Skip to main content
The Oncologist logoLink to The Oncologist
. 2014 Sep 26;19(11):1148–1155. doi: 10.1634/theoncologist.2014-0182

Novel Approaches in Anaplastic Thyroid Cancer Therapy

Kun-Tai Hsu a, Xiao-Min Yu a, Anjon W Audhya b, Juan C Jaume c, Ricardo V Lloyd d, Shigeki Miyamoto e, Tomas A Prolla f, Herbert Chen a,
PMCID: PMC4221369  PMID: 25260367

The purpose of this review is to summarize the promising preclinical and clinical studies in anaplastic thyroid cancer (ATC) treatment, as well as reveal new preclinical studies with novel approaches at genetic, organelle, cellular, and microenvironment levels. Because of the poor prognosis, all ATC patients should be referred to centers that participate in current clinical trials with new agents and delivery systems.

Keywords: Thyroid cancer, Anaplastic thyroid cancer, Notch, Histone deacetylase inhibitors, Kinase inhibitors, Clinical trials

Abstract

Anaplastic thyroid cancer (ATC), accounting for less than 2% of all thyroid cancer, is responsible for the majority of death from all thyroid malignancies and has a median survival of 6 months. The resistance of ATC to conventional thyroid cancer therapies, including radioiodine and thyroid-stimulating hormone suppression, contributes to the very poor prognosis of this malignancy. This review will cover several cellular signaling pathways and mechanisms, including RET/PTC, RAS, BRAF, Notch, p53, and histone deacetylase, which are identified to play roles in the transformation and dedifferentiation process, and therapies that target these pathways. Lastly, novel approaches and agents involving the Notch1 pathway, nuclear factor κB, Trk-fused gene, cancer stem-like cells, mitochondrial mutation, and tumor immune microenvironment are discussed. With a better understanding of the biological process and treatment modality, the hope is to improve ATC outcome in the future.

Implications for Practice:

Because of its aggressive phenotype and poor prognosis, anaplastic thyroid cancer (ATC) is automatically classified as TNM stage IV regardless of tumor burden, and survival has shown minimal improvement in the last decades. The purpose of this review is to summarize the promising preclinical and clinical studies in ATC treatment, as well as reveal new preclinical studies with novel approaches at genetic, organelle, cellular, and microenvironment levels. Because of the poor prognosis, all ATC patients should be referred to centers that participate in current clinical trials with new agents and delivery systems.

Introduction

Anaplastic thyroid carcinoma (ATC), contrary to the well differentiated thyroid cancer (DTC), is one of the most aggressive human malignancies. This undifferentiated thyroid cancer is responsible for more than half of all thyroid cancer deaths, with an overall survival rate as low as 13%, despite only accounting for <2% of thyroid cancer incidence [14]. Clinical presentation is frequently characterized by a rapidly growing neck mass with associated compressive symptoms [5]. ATC also displays highly invasive behavior, with extrathyroid extension and lymph node metastasis affecting 40% of ATC patients, whereas the remaining 60% of patients have distant metastases [6]. The aggressive phenotype and poor prognosis associated with ATC form the basis for its automatic classification as TNM stage IV regardless of tumor burden [7].

Unlike DTC, which can often be cured by surgical resection, radioiodine ablation, and thyrotropin (thyroid-stimulating hormone [TSH])-suppressive therapy, treatment options for ATC are mainly palliative because of the aggressive and resilient nature of the disease. Gross resection is recommended in nearly all cases [8], and thyroidectomy can relieve airway compression, but curative resection is often impossible [9]. Lacking the sodium-iodide symporter (NIS), ATC is resistant to therapeutic radioiodine, whereas radiotherapy and chemotherapy alone have shown limited efficacy, contributing to the limited survival improvement over the last decades [3, 10, 11]. Current recommendations support multimodal interventions that use adjuvant and neoadjuvant therapy in combination with surgery to improve control of locoregional and metastatic disease [8]. Despite these measures, ATC continues to carry a median survival of less than 6 months and a 1-year survival of less than 20% [7, 12]. Therefore, investigation of novel antiproliferative, redifferentiation, immunological, and gene therapies has been an ongoing interest [13].

Markers and Signal Pathways

During the dedifferentiation process, the thyroid carcinoma loses thyroid-specific gene expression, contributing to the lack of response to radioiodine ablation therapy in ATC [14]. Specifically, NIS, which mediates the active iodide uptake at the basolateral membrane of the thyroid follicular cell, plays a crucial role in the success of radioiodine therapy [15, 16]. In addition to RAS, BRAF, RET/PTC, and p53, the Notch family receptors and ligands were recently found to be involved in the proliferation and differentiation of thyroid cancer [17, 18]. We will cover the main pathways to date that are known to contribute to thyroid oncogenesis, tumorigenesis, and dedifferentiation, including RET-RAS-RAF, Notch1, and histone deacetylase (HDAC).

Markers

The gene mutations in DTC and ATC were found to be different; thus, the progression of indolent to aggressive thyroid cancer was thought to be a multistep tumorigenesis [19, 20]. Interestingly, ATC can develop from 1% of patients with DTC [14]. Normal thyrocytes initially undergo early mutations of ret, ras, BRAF, or a paired box homeotic gene 8 (PAX8)-peroxisome proliferator activated receptor γ (PPARγ) to transform into DTCs [2124]. Further dedifferentiation into poorly differentiated thyroid cancer (PDTC), and ATCs were found to be due to inactivation mutation of p53 [2527]. p53 tumor suppressor gene is critical in regulating cell cycle and apoptosis in response to DNA and is one of the most frequently found mutation in human malignancy. In thyroid cancer, p53 mutation appeared to be a late event responsible for transformation and dedifferentiation into more aggressive types. This is evident by the low frequency of p53 mutation in DTC but a high detection rate, up to 95%, in ATC [11]. The thyroid-specific transcription factor (TTF-1) and PAX-8 expression levels are decreased in less-differentiated papillary and follicular carcinomas and are lost in ATC, with comparable level changes of thyroglobulin (Tg), thyroperoxidase (TPO), thyrotropin receptor (TSH-R), and NIS [2831].

RET/PTC-RAS-RAF

RET/PTC is a chimeric oncogene between RET, located at chromosome 10q11.2, and the promoter of an unrelated gene, resulting in constitutive activation of the RET gene [32, 33]. To date, 13 different types of RET/PTC rearrangements have been reported [34]. RAS is the next direct downstream effector in this cascade. RAS mutation, although most significant in the diagnosis of follicular thyroid cancer (FTC), is also reported in papillary thyroid cancer (PTC) and ATC [35, 36]. From studies of different histological types of thyroid cancer, up to 60% of ATC was found to harbor RAS mutation [3638]. In a stepwise process, RAS appeared to be an “early-stage” mutation. Furthermore, RAS mutation was proposed to initiate dedifferentiation of DTC into ATC, because RAS mutation predisposes the cell to more genetic and molecular derangement, likely because of chromosome instability [39, 40].

BRAF, a member of the RAF serine/threonine-kinase family and a downstream effector of RAS, has been shown to be an important regulator in normal thyroid cell proliferation, apoptosis, and thyroid-specific gene expression [41, 42]. Liu et al. [43] showed that BRAF mutation and the subsequent activation of MAPK pathway in rat thyroid cells could silence expression of NIS, and removal of BRAF with siRNA restore the expression of thyroid-specific genes. BRAF mutation, specifically BRAFV600E mutation, was demonstrated to impede both NIS gene expression and NIS membrane localization, whereas inhibition of BRAF by smad7 reversed NIS transcription repression [4446]. In addition to promoting dedifferentiation, BRAF is also responsible in promoting migration and invasive growth [41, 47, 48]. Based on the significance of BRAF mutation in PTC and the potential of PTC to transform into ATC, BRAF is a target of investigation in treatment of ATC [49, 50].

Another translocation event underlying PTC involves a fusion between Trk-fused gene (TFG) and the receptor tyrosine kinase NTRK1 [51]. Analogous to chromosomal rearrangements involving RET, several TFG fusion proteins have been shown to result in the hyperactivation of MAPK kinase signaling [5255]. Endogenous TFG localizes to specialized subdomains of the endoplasmic reticulum that are responsible for the biogenesis of vesicles that carry secretory cargoes out of cells [55]. The TFG-NTRK1 fusion protein similarly localizes to these sites, and this distribution is key to its transforming activity [55]. Although direct targets of TFG-NTRK1 activity have yet to be defined, these studies highlight the possibility that alterations in the secretory pathway may contribute to cell transformation or oncogenesis in the thyroid.

Notch1

Notch receptors (Notch1–4) and ligands (δ-like 1, 3, and 4 and Jagged-1 and -2) were reported to regulate cell proliferation, migration, adhesion, and differentiation in various situations [56]. Depending on the cell type, Notch can function as either an oncogene or a tumor suppressor [18, 5762]. Ferretti et al. [17] first demonstrated that the expression of Notch was decreased in DTC and further in ATC when compared with normal thyroid tissue. The effect of Notch1 on cell growth and differentiation is exerted via transcription regulation [63]. Although Notch1 was studied in many other tissues and cancers, the role of Notch1 in thyroid cancer has only recently being explored. As a downstream effector of Notch1, Hes1 plays a central role in thyrocyte proliferation and differentiation, evident by a 34%–65% decrease in thyroid surface area and 69% decrease in NIS protein expression in Hes1−/− mouse embryo [64].

HDAC

Acetylation of histone lysine residue induces changes in the nucleosomal conformation caused by decreased affinity for the negatively charged DNA [65, 66]. Inhibition of HDAC was shown to induce expression of NIS, TPO, and Tg in thyroid carcinoma cell lines [6769]. Thyroid-specific gene induction resulted in increased radioiodine uptake, organification, and intratumoral radioiodine accumulation. The HDAC inhibitor induced re-expression of NIS, but TPO or Tg were not mediated by newly produced transcription factor [68]. Zhang et al. [70] found that HDAC was the link between BRAF mutation and NIS silencing. The BRAF mutation upregulates HDAC, causing an epigenetic modification via constitution histone acetylation at the NIS promoter site. The significance of HDAC in thyroid cancer resistance was demonstrated by Hou et al. [31] with a potent HDAC inhibitor, suberanilohydroxamic (SAHA or vorinostat), which restored thyroid-specific gene expression, including NIS, TSH-R, TPO, Tg, and TTF-1, as well as radioiodine uptake. Therefore, therapies that can move the balance toward histone acetylation should be able to restore thyroid gene expression and ultimately induce radioiodine avidity to improve the effectiveness of radioiodine therapy.

NF-κB

Nuclear factor κB (NF-κB) is a family of transcription factors that is held inactive in the cytoplasm of resting cells, including thyroid cancer cells, and its activation is induced by multiple proinflammatory cytokines, chemotherapeutic agents, and ionizing radiation [71]. Many NF-κB target genes are prosurvival genes and are critical for intrinsic cancer cell resistance to chemo- and radiation therapy, and therefore inhibition of NF-κB activity could lead to apoptosis or sensitization to chemo- and radiotherapy in various cancer cell, including thyroid cancer cells, in vitro and in vivo [7274]. Activation of NF-κB was observed in PTC and FTC, as well as aggressive ATC [7578]. Moreover, recent findings demonstrate that NF-κB significantly contributes to the establishment and maintenance of protumorigenic microenvironment [79, 80]. Therefore, inhibition of NF-κB activation is thought to enforce antitumorigenic effects of chemo- and radiation therapy through cancer cell-intrinsic and -extrinsic mechanisms.

Therapeutic Approach

Recent developments of treatment modalities in advance thyroid cancers include investigation of kinase inhibitors, redifferentiation therapy, statins, and gene transfer [81].

Kinase Inhibitors

Epidermal growth factor receptor (EGFR) mutation was described in thyroid cancer and contributes to RET activation [82]. Therefore, gefitinib, an EGFR inhibitor, is a drug of interest. In a phase II trial with a mixed cohort of thyroid cancer patients, including 19% as ATC, gefitinib did not induce any tumor response, but 12% of patients had stable disease at 12 months and median progression-free survival (PFS) was 16 weeks for ATC patients [83]. Other receptors are also therapeutic targets under investigation. Rugo et al. [84] established the safety and clinical activity of axitinib, an inhibitor vascular endothelial growth factor receptors 1, 2, and 3, in various advanced solid tumor, including thyroid tumor. A subsequent phase II trial from the University of Chicago with a mixed cohort of 60 patients, including 3 patients with ATC, showed that 1 ATC patient had a partial response, whereas the rest of the patients had stable disease [85]. Sorafenib is a multikinase inhibitor that targets BRAF, which showed growth inhibition in ATC xenografts and improves survival in vivo [8688]. An early phase II trial with a mixed cohort with metastatic, radioiodine-refractory disease, including one patient with PDTC and one patient with ATC, showed an overall clinical benefit rate of 77% with a median PFS of 79 weeks and a 70% mean decrease of serum Tg level [89]. However, both patients with PDTC and ATC had progressive disease despite treatment, and the treatment for the ATC patients was discontinued 4 days after initiation because of medical complications. A randomized controlled trial by Brose et al. [90], including 10% PDTC but no ATC patients, demonstrated that sorafenib elicited 12.2% partial response and was superior in prolonging PFS when compared with placebo. A recent multi-institutional phase II clinical trial with all ATC patients showed a 35% of partial response or stable disease rate in 20 patients who failed previous treatments with tolerable adverse events [91]. Because BRAF mutation was shown to be partly responsible for dedifferentiation and depressed expression of NIS [43], a combination therapy of BRAF inhibition and radioiodine therapy provides a potential future approach. Other phase II trials with various BRAF inhibitors, including vemurafenib (NCT01524978), dabrafenib (NCT02034110), and LGX818 (NCT01981187), are underway [92].

Because BRAF mutation was shown to be partly responsible for dedifferentiation and depressed expression of NIS, a combination therapy of BRAF inhibition and radioiodine therapy provides a potential future approach.

HDAC Inhibitors

Depsipeptide was able to induce re-expression of NIS, TPO, and Tg in vitro and in vivo but had no effect on re-expression of TSH-R [68]. Unfortunately, clinical experience in other cancers showed that patients treated with depsipeptide experienced severe gastrointestinal, cardiac, and cutaneous adverse events [93]. On the other hand, SAHA was shown to restore NIS, TPO, and Tg, as well as TSH-R in ATC cell lines [31]. Valproic acid (VPA), a safe and widely used anticonvulsant and mood stabilizer, was found to act as a HDAC inhibitor in addition to enhancing the apoptotic activity of paclitaxel [94, 95]. A study by Catalano et al. [96] found that when used in combination with imatinib, VPA induced cell cycle arrest via G1 cell cycle arrest by decreasing AKT phosphorylation without affecting apoptosis. Although VPA showed promise in inducing expression of NIS gene, NIS membrane localization, and radioiodine accumulation in PDTC at a safe dose, it failed to show effects in ATC beyond re-expression of NIS gene [97]. Another novel molecule, LBH589, is under phase II clinical trial at University of Wisconsin-Madison for patients with metastatic MTC and radioiodine-resistant DTC [92]. The potential of applying the results to ATC patients is promising with the inclusion of radioiodine-resistant patients in the study.

Gene Therapy

There have been many types of promising gene therapies, including corrective gene therapy, cytoreductive gene therapy, and immunomodulatory gene therapy [11]. Spitzweg et al. [10] first proposed the potential cytoreductive gene therapy to transfer NIS gene followed by radioiodine therapy. Unfortunately, transfection of the NIS gene alone does not appear sufficient in cells lacking other thyroid-specific proteins, including Tg, TPO, and TSH-R, because of efflux of radioiodine that is not organified and retained in the cells [98, 99]. Means to increase efficacy of NIS gene therapy include introducing other thyroid-specific genes to enhance radioiodine retention by organification, increasing NIS gene transduction efficiency to maximize uptake, blocking iodide efflux to minimize lose, and application of alternative radionuclides to maximize energy deposit [11, 15, 100102].

Another application for gene therapy is to allow the innate immune system to recognize and destroy the tumors via immunomodulation. Cotransfection of human interleukin-2 and the HSV-tk gene in ATC cells, showing complete elimination in ATC xenograft [103]. In a pilot study of combined suicide and cytokine gene therapy in two patients with end-stage ATC, Barzon et al. [104] discovered some promise in immune response and local tumor destruction with application of the therapy.

In a study with corrective gene therapy, Lee et al. [105] showed that neither NIS gene nor wild-type p53 gene transfer alone affect ATC cell survival in vitro or tumor growth in vivo after radionuclide treatment but found a decrease in tumor volume of 80% with NIS, wild-type p53, and radionuclide treatment. Furthermore, wild-type p53 restoration in p53 mutant ATC was associated with redifferentiation and promotes induction of thyroid-specific genes [104, 106].

Natural Compounds

Natural compounds often have relatively low toxicity and may eventually play a supporting role in drug treatments targeting ATC. We have previously shown that the plant polyphenol resveratrol suppresses ATC cell growth in a dose-dependent manner for both HTh7 and 8505C cell lines via S-phase cell-cycle arrest and apoptosis [63]. In vivo experiments showed reduced growth of ATC tumors derived from both cell lines when administered i.p. at 10 mg/kg. The expression of thyroid-specific genes including TTF1, TTF2, Pax8, and NIS was upregulated in both ATC cell lines with resveratrol treatment, and Notch1 siRNA interference abrogated the induction of some of these genes. Thus, this study links resveratrol treatment, activation of Notch1 signaling, and redifferentiation. We have also shown that chrysin (a plant flavone) treatment of ATC cells leads to induction of messenger RNA levels of Notch1 and a dose-dependent inhibition of cellular growth. Oral administration of chrysin suppressed the growth of ATC xenografts by an average of 59% compared with the vehicle control group [107]. Recently, Kang et al. [108] have shown growth-inhibiting and redifferentiation effects of several plant phytochemicals in thyroid cancer cell lines. Although the clinical experience with natural compounds in thyroid cancer is limited, these animal studies suggest potential benefits.

Other Agents and Approaches

Bortezomib, approved for treatment of advanced multiple myeloma [109], can effectively control canonical and noncanonical NF-κB signaling, and such effects are thought to contribute to antimyeloma effects [110]. Although bortezomib alone had a modest effect on advanced thyroid cancers [111], when combined with other chemotherapy agent for thyroid cancer in preclinical studies, there was some synergy, indicating the promise of bortezomib in combination therapy [112]. Inhibition of NF-κB activity in different thyroid cancer contexts, including aggressive forms of thyroid cancers, is uniformly reported to cause chemo- or radiosensitization [73, 74, 76, 113119] and is associated with reduced metastatic potential [78]. These observations provide the scientific rationale for targeting NF-κB for treatment in resistant, aggressive thyroid malignancies.

Thiazolidinediones, known most commonly for antidiabetic therapy, is a potent agonist for the PPARγ, which is a ligand-activated transcription factor responsible for cell proliferation and growth [120122]. Although ATC cell has high level of PPARγ, troglitazone was unable to inhibit cell proliferation of ATC cells until a concentration of 20 μM [121]. A later study by Antonelli et al. [123] showed that both rosiglitazone and pioglitazone inhibited ATC cells at a level of 20 μM as early as 1 hour after administration, regardless of BRAF mutation. Another study demonstrated that thiazolidinediones, with troglitazone having the greater effect, not only inhibited transformed thyroid cell proliferation but also induced redifferentiation, evident by re-expression of NIS and radioiodide uptake [122].

3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, antagonists of the rate-limiting step of cholesterol production, are frequently used for treatment of hypercholesterolemia and reducing cardiovascular morbidity and mortalities [124127]. The main effect of statin on ATC is proliferation inhibition by apoptosis via different proposed mechanisms, including inhibition of geranylgeranylation of ρ, lamin B proteolysis, and cytochrome c release from mitochondria [128131]. In an in vivo study, Wang et al. [132] demonstrated that lovastatin was able to decreased ATC tumor growth rate in xenograft by 73% at a concentration of 10 mg/kg per day. Furthermore, lovastatin exerted either apoptotic or cytomorphological differentiation effects on ATC cells at concentration [130]. However, radioiodine uptake was largely unaffected [133].

In addition to focusing on the thyroid cancer as a whole, the existence of subsets of cancer stem-like cells (CSCs) with the ability for self-renewal and unlimited growth has been postulated [134137]. Higher percentages of CSCs may be responsible for the resistance to radiation and chemotherapy, as well as the aggressive behavior of ATC [136, 138]. The inability to differentiate beyond CSC is due to genomic alterations in RET/PTC, PAX8-PPARγ rearrangement, and BRAF mutation [139]. Epithelial-mesenchymal transition is thought to be the mechanism that allows the more well-differentiated cancer cells to transform into CSCs [136, 140]. Further understanding of this mechanism and characterization of CSCs may lead to more effective therapies for more aggressive and lethal thyroid cancers [139].

Finally, modifying the tumor immune microenvironment from pro- to antitumorigenic has been an approach sought after by many [141]. It has been recently shown that thyroid inflammation affects thyroid cancer outcomes [142]. An inflammatory immune microenvironment seems to be protumorigenic, whereas an autoimmune microenvironment seems to be antitumorigenic [142, 143]. T-cell progenitors present in thyroid cancer inflammatory immune microenvironment seem to facilitate tumor development [143]. Driving these T-cell progenitors to maturity may become a therapeutic strategy in itself or in combination with other approaches.

T-cell progenitors present in thyroid cancer inflammatory immune microenvironment seem to facilitate tumor development. Driving these T-cell progenitors to maturity may become a therapeutic strategy in itself or in combination with other approaches.

Conclusion

ATC is an aggressive malignancy with extremely poor prognosis, partly because of the resistance to radioiodine uptake, which is one of the mainstay therapies besides surgery for thyroid cancer. The re-expression of thyroid-specific genes, specifically NIS, has the potential to restore the radioiodine uptake ability of the cancer. Furthermore, the ability of cells to take up radioiodine can be enhanced by TSH, which necessitates the re-expression of TSH-R. Many studies have demonstrated that redifferentiation can be achieved by modifying various pathways, including Notch1, RET/PTC-RAS-RAF-MEK, HDAC, and NF-κB with different agents or gene therapy. Clinical trials so far have only shown no or slight improvement in survival. Although a small case series from Mayo Clinic demonstrated the beneficial potential of aggressive multimodal therapy [144], further prospective trials with multimodal or multitarget approach should be performed in hopes of improving the clinical outcome of ATC. Lastly, emerging targets and approaches can not only provide better understanding of the tumor biology but also more optimistic outcome in the future.

Acknowledgments

The work was supported by an American Cancer Society Research Scholar Grant (to H.C.), an American Cancer Society MEN2 Thyroid Cancer Professorship (to H.C.), and NIH Grant R01 CA121115 (to H.C.).

Footnotes

For Further Reading:Ligy Thomas, Stephen Y. Lai, Wenli Dong et al. Sorafenib in Metastatic Thyroid Cancer: A Systematic Review. The Oncologist 2014;19:251–258.

Implications for Practice:This meta-analysis of 219 patients treated with sorafenib for metastatic thyroid cancers demonstrated that 81% of patients had either partial response or stable disease, and none had a complete response. The partial response rate was best for medullary thyroid cancer, followed by differentiated thyroid cancer. Responses in anaplastic thyroid cancer were low. The overall median progression-free survival was 18 months for all histologies. There were significant dose reductions and discontinuations as a result of toxicities, which need to be considered when treating patients who may otherwise be asymptomatic and have reasonable overall survival.

Author Contributions

Conception/Design: Kun-Tai Hsu, Xiao-Min Yu, Herbert Chen

Collection and/or assembly of data: Kun-Tai Hsu

Manuscript writing: Kun-Tai Hsu, Xiao-Min Yu

Final approval of manuscript: Kun-Tai Hsu, Xiao-Min Yu, Anjon W. Audhya, Juan C. Jaume, Ricardo V. Lloyd, Shigeki Miyamoto, Tomas A. Prolla, Herbert Chen

Disclosures

The authors indicated no financial relationships.

References

  • 1.Hundahl SA, Fleming ID, Fremgen AM, et al. A National Cancer Data Base report on 53,856 cases of thyroid carcinoma treated in the U.S., 1985–1995 [see comments] Cancer. 1998;83:2638–2648. doi: 10.1002/(sici)1097-0142(19981215)83:12<2638::aid-cncr31>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
  • 2.Kebebew E, Greenspan FS, Clark OH, et al. Anaplastic thyroid carcinoma: Treatment outcome and prognostic factors. Cancer. 2005;103:1330–1335. doi: 10.1002/cncr.20936. [DOI] [PubMed] [Google Scholar]
  • 3.Are C, Shaha AR. Anaplastic thyroid carcinoma: Biology, pathogenesis, prognostic factors, and treatment approaches. Ann Surg Oncol. 2006;13:453–464. doi: 10.1245/ASO.2006.05.042. [DOI] [PubMed] [Google Scholar]
  • 4.Sherman SI. Thyroid carcinoma. Lancet. 2003;361:501–511. doi: 10.1016/s0140-6736(03)12488-9. [DOI] [PubMed] [Google Scholar]
  • 5.Neff RL, Farrar WB, Kloos RT, et al. Anaplastic thyroid cancer. Endocrinol Metab Clin North Am. 2008;37:525–538, xi. doi: 10.1016/j.ecl.2008.02.003. [DOI] [PubMed] [Google Scholar]
  • 6.Chen J, Tward JD, Shrieve DC, et al. Surgery and radiotherapy improves survival in patients with anaplastic thyroid carcinoma: Analysis of the surveillance, epidemiology, and end results 1983-2002. Am J Clin Oncol. 2008;31:460–464. doi: 10.1097/COC.0b013e31816a61f3. [DOI] [PubMed] [Google Scholar]
  • 7.O’Neill JP, Shaha AR. Anaplastic thyroid cancer. Oral Oncol. 2013;49:702–706. doi: 10.1016/j.oraloncology.2013.03.440. [DOI] [PubMed] [Google Scholar]
  • 8.Smallridge RC, Ain KB, Asa SL, et al. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid. 2012;22:1104–1139. doi: 10.1089/thy.2012.0302. [DOI] [PubMed] [Google Scholar]
  • 9.Pinchot SN, Sippel RS, Chen H. Multi-targeted approach in the treatment of thyroid cancer. Ther Clin Risk Manag. 2008;4:935–947. doi: 10.2147/tcrm.s3062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Spitzweg C, Harrington KJ, Pinke LA, et al. Clinical review 132: The sodium iodide symporter and its potential role in cancer therapy. J Clin Endocrinol Metab. 2001;86:3327–3335. doi: 10.1210/jcem.86.7.7641. [DOI] [PubMed] [Google Scholar]
  • 11.Spitzweg C. Gene therapy in thyroid cancer. Horm Metab Res. 2009;41:500–509. doi: 10.1055/s-0029-1220744. [DOI] [PubMed] [Google Scholar]
  • 12.Smallridge RC, Copland JA. Anaplastic thyroid carcinoma: Pathogenesis and emerging therapies. Clin Oncol (R Coll Radiol) 2010;22:486–497. doi: 10.1016/j.clon.2010.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Deshpande HA, Roman S, Sosa JA. New targeted therapies and other advances in the management of anaplastic thyroid cancer. Curr Opin Oncol. 2013;25:44–49. doi: 10.1097/CCO.0b013e32835a448c. [DOI] [PubMed] [Google Scholar]
  • 14.Park JW, Clark OH. Redifferentiation therapy for thyroid cancer. Surg Clin North Am. 2004;84:921–943. doi: 10.1016/j.suc.2004.01.005. [DOI] [PubMed] [Google Scholar]
  • 15.Spitzweg C, Morris JC. The sodium iodide symporter: Its pathophysiological and therapeutic implications. Clin Endocrinol (Oxf) 2002;57:559–574. doi: 10.1046/j.1365-2265.2002.01640.x. [DOI] [PubMed] [Google Scholar]
  • 16.Dohán O, De la Vieja A, Paroder V, et al. The sodium/iodide symporter (NIS): Characterization, regulation, and medical significance. Endocr Rev. 2003;24:48–77. doi: 10.1210/er.2001-0029. [DOI] [PubMed] [Google Scholar]
  • 17.Ferretti E, Tosi E, Po A, et al. Notch signaling is involved in expression of thyrocyte differentiation markers and is down-regulated in thyroid tumors. J Clin Endocrinol Metab. 2008;93:4080–4087. doi: 10.1210/jc.2008-0528. [DOI] [PubMed] [Google Scholar]
  • 18.Kunnimalaiyaan M, Vaccaro AM, Ndiaye MA, et al. Overexpression of the NOTCH1 intracellular domain inhibits cell proliferation and alters the neuroendocrine phenotype of medullary thyroid cancer cells. J Biol Chem. 2006;281:39819–39830. doi: 10.1074/jbc.M603578200. [DOI] [PubMed] [Google Scholar]
  • 19.Moore JH, Jr, Bacharach B, Choi HY. Anaplastic transformation of metastatic follicular carcinoma of the thyroid. J Surg Oncol. 1985;29:216–221. doi: 10.1002/jso.2930290403. [DOI] [PubMed] [Google Scholar]
  • 20.Farid NR, Shi Y, Zou M. Molecular basis of thyroid cancer. Endocr Rev. 1994;15:202–232. doi: 10.1210/edrv-15-2-202. [DOI] [PubMed] [Google Scholar]
  • 21.Santoro M, Carlomagno F, Hay ID, et al. Ret oncogene activation in human thyroid neoplasms is restricted to the papillary cancer subtype. J Clin Invest. 1992;89:1517–1522. doi: 10.1172/JCI115743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ruco LP, Ranalli T, Marzullo A, et al. Expression of Met protein in thyroid tumours. J Pathol. 1996;180:266–270. doi: 10.1002/(SICI)1096-9896(199611)180:3<266::AID-PATH658>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  • 23.Bongarzone I, Pierotti MA, Monzini N, et al. High frequency of activation of tyrosine kinase oncogenes in human papillary thyroid carcinoma. Oncogene. 1989;4:1457–1462. [PubMed] [Google Scholar]
  • 24.Kroll TG, Sarraf P, Pecciarini L, et al. PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma. Science. 2000;289:1357–1360. doi: 10.1126/science.289.5483.1357. [DOI] [PubMed] [Google Scholar]
  • 25.Fagin JA, Matsuo K, Karmakar A, et al. High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas. J Clin Invest. 1993;91:179–184. doi: 10.1172/JCI116168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jossart GH, Epstein HD, Shaver JK, et al. Immunocytochemical detection of p53 in human thyroid carcinomas is associated with mutation and immortalization of cell lines. J Clin Endocrinol Metab. 1996;81:3498–3504. doi: 10.1210/jcem.81.10.8855792. [DOI] [PubMed] [Google Scholar]
  • 27.Moretti F, Nanni S, Farsetti A, et al. Effects of exogenous p53 transduction in thyroid tumor cells with different p53 status. J Clin Endocrinol Metab. 2000;85:302–308. doi: 10.1210/jcem.85.1.6295. [DOI] [PubMed] [Google Scholar]
  • 28.Ros P, Rossi DL, Acebrón A, et al. Thyroid-specific gene expression in the multi-step process of thyroid carcinogenesis. Biochimie. 1999;81:389–396. doi: 10.1016/s0300-9084(99)80086-8. [DOI] [PubMed] [Google Scholar]
  • 29.Russo D, Bulotta S, Bruno R, et al. Sodium/iodide symporter (NIS) and pendrin are expressed differently in hot and cold nodules of thyroid toxic multinodular goiter. Eur J Endocrinol. 2001;145:591–597. doi: 10.1530/eje.0.1450591. [DOI] [PubMed] [Google Scholar]
  • 30.Mian C, Lacroix L, Alzieu L, et al. Sodium iodide symporter and pendrin expression in human thyroid tissues. Thyroid. 2001;11:825–830. doi: 10.1089/105072501316973073. [DOI] [PubMed] [Google Scholar]
  • 31.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;95:820–828. doi: 10.1210/jc.2009-1888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Takahashi M, Ritz J, Cooper GM. Activation of a novel human transforming gene, ret, by DNA rearrangement. Cell. 1985;42:581–588. doi: 10.1016/0092-8674(85)90115-1. [DOI] [PubMed] [Google Scholar]
  • 33.Kimura ET, Nikiforova MN, Zhu Z, et al. High prevalence of BRAF mutations in thyroid cancer: Genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 2003;63:1454–1457. [PubMed] [Google Scholar]
  • 34.Romei C, Elisei R. Ret/ptc translocations and clinico-pathological features in human papillary thyroid carcinoma. Front Endocrinol (Lausanne) 2012;3:54. doi: 10.3389/fendo.2012.00054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gupta N, Dasyam AK, Carty SE, et al. RAS mutations in thyroid FNA specimens are highly predictive of predominantly low-risk follicular-pattern cancers. J Clin Endocrinol Metab. 2013;98:E914–E922. doi: 10.1210/jc.2012-3396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Garcia-Rostan G, Zhao H, Camp RL, et al. ras mutations are associated with aggressive tumor phenotypes and poor prognosis in thyroid cancer. J Clin Oncol. 2003;21:3226–3235. doi: 10.1200/JCO.2003.10.130. [DOI] [PubMed] [Google Scholar]
  • 37.Lemoine NR, Mayall ES, Wyllie FS, et al. High frequency of ras oncogene activation in all stages of human thyroid tumorigenesis. Oncogene. 1989;4:159–164. [PubMed] [Google Scholar]
  • 38.Basolo F, Pisaturo F, Pollina LE, et al. N-ras mutation in poorly differentiated thyroid carcinomas: Correlation with bone metastases and inverse correlation to thyroglobulin expression. Thyroid. 2000;10:19–23. doi: 10.1089/thy.2000.10.19. [DOI] [PubMed] [Google Scholar]
  • 39.Saavedra HI, Knauf JA, Shirokawa JM, et al. The RAS oncogene induces genomic instability in thyroid PCCL3 cells via the MAPK pathway. Oncogene. 2000;19:3948–3954. doi: 10.1038/sj.onc.1203723. [DOI] [PubMed] [Google Scholar]
  • 40.Howell GM, Hodak SP, Yip L. RAS mutations in thyroid cancer. The Oncologist. 2013;18:926–932. doi: 10.1634/theoncologist.2013-0072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mitsutake N, Knauf JA, Mitsutake S, et al. Conditional BRAFV600E expression induces DNA synthesis, apoptosis, dedifferentiation, and chromosomal instability in thyroid PCCL3 cells. Cancer Res. 2005;65:2465–2473. doi: 10.1158/0008-5472.CAN-04-3314. [DOI] [PubMed] [Google Scholar]
  • 42.Puxeddu E, Durante C, Avenia N, et al. Clinical implications of BRAF mutation in thyroid carcinoma. Trends Endocrinol Metab. 2008;19:138–145. doi: 10.1016/j.tem.2007.12.003. [DOI] [PubMed] [Google Scholar]
  • 43.Liu D, Hu S, Hou P, et al. Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant. Clin Cancer Res. 2007;13:1341–1349. doi: 10.1158/1078-0432.CCR-06-1753. [DOI] [PubMed] [Google Scholar]
  • 44.Riesco-Eizaguirre G, Rodríguez I, De la Vieja A, et al. The BRAFV600E oncogene induces transforming growth factor beta secretion leading to sodium iodide symporter repression and increased malignancy in thyroid cancer. Cancer Res. 2009;69:8317–8325. doi: 10.1158/0008-5472.CAN-09-1248. [DOI] [PubMed] [Google Scholar]
  • 45.Romei C, Ciampi R, Faviana P, et al. BRAFV600E mutation, but not RET/PTC rearrangements, is correlated with a lower expression of both thyroperoxidase and sodium iodide symporter genes in papillary thyroid cancer. Endocr Relat Cancer. 2008;15:511–520. doi: 10.1677/ERC-07-0130. [DOI] [PubMed] [Google Scholar]
  • 46.Riesco-Eizaguirre G, Gutiérrez-Martínez P, García-Cabezas MA, et al. The oncogene BRAF V600E is associated with a high risk of recurrence and less differentiated papillary thyroid carcinoma due to the impairment of Na+/I- targeting to the membrane. Endocr Relat Cancer. 2006;13:257–269. doi: 10.1677/erc.1.01119. [DOI] [PubMed] [Google Scholar]
  • 47.Melillo RM, Castellone MD, Guarino V, et al. The RET/PTC-RAS-BRAF linear signaling cascade mediates the motile and mitogenic phenotype of thyroid cancer cells. J Clin Invest. 2005;115:1068–1081. doi: 10.1172/JCI22758. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 48.Mesa C, Jr., Mirza M, Mitsutake N, et al. Conditional activation of RET/PTC3 and BRAFV600E in thyroid cells is associated with gene expression profiles that predict a preferential role of BRAF in extracellular matrix remodeling. Cancer Res. 2006;66:6521–6529. doi: 10.1158/0008-5472.CAN-06-0739. [DOI] [PubMed] [Google Scholar]
  • 49.Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer. 2005;12:245–262. doi: 10.1677/erc.1.0978. [DOI] [PubMed] [Google Scholar]
  • 50.Granata R, Locati L, Licitra L. Therapeutic strategies in the management of patients with metastatic anaplastic thyroid cancer: Review of the current literature. Curr Opin Oncol. 2013;25:224–228. doi: 10.1097/CCO.0b013e32835ff44b. [DOI] [PubMed] [Google Scholar]
  • 51.Greco A, Mariani C, Miranda C, et al. The DNA rearrangement that generates the TRK-T3 oncogene involves a novel gene on chromosome 3 whose product has a potential coiled-coil domain. Mol Cell Biol. 1995;15:6118–6127. doi: 10.1128/mcb.15.11.6118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hernández L, Pinyol M, Hernández S, et al. TRK-fused gene (TFG) is a new partner of ALK in anaplastic large cell lymphoma producing two structurally different TFG-ALK translocations. Blood. 1999;94:3265–3268. [PubMed] [Google Scholar]
  • 53.Hernández L, Beà S, Bellosillo B, et al. Diversity of genomic breakpoints in TFG-ALK translocations in anaplastic large cell lymphomas: Identification of a new TFG-ALK(XL) chimeric gene with transforming activity. Am J Pathol. 2002;160:1487–1494. doi: 10.1016/S0002-9440(10)62574-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Roccato E, Miranda C, Raho G, et al. Analysis of SHP-1-mediated down-regulation of the TRK-T3 oncoprotein identifies Trk-fused gene (TFG) as a novel SHP-1-interacting protein. J Biol Chem. 2005;280:3382–3389. doi: 10.1074/jbc.M407522200. [DOI] [PubMed] [Google Scholar]
  • 55.Witte K, Schuh AL, Hegermann J, et al. TFG-1 function in protein secretion and oncogenesis. Nat Cell Biol. 2011;13:550–558. doi: 10.1038/ncb2225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Bolós V, Grego-Bessa J, de la Pompa JL. Notch signaling in development and cancer. Endocr Rev. 2007;28:339–363. doi: 10.1210/er.2006-0046. [DOI] [PubMed] [Google Scholar]
  • 57.Radtke F, Raj K. The role of Notch in tumorigenesis: Oncogene or tumour suppressor? Nat Rev Cancer. 2003;3:756–767. doi: 10.1038/nrc1186. [DOI] [PubMed] [Google Scholar]
  • 58.Nickoloff BJ, Qin JZ, Chaturvedi V, et al. Jagged-1 mediated activation of notch signaling induces complete maturation of human keratinocytes through NF-kappaB and PPARgamma. Cell Death Differ. 2002;9:842–855. doi: 10.1038/sj.cdd.4401036. [DOI] [PubMed] [Google Scholar]
  • 59.Nakakura EK, Sriuranpong VR, Kunnimalaiyaan M, et al. Regulation of neuroendocrine differentiation in gastrointestinal carcinoid tumor cells by notch signaling. J Clin Endocrinol Metab. 2005;90:4350–4356. doi: 10.1210/jc.2005-0540. [DOI] [PubMed] [Google Scholar]
  • 60.Kunnimalaiyaan M, Traeger K, Chen H. Conservation of the Notch1 signaling pathway in gastrointestinal carcinoid cells. Am J Physiol Gastrointest Liver Physiol. 2005;289:G636–G642. doi: 10.1152/ajpgi.00146.2005. [DOI] [PubMed] [Google Scholar]
  • 61.Shou J, Ross S, Koeppen H, et al. Dynamics of notch expression during murine prostate development and tumorigenesis. Cancer Res. 2001;61:7291–7297. [PubMed] [Google Scholar]
  • 62.Talora C, Sgroi DC, Crum CP, et al. Specific down-modulation of Notch1 signaling in cervical cancer cells is required for sustained HPV-E6/E7 expression and late steps of malignant transformation. Genes Dev. 2002;16:2252–2263. doi: 10.1101/gad.988902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Yu XM, Jaskula-Sztul R, Ahmed K, et al. Resveratrol induces differentiation markers expression in anaplastic thyroid carcinoma via activation of Notch1 signaling and suppresses cell growth. Mol Cancer Ther. 2013;12:1276–1287. doi: 10.1158/1535-7163.MCT-12-0841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Carre A, Rachdi L, Tron E, et al. Hes1 is required for appropriate morphogenesis and differentiation during mouse thyroid gland development. PLoS One. 2011;6:e16752. doi: 10.1371/journal.pone.0016752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hong L, Schroth GP, Matthews HR, et al. Studies of the DNA binding properties of histone H4 amino terminus. Thermal denaturation studies reveal that acetylation markedly reduces the binding constant of the H4 “tail” to DNA. J Biol Chem. 1993;268:305–314. [PubMed] [Google Scholar]
  • 66.Nagy L, Kao HY, Chakravarti D, et al. Nuclear receptor repression mediated by a complex containing SMRT, mSin3A, and histone deacetylase. Cell. 1997;89:373–380. doi: 10.1016/s0092-8674(00)80218-4. [DOI] [PubMed] [Google Scholar]
  • 67.Kitazono M, Robey R, Zhan Z, et al. Low concentrations of the histone deacetylase inhibitor, depsipeptide (FR901228), increase expression of the Na(+)/I(−) symporter and iodine accumulation in poorly differentiated thyroid carcinoma cells. J Clin Endocrinol Metab. 2001;86:3430–3435. doi: 10.1210/jcem.86.7.7621. [DOI] [PubMed] [Google Scholar]
  • 68.Furuya F, Shimura H, Suzuki H, et al. Histone deacetylase inhibitors restore radioiodide uptake and retention in poorly differentiated and anaplastic thyroid cancer cells by expression of the sodium/iodide symporter thyroperoxidase and thyroglobulin. Endocrinology. 2004;145:2865–2875. doi: 10.1210/en.2003-1258. [DOI] [PubMed] [Google Scholar]
  • 69.Puppin C, D’Aurizio F, D’Elia AV, et al. Effects of histone acetylation on sodium iodide symporter promoter and expression of thyroid-specific transcription factors. Endocrinology. 2005;146:3967–3974. doi: 10.1210/en.2005-0128. [DOI] [PubMed] [Google Scholar]
  • 70.Zhang Z, Liu D, Murugan AK, et al. Histone deacetylation of NIS promoter underlies BRAF V600E-promoted NIS silencing in thyroid cancer. Endocr Relat Cancer. 2014;21:161–173. doi: 10.1530/ERC-13-0399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Miyamoto S. Nuclear initiated NF-κB signaling: NEMO and ATM take center stage. Cell Res. 2011;21:116–130. doi: 10.1038/cr.2010.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Pacifico F, Leonardi A. Role of NF-kappaB in thyroid cancer. Mol Cell Endocrinol. 2010;321:29–35. doi: 10.1016/j.mce.2009.10.010. [DOI] [PubMed] [Google Scholar]
  • 73.Meng Z, Lou S, Tan J, et al. Nuclear factor-kappa B inhibition can enhance apoptosis of differentiated thyroid cancer cells induced by 131I. PLoS One. 2012;7:e33597. doi: 10.1371/journal.pone.0033597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Meng Z, Lou S, Tan J, et al. Nuclear factor-kappa B inhibition can enhance therapeutic efficacy of 131I on the in vivo management of differentiated thyroid cancer. Life Sci. 2012;91:1236–1241. doi: 10.1016/j.lfs.2012.09.026. [DOI] [PubMed] [Google Scholar]
  • 75.Gallel P, Pallares J, Dolcet X, et al. Nuclear factor-kappaB activation is associated with somatic and germ line RET mutations in medullary thyroid carcinoma. Hum Pathol. 2008;39:994–1001. doi: 10.1016/j.humpath.2007.11.015. [DOI] [PubMed] [Google Scholar]
  • 76.Neely RJ, Brose MS, Gray CM, et al. The RET/PTC3 oncogene activates classical NF-κB by stabilizing NIK. Oncogene. 2011;30:87–96. doi: 10.1038/onc.2010.396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Liu J, Brown RE. Morphoproteomic confirmation of an activated nuclear factor-кBp65 pathway in follicular thyroid carcinoma. Int J Clin Exp Pathol. 2012;5:216–223. [PMC free article] [PubMed] [Google Scholar]
  • 78.Volpe V, Raia Z, Sanguigno L, et al. NGAL controls the metastatic potential of anaplastic thyroid carcinoma cells. J Clin Endocrinol Metab. 2013;98:228–235. doi: 10.1210/jc.2012-2528. [DOI] [PubMed] [Google Scholar]
  • 79.Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Karin M. Nuclear factor-kappaB in cancer development and progression. Nature. 2006;441:431–436. doi: 10.1038/nature04870. [DOI] [PubMed] [Google Scholar]
  • 81.Kapiteijn E, Schneider TC, Morreau H, et al. New treatment modalities in advanced thyroid cancer. Ann Oncol. 2012;23:10–18. doi: 10.1093/annonc/mdr117. [DOI] [PubMed] [Google Scholar]
  • 82.Kogan EA, Rozhkova EB, Seredin VP, et al. [Prognostic value of the expression of thyreoglobulin and oncomarkers (p53, EGFR, ret-oncogene) in different types of papillary carcinoma of the thyroid: Clinicomorphological and immunohistochemical studies] Arkh Patol. 2006;68:8–11. [PubMed] [Google Scholar]
  • 83.Pennell NA, Daniels GH, Haddad RI, et al. A phase II study of gefitinib in patients with advanced thyroid cancer. Thyroid. 2008;18:317–323. doi: 10.1089/thy.2007.0120. [DOI] [PubMed] [Google Scholar]
  • 84.Rugo HS, Herbst RS, Liu G, et al. Phase I trial of the oral antiangiogenesis agent AG-013736 in patients with advanced solid tumors: Pharmacokinetic and clinical results. J Clin Oncol. 2005;23:5474–5483. doi: 10.1200/JCO.2005.04.192. [DOI] [PubMed] [Google Scholar]
  • 85.Cohen EE, Rosen LS, Vokes EE, et al. Axitinib is an active treatment for all histologic subtypes of advanced thyroid cancer: Results from a phase II study. J Clin Oncol. 2008;26:4708–4713. doi: 10.1200/JCO.2007.15.9566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Salvatore G, De Falco V, Salerno P, et al. BRAF is a therapeutic target in aggressive thyroid carcinoma. Clin Cancer Res. 2006;12:1623–1629. doi: 10.1158/1078-0432.CCR-05-2378. [DOI] [PubMed] [Google Scholar]
  • 87.Ouyang B, Knauf JA, Smith EP, et al. Inhibitors of Raf kinase activity block growth of thyroid cancer cells with RET/PTC or BRAF mutations in vitro and in vivo. Clin Cancer Res. 2006;12:1785–1793. doi: 10.1158/1078-0432.CCR-05-1729. [DOI] [PubMed] [Google Scholar]
  • 88.Kim S, Yazici YD, Calzada G, et al. Sorafenib inhibits the angiogenesis and growth of orthotopic anaplastic thyroid carcinoma xenografts in nude mice. Mol Cancer Ther. 2007;6:1785–1792. doi: 10.1158/1535-7163.MCT-06-0595. [DOI] [PubMed] [Google Scholar]
  • 89.Gupta-Abramson V, Troxel AB, Nellore A, et al. Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol. 2008;26:4714–4719. doi: 10.1200/JCO.2008.16.3279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Brose MS, Nutting C, Jarzab B, et al. Sorafenib in locally advanced or metastatic patients with radioactive iodine-refractory differentiated thyroid cancer: The phase III decision trial. A. J Clin Oncol. 2013;XXX(suppl):31. doi: 10.3978/j.issn.2304-3865.2014.01.02. [DOI] [PubMed] [Google Scholar]
  • 91.Savvides P, Nagaiah G, Lavertu P, et al. Phase II trial of sorafenib in patients with advanced anaplastic carcinoma of the thyroid. Thyroid. 2013;23:600–604. doi: 10.1089/thy.2012.0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.U.S. National Institutes of Health. NCT01013597. http://www.clinicaltrials.gov. Accessed March 7, 2014.
  • 93.Marks P, Rifkind RA, Richon VM, et al. Histone deacetylases and cancer: Causes and therapies. Nat Rev Cancer. 2001;1:194–202. doi: 10.1038/35106079. [DOI] [PubMed] [Google Scholar]
  • 94.Phiel CJ, Zhang F, Huang EY, et al. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276:36734–36741. doi: 10.1074/jbc.M101287200. [DOI] [PubMed] [Google Scholar]
  • 95.Catalano MG, Poli R, Pugliese M, et al. Valproic acid enhances tubulin acetylation and apoptotic activity of paclitaxel on anaplastic thyroid cancer cell lines. Endocr Relat Cancer. 2007;14:839–845. doi: 10.1677/ERC-07-0096. [DOI] [PubMed] [Google Scholar]
  • 96.Catalano MG, Pugliese M, Poli R, et al. Effects of the histone deacetylase inhibitor valproic acid on the sensitivity of anaplastic thyroid cancer cell lines to imatinib. Oncol Rep. 2009;21:515–521. [PubMed] [Google Scholar]
  • 97.Fortunati N, Catalano MG, Arena K, et al. Valproic acid induces the expression of the Na+/I- symporter and iodine uptake in poorly differentiated thyroid cancer cells. J Clin Endocrinol Metab. 2004;89:1006–1009. doi: 10.1210/jc.2003-031407. [DOI] [PubMed] [Google Scholar]
  • 98.Lee WW, Lee B, Kim SJ, et al. Kinetics of iodide uptake and efflux in various human thyroid cancer cells by expressing sodium iodide symporter gene via a recombinant adenovirus. Oncol Rep. 2003;10:845–849. [PubMed] [Google Scholar]
  • 99.Haberkorn U, Beuter P, Kübler W, et al. Iodide kinetics and dosimetry in vivo after transfer of the human sodium iodide symporter gene in rat thyroid carcinoma cells. J Nucl Med. 2004;45:827–833. [PubMed] [Google Scholar]
  • 100.Furuya F, Shimura H, Miyazaki A, et al. Adenovirus-mediated transfer of thyroid transcription factor-1 induces radioiodide organification and retention in thyroid cancer cells. Endocrinology. 2004;145:5397–5405. doi: 10.1210/en.2004-0631. [DOI] [PubMed] [Google Scholar]
  • 101.Elisei R, Vivaldi A, Ciampi R, et al. Treatment with drugs able to reduce iodine efflux significantly increases the intracellular retention time in thyroid cancer cells stably transfected with sodium iodide symporter complementary deoxyribonucleic acid. J Clin Endocrinol Metab. 2006;91:2389–2395. doi: 10.1210/jc.2005-2480. [DOI] [PubMed] [Google Scholar]
  • 102.Petrich T, Helmeke HJ, Meyer GJ, et al. Establishment of radioactive astatine and iodine uptake in cancer cell lines expressing the human sodium/iodide symporter. Eur J Nucl Med Mol Imaging. 2002;29:842–854. doi: 10.1007/s00259-002-0784-7. [DOI] [PubMed] [Google Scholar]
  • 103.Barzon L, Bonaguro R, Castagliuolo I, et al. Gene therapy of thyroid cancer via retrovirally-driven combined expression of human interleukin-2 and herpes simplex virus thymidine kinase. Eur J Endocrinol. 2003;148:73–80. doi: 10.1530/eje.0.1480073. [DOI] [PubMed] [Google Scholar]
  • 104.Barzon L, Gnatta E, Castagliuolo I, et al. Modulation of retrovirally driven therapeutic genes by mutant TP53 in anaplastic thyroid carcinoma. Cancer Gene Ther. 2005;12:381–388. doi: 10.1038/sj.cgt.7700789. [DOI] [PubMed] [Google Scholar]
  • 105.Lee YJ, Chung JK, Kang JH, et al. 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;37:235–241. doi: 10.1007/s00259-009-1251-5. [DOI] [PubMed] [Google Scholar]
  • 106.Moretti F, Farsetti A, Soddu S, et al. p53 re-expression inhibits proliferation and restores differentiation of human thyroid anaplastic carcinoma cells. Oncogene. 1997;14:729–740. doi: 10.1038/sj.onc.1200887. [DOI] [PubMed] [Google Scholar]
  • 107.Yu XM, Phan T, Patel PN, et al. Chrysin activates Notch1 signaling and suppresses tumor growth of anaplastic thyroid carcinoma in vitro and in vivo. Cancer. 2013;119:774–781. doi: 10.1002/cncr.27742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kang HJ, Youn YK, Hong MK, et al. Antiproliferation and redifferentiation in thyroid cancer cell lines by polyphenol phytochemicals. J Korean Med Sci. 2011;26:893–899. doi: 10.3346/jkms.2011.26.7.893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Richardson PG, Mitsiades C, Hideshima T, et al. Bortezomib: Proteasome inhibition as an effective anticancer therapy. Annu Rev Med. 2006;57:33–47. doi: 10.1146/annurev.med.57.042905.122625. [DOI] [PubMed] [Google Scholar]
  • 110.Hideshima T, Mitsiades C, Tonon G, et al. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 2007;7:585–598. doi: 10.1038/nrc2189. [DOI] [PubMed] [Google Scholar]
  • 111.Putzer D, Gabriel M, Kroiss A, et al. First experience with proteasome inhibitor treatment of radioiodine nonavid thyroid cancer using bortezomib. Clin Nucl Med. 2012;37:539–544. doi: 10.1097/RLU.0b013e31824c5f24. [DOI] [PubMed] [Google Scholar]
  • 112.Mitsiades CS, McMillin D, Kotoula V, et al. Antitumor effects of the proteasome inhibitor bortezomib in medullary and anaplastic thyroid carcinoma cells in vitro. J Clin Endocrinol Metab. 2006;91:4013–4021. doi: 10.1210/jc.2005-2472. [DOI] [PubMed] [Google Scholar]
  • 113.Starenki DV, Namba H, Saenko VA, et al. Induction of thyroid cancer cell apoptosis by a novel nuclear factor kappaB inhibitor, dehydroxymethylepoxyquinomicin. Clin Cancer Res. 2004;10:6821–6829. doi: 10.1158/1078-0432.CCR-04-0463. [DOI] [PubMed] [Google Scholar]
  • 114.Starenki D, Namba H, Saenko V, et al. Inhibition of nuclear factor-kappaB cascade potentiates the effect of a combination treatment of anaplastic thyroid cancer cells. J Clin Endocrinol Metab. 2004;89:410–418. doi: 10.1210/jc.2003-031216. [DOI] [PubMed] [Google Scholar]
  • 115.Pacifico F, Mauro C, Barone C, et al. Oncogenic and anti-apoptotic activity of NF-kappa B in human thyroid carcinomas. J Biol Chem. 2004;279:54610–54619. doi: 10.1074/jbc.M403492200. [DOI] [PubMed] [Google Scholar]
  • 116.Iannetti A, Pacifico F, Acquaviva R, et al. The neutrophil gelatinase-associated lipocalin (NGAL), a NF-kappaB-regulated gene, is a survival factor for thyroid neoplastic cells. Proc Natl Acad Sci USA. 2008;105:14058–14063. doi: 10.1073/pnas.0710846105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Bauerle KT, Schweppe RE, Haugen BR. Inhibition of nuclear factor-kappa B differentially affects thyroid cancer cell growth, apoptosis, and invasion. Mol Cancer. 2010;9:117. doi: 10.1186/1476-4598-9-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Cras A, Politis B, Balitrand N, et al. Bexarotene via CBP/p300 induces suppression of NF-κB-dependent cell growth and invasion in thyroid cancer. Clin Cancer Res. 2012;18:442–453. doi: 10.1158/1078-0432.CCR-11-0510. [DOI] [PubMed] [Google Scholar]
  • 119.Rosato RR, Kolla SS, Hock SK, et al. Histone deacetylase inhibitors activate NF-kappaB in human leukemia cells through an ATM/NEMO-related pathway. J Biol Chem. 2010;285:10064–10077. doi: 10.1074/jbc.M109.095208. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 120.Grommes C, Landreth GE, Heneka MT. Antineoplastic effects of peroxisome proliferator-activated receptor gamma agonists. Lancet Oncol. 2004;5:419–429. doi: 10.1016/S1470-2045(04)01509-8. [DOI] [PubMed] [Google Scholar]
  • 121.Park JW, Zarnegar R, Kanauchi H, et al. Troglitazone, the peroxisome proliferator-activated receptor-gamma agonist, induces antiproliferation and redifferentiation in human thyroid cancer cell lines. Thyroid. 2005;15:222–231. doi: 10.1089/thy.2005.15.222. [DOI] [PubMed] [Google Scholar]
  • 122.Fröhlich E, Machicao F, Wahl R. Action of thiazolidinediones on differentiation, proliferation and apoptosis of normal and transformed thyrocytes in culture. Endocr Relat Cancer. 2005;12:291–303. doi: 10.1677/erc.1.00973. [DOI] [PubMed] [Google Scholar]
  • 123.Antonelli A, Ferrari SM, Fallahi P, et al. Thiazolidinediones and antiblastics in primary human anaplastic thyroid cancer cells. Clin Endocrinol (Oxf) 2009;70:946–953. doi: 10.1111/j.1365-2265.2008.03415.x. [DOI] [PubMed] [Google Scholar]
  • 124.Shepherd J, Cobbe SM, Ford I, et al. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. N Engl J Med. 1995;333:1301–1307. doi: 10.1056/NEJM199511163332001. [DOI] [PubMed] [Google Scholar]
  • 125.Davidson MH. Safety profiles for the HMG-CoA reductase inhibitors: Treatment and trust. Drugs. 2001;61:197–206. doi: 10.2165/00003495-200161020-00005. [DOI] [PubMed] [Google Scholar]
  • 126.Eisenberg DA. Cholesterol lowering in the management of coronary artery disease: The clinical implications of recent trials. Am J Med. 1998;104:2S–5S. doi: 10.1016/s0002-9343(98)00038-2. [DOI] [PubMed] [Google Scholar]
  • 127.Sacks FM, Pfeffer MA, Moye LA, et al. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events Trial investigators. N Engl J Med. 1996;335:1001–1009. doi: 10.1056/NEJM199610033351401. [DOI] [PubMed] [Google Scholar]
  • 128.Zhong WB, Wang CY, Chang TC, et al. Lovastatin induces apoptosis of anaplastic thyroid cancer cells via inhibition of protein geranylgeranylation and de novo protein synthesis. Endocrinology. 2003;144:3852–3859. doi: 10.1210/en.2003-0098. [DOI] [PubMed] [Google Scholar]
  • 129.Di Matola T, D’Ascoli F, Luongo C, et al. Lovastatin-induced apoptosis in thyroid cells: Involvement of cytochrome c and lamin B. Eur J Endocrinol. 2001;145:645–650. doi: 10.1530/eje.0.1450645. [DOI] [PubMed] [Google Scholar]
  • 130.Wang CY, Zhong WB, Chang TC, et al. Lovastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, induces apoptosis and differentiation in human anaplastic thyroid carcinoma cells. J Clin Endocrinol Metab. 2003;88:3021–3026. doi: 10.1210/jc.2002-021834. [DOI] [PubMed] [Google Scholar]
  • 131.Zhong WB, Liang YC, Wang CY, et al. Lovastatin suppresses invasiveness of anaplastic thyroid cancer cells by inhibiting Rho geranylgeranylation and RhoA/ROCK signaling. Endocr Relat Cancer. 2005;12:615–629. doi: 10.1677/erc.1.01012. [DOI] [PubMed] [Google Scholar]
  • 132.Wang CY, Shui HA, Chang TC. In vivo evidence of duality effects for lovastatin in a nude mouse cancer model. Int J Cancer. 2010;126:578–582. doi: 10.1002/ijc.24760. [DOI] [PubMed] [Google Scholar]
  • 133.Bifulco M, Laezza C, Aloj SM. Inhibition of farnesylation blocks growth but not differentiation in FRTL-5 thyroid cells. Biochimie. 1999;81:287–290. doi: 10.1016/s0300-9084(99)80072-8. [DOI] [PubMed] [Google Scholar]
  • 134.Takano T. Fetal cell carcinogenesis of the thyroid: Theory and practice. Semin Cancer Biol. 2007;17:233–240. doi: 10.1016/j.semcancer.2006.02.001. [DOI] [PubMed] [Google Scholar]
  • 135.Todaro M, Iovino F, Eterno V, et al. Tumorigenic and metastatic activity of human thyroid cancer stem cells. Cancer Res. 2010;70:8874–8885. doi: 10.1158/0008-5472.CAN-10-1994. [DOI] [PubMed] [Google Scholar]
  • 136.Hardin H, Montemayor-Garcia C, Lloyd RV. Thyroid cancer stem-like cells and epithelial-mesenchymal transition in thyroid cancers. Hum Pathol. 2013;44:1707–1713. doi: 10.1016/j.humpath.2013.01.009. [DOI] [PubMed] [Google Scholar]
  • 137.Lloyd RV, Hardin H, Montemayor-Garcia C, et al. Stem cells and cancer stem-like cells in endocrine tissues. Endocr Pathol. 2013;24:1–10. doi: 10.1007/s12022-013-9235-1. [DOI] [PubMed] [Google Scholar]
  • 138.Buehler D, Hardin H, Shan W, et al. Expression of epithelial-mesenchymal transition regulators SNAI2 and TWIST1 in thyroid carcinomas. Mod Pathol. 2013;26:54–61. doi: 10.1038/modpathol.2012.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Takano T. Fetal cell carcinogenesis of the thyroid: A modified theory based on recent evidence. Endocr J. 2014;61:311–320. doi: 10.1507/endocrj.ej13-0517. [DOI] [PubMed] [Google Scholar]
  • 140.Montemayor-Garcia C, Hardin H, Guo Z, et al. The role of epithelial mesenchymal transition markers in thyroid carcinoma progression. Endocr Pathol. 2013;24:206–212. doi: 10.1007/s12022-013-9272-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: Harnessing the T cell response. Nat Rev Immunol. 2012;12:269–281. doi: 10.1038/nri3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Paparodis R, Imam S, Todorova-Koteva K, et al. Hashimoto’s thyroiditis pathology and risk for thyroid cancer. Thyroid. 2014;24:1107–1114. doi: 10.1089/thy.2013.0588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Imam S, Paparodis R, Sharma D, et al. Lymphocytic profiling in thyroid cancer provides clues for failure of tumor immunity. Endocr Relat Cancer. 2014;21:505–516. doi: 10.1530/ERC-13-0436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Foote RL, Molina JR, Kasperbauer JL, et al. Enhanced survival in locoregionally confined anaplastic thyroid carcinoma: A single-institution experience using aggressive multimodal therapy. Thyroid. 2011;21:25–30. doi: 10.1089/thy.2010.0220. [DOI] [PubMed] [Google Scholar]

Articles from The Oncologist are provided here courtesy of Oxford University Press

RESOURCES