Skip to main content
Erschienen in: Molecular Cancer 1/2018

Open Access 01.12.2018 | Review

Therapeutic advances in anaplastic thyroid cancer: a current perspective

verfasst von: Shikha Saini, Kiara Tulla, Ajay V. Maker, Kenneth D. Burman, Bellur S. Prabhakar

Erschienen in: Molecular Cancer | Ausgabe 1/2018

Abstract

Thyroid cancer incidence is increasing at an alarming rate, almost tripling every decade. In 2017, it was the fifth most common cancer in women. Although the majority of thyroid tumors are curable, about 2–3% of thyroid cancers are refractory to standard treatments. These undifferentiated, highly aggressive and mostly chemo-resistant tumors are phenotypically-termed anaplastic thyroid cancer (ATC). ATCs are resistant to standard therapies and are extremely difficult to manage. In this review, we provide the information related to current and recently emerged first-line systemic therapy (Dabrafenib and Trametinib) along with promising therapeutics which are in clinical trials and may be incorporated into clinical practice in the future. Different categories of promising therapeutics such as Aurora kinase inhibitors, multi-kinase inhibitors, epigenetic modulators, gene therapy using oncolytic viruses, apoptosis-inducing agents, and immunotherapy are reviewed. Combination treatment options that showed synergistic and antagonistic effects are also discussed. We highlight ongoing clinical trials in ATC and discuss how personalized medicine is crucial to design the second line of treatment. Besides using conventional combination therapy, embracing a personalized approach based on advanced genomics and proteomics assessment will be crucial to developing a tailored treatment plan to improve the chances of clinical success.
Abkürzungen
ALK
Anaplastic Lymphoma Kinase
ATA
American Thyroid Association
BET
Bromodomain and extra-terminal containing protein
CDK
Cyclin-dependent kinase
DTC
Differentiated Thyroid Cancer
EGFR
Epidermal Growth Factor Receptor
FDA
Food and Drug Administration
FGFR
Fibroblast Growth Factor Receptor
FLT-3
FMS Related Tyrosine Kinase 3
FNA
Fine Needle Aspiration
HDAC
Histone Deacetylase
Hsp90
Heat Shock Protein 90
HSPs
Heat Shock Proteins
LPEI
Linear Polyethylenimine
NIS
Sodium iodide Symporter
ORR
Objective Response Rate
OS
Overall Survival
PARP
Poly ADP ribose polymerase
PD-1
Programmed Cell Death Protein-1
PDGFR
Platelet-Derived Growth Factor Receptor
PDL-1
Programmed Death Ligand-1
PEG
Polyethylene Glycol
PLGA
Poly-lactic-co-glycolic acid
PLK-1
Polo-like kinase-1
PPARγ
Peroxisome Proliferator-Activated Receptor gamma;
PR
Partial Response
PTC
Papillary Thyroid Cancer
PTDC
Poorly Differentiated Thyroid Cancer
RR
Response Rate
SAHA
Suberoylanilide Hydroxamic Acid
TAM
Tumor-Associated Macrophages
TDP-A
Thailandepsin A
TG
Thyroglobulin
TPO
Thyroid Peroxidase
TRAIL
TNF-Related Apoptosis-Inducing Ligand (TRAIL)
TSHR
Thyroid-Stimulating Hormone Receptor
TTF-1
Thyroid Transcription Factor-1
TTP
Time to Progression
UICC
Union for International Cancer Control
VE-Cadherin
Vascular Endothelial Cadherin
VEGFR
Vascular Endothelial Growth Factor Receptor
VPA
Valproic acid

Background

Thyroid cancer is the most common endocrine-related malignancy, accounting for more than 90% of endocrine cancers [1]. In 2017, more than 56,870 new cases were diagnosed in the United States constituting 3.4% of all new cancer cases [2]. The majority of thyroid tumors are pathologically differentiated cancers and exhibit good prognosis with > 98% five-year survival. Among these differentiated thyroid cancers (DTC), papillary thyroid cancer (PTC) is the most common, comprising about 80% of all thyroid cancers. The other DTCs include follicular thyroid cancer (FTC), and medullary thyroid cancers (MTC). These malignancies originate from follicular and parafollicular cells, respectively. A small subset of thyroid cancer, known as anaplastic thyroid cancer (ATC), is undifferentiated, and nearly incurable with a median survival of only six months. Because of its dismal prognosis, it is responsible for 40–50% of total thyroid cancer-related deaths in the United States. Poorly differentiated thyroid cancer (PDTC) resembles ATC due to its aggressive nature but has partial overlap with FTC/PTC, retaining follicular elements and thyroglobulin production [3]. Thus, as opposed to most DTCs for which surgery and radioiodine therapy will result in an excellent prognosis, ATC poses a significant clinical challenge as it is highly aggressive and with comparably no effective therapeutic options.
According to the American Thyroid Association (ATA) guidelines, first-line treatment for ATC includes surgical resection, if possible, and external beam radiation therapy for local control. Though total thyroidectomy with high-dose radiation therapy is associated with improved survival [4]; second-line treatment with targeted therapies, single or in combination, are often employed. Thus, current clinical ATC management is still evolving, and new treatments are being urgently developed. This review provides comprehensive information related to therapeutic options that are available or in the pipeline. A comprehensive review of original research articles, reviews, clinical investigations, and editorials published in the last ten years from Medline/PubMed, Google Scholar and SciFinder was conducted. Clinical trials related information was assessed from https://​clinicaltrials.​gov/​. The purpose of this study is to highlight current treatment strategies and their limitations, improve our understanding of their pitfalls, and to propose ways to overcome these hurdles.

Clinical manifestation and diagnosis

ATC often presents as a neck mass causing dysphagia, dysphonia or hoarseness, stridor, and dyspnea due to mass effect on the esophagus and the trachea [5, 6]. According to the Union for International Cancer Control (UICC) staging system, ATC tumors are automatically designated as stage IV disease irrespective of tumor burden and presence or absence of metastasis. They are sub-classified as IVa, IVb, IVc and IVd depending on the extent of invasion of the surrounding tissue layers [7]. Pathologically, ATC cells are spindle-shaped, giant and squamoid cells, with a high mitotic index, necrosis, hemorrhage and vascular invasion [8]. Due to their vascularity, misdiagnosis with angiosarcoma is common. About 70% of ATCs invade surrounding tissues including fat, trachea, esophagus and larynx. The most common metastatic sites in ATC patients are the lungs, bone, and brain [8].
The diagnosis of ATC is confirmed via fine needle aspiration (FNA). However, false-negatives are common due to low cellularity and inflammatory and necrotic debris. At the same time, there is a lack of definitive molecular biomarkers to detect ATC in FNA biopsies with high sensitivity. To improve the sensitivity and specificity of diagnosis, molecular diagnostics using customized panels based on next-generation sequencing holds promise for the future [9, 10].

Current therapeutic regimes

For ATC, a multimodal therapeutic approach is employed which includes surgical resection, hyper-fractionated accelerated external beam radiotherapy, followed by a combination of chemotherapies and/or palliative care [11]. Despite multi-modality treatment, the prognosis for ATC patients is poor. Due to the failure of single-agent chemotherapy, a combination of two or more drugs such as Paclitaxel, Cisplatin, Doxorubicin, Pegfilgrastim and Docetaxel is administered to ATC patients [12]. Second-line treatments include more targeted therapies such as tyrosine kinase inhibitors, anti-angiogenic drugs, and agonists and multi-kinase inhibitors targeting hyperactive BRAF, mTOR, and/or BCR-ABL. In the past decade, several novel drugs that target proliferation, angiogenesis, immunosuppression, metabolomic changes and epigenetic reprogramming have been evaluated (Fig. 1). However, it is pertinent to note that clinical trials of potentially effective treatments for ATC are hampered by its low incidence and aggressiveness that limits enrollment leading to poor statistical power and a limited treatment time-frame. Current mainstays of ATC therapeutic management include:

Surgical resection

Surgical management is generally precluded for patients with ATC due to extensive metastasis [13]; however, complete surgical resection when achievable with limited morbidity is recommended for localized disease. If the tumor is limited to the thyroid parenchyma, thyroidectomy with wide margins is recommended, especially if the tumor reflects good prognostic features including unilobar disease, diameter < 5 cm, and without nodal spread, for which lobectomy alone may be performed [14]. In fact, in the rare instance of a small, unilocular, contained mass, total thyroidectomy appears to offer no survival advantage over lobectomy while carrying greater operative risks [15, 16]. Interestingly, in a Mayo Clinic series over 50-years, incomplete resection was no worse than negative margins, in regard to overall patient survival. This is likely due to the overall poor prognosis and difficulty in local control even with surgery for this aggressive disease. Regardless, many still advocate for total thyroidectomy and central node dissection, and current guidelines recommend this approach if R0 (microscopically negative resection) or R1 (grossly negative, microscopic positive) resection can be achieved [12, 16].
Complete resection is a challenge, often due to tumor size, extra organ growth extension, and local invasion. The goal of surgery is for a margin-negative R0 resection. Pre-operative evaluation with high-quality fine-cut cross-sectional imaging and ultrasound is necessary to determine tumor extent and the possible local involvement of the carotid artery, jugular vein, vagus nerve and its branches, trachea, esophagus, sternocleidomastoid and strap muscles. Speech changes may already be evident at the time of presentation, which is concerning for recurrent laryngeal nerve involvement. Laryngoscopy can be an invaluable tool to assess vocal cord mobility while bronchoscopy allows visual assessment for tracheal involvement. If one recurrent laryngeal nerve is already involved, then it is of utmost importance that the contralateral nerve is preserved and/or only ipsilateral lobectomy is performed, if possible. If a contralateral resection is necessary to obtain negative margins, the use of a nerve stimulator may improve recurrent laryngeal nerve identification during surgery, although this is surgeon-dependent. Concurrently, imaging is critical in the assessment of metastatic disease, in which case resection should be reserved purely for cases of airway compromise.
After a proper staging assessment, if resection can be performed, patients have improved outcomes even with margin positivity. However, laryngectomy is usually discouraged due to the likelihood of persistent disease and the severe morbidity associated with this procedure. Current recommendations, based on moderate quality evidence, advise for lobectomy, total, or near-total thyroidectomy, with therapeutic lymph node dissection [12]. This approach involves the standard lifting of sub-platysmal flaps and division of the deep cervical fascia in the midline to expose the underlying gland. Involvement of the strap muscles necessitates the division of the muscles, which should be performed en bloc with the thyroid. Due to the desire to obtain wide negative margins, and because of the infiltrative nature of this cancer, the parathyroid glands may need to be sacrificed. In these instances, efforts should be made to identify and preserve all uninvolved parathyroid glands, and to confirm histologically, mince, and re-implant the tissue to prevent severe hypocalcemia. Central and lateral lymphadenectomy is performed at the same time of surgical resection, and additional soft tissue, fascia, muscle, or veins should be taken en bloc if invaded.
Tracheostomy or palliative resection is sometimes necessary due to tracheal involvement in order to manage airway compromise or esophageal obstruction. As has been mentioned, chemotherapy alone is ineffective in controlling this disease. However, a study by the Swedish Anaplastic Thyroid Cancer Group combined neoadjuvant Adriamycin and 3 weeks of radiation followed by debulking surgery and adjuvant radiation [17]. With this protocol, some previously unresectable tumors became resectable. Though there were individual patient successes, overall improvement in local control and overall survival remains to be demonstrated. Clearly, surgery has a role in the treatment of this disease, particularly for small, localized tumors, and in the palliative setting to avoid suffocation; however, optimal outcomes involve multimodality treatment strategies.

External beam radiation therapy

Due to their undifferentiated phenotype and lack of sodium-iodine symporters, radioactive iodine (I131) ablation therapy is ineffective in localizing radiation to the gland and therefore, external beam radiation therapy is utilized. Due to the aggressive time-course of the disease, accelerated, hypo-fractionated radiation treatment is preferred. However, it can induce acute toxicity. The dosing regimens utilized are based on case studies, non-randomized trials, and institutional experience. The contribution of high-intensity external beam radiation therapy to improve survival relies on the ability to adequately resect the tumor, which as mentioned above, is only possible in a minority of cases. A recent meta-analysis including 17 retrospective studies of 1147 patients, showed that postoperative radiotherapy significantly improved survival in patients with resected tumors versus patients with surgery alone (HR, 0.556; 95% confidence interval, 0.419–0.737; p < 0.001). Moreover, it was also found that patients with stage IVa (HR, 0.364; p = 0.012) and IVb (HR, 0.460; p = 0.059) disease may derive a survival benefit from post-operative radiotherapy, whereas stage IVc patients may not [18]. A meta-analysis of 1288 ATC patients from National Cancer Database revealed that patients with unresected ATC might benefit from multimodal loco-regional treatment that incorporates higher (60–75 Gy) versus lower (45–59.9 Gy) radiation doses [19]. The major challenge in analyzing the advantages of adjuvant radiation is the inherent time bias, as many patients studied already had extremely limited survival at the time of diagnosis. Recently, a unique single platform dual therapy based on polyethylene glycol-coated [64Cu] CuS nanoparticles was developed, which combines radiation and photothermal therapy. This investigation showed promising improvement in overall survival in a preclinical orthotopic mouse model and has the potential for clinical translation [20].
Attempts have also been made to re-sensitize ATC cells to radioiodine therapy by reintroducing sodium iodide symporters. In this context, a recent study showed promising results by employing nanoparticle vectors (polyplexes) based on linear polyethylenimine (LPEI) and polyethylene glycol (PEG). These nanoparticles were coupled to the synthetic peptide GE11, which acts as an EGFR ligand and provides the basis for directed delivery. These nanoparticles effectively induced sodium-iodide symporters in ATC cells with high EGFR expression [21].

Chemotherapy

As per National Comprehensive Cancer Network guidelines, for stage IVa/ IVb ATC tumors total thyroidectomy (if possible) should be followed by external beam radiotherapy and systemic therapy with genotoxic drugs such as Paclitaxel, Doxorubicin, Docetaxel, Carboplatin, Dabrafenib and Trametinib. Recommended regimens include Paclitaxel and Carboplatin combinations, Docetaxel and Doxorubicin combinations, Paclitaxel alone, or Doxorubicin alone [12]. For BRAFV600E mutation-positive tumors, a combination of Dabrafenib and Trametinib has shown promising clinical responses and is recommended [22]. For stage IVc patients, in addition to these regimens, palliative radiation therapy for locoregional metastases and second-line treatment with various systemic chemotherapies are often considered. Though Paclitaxel is the most effective chemotherapeutic drug, chemo-resistance is common, mainly due to tumor-associated macrophages (TAMs) in ATC. TAMs occupy 50% of the tumor volume and provides paracrine signals via CSF-1/CSF-1R axis, which promotes tumor progression [23]. Thus, targeting CSF-1/CSF-1R pathway in TAMs was shown to restore the sensitivity of thyroid cancer cells to Paclitaxel [24]. Along similar lines, JAK/STAT inhibitors can also be employed for Paclitaxel-resistant tumors [25]. Such therapeutics based on the mutational and proteomic landscape of tumors might be helpful in overcoming therapeutic resistance in ATC.

Targeted/multi-targeted inhibitors

Targeted inhibitors act on a specific target molecule and prevent cancer growth and progression. These inhibitors usually target hyperactive or mutant molecules active in signaling pathways in cancer cells. RAF/MAPK signaling is integral to ATC progression and some of the molecular therapeutics including the BRAF inhibitor (Vemurafenib/PLX4032) and the MEK inhibitor (Selumetinib/AZD6244) target this signaling cascade. In general, these single-targeted inhibitors failed to show significant therapeutic responses in ATC patients leading to the use of multi-kinase inhibitors (MKIs). MKIs can simultaneously act on two or more targets. MKIs such as Sorafenib, Axitinib, Pazopanib and Sunitinib has been evaluated in preclinical models, as well as in clinical trials, and have shown some encouraging results (Table 1). Sorafenib demonstrated progression-free survival (PFS) of about five months in phase III clinical trial with patients exhibiting manageable toxicities when compared to the placebo group [26]. However, most of the patients experienced disease relapse indicating the need for a combination of MKIs to improve outcomes. In this context, in advanced DTC patients who were unsuccessfully treated with Sorafenib, salvage therapy using Sunitinib, Pazopanib, Cabozantinib, Lenvatinib, and Vemurafenib was tested. With salvage therapy, a partial response (PR) was observed in 41% (7/17) patients suggesting a synergy of these drugs with Sorafenib [27]. In an attempt to improve the specificity of Sorafenib, it was chemically loaded in Poly-lactic-co-glycolic acid (PLGA) nanoparticles and combined with Cetuximab (EGFR inhibitor) [28]. This formulation significantly improved cytotoxicity against ATC cells without affecting normal thyroid cells.
Table 1
Results from clinical trials conducted in advanced, metastatic, radioiodine-refractory and anaplastic thyroid cancers conducted between 2013 and 2017 in the United States (Source: https://​clinicaltrials.​gov/​)
No.
Drug
Phase
Cancer
Number of patients
Response Rate
Progression free survival
Overall survival
Reference
1
Sorafenib (Bay43–9006, Nexavar)
II
ATC
20
PR in 2/20 (10%); Stable disease in 5/20 (25%)
1.9 months
[107]
2
Carbozantinib
III
MTC
330
28%
11.2
[108]
3
Efatutazone+ Paclitaxel
I
ATC
15
PR = 1; SD = 7
3.3 months
[39]
4
Pazopanib
II
Advanced and progressive medullary
35
5/35
9.4
19.9
[109]
5
Fosbretabulin + Paclitaxel/Carboplatin
II
ATC
8
20%
3.3
5.2 months
[110]
6
Vemurafenib
 
BRAFV600E positive, metastatic, radio-iodine refractory PTC
26
10/26
[110]
7
Axitinib
II
Advanced thyroid cancer
52
35%
16.1
23.2
[111]
8
Levatinib
III
Iodine refractor TC
261
64.8%
18.3
[112]
9
Sunitinib (second line of therapy)
II
Progressive, radio-iodine Refractory thyroid cancer
25
5/20 (25%)
6 months
13 months
[113]
10
Cabozantinib (XL-184)
III
Advanced MTC
  
11.2 months
[114]
11
Dabrafenib plus trametinib
II
BRAF V600E–mutated anaplastic thyroid cancer
16
69%
[23]
Another MKI, Lenvatinib, was evaluated in a phase III trial in metastatic DTCs with a 65% response rate (RR), and a median PFS of 18.3 months versus 3.6 months in placebo-treated patients [29]. A retrospective analysis of ATC patients treated with Lenvatinib resulted in partial responses (PR) in 60% (3/5) of patients whereas 40% (2/5) experienced stable disease with manageable toxicity. In addition, the observed objective response rate (ORR) was 60% with median time to progression (TTP) and overall survival (OS) as 88 and 165 days respectively [30].
A very recent clinical trial in BRAFV600E–mutated ATCs showed positive results by using a combination of Dabrafenib (150 mg twice daily) and Trametinib (2 mg once daily). Prior to the treatment, all patients received either surgery or radiation therapy. The overall response rate (RR) was 69% (11 of 16; 95% CI, 41% to 89%). Predicted 12-month estimates of median duration of response, PFS and OS were 90%, 79%, and 80%, respectively. So far, this is the first clinical trial that showed a high clinical response for this orphan disease [22].

Ongoing clinical trials

Some potential MKIs have been tested in clinical trials and a list of recently completed clinical trials in advanced thyroid cancers is outlined in Table 1. In advanced FTC, encouraging results were obtained by using the selective allosteric MEK1 and MEK2 inhibitor Selumetinib (AZD6244, ARRY-142886). Particularly, Selumetinib resensitized radio-iodine refractory ATC to uptake radio-iodine and can be used as a complementary therapy [31]. Targeting mTOR signaling holds promise and a phase I/II clinical trial evaluating the efficacy of mTOR inhibitor, Sapanisertib is underway. Several clinical trials are also underway to assess the combination of Selumetinib, Everolimus, Lenvatinib, Cabozantinib, Vandetanib and Vatalanib in ATC patients [32]. A recently completed phase II/III clinical trial using a combination of Paclitaxel and Valproic acid (VPA) showed no benefit in overall survival and disease progression [33]. Four drugs that have been approved for advanced thyroid cancer treatment after phase III clinical trial completion include Vandetanib (ZETA), Cabozantinib (EXAM), Sorafenib (DECISION) and Lenvatinib (SELECT) [34, 35]. Of note, several MKIs such as Sorafenib, Axitinib, and Sunitinib exhibited limited efficacy [36, 37]. Aimed at improving the Paclitaxel efficacy, a phase I clinical trial using a combination of Paclitaxel and the PPARγ agonist, Efatutazone demonstrated safety and merited further evaluation in phase II in ATC patients which is ongoing (Table 2) [38].
Table 2
Ongoing Clinical Trials in Anaplastic Thyroid Cancer (as on July 15, 2018), listed from https://​clinicaltrials.​gov/​
S. No
Phase
Drug
Drug Action
Clinical Trial No.
Status
Sponsors
1
II
MLN0128
mTOR kinase inhibitor
NCT02244463
Recruiting
Dana-Farber Cancer Institute, USA
2
II
Lenvatinib
MKI against VEGFR1, 2, and 3
NCT02726503
Recruiting
Translational Research Informatics Center, Kobe, Hyogo, Japan
NCT02657369
Recruiting
Eisai Inc. USA
3
Early phase I
Trametinib in combination with Paclitaxel
MEK inhibitor (Trametinib) with chemotherapy
NCT03085056
Recruiting
Memorial Sloan Kettering Cancer Center, USA
4
II
Pembrolizumab
Antibody against PD-1 receptor
NCT02688608
Recruiting
University of Texas Southwestern Medical Center, USA
5
II
Inolitazone Dihydrochloride (Efutazone) and Paclitaxel
PPAR-γ agonist (Efutazone) with chemotherapy
NCT02152137
Recruiting
Alliance for Clinical Trials in Oncology, USA
6
I
Combination of Durvalumab (MEDI4736) or Tremelimumab with Stereotactic Body Radiotherapy (SBRT)
Checkpoint inhibitor drugs: Durvalumab (PD-1/PDL-1 interaction blocker) and Tramelimumab (anti-CTLA4 antibody) with radiations
NCT03122496
Recruiting
Memorial Sloan Kettering Cancer Center, USA
7
II
Intensity-Modulated Radiation Therapy and Paclitaxel with or Without Pazopanib Hydrochloride
Pazopanib is a MKI against c-kit, FGFR, PDGFR and VEGFR
NCT01236547
Ongoing but not yet recruiting participants
National Cancer Institute (NCI), USA
8.
II
Ceritinib
ALK inhibitor
NCT02289144
Recruiting
University of Texas Southwestern Medical Center, USA
9
II
Atezolizumab Combinations with or without chemotherapy such as paclitaxel, Vemurifinib, Nab-paclitaxel, Cobimetinib and Bevacizumab
anti-PDL-1 antibody (Atezolizumab)
NCT03181100
Recruiting
M.D. Anderson Cancer Center, USA
10
I
FAZ053 as Single Agent and in combination with PDR001
FAZ053 is anti-PDL-1 antibody and PDR001 is monoclonal antibody against PD-1.
NCT02936102
Recruiting
Novartis Pharmaceuticals, USA
11
II
Dabrafenib and Trametinib
Dabrafenib acts against BRAFV600E mutations and Trametinib is MEK (1 and 2) inhibitor
NCT02034110
Recruiting
GlaxoSmithKline, USA
12
II
GW 786034 (Pazopanib Hydrochloride)
Pazopanib is a MKI against c-kit, FGFR, PDGFR and VEGFR
NCT00625846
Active, not recruiting
National Cancer Institute (NCI), USA
13
II
Pembrolizumab, Chemotherapy,and Radiation Therapy With or Without Surgery
anti-PD1 immunotherapy
NCT03211117
Active, not recruiting
Mayo Clinic,National Cancer Institute (NCI), USA
14
I/II
PDR001
anti-PD1 monoclonal antibody
NCT02404441
Recruiting
Novartis Pharmaceuticals

Promising therapeutic options

Therapeutic success in ATC patients has been very limited and thus, there is a continuing need to develop novel therapies. Several pre-clinical investigations have been carried out to explore the potential of various drugs, and some of the promising categories are discussed below:

Aurora kinase inhibitors

Aurora kinases are serine/threonine kinases involved in chromosomal segregation and cytokinesis during mitosis. These kinases include three members: Aurora A, B and C. Besides mitosis, these are involved in determining cell polarity, migration and invasion, and telomerase activity [39]. Their dysregulation is frequently noted in ATC as compared to PTC or normal thyroid tissues. [40]. Several Aurora kinase inhibitors including MK-0457 (VX-680), SNS-314 Mesylate, ZM447439, and AZD1152 have been tested and have shown significant cell cycle arrest and subsequent reduction in growth and proliferation in vitro. Particularly, administration of MLN8054 reduced tumor volume by 86% in an ATC xenograft mouse model [41]. In a different study, a combination of MLN8054 with Bortezomib (proteasome inhibitor) induced apoptosis and cell cycle arrest in ATC cells [42]. Another Aurora kinase inhibitor, Pazopanib showed synergistic cytotoxicity with Paclitaxel [43].
Another member of mitosis-related kinases, Polo-like kinase-1 (PLK-1), which regulates chromosomal segregation, is highly active in ATC. Its inhibitor, GSK461364 induced apoptosis in both ATC allograft mouse model and PDTC-derived cell lines [44].

Natural/synthetic compounds

Screening of potential drugs using compound libraries has resulted in the identification of several novel inhibitors. In a high throughput screening of 3282 drugs targeting mTOR, Torin2 showed a remarkable reduction in cellular proliferation in vitro and inhibition of tumor growth and metastasis in vivo [45]. In another study, pretreatment with Carfilzomib (Proteasome inhibitor) resulted in the reduced metastatic spread and disease progression in mice [46]. Similarly, administration of YM155 (Survivin inhibitor) and CUDC-101 (Histone Deacetylase and EGFR inhibitor) showed a significantly suppressed tumor growth and reduced metastasis in vivo [47, 48]. CUDC-101 is currently underway for testing in phase II clinical trial in ATC patients (Table 2).
As these tumors do not respond to radio-iodine ablation therapy, several compounds have been identified by high-throughput screening that can induce re-differentiation programme. An example of such compounds, Resveratrol can induce functional Notch1 protein expression and activate transcription of thyroid-specific genes including TTF1, TTF2, Pax8, and NIS [49]. In addition, it can also reduce stem cell markers confirming its potential to induce differentiation [50]. Other examples of such re-differentiation-inducing compounds are 1, 25 dihydroxy vitamin D3 (Calcitriol), Hesperetin and VPA [5052]. Similarly, Chrysin can upregulate the expression of NIS by activating Notch and its downstream effector, HES1. Also, treatment with Chrysin resulted in diminished cellular growth in vitro and tumor growth in vivo [53]. Collectively, these compounds may complement radioiodine therapy and warrant more comprehensive assessment to explore their clinical efficacy.

Gene therapy using oncolytic viruses

This therapy is a promising approach to restore the expression of tumor suppressor genes and to target oncogenes. Restoration of NIS and p53 expression using an adenovirus-5 vector showed a significant increase in uptake of radioactive iodine (I131) and improved cytotoxicity in vitro and in vivo [54]. The combination of oncolytic viruses with small molecule inhibitors yielded promising preclinical results. For instance, a combination of the ATM (a Ser/Thr kinase involved in DNA replication) inhibitor, KU55933 with oncolytic adenovirus, dl922–947 improved the efficacy of ionizing radiation treatment in ATC cells [55]. Recently, a novel approach using an oncolytic virus (dl922–947) showed a remarkable increase in the efficacy of PARP inhibitor, Olaparib [56]. Additionally, the oncolytic adenovirus, dl922–947 was shown to modulate tumor microenvironment by decreasing IL-8/CXCL8 and MCP-1/CCL2 expression which resulted in compromised angiogenesis and macrophage infiltration [57]. Although preclinical studies demonstrated encouraging results, the clinical implications of oncolytic viruses is still evolving [58].

Novel targeted inhibitors

Specific molecules that can inhibit the key signaling cascades in ATC are of significant interest. In this regard, some of the targeted tyrosine kinases, which are targeted in ATC include EGFR, PDGFR, VEGFR, cMET (Hepatocyte Growth Factor Receptor) and RET [59, 60]. All these molecules have been targeted and resulted in a semi-favorable response in ATC patients. A list of target-specific inhibitors is given in Table 3. Several PI3k/Akt/mTOR pathway inhibitors such as Everolimus, Temsirolimus, GSK69093, MK-2206, PX866, and ZSTK474 have been tested in preclinical and clinical studies. Everolimus showed safety in phase I clinical trial and is currently in phase II clinical trial (NCT02143726) in combination with Sorafenib [61] for the treatment of ATC.
Table 3
Different categories of drugs used in preclinical and clinical studies in ATC
Chemotherapeutic agents
 Topoisomerase inhibitor
Doxorubicin, Etoposide
 Microtubule assembly
Paclitaxel, Vinorelbine, Docetaxel
 DNA crosslinking agents
Cisplatin, Carboplatin, Cyclophosphamide, Neoplatin
 Nucleoside Analog
Gemcitabine, 5- fluorouracil
Targeted inhibitors/antibodies
 ALK1
GSK461364A
 Akt
MK-2206 2HCL, Perifosine, GSK690693, GDC-0068, AT7867
 Aurora Kinases
MK-0457 (VX-680), SNS-314 mesylate, ZM447439, AZD1152 and MLN8054
 Bcl2
Obatoclax
 CDK
BP14
 EGFR
Cetuximab (C225), Manumycin A, Geldanamycin, Gefitinib (ZD1839)
 HSP90
Tanespimycin (17-N-allylamino-17-demethoxygeldanamycin, NVP-A0Y922, SNX5422
 I-κB
Ciglitazone (upregulates TrailR1, −R2)
 PARP
Olaparib
 PD-1 receptor
Pembrolizumab, PDR001
 PDL-1
Durvalumab, Atezolizumab, FAZ053
 CTLA4
Tramelimumab
 TGF-β
LY2157299, SB 525334, LY2109761, Perfenidone, GW788388
 SMO (Wnt signaling pathway)
LDE225, LY2940680, PF-5274857, SANT-1
 γ-secretase
RO4929097, LY-411575
Anti-angiogenic agents
 Vascular disrupting agent
Combretastatin A4 phosphate (CA4P), Fosbretabulin
 VEGF
Bevacizumab, AZD2171, Cediranib
Multi-Kinase inhibitors
 VEGF 1, 2 and 3, PDGFR and c-KIT
Axitinib (AG-013736), Pazopanib
 VEGFR1, 2 and 3, EGFR and RET kinases
Vandetanib
 VEGFR-1, PDGFR, RET, FLT-3 and CSF-1R
Sunitinib
 VEGFR2, EGFR and RET
CLM94
 BCR-ABL, PDGFR and c-kit
Imatinib
 VEGFR 1, 2, PDGFRβ, RET, BRAF and c-Kit
Sorafenib (Bay43–9006, Nexavar)
 VEGFR-1, −2 and − 3, PDGFRβ, RET, FGFR −1, − 2, −3, −4 and c-KIT
Lenvatinib (E7080)
 VEGFR 2, RET, MET, kit
Cabozantinib
 VEGFR −1, − 2, −3, RET, kit, PDGFR
Motesanib
 VEGFR − 1, −3, PDGFR, FGFR1–3
Ninetedanib
 RET, PDGFR, FGFR, FLT3, kit
Ponatinib
 MET, ALK, ROS1
Crizotinib
Epigenetic modifiers
 HDAC inhibitors
Valproic acid, Thailandepsin A (TDP-A), Trichostatin A (TSA), Suberoyl Amide Hydroxamic Acid (SAHA), N-hydroxy-7-(2-naphthylthio)heptanomide (HNHA)
 BET inhibitors
JQ1, I-BET762
Miscellaneous
 HDACs, EGFR (dual inhibitor)
CUDC-101
 Proteosome inhibitors
Carfilzomib, Bortezomib (PS-341)
 PPARγ agonists
Rosiglitazone, RS5444, Pioglitazone, Troglitazone
BRAFV600E inhibitor, Vemurafenib, also showed limited efficacy [62], due to activation of downstream PI3k/Akt and MAPK pathways by alternate mechanisms. For example, cMET can directly activate PDK-1 and Ras thereby, bypassing BRAF mediated signaling and avoiding the current PI3k/Akt/mTOR pathway targets. In an effort to overcome this impediment, downstream pathway inhibitors were explored. An example of such inhibitors is OSU-53 which targets mTOR and activates AMPK. This molecule effectively inhibited cellular growth in ATC cells lines with activating mutations in Ras or BRAF [63]. Among the newly discovered MKIs, a class of “pyrazolo[3,4-d]pyrimidine” compounds (CLM29 and CLM24) that inhibit several targets such as EGFR and VEGFR, showed anti-proliferative and anti-metastatic effects in ATC-derived primary cells and established cell lines [64].
NF-κB signaling is crucial in ATC progression and can be targeted by employing proteasome inhibitors. Carfilzomib, a potent proteasome inhibitor, induced apoptosis in ATC cells by upregulating p27 and downregulating the anti-apoptotic molecule ATF4 [46]. Likewise, Bortezomib in combination with MLN8054 (Aurora kinase inhibitor) showed reduced cellular growth and induced apoptosis in ATC cells [42] Another promising HIV protease inhibitor, Nelfinavir, which blocks both MAPK and PI3k/Akt signaling pathways, exhibited induction of DNA damage and inhibition of cell proliferation in ATC cells in vitro [65].
Heat Shock Proteins (HSPs) represent another class of promising molecular targets for therapeutic purposes. These proteins are expressed in stressful conditions in normal cells but are aberrantly expressed in cancer cells. Combined inhibition of HSP90 (by Radicicol) and HSP70 resulted in significant induction of apoptosis in ATC cells [66]. Two newly discovered HSP90 inhibitors, KU711 and WGA-TA, showed a remarkable reduction in stemness (i.e. aldehyde dehydrogenase (ALDH)+ and CD44+), migration and invasion of ATC cells in vitro, and downregulation of β-Catenin, BRAF, Akt, and phosphoAkt [67]. These promising findings provide the framework for another therapeutic option to attack this aggressive and debilitating disease.

Epigenetic silencing

In an intricate process of carcinogenesis, epigenetic reprogramming can lead to activation of tumor-promoting genes and de-activation of tumor-suppressing genes. Epigenetic silencing represents a promising approach to induce cytotoxicity in ATC. Broadly, two categories of epigenetic modulators have been tested: Histone Deacetylases (HDACs) inhibitors and Bromodomain and Extra-Terminal (BET) inhibitors. Trichostatin-A (TSA) and Suberanilohydroxamic (SAHA or Vorinostat) are two well-characterized HDAC inhibitors, which restored the expression of the thyroid-specific genes including NIS, TSHR, TPO, TG, and TTF-1 in ATC cells and increased their radioiodine uptake [68]. Treatment with these HDAC inhibitors also showed diminished CD33 expression and increased expression of NIS, Tg, and TTF1 in ARO cells. However, these HDACs inhibitors resulted in increased expression of stem cells markers Oct4, Nanog, Sox2, Klf4, and c-Myc, suggesting significant off-target effects [69]. Several other HDAC inhibitors exhibited promising results without such side-effects. Thailandepsin A (TDP-A) showed promising antiproliferative effects concomitant with cell cycle arrest and apoptosis activation in ATC cells [70]. N-hydroxy-7-(2-naphthylthio) heptoxide (HNHA) is a recently discovered HDAC inhibitor that showed promising results in PTC and ATC cell lines by inducing caspase-dependent and ER stress-mediated apoptosis [71]. Similarly, Pugliese et al. showed that treatment with LBH589 can induce re-differentiation in the ATC cell lines BHT-101 and Cal-62, marked by an increased NIS expression and radioiodine uptake [72]. This class of inhibitors warrants further investigation to explore the therapeutic implications in preclinical and clinical settings.
A recently emerging category of epigenetic modulators, BET inhibitors exert their biological function by targeting the bromodomain and extra-terminal of BET proteins, which interact with HDACs and regulate gene expression. Two recently discovered BET inhibitors, JQ1 and I-BET762 blocked cell cycle arrest in ATC cells by targeting MCM5. MCM5 is highly over-expressed in PTC and ATC tissue specimens indicating its potential as a molecular target [73]. In particular, JQ1 was evaluated in an ATC mouse model, ThrbPV/PVKrasG12D, and exhibited significant tumor reduction and improved survival which is mediated by reduced MYC expression and disrupted cyclin-CDK4/RB/E2F3 signaling, indicating its promising applications as an anti-cancer drug [74].

Metabolic pathway targeting

Cancer cells have a high proliferation rate and altered metabolomic landscape which can be employed in therapeutics. Very few studies have been done towards targeting metabolomic pathways in ATC. A glucose analog, 2-deoxyglucose (2DG) was shown to re-sensitize ATC cells to radiation and chemotherapy (Cisplatin), but the observed effect was transient [75]. Also, an analog of vitamin D3, 19-nor-2α-(3-hydroxypropyl)-1α,25-dihydroxy vitamin D3 (MART-10) was demonstrated to inhibit migration and invasion of ATC cells by blocking the EMT pathway [76]. Microarray analysis of ATC versus normal thyroid tissues revealed significant distortion of fatty acid metabolism, and Stearoyl-CoA desaturase 1 (SCD1) was identified as a differentially expressed enzyme in ATC. SCD1 targeting induced endoplasmic reticulum stress and consequently apoptosis in ATC cells, both in vitro and in vivo [77]. Hence, this approach might help in improving the existing therapeutic interventions utilized today.

Apoptosis enhancing strategies

Cancer cells bypass apoptotic signals and often display insensitivity to apoptosis-inducing agents. In thyroid cancer, an important apoptosis-inducing molecule, TNF-related apoptosis-inducing ligand (TRAIL), has been shown to potently and selectively kill cancer cells. TRAIL has emerged as an attractive molecular target owing to its cancer cell specificity and lack of toxicity to normal cells. TRAIL resistance factors include activation of c-FLICE-like inhibitory protein (c-Flip) and reduced expression of Trail-R1 and Trail-R2 receptors on the tumor cell surface. Interestingly, gene silencing of c-Flip and MADD, a key player in TRAIL-induced apoptosis, can significantly improve the TRAIL sensitivity [78]. Further, MADD knock-down and/or MADD dephosphorylation can also render differentiated thyroid cancer cells susceptible to TRAIL [79]. Thus, targeting these TRAIL resistance factors can be used to improve TRAIL sensitivity. In an independent study conducted by Gunda et al., the TRAIL-R2 receptor agonistic antibody, Lexatumumab, was shown to induce apoptosis in HTH7 (ATC), BCPAP and TCP-1 cells. Interestingly, in Lexatumumab resistant cells harboring a BRAFV600E mutation, a combination of BRAF inhibitor (PLX4720) or PI3k inhibitor (LY294002) can be employed to overcome apoptotic resistance [80]. Several HDAC inhibitors were shown to synergize with TRAIL activity and can be employed to overcome its resistance. For instance, combining TRAIL with the HDAC inhibitor Vorinostat (SAHA) resulted in increased DR5 expression and cell death [81]. Combination of TRAIL and the HDAC inhibitor VPA also induced significant apoptosis in TRAIL-resistant 8505C (ATC) cells by activating Jnk and phosphorylating FADD and c-jun, but not p38 [82]. HDAC inhibitors such as SAHA and MS-275 promoted apoptosis by preventing TRAIL degradation in thyroid cancer cells [83]. Another apoptosis-inducing agent, Obatoclax (BCL inhibitor) induced significant cell death through necrosis and lysosome neutralization in ATC cells [84].

Immunotherapy

The presence of TAMs, NK cells and other TILs within ATC tissues highlight the relevance of tumor-immune cell interaction [85]. TAMs (type M2) promote tumor growth in ATC by expressing high levels of immunosuppressive cytokines such as IL-10 and TGF-β1 [86]. Other immunosuppressive mechanisms include binding of Programmed Death Ligand-1 (PD-L1) with its cognate receptor PD1 expressed on T cells, which down-modulates effector T cell function. In ATC, BRAFV600E mutation is strongly associated with the expression of PD-L1 (P = 0.015) [87]. In a recent retrospective study, high PD-1 expression (> 40% staining) in inflammatory cells was associated with worse overall survival (OS; hazard ratio, 3.36; 95% confidence interval, 1.00 to 12.96; P < 0.05) and trended towards worse PFS, whereas high PD-L1 expression in tumor cells (> 33% staining) trended towards worse PFS and OS indicating the crucial role of PD-1/PD-L-1 pathway in ATC [88]. In a preclinical BRAFV600E/WT;p53−/− mouse model, treatment with a BRAF inhibitor (PLX4720) and an anti-PD-L1 antibody resulted in a significant tumor regression and strong anti-tumor immune response [89]. The potential use of immunotherapy was exemplified by an exceptional response observed upon treatment with Vemurafenib (BRAFV600E inhibitor) and Nivolumab (human IgG4 anti-monoclonal PD-1 antibody) for tumor harboring BRAF mutation and PDL-1 positivity [90]. Several clinical trials using inhibitors/antibodies targeting PD-1 and PD-L1 are underway for clinical trials in ATC patients (Table 2) and hold promise.
Like PD-1/PD-L1, another crucial tumor-immune cell interaction that can be a potential immunotarget for ATC is CD70-CD27 as it is found in 49% of ATC specimens [91]. A clinical study showed that CD70 expression was associated with BRAFV600E mutation in ATC lesions and remained stable throughout the disease progression. However, no correlation was observed between CD70 and PD-L1 in ATC [91]. In addition, NK cell-based adoptive cellular therapy showed promising results in a preclinical mouse model of ATC pulmonary metastasis [92]. The major hurdle with immunotherapy is the low number of TILs, thus agents that can improve TIL trafficking needs to be explored.

Combination treatment

Owing to inherent and acquired chemoresistance, a combination of different drugs is often used in preclinical and clinical trials to improve therapeutic efficacy. However, it is important to establish a synergistic relationship between two drugs for their implications as a combination therapy. Several investigations were conducted to determine the relationship between drugs and some of them revealed crucial information about their behavior in combination. In this context, Allegri et al. have shown a synergistic effect between a CDK inhibitor (BP-14) and a mTOR inhibitor (Everolimus) by demonstrating loss of cell viability and down-regulation of EMT-related genes [93]. Combination of the NF-κB inhibitor (Quinacrine) and Sorafenib showed improved survival in an orthotopic mouse model in comparison to vehicle-treated and Doxorubicin-treated mice [94]. Combining MEK inhibitor (Trametinib) and multi-kinase inhibitor (Pazopanib) showed a significant reduction in the growth of xenografted tumors containing KRASG12R and BRAFV600E mutations [95]. Treatment with a combination of Carboplatin (CBDCA) and Radachlorin-photodynamic therapy (PDT) resulted in a significant tumor reduction due to activation of intrinsic apoptosis [96]. Combination of BRAF inhibitor (PLX4720) and Src tyrosine receptor/Bcr-Abl family inhibitor (Dasatinib) showed reduced tumor size, increased immune cell infiltration and induced apoptosis in an orthotopic ATC mouse model [97]. Combining PPARγ ligand (Troglitazone) and cholesterol-lowering drug (Lovastatin) demonstrated a significant suppression of EGF-induced migration in ATC cells, marked by the reduction of Vimentin and N-cadherin [98]. Synergistic cytotoxicity with Doxorubicin and Cucurbitacin B was observed in ATC cells in vitro and this effect was modulated by JNK2/STAT3 and ERK1/2 [99]. However, this effect is yet to be demonstrated in vivo. A combination of HDAC inhibitor (SAHA) and the PARP inhibitor (PJ34) exhibited a synergistic effect against SW1736 cell growth in vitro. This combination treatment also caused induction of TSHR, but not of NIS, TTF1, TTF2, and PAX8 mRNA levels [100]. Similar synergistic effects were observed with HDAC inhibitor (PXD101) and HSP90 inhibitor (NVP-AUY922), concomitant with the inactivation of PI3k/Akt signaling and activation of DNA damage response in ATC cells [101]. An HSP90 inhibitor, SNX5422 revealed synergy with many HDAC inhibitors including PXD101, SAHA, and TSA [102]. Combination of CUDC-101 with a second-generation proteasome inhibitor, Carfilzomib, yielded synergistic effect by affecting cell cycle at G2/M phase and activating apoptosis depicted by PARP cleavage and Caspase-3 activation [103].
In contrast, some of the drug combinations exhibited an antagonistic or non-synergistic relationship. For instance, combining NF-κB inhibitors with taxane cytotoxic drugs and/or radiation therapy did not show any synergistic effect in ATC cells [104]. Similarly, a Pan MEK inhibitor (U0126) and BRAF inhibitor (PLX4720) did not show any inhibition of invasive potential of ATC cells suggesting that migration and invasion in ATC cells are mediated by other non-MEK mechanisms [104]. Hence, careful selection of a combination of drugs based on genomic and proteomic profiling of tumors is crucial for strategically improving the therapeutic efficacy.

Conclusion

ATC remains a clinical challenge because of its de-differentiated phenotype and highly aggressive features. Several pre-clinical therapeutic studies including combinations of MKIs and HDAC inhibitors have shown encouraging results and hold promise for further investigation in clinical trials. Several clinical studies are ongoing to determine the safety and efficiency of novel drugs, but low patient accrual and limited long-term survival limit their translational ability. Combining several MKIs or using different salvage therapies might improve therapeutic outcomes. Evaluating drug treatment responses in primary cell cultures of patient tumors might help in guiding second-line treatment to develop precision medicine. An excellent case of such personalized therapy was demonstrated by Eckhard et al., wherein a patient’s tumor cells were cultured with Sorafenib, Vandetanib and MLN8054 (Aurora kinase inhibitor) in vitro while the patient was undergoing radiation and chemotherapy (Docetaxel and Cisplatin). Based on the in vitro data, the patient was subsequently treated with Sorafenib and achieved 43-month disease-free survival [105]. Analyzing patient biopsies during treatment might guide in understanding the mechanism of ATC progression, drug sensitivity, and chemoresistance leading to the selection of appropriate second-line of treatment. This can be exemplified by an unusual ATC case report in which an extraordinary response was achieved with Everolimus (FDA approved mTOR inhibitor) for the first 18 months with subsequent development of progressive disease. Comparison of the genomic sequences of sensitive tumors and drug-resistant tumors from the same patient revealed a nonsense mutation in TSC2 (a negative regulator of mTOR), which imparted drug sensitivity. The acquired resistance to Everolimus was due to a mutation in the mTOR gene which prevented binding of Everolimus to mTOR (allosteric inhibition). However, mutated mTOR could still be targeted by other direct inhibitors [106]. This substantiates the fact that comprehensive genomic analysis of serial biopsies during the treatment might help in deciding the follow-up treatment strategy in an effective manner (Fig. 2).
Additionally, gaining insights into the tumor-specific mutational landscape will not only help in developing a tailored treatment plan but also paves the way to design novel therapeutics. Although the presence of TAMs within ATC substantially hinders the capturing of ATC specific transcriptomes via Next-Generation Sequencing, significant information can be derived by using this method. Also, there is a need to develop better deep sequencing and bioinformatic algorithms that account for TAMs background noise. Undoubtedly, assessment of cellular, genomic, and molecular data is critical to developing better diagnostic and therapeutic approaches to this disease. Development of precision medicine will benefit from comprehensive analysis of pharmacological markers to predict the course of treatment. Including such efforts might help better manage this lethal malignancy.

Funding

This work was supported by the VA Merit Review (Project # 1I01BX002285-01A1).

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
Not applicable.
All authors consented for publication in its present form.

Competing interests

The authors declare that they have no competing interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
3.
Zurück zum Zitat Burman KD. Is poorly differentiated thyroid Cancer poorly characterized? J Clin Endocrinol Metab. 2014;99(4):1167–9.PubMedCrossRef Burman KD. Is poorly differentiated thyroid Cancer poorly characterized? J Clin Endocrinol Metab. 2014;99(4):1167–9.PubMedCrossRef
5.
Zurück zum Zitat Sun C, Li Q, Hu Z, He J, Li C, Li G, et al. Treatment and prognosis of anaplastic thyroid carcinoma: experience from a single institution in China. PLoS One. 2013;8(11):e80011.PubMedPubMedCentralCrossRef Sun C, Li Q, Hu Z, He J, Li C, Li G, et al. Treatment and prognosis of anaplastic thyroid carcinoma: experience from a single institution in China. PLoS One. 2013;8(11):e80011.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Haddad RI, Lydiatt WM, Ball DW, Busaidy NL, Byrd D, Callender G, et al. Anaplastic thyroid carcinoma, version 2.2015. J Natl Compr Cancer Netw. 2015;13(9):1140–50.CrossRef Haddad RI, Lydiatt WM, Ball DW, Busaidy NL, Byrd D, Callender G, et al. Anaplastic thyroid carcinoma, version 2.2015. J Natl Compr Cancer Netw. 2015;13(9):1140–50.CrossRef
7.
9.
Zurück zum Zitat Nikiforova MN, Wald AI, Roy S, Durso MB, Nikiforov YE. Targeted next-generation sequencing panel (ThyroSeq) for detection of mutations in thyroid Cancer. J Clin Endocrinol Metabol. 2013;98(11):E1852–E60.CrossRef Nikiforova MN, Wald AI, Roy S, Durso MB, Nikiforov YE. Targeted next-generation sequencing panel (ThyroSeq) for detection of mutations in thyroid Cancer. J Clin Endocrinol Metabol. 2013;98(11):E1852–E60.CrossRef
10.
Zurück zum Zitat Cantara S, Marzocchi C, Pilli T, Cardinale S, Forleo R, Castagna M, et al. Molecular signature of indeterminate thyroid lesions: current methods to improve fine needle aspiration cytology (FNAC) diagnosis. Int J Mol Sci. 2017;18(4):775.PubMedCentralCrossRef Cantara S, Marzocchi C, Pilli T, Cardinale S, Forleo R, Castagna M, et al. Molecular signature of indeterminate thyroid lesions: current methods to improve fine needle aspiration cytology (FNAC) diagnosis. Int J Mol Sci. 2017;18(4):775.PubMedCentralCrossRef
12.
Zurück zum Zitat Smallridge RC, Ain KB, Asa SL, Bible KC, Brierley JD, Burman KD, et al. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid. 2012;22(11):1104–39.PubMedCrossRef Smallridge RC, Ain KB, Asa SL, Bible KC, Brierley JD, Burman KD, et al. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid. 2012;22(11):1104–39.PubMedCrossRef
13.
Zurück zum Zitat Sherman SI. Thyroid carcinoma. Lancet (London, England). 2003;361(9356):501–11.CrossRef Sherman SI. Thyroid carcinoma. Lancet (London, England). 2003;361(9356):501–11.CrossRef
14.
Zurück zum Zitat Nel CJC, van Heerden JA, Goellner JR, Gharib H, McConahey WM, Taylor WF, et al. Anaplastic carcinoma of the thyroid: a Clinicopathologic study of 82 cases. Mayo Clin Proc. 1985;60(1):51–8.PubMedCrossRef Nel CJC, van Heerden JA, Goellner JR, Gharib H, McConahey WM, Taylor WF, et al. Anaplastic carcinoma of the thyroid: a Clinicopathologic study of 82 cases. Mayo Clin Proc. 1985;60(1):51–8.PubMedCrossRef
15.
Zurück zum Zitat Venkatesh YS, Ordonez NG, Schultz PN, Hickey RC, Goepfert H, Samaan NA. Anaplastic carcinoma of the thyroid. A clinicopathologic study of 121 cases. Cancer. 1990;66(2):321–30.PubMedCrossRef Venkatesh YS, Ordonez NG, Schultz PN, Hickey RC, Goepfert H, Samaan NA. Anaplastic carcinoma of the thyroid. A clinicopathologic study of 121 cases. Cancer. 1990;66(2):321–30.PubMedCrossRef
16.
Zurück zum Zitat McIver B, Hay ID, Giuffrida DF, Dvorak CE, Grant CS, Thompson GB, et al. Anaplastic thyroid carcinoma: a 50-year experience at a single institution. Surgery. 2001;130(6):1028–34.PubMedCrossRef McIver B, Hay ID, Giuffrida DF, Dvorak CE, Grant CS, Thompson GB, et al. Anaplastic thyroid carcinoma: a 50-year experience at a single institution. Surgery. 2001;130(6):1028–34.PubMedCrossRef
17.
Zurück zum Zitat Tennvall J, Lundell G, Hallquist A, Wahlberg P, Wallin G, Tibblin S. Combined doxorubicin, hyperfractionated radiotherapy, and surgery in anaplastic thyroid carcinoma. Report on two protocols. The Swedish anaplastic thyroid Cancer group. Cancer. 1994;74(4):1348–54.PubMedCrossRef Tennvall J, Lundell G, Hallquist A, Wahlberg P, Wallin G, Tibblin S. Combined doxorubicin, hyperfractionated radiotherapy, and surgery in anaplastic thyroid carcinoma. Report on two protocols. The Swedish anaplastic thyroid Cancer group. Cancer. 1994;74(4):1348–54.PubMedCrossRef
18.
Zurück zum Zitat Kwon J, Kim BH, Jung H-W, Besic N, Sugitani I, Wu H-G. The prognostic impacts of postoperative radiotherapy in the patients with resected anaplastic thyroid carcinoma: a systematic review and meta-analysis. Eur J Cancer. 2016;59:34–45.PubMedCrossRef Kwon J, Kim BH, Jung H-W, Besic N, Sugitani I, Wu H-G. The prognostic impacts of postoperative radiotherapy in the patients with resected anaplastic thyroid carcinoma: a systematic review and meta-analysis. Eur J Cancer. 2016;59:34–45.PubMedCrossRef
19.
Zurück zum Zitat Pezzi TA, Mohamed ASR, Sheu T, Blanchard P, Sandulache VC, Lai SY, et al. Radiation therapy dose is associated with improved survival for unresected anaplastic thyroid carcinoma: outcomes from the National Cancer Data Base. Cancer. 2017;123(9):1653–61.PubMedCrossRef Pezzi TA, Mohamed ASR, Sheu T, Blanchard P, Sandulache VC, Lai SY, et al. Radiation therapy dose is associated with improved survival for unresected anaplastic thyroid carcinoma: outcomes from the National Cancer Data Base. Cancer. 2017;123(9):1653–61.PubMedCrossRef
20.
Zurück zum Zitat Zhou M, Chen Y, Adachi M, Wen X, Erwin B, Mawlawi O, et al. Single agent nanoparticle for radiotherapy and radio-photothermal therapy in anaplastic thyroid cancer. Biomaterials. 2015;57:41–9.PubMedPubMedCentralCrossRef Zhou M, Chen Y, Adachi M, Wen X, Erwin B, Mawlawi O, et al. Single agent nanoparticle for radiotherapy and radio-photothermal therapy in anaplastic thyroid cancer. Biomaterials. 2015;57:41–9.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat SK A, Patrick D, Christina S, Kerstin K, Kathrin K, Nathalie S, et al. Reintroducing the sodium–iodide symporter to anaplastic thyroid carcinoma. Thyroid. 2017;27(12):1534–43.CrossRef SK A, Patrick D, Christina S, Kerstin K, Kathrin K, Nathalie S, et al. Reintroducing the sodium–iodide symporter to anaplastic thyroid carcinoma. Thyroid. 2017;27(12):1534–43.CrossRef
22.
Zurück zum Zitat Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, et al. Dabrafenib and Trametinib treatment in patients with locally advanced or metastatic BRAF V600–mutant anaplastic thyroid Cancer. J Clin Oncol. 2018;36(1):7–13.PubMedCrossRef Subbiah V, Kreitman RJ, Wainberg ZA, Cho JY, Schellens JHM, Soria JC, et al. Dabrafenib and Trametinib treatment in patients with locally advanced or metastatic BRAF V600–mutant anaplastic thyroid Cancer. J Clin Oncol. 2018;36(1):7–13.PubMedCrossRef
23.
Zurück zum Zitat Ryder M, Gild M, Hohl TM, Pamer E, Knauf J, Ghossein R, et al. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid Cancer progression. PLoS One. 2013;8(1):e54302.PubMedPubMedCentralCrossRef Ryder M, Gild M, Hohl TM, Pamer E, Knauf J, Ghossein R, et al. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid Cancer progression. PLoS One. 2013;8(1):e54302.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Ryder M, Gild M, Hohl TA, Pamer E, Ghossein R, Knauf JA, Bollag G, Joyce J, Fagin, JA. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid cancer progression. PLOS 1. 2013;8(1):e54302.PubMedPubMedCentralCrossRef Ryder M, Gild M, Hohl TA, Pamer E, Ghossein R, Knauf JA, Bollag G, Joyce J, Fagin, JA. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid cancer progression. PLOS 1. 2013;8(1):e54302.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Fujita T, Liu J, Myoujin M, Shimada S, Ishikawa Y, Fujimori M. JAK inhibitors to suppress paclitaxel-resistant anaplastic thyroid cancer via IL-6 reduction. J Clin Oncol. 2018;36(15_suppl):e18093-e.CrossRef Fujita T, Liu J, Myoujin M, Shimada S, Ishikawa Y, Fujimori M. JAK inhibitors to suppress paclitaxel-resistant anaplastic thyroid cancer via IL-6 reduction. J Clin Oncol. 2018;36(15_suppl):e18093-e.CrossRef
27.
Zurück zum Zitat Dadu R, Devine C, Hernandez M, Waguespack SG, Busaidy NL, Hu MI, et al. Role of salvage targeted therapy in differentiated thyroid cancer patients who failed first-line sorafenib. J Clin Endocrinol Metab. 2014;99(6):2086–94.PubMedPubMedCentralCrossRef Dadu R, Devine C, Hernandez M, Waguespack SG, Busaidy NL, Hu MI, et al. Role of salvage targeted therapy in differentiated thyroid cancer patients who failed first-line sorafenib. J Clin Endocrinol Metab. 2014;99(6):2086–94.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55.PubMedCrossRef Cabanillas ME, Habra MA. Lenvatinib: role in thyroid cancer and other solid tumors. Cancer Treat Rev. 2016;42:47–55.PubMedCrossRef
30.
Zurück zum Zitat Koyama S, Miyake N, Fujiwara K, Morisaki T, Fukuhara T, Kitano H, et al. Lenvatinib for anaplastic thyroid Cancer and Lenvatinib-induced thyroid dysfunction. Eur Thyroid J. 2018;7(3):139–44.PubMedCrossRefPubMedCentral Koyama S, Miyake N, Fujiwara K, Morisaki T, Fukuhara T, Kitano H, et al. Lenvatinib for anaplastic thyroid Cancer and Lenvatinib-induced thyroid dysfunction. Eur Thyroid J. 2018;7(3):139–44.PubMedCrossRefPubMedCentral
31.
Zurück zum Zitat Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid Cancer. N Engl J Med. 2013;368(7):623–32.PubMedPubMedCentralCrossRef Ho AL, Grewal RK, Leboeuf R, Sherman EJ, Pfister DG, Deandreis D, et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid Cancer. N Engl J Med. 2013;368(7):623–32.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Viola D, Valerio L, Molinaro E, Agate L, Bottici V, Biagini A, et al. Treatment of advanced thyroid cancer with targeted therapies: ten years of experience. Endocr Relat Cancer. 2016;23(4):R185–205.PubMedCrossRef Viola D, Valerio L, Molinaro E, Agate L, Bottici V, Biagini A, et al. Treatment of advanced thyroid cancer with targeted therapies: ten years of experience. Endocr Relat Cancer. 2016;23(4):R185–205.PubMedCrossRef
33.
Zurück zum Zitat Catalano MG, Pugliese M, Gallo M, Brignardello E, Milla P, Orlandi F, et al. Valproic acid, a histone deacetylase inhibitor, in combination with paclitaxel for anaplastic thyroid Cancer: results of a multicenter randomized controlled phase II/III trial. Int J Endocrinol. 2016;2016:2930414.PubMedPubMedCentralCrossRef Catalano MG, Pugliese M, Gallo M, Brignardello E, Milla P, Orlandi F, et al. Valproic acid, a histone deacetylase inhibitor, in combination with paclitaxel for anaplastic thyroid Cancer: results of a multicenter randomized controlled phase II/III trial. Int J Endocrinol. 2016;2016:2930414.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Antonelli A, Fallahi P, Ulisse S, Ferrari SM, Minuto M, Saraceno G, et al. New targeted therapies for anaplastic thyroid cancer. Anti Cancer Agents Med Chem. 2012;12(1):87–93.CrossRef Antonelli A, Fallahi P, Ulisse S, Ferrari SM, Minuto M, Saraceno G, et al. New targeted therapies for anaplastic thyroid cancer. Anti Cancer Agents Med Chem. 2012;12(1):87–93.CrossRef
37.
Zurück zum Zitat Antonelli A, Fallahi P, Ulisse S, Ferrari SM, Mazzi V, Domenicantonio AD, et al. Tyrosine kinase inhibitors for the therapy of anaplastic thyroid cancer. Int J Endocr Oncol. 2015;2(2):135–42.CrossRef Antonelli A, Fallahi P, Ulisse S, Ferrari SM, Mazzi V, Domenicantonio AD, et al. Tyrosine kinase inhibitors for the therapy of anaplastic thyroid cancer. Int J Endocr Oncol. 2015;2(2):135–42.CrossRef
38.
Zurück zum Zitat Smallridge RC, Copland JA, Brose MS, Wadsworth JT, Houvras Y, Menefee ME, et al. Efatutazone, an oral PPAR-gamma agonist, in combination with paclitaxel in anaplastic thyroid cancer: results of a multicenter phase 1 trial. J Clin Endocrinol Metab. 2013;98(6):2392–400.PubMedPubMedCentralCrossRef Smallridge RC, Copland JA, Brose MS, Wadsworth JT, Houvras Y, Menefee ME, et al. Efatutazone, an oral PPAR-gamma agonist, in combination with paclitaxel in anaplastic thyroid cancer: results of a multicenter phase 1 trial. J Clin Endocrinol Metab. 2013;98(6):2392–400.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Baldini E, D'Armiento M, Ulisse S. A New Aurora in Anaplastic Thyroid Cancer Therapy. Int J Endocrinol. 2014;2014:11.CrossRef Baldini E, D'Armiento M, Ulisse S. A New Aurora in Anaplastic Thyroid Cancer Therapy. Int J Endocrinol. 2014;2014:11.CrossRef
40.
Zurück zum Zitat Nikonova AS, Astsaturov I, Serebriiskii IG, Dunbrack RL Jr, Golemis EA. Aurora a kinase (AURKA) in normal and pathological cell division. Cell Mol Life Sci. 2013;70(4):661–87.PubMedCrossRef Nikonova AS, Astsaturov I, Serebriiskii IG, Dunbrack RL Jr, Golemis EA. Aurora a kinase (AURKA) in normal and pathological cell division. Cell Mol Life Sci. 2013;70(4):661–87.PubMedCrossRef
41.
Zurück zum Zitat Wunderlich A, Fischer M, Schloßhauer T, Ramaswamy A, Greene BH, Brendel C, et al. Evaluation of Aurora kinase inhibition as a new therapeutic strategy in anaplastic and poorly differentiated follicular thyroid cancer. Cancer Sci. 2011;102(4):762–8.PubMedCrossRef Wunderlich A, Fischer M, Schloßhauer T, Ramaswamy A, Greene BH, Brendel C, et al. Evaluation of Aurora kinase inhibition as a new therapeutic strategy in anaplastic and poorly differentiated follicular thyroid cancer. Cancer Sci. 2011;102(4):762–8.PubMedCrossRef
42.
Zurück zum Zitat Wunderlich A, Roth S, Ramaswamy A, Greene BH, Brendel C, Hinterseher U, et al. Combined inhibition of cellular pathways as a future therapeutic option in fatal anaplastic thyroid cancer. Endocrine. 2012;42(3):637–46.PubMedCrossRef Wunderlich A, Roth S, Ramaswamy A, Greene BH, Brendel C, Hinterseher U, et al. Combined inhibition of cellular pathways as a future therapeutic option in fatal anaplastic thyroid cancer. Endocrine. 2012;42(3):637–46.PubMedCrossRef
43.
Zurück zum Zitat Isham CR, Bossou AR, Negron V, Fisher KE, Kumar R, Marlow L, et al. Pazopanib Enhances Paclitaxel-Induced Mitotic Catastrophe in Anaplastic Thyroid Cancer. Sci Transl Med. 2013;5(166):166ra3.PubMedPubMedCentralCrossRef Isham CR, Bossou AR, Negron V, Fisher KE, Kumar R, Marlow L, et al. Pazopanib Enhances Paclitaxel-Induced Mitotic Catastrophe in Anaplastic Thyroid Cancer. Sci Transl Med. 2013;5(166):166ra3.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Russo MA, Kang KS, Di Cristofano A. The PLK1 inhibitor GSK461364A is effective in poorly differentiated and anaplastic thyroid carcinoma cells, independent of the nature of their driver mutations. Thyroid. 2013;23(10):1284–93.PubMedPubMedCentralCrossRef Russo MA, Kang KS, Di Cristofano A. The PLK1 inhibitor GSK461364A is effective in poorly differentiated and anaplastic thyroid carcinoma cells, independent of the nature of their driver mutations. Thyroid. 2013;23(10):1284–93.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Sadowski SM, Boufraqech M, Zhang L, Mehta A, Kapur P, Zhang Y, et al. Torin2 targets dysregulated pathways in anaplastic thyroid cancer and inhibits tumor growth and metastasis. Oncotarget. 2015;6(20):18038–49.PubMedPubMedCentralCrossRef Sadowski SM, Boufraqech M, Zhang L, Mehta A, Kapur P, Zhang Y, et al. Torin2 targets dysregulated pathways in anaplastic thyroid cancer and inhibits tumor growth and metastasis. Oncotarget. 2015;6(20):18038–49.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Mehta A, Zhang L, Boufraqech M, Zhang Y, Patel D, Shen M, et al. Carfilzomib is an effective anticancer agent in anaplastic thyroid cancer. Endocr Relat Cancer. 2015;22(3):319–29.PubMedCrossRef Mehta A, Zhang L, Boufraqech M, Zhang Y, Patel D, Shen M, et al. Carfilzomib is an effective anticancer agent in anaplastic thyroid cancer. Endocr Relat Cancer. 2015;22(3):319–29.PubMedCrossRef
47.
Zurück zum Zitat Mehta A, Zhang L, Boufraqech M, Liu-Chittenden Y, Zhang Y, Patel D, et al. Inhibition of Survivin with YM155 induces durable tumor response in anaplastic thyroid Cancer. Clin Cancer Res. 2015;21(18):4123–32.PubMedPubMedCentralCrossRef Mehta A, Zhang L, Boufraqech M, Liu-Chittenden Y, Zhang Y, Patel D, et al. Inhibition of Survivin with YM155 induces durable tumor response in anaplastic thyroid Cancer. Clin Cancer Res. 2015;21(18):4123–32.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Zhang L, Zhang Y, Mehta A, Boufraqech M, Davis S, Wang J, et al. Dual inhibition of HDAC and EGFR signaling with CUDC-101 induces potent suppression of tumor growth and metastasis in anaplastic thyroid cancer. Oncotarget. 2015;6(11):9073–85.PubMedPubMedCentral Zhang L, Zhang Y, Mehta A, Boufraqech M, Davis S, Wang J, et al. Dual inhibition of HDAC and EGFR signaling with CUDC-101 induces potent suppression of tumor growth and metastasis in anaplastic thyroid cancer. Oncotarget. 2015;6(11):9073–85.PubMedPubMedCentral
49.
Zurück zum Zitat Yu X-M, Jaskula-Sztul R, Ahmed K, Harrison AD, Kunnimalaiyaan M, Chen H. Resveratrol induces differentiation markers expression in anaplastic thyroid carcinoma via activation of Notch1 signaling and suppresses cell growth. Mol Cancer Ther. 2013;12(7):1276–87.PubMedPubMedCentralCrossRef Yu X-M, Jaskula-Sztul R, Ahmed K, Harrison AD, Kunnimalaiyaan M, Chen H. Resveratrol induces differentiation markers expression in anaplastic thyroid carcinoma via activation of Notch1 signaling and suppresses cell growth. Mol Cancer Ther. 2013;12(7):1276–87.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Hardin H, Yu XM, Harrison AD, Larrain C, Zhang R, Chen J, et al. Generation of novel thyroid Cancer stem-like cell clones: effects of resveratrol and Valproic acid. Am J Pathol. 2016;186(6):1662–73.PubMedPubMedCentralCrossRef Hardin H, Yu XM, Harrison AD, Larrain C, Zhang R, Chen J, et al. Generation of novel thyroid Cancer stem-like cell clones: effects of resveratrol and Valproic acid. Am J Pathol. 2016;186(6):1662–73.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Peng W, Wang K, Zheng R, Derwahl M. 1,25 dihydroxyvitamin D3 inhibits the proliferation of thyroid cancer stem-like cells via cell cycle arrest. Endocr Res. 2016;41(2):71–80.PubMedCrossRef Peng W, Wang K, Zheng R, Derwahl M. 1,25 dihydroxyvitamin D3 inhibits the proliferation of thyroid cancer stem-like cells via cell cycle arrest. Endocr Res. 2016;41(2):71–80.PubMedCrossRef
52.
Zurück zum Zitat Patel PN, Yu XM, Jaskula-Sztul R, Chen H. Hesperetin activates the Notch1 signaling cascade, causes apoptosis, and induces cellular differentiation in anaplastic thyroid cancer. Ann Surg Oncol. 2014;21(Suppl 4):S497–504.PubMedCrossRef Patel PN, Yu XM, Jaskula-Sztul R, Chen H. Hesperetin activates the Notch1 signaling cascade, causes apoptosis, and induces cellular differentiation in anaplastic thyroid cancer. Ann Surg Oncol. 2014;21(Suppl 4):S497–504.PubMedCrossRef
53.
Zurück zum Zitat Yu X-M, Phan T, Patel PN, Jaskula-Sztul R, Chen H. Chrysin activates Notch1 signaling and suppresses tumor growth of anaplastic thyroid carcinoma in vitro and in vivo. Cancer. 2013;119(4):774–81.PubMedCrossRef Yu X-M, Phan T, Patel PN, Jaskula-Sztul R, Chen H. Chrysin activates Notch1 signaling and suppresses tumor growth of anaplastic thyroid carcinoma in vitro and in vivo. Cancer. 2013;119(4):774–81.PubMedCrossRef
54.
Zurück zum Zitat Lee YJ, Chung JK, Kang JH, Jeong JM, Lee DS, Lee MC. Wild-type p53 enhances the cytotoxic effect of radionuclide gene therapy using sodium iodide symporter in a murine anaplastic thyroid cancer model. Eur J Nucl Med Mol Imaging. 2010;37(2):235–41.PubMedCrossRef Lee YJ, Chung JK, Kang JH, Jeong JM, Lee DS, Lee MC. Wild-type p53 enhances the cytotoxic effect of radionuclide gene therapy using sodium iodide symporter in a murine anaplastic thyroid cancer model. Eur J Nucl Med Mol Imaging. 2010;37(2):235–41.PubMedCrossRef
55.
Zurück zum Zitat Passaro C, Abagnale A, Libertini S, Volpe M, Botta G, Cella L, et al. Ionizing radiation enhances dl922-947-mediated cell death of anaplastic thyroid carcinoma cells. Endocr Relat Cancer. 2013;20(5):633–47.PubMedCrossRef Passaro C, Abagnale A, Libertini S, Volpe M, Botta G, Cella L, et al. Ionizing radiation enhances dl922-947-mediated cell death of anaplastic thyroid carcinoma cells. Endocr Relat Cancer. 2013;20(5):633–47.PubMedCrossRef
56.
Zurück zum Zitat Passaro C, Volpe M, Botta G, Scamardella E, Perruolo G, Gillespie D, et al. PARP inhibitor olaparib increases the oncolytic activity of dl922-947 in in vitro and in vivo model of anaplastic thyroid carcinoma. Mol Oncol. 2015;9(1):78–92.PubMedCrossRef Passaro C, Volpe M, Botta G, Scamardella E, Perruolo G, Gillespie D, et al. PARP inhibitor olaparib increases the oncolytic activity of dl922-947 in in vitro and in vivo model of anaplastic thyroid carcinoma. Mol Oncol. 2015;9(1):78–92.PubMedCrossRef
57.
Zurück zum Zitat Passaro C, Borriello F, Vastolo V, Di Somma S, Scamardella E, Gigantino V, et al. The oncolytic virus dl922-947 reduces IL-8/CXCL8 and MCP-1/CCL2 expression and impairs angiogenesis and macrophage infiltration in anaplastic thyroid carcinoma. Oncotarget. 2016;7(2):1500–15.PubMedCrossRef Passaro C, Borriello F, Vastolo V, Di Somma S, Scamardella E, Gigantino V, et al. The oncolytic virus dl922-947 reduces IL-8/CXCL8 and MCP-1/CCL2 expression and impairs angiogenesis and macrophage infiltration in anaplastic thyroid carcinoma. Oncotarget. 2016;7(2):1500–15.PubMedCrossRef
58.
Zurück zum Zitat Passaro C, Portella G. Oncolytic virotherapy for thyroid cancer: will it translate to the clinic? Int J Endocr Oncol. 2015;2(1):5–8.CrossRef Passaro C, Portella G. Oncolytic virotherapy for thyroid cancer: will it translate to the clinic? Int J Endocr Oncol. 2015;2(1):5–8.CrossRef
59.
Zurück zum Zitat Wiseman SM, Masoudi H, Niblock P, Turbin D, Rajput A, Hay J, et al. Anaplastic thyroid carcinoma: expression profile of targets for therapy offers new insights for disease treatment. Ann Surg Oncol. 2007;14(2):719–29.PubMedCrossRef Wiseman SM, Masoudi H, Niblock P, Turbin D, Rajput A, Hay J, et al. Anaplastic thyroid carcinoma: expression profile of targets for therapy offers new insights for disease treatment. Ann Surg Oncol. 2007;14(2):719–29.PubMedCrossRef
60.
Zurück zum Zitat Abate EG, Smallridge RC. Managing anaplastic thyroid carcinoma. Expert Rev Endocrinol Metab. 2011;6(6):793–809.CrossRefPubMed Abate EG, Smallridge RC. Managing anaplastic thyroid carcinoma. Expert Rev Endocrinol Metab. 2011;6(6):793–809.CrossRefPubMed
61.
Zurück zum Zitat Lim SM, Chang H, Yoon MJ, Hong YK, Kim H, Chung WY, et al. A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Annals Oncol. 2013;24(12):3089–94.CrossRef Lim SM, Chang H, Yoon MJ, Hong YK, Kim H, Chung WY, et al. A multicenter, phase II trial of everolimus in locally advanced or metastatic thyroid cancer of all histologic subtypes. Annals Oncol. 2013;24(12):3089–94.CrossRef
62.
Zurück zum Zitat Nehs MA, Nucera C, Nagarkatti SS, Sadow PM, Morales-Garcia D, Hodin RA, et al. Late intervention with anti-BRAF(V600E) therapy induces tumor regression in an orthotopic mouse model of human anaplastic thyroid cancer. Endocrinology. 2012;153(2):985–94.PubMedCrossRef Nehs MA, Nucera C, Nagarkatti SS, Sadow PM, Morales-Garcia D, Hodin RA, et al. Late intervention with anti-BRAF(V600E) therapy induces tumor regression in an orthotopic mouse model of human anaplastic thyroid cancer. Endocrinology. 2012;153(2):985–94.PubMedCrossRef
63.
Zurück zum Zitat Plews RL, Mohd Yusof A, Wang C, Saji M, Zhang X, Chen C-S, et al. A novel dual AMPK activator/mTOR inhibitor inhibits thyroid Cancer cell growth. J Clin Endocrinol Metab. 2015;100(5):E748–E56.PubMedPubMedCentralCrossRef Plews RL, Mohd Yusof A, Wang C, Saji M, Zhang X, Chen C-S, et al. A novel dual AMPK activator/mTOR inhibitor inhibits thyroid Cancer cell growth. J Clin Endocrinol Metab. 2015;100(5):E748–E56.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Jensen K, Bikas A, Patel A, Kushchayeva Y, Costello J, McDaniel D, et al. Nelfinavir inhibits proliferation and induces DNA damage in thyroid cancer cells. Endocr Relat Cancer. 2017;24(3):147–56.PubMedCrossRef Jensen K, Bikas A, Patel A, Kushchayeva Y, Costello J, McDaniel D, et al. Nelfinavir inhibits proliferation and induces DNA damage in thyroid cancer cells. Endocr Relat Cancer. 2017;24(3):147–56.PubMedCrossRef
66.
Zurück zum Zitat Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. Hsp70 inhibition potentiates radicicol-induced cell death in anaplastic thyroid carcinoma cells. Anticancer Res. 2014;34(9):4829–37.PubMed Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. Hsp70 inhibition potentiates radicicol-induced cell death in anaplastic thyroid carcinoma cells. Anticancer Res. 2014;34(9):4829–37.PubMed
67.
Zurück zum Zitat White PT, Subramanian C, Zhu Q, Zhang H, Zhao H, Gallagher R, et al. Novel HSP90 inhibitors effectively target functions of thyroid cancer stem cell preventing migration and invasion. Surgery. 2016;159(1):142–51.PubMedCrossRef White PT, Subramanian C, Zhu Q, Zhang H, Zhao H, Gallagher R, et al. Novel HSP90 inhibitors effectively target functions of thyroid cancer stem cell preventing migration and invasion. Surgery. 2016;159(1):142–51.PubMedCrossRef
68.
Zurück zum Zitat Hou P, Bojdani E, Xing M. Induction of thyroid gene expression and radioiodine uptake in thyroid cancer cells by targeting major signaling pathways. J Clin Endocrinol Metab. 2010;95(2):820–8.PubMedCrossRef Hou P, Bojdani E, Xing M. Induction of thyroid gene expression and radioiodine uptake in thyroid cancer cells by targeting major signaling pathways. J Clin Endocrinol Metab. 2010;95(2):820–8.PubMedCrossRef
69.
Zurück zum Zitat Ke C-C, Liu R-S, Chi C-W, Lee C-H. HDAC inhibitor induces re-expression of thyroid specific genes as well as differentiating in anaplastic thyroid cancer. J Nucl Med. 2013;54(supplement 2):1341. Ke C-C, Liu R-S, Chi C-W, Lee C-H. HDAC inhibitor induces re-expression of thyroid specific genes as well as differentiating in anaplastic thyroid cancer. J Nucl Med. 2013;54(supplement 2):1341.
70.
Zurück zum Zitat Weinlander E, Somnay Y, Harrison AD, Wang C, Cheng Y-Q, Jaskula-Sztul R, et al. The novel histone deacetylase inhibitor thailandepsin a inhibits anaplastic thyroid cancer growth. J Surg Res. 2014;190(1):191–7.PubMedPubMedCentralCrossRef Weinlander E, Somnay Y, Harrison AD, Wang C, Cheng Y-Q, Jaskula-Sztul R, et al. The novel histone deacetylase inhibitor thailandepsin a inhibits anaplastic thyroid cancer growth. J Surg Res. 2014;190(1):191–7.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Kim S-M, Park K-C, Jeon J-Y, Kim B-W, Kim H-K, Chang H-J, et al. Potential anti-cancer effect of N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA), a novel histone deacetylase inhibitor, for the treatment of thyroid cancer. BMC Cancer. 2015;15(1):1–11.CrossRef Kim S-M, Park K-C, Jeon J-Y, Kim B-W, Kim H-K, Chang H-J, et al. Potential anti-cancer effect of N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA), a novel histone deacetylase inhibitor, for the treatment of thyroid cancer. BMC Cancer. 2015;15(1):1–11.CrossRef
72.
Zurück zum Zitat Pugliese M, Fortunati N, Germano A, Asioli S, Marano F, Palestini N, et al. Histone deacetylase inhibition affects sodium iodide symporter expression and induces 131I cytotoxicity in anaplastic thyroid cancer cells. Thyroid. 2013;23(7):838–46.PubMedCrossRef Pugliese M, Fortunati N, Germano A, Asioli S, Marano F, Palestini N, et al. Histone deacetylase inhibition affects sodium iodide symporter expression and induces 131I cytotoxicity in anaplastic thyroid cancer cells. Thyroid. 2013;23(7):838–46.PubMedCrossRef
73.
Zurück zum Zitat Mio C, Lavarone E, Conzatti K, Baldan F, Toffoletto B, Puppin C, et al. MCM5 as a target of BET inhibitors in thyroid cancer cells. Endocr Relat Cancer. 2016;23(4):335–47.PubMedPubMedCentralCrossRef Mio C, Lavarone E, Conzatti K, Baldan F, Toffoletto B, Puppin C, et al. MCM5 as a target of BET inhibitors in thyroid cancer cells. Endocr Relat Cancer. 2016;23(4):335–47.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Zhu X, Enomoto K, Zhao L, Zhu YJ, Willingham MC, Meltzer PS, et al. Bromodomain and extraterminal protein inhibitor JQ1 suppresses thyroid tumor growth in a mouse model. Clin Cancer Res. 2017;23(2):430–40.PubMedCrossRef Zhu X, Enomoto K, Zhao L, Zhu YJ, Willingham MC, Meltzer PS, et al. Bromodomain and extraterminal protein inhibitor JQ1 suppresses thyroid tumor growth in a mouse model. Clin Cancer Res. 2017;23(2):430–40.PubMedCrossRef
75.
Zurück zum Zitat Sandulache VC, Skinner HD, Wang Y, Chen Y, Dodge CT, Ow TJ, et al. Glycolytic inhibition alters anaplastic thyroid carcinoma tumor metabolism and improves response to conventional chemotherapy and radiation. Mol Cancer Ther. 2012;11(6):1373–80.PubMedCrossRef Sandulache VC, Skinner HD, Wang Y, Chen Y, Dodge CT, Ow TJ, et al. Glycolytic inhibition alters anaplastic thyroid carcinoma tumor metabolism and improves response to conventional chemotherapy and radiation. Mol Cancer Ther. 2012;11(6):1373–80.PubMedCrossRef
76.
Zurück zum Zitat Chiang K-C, Kuo S-F, Chen C-H, Ng S, Lin S-F, Yeh C-N, et al. MART-10, the vitamin D analog, is a potent drug to inhibit anaplastic thyroid cancer cell metastatic potential. Cancer Lett. 2015;369(1):76–85.PubMedCrossRef Chiang K-C, Kuo S-F, Chen C-H, Ng S, Lin S-F, Yeh C-N, et al. MART-10, the vitamin D analog, is a potent drug to inhibit anaplastic thyroid cancer cell metastatic potential. Cancer Lett. 2015;369(1):76–85.PubMedCrossRef
77.
Zurück zum Zitat CAv R, Marlow LA, Pinkerton AB, Crist A, Miller J, Tun HW, et al. Aberrant lipid metabolism in anaplastic thyroid carcinoma reveals Stearoyl CoA desaturase 1 as a novel therapeutic target. J Clin Endocrinol Metab. 2015;100(5):E697–709.CrossRef CAv R, Marlow LA, Pinkerton AB, Crist A, Miller J, Tun HW, et al. Aberrant lipid metabolism in anaplastic thyroid carcinoma reveals Stearoyl CoA desaturase 1 as a novel therapeutic target. J Clin Endocrinol Metab. 2015;100(5):E697–709.CrossRef
78.
Zurück zum Zitat Li L-C, Jayaram S, Ganesh L, Qian L, Rotmensch J, Maker AV, et al. Knockdown of MADD and c-FLIP overcomes resistance to TRAIL-induced apoptosis in ovarian cancer cells. Am J Obstet Gynecol. 2011;205(4):362 e12-.e25.PubMedPubMedCentralCrossRef Li L-C, Jayaram S, Ganesh L, Qian L, Rotmensch J, Maker AV, et al. Knockdown of MADD and c-FLIP overcomes resistance to TRAIL-induced apoptosis in ovarian cancer cells. Am J Obstet Gynecol. 2011;205(4):362 e12-.e25.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Li L-C, Jayarama S, Pilli T, Qian L, Pacini F, Prabhakar BS. Down-modulation of expression, or Dephosphorylation, of IG20/MADD in tumor necrosis factor–related apoptosis-inducing ligand–resistant thyroid Cancer cells makes them susceptible to treatment with this ligand. Thyroid. 2012;23(1):70–8.CrossRef Li L-C, Jayarama S, Pilli T, Qian L, Pacini F, Prabhakar BS. Down-modulation of expression, or Dephosphorylation, of IG20/MADD in tumor necrosis factor–related apoptosis-inducing ligand–resistant thyroid Cancer cells makes them susceptible to treatment with this ligand. Thyroid. 2012;23(1):70–8.CrossRef
80.
Zurück zum Zitat Gunda V, Bucur O, Varnau J, Vanden Borre P, Bernasconi MJ, Khosravi-Far R, et al. Blocks to thyroid cancer cell apoptosis can be overcome by inhibition of the MAPK and PI3K/AKT pathways. Cell Death Dis. 2014;5(3):e1104.PubMedPubMedCentralCrossRef Gunda V, Bucur O, Varnau J, Vanden Borre P, Bernasconi MJ, Khosravi-Far R, et al. Blocks to thyroid cancer cell apoptosis can be overcome by inhibition of the MAPK and PI3K/AKT pathways. Cell Death Dis. 2014;5(3):e1104.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Cha HY, Lee BS, Kang S, Shin YS, Chang JW, Sung ES, et al. Valproic acid sensitizes TRAIL-resistant anaplastic thyroid carcinoma cells to apoptotic cell death. Ann Surg Oncol. 2013;20(Suppl 3):S716–24.PubMedCrossRef Cha HY, Lee BS, Kang S, Shin YS, Chang JW, Sung ES, et al. Valproic acid sensitizes TRAIL-resistant anaplastic thyroid carcinoma cells to apoptotic cell death. Ann Surg Oncol. 2013;20(Suppl 3):S716–24.PubMedCrossRef
83.
Zurück zum Zitat Borbone E, Berlingieri M, De Bellis F, Nebbioso A, Chiappetta G, Mai A, et al. Histone deacetylase inhibitors induce thyroid cancer-specific apoptosis through proteasome-dependent inhibition of TRAIL degradation. Oncogene. 2010;29(1):105–16.PubMedCrossRef Borbone E, Berlingieri M, De Bellis F, Nebbioso A, Chiappetta G, Mai A, et al. Histone deacetylase inhibitors induce thyroid cancer-specific apoptosis through proteasome-dependent inhibition of TRAIL degradation. Oncogene. 2010;29(1):105–16.PubMedCrossRef
84.
Zurück zum Zitat Champa D, Orlacchio A, Patel B, Ranieri M, Shemetov AA, Verkhusha VV, et al. Obatoclax kills anaplastic thyroid cancer cells by inducing lysosome neutralization and necrosis. Oncotarget. 2016;7(23):34453–71.PubMedPubMedCentralCrossRef Champa D, Orlacchio A, Patel B, Ranieri M, Shemetov AA, Verkhusha VV, et al. Obatoclax kills anaplastic thyroid cancer cells by inducing lysosome neutralization and necrosis. Oncotarget. 2016;7(23):34453–71.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat French JD, Bible K, Spitzweg C, Haugen BR, Ryder M. Leveraging the immune system to treat advanced thyroid cancers. Lancet Diabetes Endocrinol. 2017;5(6):469–81.PubMedCrossRef French JD, Bible K, Spitzweg C, Haugen BR, Ryder M. Leveraging the immune system to treat advanced thyroid cancers. Lancet Diabetes Endocrinol. 2017;5(6):469–81.PubMedCrossRef
86.
Zurück zum Zitat Caillou B, Talbot M, Weyemi U, Pioche-Durieu C, Al Ghuzlan A, Bidart JM, et al. Tumor-associated macrophages (TAMs) form an interconnected cellular supportive network in anaplastic thyroid carcinoma. PLoS One. 2011;6(7):e22567.PubMedPubMedCentralCrossRef Caillou B, Talbot M, Weyemi U, Pioche-Durieu C, Al Ghuzlan A, Bidart JM, et al. Tumor-associated macrophages (TAMs) form an interconnected cellular supportive network in anaplastic thyroid carcinoma. PLoS One. 2011;6(7):e22567.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Bastman JJ, Serracino HS, Zhu Y, Koenig MR, Mateescu V, Sams SB, et al. Tumor-infiltrating T cells and the PD-1 checkpoint pathway in advanced differentiated and anaplastic thyroid Cancer. J Clin Endocrinol Metab. 2016;101(7):2863–73.PubMedPubMedCentralCrossRef Bastman JJ, Serracino HS, Zhu Y, Koenig MR, Mateescu V, Sams SB, et al. Tumor-infiltrating T cells and the PD-1 checkpoint pathway in advanced differentiated and anaplastic thyroid Cancer. J Clin Endocrinol Metab. 2016;101(7):2863–73.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Chintakuntlawar AV, Rumilla KM, Smith CY, Jenkins SM, Foote RL, Kasperbauer JL, et al. Expression of PD-1 and PD-L1 in anaplastic thyroid Cancer patients treated with multimodal therapy: results from a retrospective study. J Clin Endocrinol Metab. 2017;102(6):1943–50.PubMedCrossRef Chintakuntlawar AV, Rumilla KM, Smith CY, Jenkins SM, Foote RL, Kasperbauer JL, et al. Expression of PD-1 and PD-L1 in anaplastic thyroid Cancer patients treated with multimodal therapy: results from a retrospective study. J Clin Endocrinol Metab. 2017;102(6):1943–50.PubMedCrossRef
89.
Zurück zum Zitat Brauner E, Gunda V, Borre PV, Zurakowski D, Kim YS, Dennett KV, et al. Combining BRAF inhibitor and anti PD-L1 antibody dramatically improves tumor regression and anti tumor immunity in an immunocompetent murine model of anaplastic thyroid cancer. Oncotarget. 2016;7(13):17194–211.PubMedPubMedCentralCrossRef Brauner E, Gunda V, Borre PV, Zurakowski D, Kim YS, Dennett KV, et al. Combining BRAF inhibitor and anti PD-L1 antibody dramatically improves tumor regression and anti tumor immunity in an immunocompetent murine model of anaplastic thyroid cancer. Oncotarget. 2016;7(13):17194–211.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Kollipara R, Schneider B, Radovich M, Babu S, Kiel PJ. Exceptional response with immunotherapy in a patient with anaplastic thyroid Cancer. Oncologist. 2017;22(10):1149–51.PubMedPubMedCentralCrossRef Kollipara R, Schneider B, Radovich M, Babu S, Kiel PJ. Exceptional response with immunotherapy in a patient with anaplastic thyroid Cancer. Oncologist. 2017;22(10):1149–51.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Zwaenepoel K, Jacobs J, De Meulenaere A, Silence K, Smits E, Siozopoulou V, et al. CD70 and PDL1 in anaplastic thyroid cancer -NDASH-promising targets for immunotherapy. Histopathology. 2017;71(3):357–65.PubMedCrossRef Zwaenepoel K, Jacobs J, De Meulenaere A, Silence K, Smits E, Siozopoulou V, et al. CD70 and PDL1 in anaplastic thyroid cancer -NDASH-promising targets for immunotherapy. Histopathology. 2017;71(3):357–65.PubMedCrossRef
92.
93.
Zurück zum Zitat Allegri L, Baldan F, Mio C, Puppin C, Russo D, Krystof V, et al. Effects of BP-14, a novel cyclin-dependent kinase inhibitor, on anaplastic thyroid cancer cells. Oncol Rep. 2016;35(4):2413–8.PubMedCrossRef Allegri L, Baldan F, Mio C, Puppin C, Russo D, Krystof V, et al. Effects of BP-14, a novel cyclin-dependent kinase inhibitor, on anaplastic thyroid cancer cells. Oncol Rep. 2016;35(4):2413–8.PubMedCrossRef
94.
Zurück zum Zitat Abdulghani J, Gokare P, Gallant J-N, Dicker D, Whitcomb T, Cooper T, et al. Sorafenib and Quinacrine target anti-apoptotic protein MCL1: a poor prognostic marker in anaplastic thyroid Cancer (ATC). Clin Cancer Res. 2016;22(24):6192.PubMedCrossRefPubMedCentral Abdulghani J, Gokare P, Gallant J-N, Dicker D, Whitcomb T, Cooper T, et al. Sorafenib and Quinacrine target anti-apoptotic protein MCL1: a poor prognostic marker in anaplastic thyroid Cancer (ATC). Clin Cancer Res. 2016;22(24):6192.PubMedCrossRefPubMedCentral
95.
Zurück zum Zitat Ball DW, Jin N, Xue P, Bhan S, Ahmed SR, Rosen DM, et al. Trametinib with and without pazopanib has potent preclinical activity in thyroid cancer. Oncol Rep. 2015;34(5):2319–24.PubMedPubMedCentralCrossRef Ball DW, Jin N, Xue P, Bhan S, Ahmed SR, Rosen DM, et al. Trametinib with and without pazopanib has potent preclinical activity in thyroid cancer. Oncol Rep. 2015;34(5):2319–24.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Biswas R, Mondal A, Ahn J-C. Deregulation of EGFR/PI3K and activation of PTEN by photodynamic therapy combined with carboplatin in human anaplastic thyroid cancer cells and xenograft tumors in nude mice. J Photochem Photobiol B Biol. 2015;148:118–27.CrossRef Biswas R, Mondal A, Ahn J-C. Deregulation of EGFR/PI3K and activation of PTEN by photodynamic therapy combined with carboplatin in human anaplastic thyroid cancer cells and xenograft tumors in nude mice. J Photochem Photobiol B Biol. 2015;148:118–27.CrossRef
97.
Zurück zum Zitat Vanden Borre P, Gunda V, McFadden DG, Sadow PM, Varmeh S, Bernasconi M, et al. Combined BRAF(V600E)- and SRC-inhibition induces apoptosis, evokes an immune response and reduces tumor growth in an immunocompetent orthotopic mouse model of anaplastic thyroid cancer. Oncotarget. 2014;5(12):3996–4010.PubMed Vanden Borre P, Gunda V, McFadden DG, Sadow PM, Varmeh S, Bernasconi M, et al. Combined BRAF(V600E)- and SRC-inhibition induces apoptosis, evokes an immune response and reduces tumor growth in an immunocompetent orthotopic mouse model of anaplastic thyroid cancer. Oncotarget. 2014;5(12):3996–4010.PubMed
98.
Zurück zum Zitat Chin LH, Hsu SP, Zhong WB, Liang YC. Combined treatment with troglitazone and lovastatin inhibited epidermal growth factor-induced migration through the downregulation of cysteine-rich protein 61 in human anaplastic thyroid cancer cells. PLoS One. 2015;10(3):e0118674.PubMedPubMedCentralCrossRef Chin LH, Hsu SP, Zhong WB, Liang YC. Combined treatment with troglitazone and lovastatin inhibited epidermal growth factor-induced migration through the downregulation of cysteine-rich protein 61 in human anaplastic thyroid cancer cells. PLoS One. 2015;10(3):e0118674.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. Doxorubicin has a synergistic cytotoxicity with cucurbitacin B in anaplastic thyroid carcinoma cells. Tumour Biol. 2017;39(2):1010428317692252.PubMedCrossRef Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. Doxorubicin has a synergistic cytotoxicity with cucurbitacin B in anaplastic thyroid carcinoma cells. Tumour Biol. 2017;39(2):1010428317692252.PubMedCrossRef
100.
Zurück zum Zitat Baldan F, Mio C, Allegri L, Puppin C, Russo D, Filetti S, et al. Synergy between HDAC and PARP inhibitors on proliferation of a human anaplastic thyroid Cancer-derived cell line. Int J Endocrinol. 2015;2015:7.CrossRef Baldan F, Mio C, Allegri L, Puppin C, Russo D, Filetti S, et al. Synergy between HDAC and PARP inhibitors on proliferation of a human anaplastic thyroid Cancer-derived cell line. Int J Endocrinol. 2015;2015:7.CrossRef
101.
Zurück zum Zitat Kim SH, Kang JG, Kim CS, Ihm S-H, Choi MG, Yoo HJ, et al. Novel heat shock protein 90 inhibitor NVP-AUY922 synergizes with the histone deacetylase inhibitor PXD101 in induction of death of anaplastic thyroid carcinoma cells. J Clin Endocrinol Metab. 2015;100(2):E253–E61.PubMedCrossRef Kim SH, Kang JG, Kim CS, Ihm S-H, Choi MG, Yoo HJ, et al. Novel heat shock protein 90 inhibitor NVP-AUY922 synergizes with the histone deacetylase inhibitor PXD101 in induction of death of anaplastic thyroid carcinoma cells. J Clin Endocrinol Metab. 2015;100(2):E253–E61.PubMedCrossRef
102.
Zurück zum Zitat Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. The heat shock protein 90 inhibitor SNX5422 has a synergistic activity with histone deacetylase inhibitors in induction of death of anaplastic thyroid carcinoma cells. Endocrine. 2016;51(2):274–82.PubMedCrossRef Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, et al. The heat shock protein 90 inhibitor SNX5422 has a synergistic activity with histone deacetylase inhibitors in induction of death of anaplastic thyroid carcinoma cells. Endocrine. 2016;51(2):274–82.PubMedCrossRef
103.
Zurück zum Zitat Zhang L, Boufraqech M, Lake R, Kebebew E. Carfilzomib potentiates CUDC-101-induced apoptosis in anaplastic thyroid cancer. Oncotarget. 2016;7(13):16517–28.PubMedPubMedCentral Zhang L, Boufraqech M, Lake R, Kebebew E. Carfilzomib potentiates CUDC-101-induced apoptosis in anaplastic thyroid cancer. Oncotarget. 2016;7(13):16517–28.PubMedPubMedCentral
104.
Zurück zum Zitat Ingeson-Carlsson C, Martinez-Monleon A, Nilsson M. Differential effects of MAPK pathway inhibitors on migration and invasiveness of BRAFV600E mutant thyroid cancer cells in 2D and 3D culture. Exp Cell Res. 2015;338(2):127–35.PubMedCrossRef Ingeson-Carlsson C, Martinez-Monleon A, Nilsson M. Differential effects of MAPK pathway inhibitors on migration and invasiveness of BRAFV600E mutant thyroid cancer cells in 2D and 3D culture. Exp Cell Res. 2015;338(2):127–35.PubMedCrossRef
105.
Zurück zum Zitat Eckhardt S, Hoffmann S, Damanakis AI, Di Fazio P, Pfestroff A, Luster M, et al. Individualized multimodal treatment strategy for anaplastic thyroid carcinoma—case report of long-term remission and review of literature. Int J Surg Case Rep. 2016;25:174–8.PubMedPubMedCentralCrossRef Eckhardt S, Hoffmann S, Damanakis AI, Di Fazio P, Pfestroff A, Luster M, et al. Individualized multimodal treatment strategy for anaplastic thyroid carcinoma—case report of long-term remission and review of literature. Int J Surg Case Rep. 2016;25:174–8.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Wagle N, Grabiner BC, Van Allen EM, Amin-Mansour A, Taylor-Weiner A, Rosenberg M, et al. Response and acquired resistance to Everolimus in anaplastic thyroid Cancer. N Engl J Med. 2014;371(15):1426–33.PubMedPubMedCentralCrossRef Wagle N, Grabiner BC, Van Allen EM, Amin-Mansour A, Taylor-Weiner A, Rosenberg M, et al. Response and acquired resistance to Everolimus in anaplastic thyroid Cancer. N Engl J Med. 2014;371(15):1426–33.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Savvides P, Nagaiah G, Lavertu P, Fu P, Wright JJ, Chapman R, et al. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid. 2013;23(5):600–4.PubMedPubMedCentralCrossRef Savvides P, Nagaiah G, Lavertu P, Fu P, Wright JJ, Chapman R, et al. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid. 2013;23(5):600–4.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, et al. Cabozantinib in Progressive Medullary Thyroid Cancer. Journal of Clinical Oncology. 2013. Elisei R, Schlumberger MJ, Müller SP, Schöffski P, Brose MS, Shah MH, et al. Cabozantinib in Progressive Medullary Thyroid Cancer. Journal of Clinical Oncology. 2013.
109.
Zurück zum Zitat Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, et al. A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: MC057H. The Journal of clinical endocrinology and metabolism. 2014;99(5):1687–93CrossRef Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, et al. A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: MC057H. The Journal of clinical endocrinology and metabolism. 2014;99(5):1687–93CrossRef
110.
Zurück zum Zitat Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid. 2014;24PubMedCrossRef Sosa JA, Elisei R, Jarzab B, Balkissoon J, Lu SP, Bal C. Randomized safety and efficacy study of fosbretabulin with paclitaxel/carboplatin against anaplastic thyroid carcinoma. Thyroid. 2014;24PubMedCrossRef
111.
Zurück zum Zitat Locati LD, Licitra L, Agate L, Ou SH, Boucher A, Jarzab B, et al. Treatment of advanced thyroid cancer with axitinib: Phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments. Cancer. 2014;120(17):2694–703.PubMedCrossRef Locati LD, Licitra L, Agate L, Ou SH, Boucher A, Jarzab B, et al. Treatment of advanced thyroid cancer with axitinib: Phase 2 study with pharmacokinetic/pharmacodynamic and quality-of-life assessments. Cancer. 2014;120(17):2694–703.PubMedCrossRef
112.
Zurück zum Zitat Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, et al. A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: MC057H. The Journal of clinical endocrinology and metabolism. 2014;99(5):1687–93.CrossRef Bible KC, Suman VJ, Molina JR, Smallridge RC, Maples WJ, Menefee ME, et al. A multicenter phase 2 trial of pazopanib in metastatic and progressive medullary thyroid carcinoma: MC057H. The Journal of clinical endocrinology and metabolism. 2014;99(5):1687–93.CrossRef
113.
Zurück zum Zitat Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus Placebo in Radioiodine-Refractory Thyroid Cancer. New England Journal of Medicine. 2015;372(7):621-30.CrossRef Schlumberger M, Tahara M, Wirth LJ, Robinson B, Brose MS, Elisei R, et al. Lenvatinib versus Placebo in Radioiodine-Refractory Thyroid Cancer. New England Journal of Medicine. 2015;372(7):621-30.CrossRef
114.
Zurück zum Zitat Krajewska J, Olczyk T, Jarzab B. Cabozantinib for the treatment of progressive metastatic medullary thyroid cancer. Expert Review of Clinical Pharmacology. 2016;9(1):69–79.PubMedCrossRef Krajewska J, Olczyk T, Jarzab B. Cabozantinib for the treatment of progressive metastatic medullary thyroid cancer. Expert Review of Clinical Pharmacology. 2016;9(1):69–79.PubMedCrossRef
Metadaten
Titel
Therapeutic advances in anaplastic thyroid cancer: a current perspective
verfasst von
Shikha Saini
Kiara Tulla
Ajay V. Maker
Kenneth D. Burman
Bellur S. Prabhakar
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2018
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-018-0903-0

Weitere Artikel der Ausgabe 1/2018

Molecular Cancer 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.