing anaplastic thyroid carcinoma 51. Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. The
cell viability and motility. Onco Targets Ther. 2014;7:425–32. heat shock protein 90 inhibitor SNX5422 has a synergistic activity
https​://doi.org/10.2147/ott.s5732​4. with histone deacetylase inhibitors in induction of death of ana-
39. Brauner E, Gunda V, Vanden Borre P, Zurakowski D, Kim YS, plastic thyroid carcinoma cells. Endocrine. 2016;51(2):274–82.
Dennett KV, et al. Combining BRAF inhibitor and anti PD-L1 https​://doi.org/10.1007/s1202​0-015-0706-7.
antibody dramatically improves tumor regression and anti tumor 52. Zhang L, Boufraqech M, Lake R, Kebebew E. Carfilzomib poten-
immunity in an immunocompetent murine model of anaplastic tiates CUDC-101-induced apoptosis in anaplastic thyroid cancer.
thyroid cancer. Oncotarget. 2016;7(13):17194–211. https​://doi. Oncotarget. 2016;7(13):16517–28.
org/10.18632​/oncot​arget​.7839. 53. Catalano MG, Pugliese M, Gallo M, Brignardello E, Milla P,
40. Gunda V, Gigliotti B, Ndishabandi D, Ashry T, McCarthy M, Orlandi F, et al. Valproic acid, a histone deacetylase inhibitor, in
Zhou Z, et al. Combinations of BRAF inhibitor and anti-PD-1/ combination with paclitaxel for anaplastic thyroid cancer: results
PD-L1 antibody improve survival and tumour immunity in an of a multicenter randomized controlled phase II/III trial. Int J
immunocompetent model of orthotopic murine anaplastic thy- Endocrinol. 2016;2930414(10):27.
roid cancer. Br J Cancer. 2018;119(10):1223–322. https​://doi. 54. Gottlicher M, Minucci S, Zhu P, Kramer OH, Schimpf A,
org/10.1038/s4141​6-018-0296-2. Giavara S, et al. Valproic acid defines a novel class of HDAC
41. Vanden Borre P, Gunda V, McFadden DG, Sadow PM, Varmeh S, inhibitors inducing differentiation of transformed cells. EMBO J.
Bernasconi M, et al. Combined BRAF(V600E)- and SRC-inhibi- 2001;20(24):6969–78. https:​ //doi.org/10.1093/emboj/​ 20.24.6969.
tion induces apoptosis, evokes an immune response and reduces 55. Sherman EJ, Dunn LA, Ho AL, Baxi SS, Ghossein RA, Fury
tumor growth in an immunocompetent orthotopic mouse model MG, et al. Phase 2 study evaluating the combination of sorafenib
of anaplastic thyroid cancer. Oncotarget. 2014;5(12):3996–4010. and temsirolimus in the treatment of radioactive iodine-refractory
https​://doi.org/10.18632​/oncot​arget​.2130. thyroid cancer. Cancer. 2017;123(21):4114–211.
42. Ingeson-Carlsson C, Martinez-Monleon A, Nilsson M. Dif- 56. Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM,
ferential effects of MAPK pathway inhibitors on migration and Soria JC, et al. Dabrafenib and trametinib treatment in patients
invasiveness of BRAF(V600E) mutant thyroid cancer cells in 2D with locally advanced or metastatic BRAF V600-mutant ana-
and 3D culture. Exp Cell Res. 2015;338(2):127–35. https​://doi. plastic thyroid cancer. J Clin Oncol. 2018;36(1):7–13. https​://
org/10.1016/j.yexcr​.2015.08.003. doi.org/10.1200/jco.2017.73.6785.
43. Chin LH, Hsu SP, Zhong WB, Liang YC. Combined treatment 57. Murakami H, Ono A, Takahashi T, Onozawa Y, Tsushima T,
with troglitazone and lovastatin inhibited epidermal growth factor- Yamazaki K, et al. Phase I study of Efatutazone, an oral PPAR-
induced migration through the downregulation of cysteine-rich gamma agonist, in patients with metastatic solid tumors. Antican-
protein 61 in human anaplastic thyroid cancer cells. PLoS ONE. cer Res. 2014;34(9):5133–41.
2015;10(3):e0118674. https:​ //doi.org/10.1371/journa​ l.pone.01186​ 58. Smallridge RC, Copland JA, Brose MS, Wadsworth JT, Houvras
74. Y, Menefee ME, et al. Efatutazone, an oral PPAR-gamma ago-
44. Lin BR, Chang CC, Chen LR, Wu MH, Wang MY, Kuo IH, nist, in combination with paclitaxel in anaplastic thyroid cancer:
et al. Cysteine-rich 61 (CCN1) enhances chemotactic migration, results of a multicenter phase 1 trial. J Clin Endocrinol Metab.
transendothelial cell migration, and intravasation by concomi- 2013;98(6):2392–400.
tantly up-regulating chemokine receptor 1 and 2. Mol Cancer 59. Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C, et al.
Res. 2007;5(11):1111–23. https​://doi.org/10.1158/1541-7786. Randomized safety and efficacy study of fosbretabulin with pacli-
mcr-06-0289. taxel/carboplatin against anaplastic thyroid carcinoma. Thyroid.
45. Zhong WB, Tsai YC, Chin LH, Tseng JH, Tang LW, Horng S, 2014;24(2):232–40. https​://doi.org/10.1089/thy.2013.0078.
et al. A synergistic anti-cancer effect of troglitazone and lovasta- 60. Gramza A, Balasubramaniam S, Fojo AT, Ward J, Wells SA.
tin in a human anaplastic thyroid cancer cell line and in a mouse Phase I/II trial of crolibulin and cisplatin in solid tumors with a
xenograft model. Int J Mol Sci. 2018;19(7):1834. https​://doi. focus on anaplastic thyroid cancer: phase I results. J Clin Oncol.
org/10.3390/ijms1​90718​34. 2013;31:6074. https​: //doi.org/10.1200/jco.2013.31.15_suppl​
46. Wong K, Di Cristofano F, Ranieri M, De Martino D, Di Cristo- .6074.
fano A. PI3K/mTOR inhibition potentiates and extends palboci- 61. Zhang Q, Xiao X, Li M, Yu M, Ping F, Zheng J, et al. Vildagliptin
clib activity in anaplastic thyroid cancer. Endocr Relat Cancer. increases butyrate-producing bacteria in the gut of diabetic rats.
2019;26(4):425–36. https​://doi.org/10.1530/erc-19-0011. PLoS ONE. 2017;12(10):e0184735. https:​ //doi.org/10.1371/journ​
47. Allegri L, Baldan F, Mio C, Puppin C, Russo D, Krystof V, et al. al.pone.01847​35.
Effects of BP-14, a novel cyclin-dependent kinase inhibitor, on 62. Nomura M, Otsuka A, Kondo T, Nagai H, Nonomura Y, Kaku
anaplastic thyroid cancer cells. Oncol Rep. 2016;35(4):2413–8. Y, et al. Efficacy and safety of retreatment with nivolumab in
https​://doi.org/10.3892/or.2016.4614. metastatic melanoma patients previously treated with nivolumab.
48. De Martino D, Yilmaz E, Orlacchio A, Ranieri M, Zhao K, Di Cancer Chemother Pharmacol. 2017;80(5):999–1004. https​://doi.
Cristofano A. PI3K blockage synergizes with PLK1 inhibition org/10.1007/s0028​0-017-3444-0.
preventing endoreduplication and enhancing apoptosis in ana- 63. Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance
plastic thyroid cancer. Cancer Lett. 2018;439:56–655. https​://doi. AM, Cowey CL, et  al. Adjuvant Nivolumab versus ipili-
org/10.1016/j.canle​t.2018.09.024. mumab in resected stage III or IV melanoma. N Engl J Med.
49. Baldan F, Mio C, Allegri L, Puppin C, Russo D, Filetti S, et al. 2017;377(19):1824–35. https:​ //doi.org/10.1056/NEJMoa​ 17090​ 30.
Synergy between HDAC and PARP inhibitors on proliferation of 64. Marconcini R, Spagnolo F, Stucci LS, Ribero S, Marra E, Rosa
a human anaplastic thyroid cancer-derived cell line. Int J Endo- F, et al. Current status and perspectives in immunotherapy for
crinol. 2015;2015:978371. https​://doi.org/10.1155/2015/97837​1. metastatic melanoma. Oncotarget. 2018;9(15):12452–70. https​://
50. Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. doi.org/10.18632​/oncot​arget​.23746​.
Novel heat shock protein 90 inhibitor NVP-AUY922 synergizes 65. Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD-L1
with the histone deacetylase inhibitor PXD101 in induction of as cancer therapeutics. J Hematol Oncol. 2019;12(1):92. https​://
death of anaplastic thyroid carcinoma cells. J Clin Endocrinol doi.org/10.1186/s1304​5-019-0779-5.

13
Medical Oncology (2020) 37:19 Page 15 of 15  19

66. Salehian B, Liem SY, Mojazi Amiri H, Maghami E. Clinical Publisher’s Note Springer Nature remains neutral with regard to
trials in management of anaplastic thyroid carcinoma; progres- jurisdictional claims in published maps and institutional affiliations.
sions and set backs: a systematic review. Int J Endocrinol Metab.
2019;17(1):e67759. https​://doi.org/10.5812/ijem.67759​.
67. Ferrari SM, Fallahi P, La Motta C, Elia G, Ragusa F, Ruffilli I,
et al. Recent advances in precision medicine for the treatment
of anaplastic thyroid cancer. Expert Rev Precis Med Drug Dev.
2019;4(1):37–49. https​://doi.org/10.1080/23808​993.2019.15659​
40.

13

You might also like