Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer

Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer

Accepted Manuscript Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer Thanyawat Sasanakietkul, Timothy D. Murtha, Mahsa ...

942KB Sizes 6 Downloads 115 Views

Accepted Manuscript Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer Thanyawat Sasanakietkul, Timothy D. Murtha, Mahsa Javid, Reju Korah, Tobias Carling PII:

S0303-7207(17)30291-5

DOI:

10.1016/j.mce.2017.05.022

Reference:

MCE 9953

To appear in:

Molecular and Cellular Endocrinology

Received Date: 16 March 2017 Revised Date:

12 May 2017

Accepted Date: 21 May 2017

Please cite this article as: Sasanakietkul, T., Murtha, T.D., Javid, M., Korah, R., Carling, T., Epigenetic modifications in poorly differentiated and anaplastic thyroid cancer, Molecular and Cellular Endocrinology (2017), doi: 10.1016/j.mce.2017.05.022. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Review Article Epigenetic Modifications in Poorly Differentiated and Anaplastic

RI PT

Thyroid Cancer

Thanyawat Sasanakietkul1,2, Timothy D. Murtha1,2, Mahsa Javid1,2, Reju Korah1,2, Tobias Carling1,2*

Yale Endocrine Neoplasia Laboratory, 2Department of Surgery, Section of Endocrine Surgery,

TE D

M AN U

Yale School of Medicine, New Haven, CT 06520, USA

SC

1

*To whom correspondence should be addressed

EP

Tobias Carling, MD, PhD, FACS Section of Endocrine Surgery

AC C

Yale Endocrine Neoplasia Laboratory Department of Surgery

Yale University School of Medicine 333 Cedar Street, FMB130A New Haven, CT 06520

+1-203-737-2036 (phone) +1-203 737-4067 (fax) E-Mail: [email protected]

ACCEPTED MANUSCRIPT

ABSTRACT Well-differentiated thyroid cancer accounts for the majority of endocrine malignancies and, in general, has an excellent prognosis. In contrast, the less common poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid carcinoma (ATC) are two of the most

RI PT

aggressive human malignancies. Recently, there has been an increased focus on the epigenetic alterations underlying thyroid carcinogenesis, including those that drive PDTC and ATC. Dysregulated epigenetic candidates identified include the Aurora group, KMT2D, PTEN,

RASSF1A, multiple non-coding RNAs (ncRNA), and the SWI/SNF chromatin-remodeling

SC

complex. A deeper understanding of the signaling pathways affected by epigenetic dysregulation may improve prognostic testing and support the advancement of thyroid-specific epigenetic

M AN U

therapies. This review outlines the current understanding of epigenetic alterations observed in PDTC and ATC and explores the potential for exploiting this understanding in developing novel therapeutic strategies.

Keywords: Epigenetics, Poorly differentiated thyroid carcinoma, Anaplastic thyroid carcinoma,

1. Introduction

TE D

MicroRNA, Non-coding RNA

Thyroid cancer is the most common endocrine malignancy and has had a consistently increasing incidence for the past four decades (1-3). The vast majority (> 95%) of thyroid

EP

cancers are derived from follicular epithelial cells while the remainder arises from parafollicular cells (3). Follicular cell-based thyroid cancer is divided into three major categories: well-

AC C

differentiated thyroid carcinoma (WDTC), poorly differentiated thyroid carcinoma (PDTC), and undifferentiated or anaplastic thyroid carcinoma (ATC). WDTC includes follicular thyroid carcinoma (FTC) and papillary thyroid carcinoma (PTC). Among the major thyroid cancer types, ATC is the most rare and aggressive variant (4). Because of its high proliferative index and invasive behavior, the disease-specific mortality of ATC is 69.4% at 6 months and 80.7% at 12 months (5). The dismal prognosis of ATC is partially due to its inherent resistance to both radioactive iodine and conventional chemotherapy. As the tumor cells of PDTC and ATC lack appreciable expression of the sodium-iodide symporter (NIS), they are incapable of iodine uptake (6-8). Moreover, ATC cells may not secrete thyroglobulin and are potentially refractory

ACCEPTED MANUSCRIPT

to thyroid stimulating hormone (TSH) because of thyrotropin receptor deficiency on their plasma cell membrane. The combination of these tumor characteristics significantly limits the efficacy

2. Poorly differentiated and anaplastic thyroid carcinoma

RI PT

of conventional radioactive iodine therapy (6, 7).

Since Sakamoto et al. (1983) first described PDTC, there has been contention regarding its characterization and diagnosis (9). Currently, several distinct criteria for diagnosing PDTC are in use. The 2004 World Health Organization Classification of Tumors describes PDTC as,

SC

“…follicular-cell neoplasms that show limited evidence of structural follicular cell

differentiation and occupy both morphologically and behaviorally an intermediate position

M AN U

between differentiated (follicular and papillary) and undifferentiated (anaplastic) carcinomas.” (10). The histologic features described are in part due to the dedifferentiation typically observed in PDTC.

Alternatively, the Turin proposal described PDTC as having, “(1) a solid/trabecular/insular pattern of growth, (2) absence of conventional nuclear features of PTC, and (3) presence of at least one of the following features: convoluted nuclei, mitotic activity

TE D

(≥3×10 HPF), necrosis.” (11). Similarly, a Memorial Sloan-Kettering Cancer Center (MSKCC) study defined PDTC based on, “…the presence of ≥ 5 mitosis per 10 high-power microscopic fields (HPF)(x400) and/or fresh tumor necrosis in thyroid carcinomas demonstrating definite evidence of follicular cell differentiation on routine microscopy and/or by immunohistochemistry

EP

(i.e., thyroglobulin positivity).” (12). These definitions highlight PDTC’s association with necrosis and high mitotic indices, consistent with their aggressive phenotype. It is important to

AC C

note that the MSKCC diagnostic criteria distinguish PDTC from other thyroid cancer subtypes including ATC (Table 1), irrespective of tumor growth patterns and nuclear features. PDTC accounts for approximately 0.5–7% of all thyroid cancers and has a mean age at

diagnosis of 60.6 years (13). ATC accounts for less than 1% of thyroid cancers and is predominately found in elderly patients with a mean age of 71 years (5). ATC also has a distinct female preponderance (60–70%) (5). On gross examination, ATCs are typically large, necrotic, and exhibit widespread hemorrhagic changes (14). Individual tumor morphology is influenced by the unique combination of spindle, giant, and squamoid cells (14). High mitotic rates and atypical mitoses are generally present, and necrosis is often observed in ATCs (Figure 1). On

ACCEPTED MANUSCRIPT

immunohistochemical analysis, the majority of ATCs are negative for characteristic transcription factors, such as thyroid transcription factor-1 (TTF-1), and thyroglobulin (14, 15). In contrast, PDTC is often positive for thyroglobulin and may display nuclear positivity for TTF-1 (13). In the diagnosis of equivocal cases, these immunohistochemistry features aid in differentiating

RI PT

between PDTC and ATC. However, molecular profiling using next generation genetic/epigenetic analysis could improve the process of establishing a definitive diagnostic distinction between these aggressive tumors. PDTC exhibits the intermediate aggressive clinical characteristics

between WDTC and ATC, such as tumor growth rate, lymph node metastasis (WDTC = 47%,

SC

PDTC = 64%, ATC = 100%), distant metastatic (WDTC = 0%, PDTC = 50%, ATC = 87%) 5year disease free survival (WDTC = 91%, PDTC = 51%, ATC = 0%) and overall survival in 43

M AN U

months (WDTC = 100%, PDTC = 70%, ATC = 0%) (16, 17). Currently, treatment for both PDTC and ATC are still challenging. Surgery (total thyroidectomy and cervical lymph node dissection) is the principle therapeutic option. Although PDTC and ATC are very aggressive subtypes of thyroid cancer, a unified single therapy usually will not achieve the desired curative goals to these thyroid cancers that run distinct clinical courses. The multimodality treatments (radioactive iodine ablation, external beam radiation, chemotherapy, and targeted therapy) have

17).

TE D

been in use to control the extent of disease but the outcomes remain unsatisfactory (8, 9, 12, 16,

AC C

EP

Figure 1. Histopathologic pictures (H&E) of (A) PDTC and (B) ATC

Table 1. Characteristics of PDTC and ATC

ACCEPTED MANUSCRIPT

Disease Characteristics

Poorly differentiated thyroid carcinoma

Anaplastic thyroid carcinoma

Proportion of all thyroid

0.5-7%

< 1%

Mean age at diagnosis

60 years

71 years

Histopathology

Follicular cell origin

Follicular cell origin

- Limited evidence of structural follicular cell

- Severe nuclear atypia

differentiation a

- Numerous mitotic figures, atypical mitoses, extensive necrosis and, often mixed morphology

- Morphologically and behaviorally intermediate between differentiated and undifferentiated carcinoma b

with spindle cells and giant cells. - Three patterns:

- Absence of conventional nuclear features of PTC

b

- Presence of at least one of the following: convoluted b

1. Spindle cells resembling sarcoma 2. Large, pleomorphic giant cells resembling osteoclasts with cellular connective tissue septa

M AN U

nuclei, mitotic activity (≥3×10 HPF), or necrosis

a

SC

- A solid/trabecular/insular pattern of growth

RI PT

cancers

- Presence of ≥ 5/10 (HPF)(x400) and/or fresh tumor

3. Squamoid pattern

necrosis in demonstrating definite evidence of follicular cell differentiation on routine microscopy and/or by immunohistochemistry c

Positive: Cytokeratins (CK7, CK18, CK 19), TTF-1,

Positive: Cytokeratins (CK7, CK18, CK 19, CK10/13),

markers

Thyroglobulin, Ki-67, Cyclin D1, IMP3

Ki-67, p53, Vimentin, Cyclin D1, EMA

Negative: Calcitonin, Chromogranin, CEA,

Negative: Calcitonin, Chromogranin, TTF-1,

Synaptophysin

RET/PTC oncoprotein, Thyroglobulin, Synaptophysin

TE D

Immunohistochemical

a. World Health Organization classification; b. Turin conference; c. Memorial Sloan-Kettering Cancer Center

3. PDTC and ATC’ alterated pathways and genetic alterations

EP

3.1 Dysregulated signaling pathways in PDTC and ATC WDTCs commonly exhibit dysregulated activity of the mitogen-activated protein kinase

AC C

(MAPK) pathway, the phosphoinositide 3-kinase (PI3K) signaling pathway, or a combination of the two (18-20). These alterations occur through direct genomic/epigenomic mutations or indirectly via epigenetic deregulation (21-23). Although the molecular basis for disease progression from WDTC to PDTC and/or ATC is debatable, the overlap of dysregulated signaling pathways among these entities may support a continuum of PDTC and ATC from WDTC (18, 24-26). Another commonality between WDTC and PDTC/ATC is the observation that the dysfunctions identified in the ERK1/2-MEK1/2 and PI3K-Akt signaling pathways are due to common genetic alterations such as BRAFV600E (18, 19, 27). Similarly, mutations in NRAS and HRAS, which have the potential to activate the ERK1/2-MEK1/2 and the PI3K-Akt

ACCEPTED MANUSCRIPT

pathway, are often found in both FTC and ATC (18, 24, 28-30). Furthermore, PTEN silencing, found in both PTC and ATC, may result in constitutive activation of PI3K-Akt anti-apoptotic pathways (18, 19, 24, 27, 31). Despite the scarcity of studies focusing on signaling disarray in PDTC, the available data

RI PT

suggest a clear continuum of constitutively active MAPK signaling—potentially through the disease progression route or de novo activation—in PDTC (26, 32). In addition to the commonly dysregulated RAS-RAF-MAPK pathways, PDTC and ATC were also found to harbor aberrant PIK3CA-PTEN-Akt-mTOR pathways (24). Alternatively, in undifferentiated ATC, there is

SC

evidence of deregulated WNT and NOTCH pathways, in addition to the constitutively active MAPK and PI3K pathways, potentially contributing to their highly aggressive behavior (26, 33-

M AN U

37).

Both PDTC and ATC also exhibit deregulated expression of a variety of genes involved in cell cycle regulation, focal adhesion signaling, actin cytoskeleton dynamics, and the TGF-β signaling pathways (38). Moreover, the interruption of the thyroid-stimulating hormone receptor (TSHR) pathway and the consequent dedifferentiation signaling in PDTC and ATC has been found mediated by the silencing of hematopoietically-expressed homeobox (HHEX) protein (38).

TE D

Similarly, nuclear factor kappa B (NF- B) activation has been observed in the majority of ATCs. In vitro inhibition of NF- B in ATC cell lines appears to block cellular proliferation and invasive behavior (39, 40) suggesting its potential contribution to the invasive phenotype of ATC. Genetic disruption of the isocitrate dehydrogenase (IDH1) pathway through mutation of IDH1 has been

EP

shown to be associated with a variety of aggressive tumors including ATC (41-43).

AC C

3.2 Genetic alterations in PDTC and ATC The major genetic alterations described in PDTC and ATC include frequent somatic

mutations in BRAF and the RAS family, as well as additional recurrent mutations in TP53, CTNNB1, PI3KCA, ALK, PTEN, and TERT promoter (14, 25, 30, 37, 44-48). The BRAFV600E mutation, found in > 70% of well-differentiated PTCs, is also found in 33% of PDTCs and 1945% of ATCs. Presence of the BRAFV600E mutation in PDTC and ATC is associated with worse prognosis, more aggressive phenotype, a higher rate of recurrence, and an increased likelihood of treatment resistance (37, 48-50). Interestingly, mutations in the ubiquitous tumor suppressor gene TP53 are found in only 8% of PDTC and a variable number of ATCs (29–73%) (37, 48, 51, 52).

ACCEPTED MANUSCRIPT

Activating mutations of CTNNB1, found in 65% of ATCs, may stimulate the WNT-β-catenin signaling pathway (33, 34). Similarly, mutational activation of the p110α catalytic subunit of PI3K is reported in 18–23% of ATCs. Both WNT and PI3K signaling activation has been shown to have the potential to manipulate cancer cell cycle progression and cell motility (37, 53). Other

RI PT

somatic gene mutations reported in PDTC, include anaplastic lymphoma kinase (ALK; 4%), PTEN (4%), NRAS (21%), HRAS (5%), and KRAS (2%), while mutations in ATC include those in ALK gene (0–11%), PTEN (10–20%), NRAS (17–18%) and HRAS (5–6%) (28, 37, 46, 48, 51, 52). The promoter mutations in human telomerase reverse transcriptase (hTERT) have been

SC

recently reported in ATC as well (54). Of the two common TERT promoter mutations (C228T and C250T), 33–50% of hTERT promoter mutations in PDTC and ATC occur in C228T. This

M AN U

mutation is believed to increase telomerase activity, possibly contributing to the aggressive behavior of PDTC and ATC (54). The first comprehensive whole-exome sequencing study of ATC revealed that its genetic landscape is highly heterogeneous. Somatic mutations were detected in 64% of ATC cases which included multiple novel mutations including those in mTOR, NF1, NF2, ERBB2, EIF1AX, MLH1, MLH3, MSH5, MSH6 and USH2A (48). Other genomic alterations that may influence PDTC and ATC tumorigenesis include

TE D

copy number variations (CNV), gene fusions, and chromosomal translocations (Table 2). The receptor tyrosine kinase group of genes, composed of EGFR, PDGFRα, PDGFRβ, PIK3Ca, PIK3Cb, PDK1 VEGFR1, VEGFR2, KIT, and MET, has been reported to have substantial copy number gains in ATC, implicating its potential role in promoting ATC tumorigenesis (18). Large

EP

chromosomal gains involving 7p, 8q, 9q, 3p13-14, and 11q13 were found in ATC, while chromosomal losses involving 5q11-31 were frequently observed (17, 55, 56). Gene fusions,

AC C

such as RET-PTC and STRN/ALK (9% of PDTCs and 4% of ATCs), are found in a small proportion of PDTCs and ATCs and may also contribute to their aggressive behaviors (30, 57, 58).

Table 2. Major Signaling and Mutational landscapes of PDTC and ATC Poorly differentiated thyroid cancer

Anaplastic thyroid cancer

ACCEPTED MANUSCRIPT

MAPK



MAPK



PI3K-Akt



PI3K-Akt



PI3K/Akt/mTOR inactivation



PI3K/Akt/mTOR inactivation



WNT-β-catenin



WNT-β-catenin



TSHR



NOTCH*



p53



TSHR



NF B*



p53



IDH1*

RI PT

Gene alterations



BRAF, RAS, TP53, CTNNB1, PIK3CA, PTEN,

BRAF, RAS, TP53, CTNNB1, PI3KCA, PTEN, AKT1, TERT,

AKT1, TERT, TSHR, ALK, STRN/ALK

TSHR, ALK, mTOR*, NF1*, NF2*, MLH1*, MLH3*,

SC

Signaling pathways

MSH5*, MSH6*, ERBB2*, EIF1AX*, USH2A*, RTK*, IDH1*, STRN/ALK, RET-PTC*

M AN U

*Identified only in ATC, not in PDTC

4. Epigenetic mechanisms and classifications in cancer

Tightly controlled epigenetic regulation is essential for normal growth, development, acquisition, and maintenance of unique functions of organs and organ systems. Deregulation of epigenetic controls, on the other hand, may play crucial roles in the origin and/or progression of

TE D

various disease processes including cancer. Cancer-specific, as well as global epigenetic aberrations, are reported in a variety of malignancies including thyroid cancers (59). The four major epigenetic mechanisms that are found deregulated during various stages of carcinogenesis are: (1) DNA methylation (the tumor suppressor genes or immune response genes are aberrantly

EP

silenced by DNA methylation in their promoter regions), (2) histone modification (a posttranslational modification of histone proteins that regulates gene expression which includes

AC C

methylation, phosphorylation, acetylation, ubiquitylation, and sumoylation), (3) chromatin remodeling (the chromatin architectural rearrangement or modification that allow transcription factors to have atypical access to the condensed genomic DNA resulting in aberrant control of gene expression), and (4) deregulation of ncRNAs which constitutes small ncRNAs (microRNAs, Piwi-interacting RNA, transcription initiation RNA, tRNA-derived small RNA, etc.) and long ncRNAs (lncRNAs) (60-62). MicroRNAs (miRNAs) can impair RNA translation by hybridizing to specific domains of the untranslated region of target mRNAs (63, 64) while lncRNAs can play a role in tumorigenesis by aberrantly controlling the transcription and/or posttranscriptional regulation of gene expression (65).

ACCEPTED MANUSCRIPT

Based on their specific roles, epigenetic deregulators can be classified into: (a) epigenetic modifiers, (b) mediators, and (c) modulators (66). Epigenetic modifiers are genes that abrogate gene expression control designed for structural and/or functional differentiation of the cells through DNA methylation, histone modification, or chromatin structure alterations. Epigenetic

RI PT

mediators are genes and their products that can target epigenetic modifiers and mediate the dedifferentiation process of tumor cells or their progenitors towards a stem cell-like pre-

differentiated status. Epigenetic modulators are genes and effectors that can influence the

epigenetic modifiers and epigenetic mediators in executing their respective signaling pathways.

SC

Environmental factors, such as injury, inflammation, or other forms of stress, are examples of

M AN U

epigenetic modulators that can push tissues to acquire cancer-promoting alterations (66).

5. Epigenetic modifications in thyroid cancer

Aberrant DNA methylation leading to the activation of proto-oncogenes or silencing of tumor suppressor genes, is common in thyroid tumors. Tumor suppressor genes that are epigenetically inactivated in thyroid cancers include DAPK, PTEN, RASSF1A, RAPβ2, RAP1GAP, SLC5A8, and TIMP3 (23). Aberrant promotor methylation of TIMP3, SLC5A8,

TE D

DAPK, and RARb2 also overlap with BRAF mutations in PTC and may contribute to the tumor behavior (67, 68). The PTEN promoter hypermethylation rate in PTC and FTC are 50% and 100% respectively, suggesting their role in abrogating the PI3K-Akt pathway and leading to the development of thyroid tumors (31). RAS association domain family 1A (RASSF1A) promotor

EP

hypermethylation is seen in approximately 30% of thyroid cancers and may alter the RASSF1MST1-FOXO3 apoptosis pathway (30, 69, 70). In PTC, promoter hypermethylation of RASSF1A

AC C

is significantly correlated with tumor multifocality, while RASSF1A hypomethylation appears to be compatible with extracapsular extension (71). The tissue inhibitor of metalloproteinase 3 (TIMP3) gene, silenced by promoter hypermethylation in a large number of PTCs, results in the promotion of tumor aggressiveness, including lymph node metastasis and tumor multifocality (21, 67). Similarly, downregulation of the death-associated protein kinase (DAPK), a calcium/calmodulin-dependent serine-threonine kinase which controls cell death, has been observed in 34% of PTCs, potentially increasing cell survival and invasive behavior (21, 67). Although not as extensively studied as promoter methylation, the potential contributions of histone modifications toward thyroid carcinogenesis, are gaining attention (22). Based on the

ACCEPTED MANUSCRIPT

reported efficacy of histone deacetylase (HDAC) inhibitors in other cancer types, recent exploratory studies have demonstrated significant therapeutic potential of HDAC inhibitors in the treatment of thyroid cancers as well (72-75). HDAC inhibitors partially restored radioactive iodine uptake and, combined with chemotherapy, radiation therapy, and surgery, showed

RI PT

improvement in the curative rate of ATC and increased ATC cell death in vivo and in vitro (73, 76-79).

Several recent studies suggest an association between deregulated miRNA expression and thyroid cancers. Upregulation of the miR221-222 cluster, located on the X chromosome, has

SC

been implicated to regulate cell cycle progression and apoptosis in differentiated thyroid cancers (80, 81). MicroRNA miR-146 is involved in multiple cancer types and has been found to be

M AN U

overexpressed in all follicular cell-derived cancers (80). Other miRNAs that are commonly overexpressed in PTC include miR-21 and miR-181 (23, 82-84). In FTC, miR-192, miR-197, miR-328, and miR-346 expression have been reported to be downregulated when compared to follicular adenoma (85). These dysregulated miRNAs may contribute individually or in combination with other genetic or epigenetic events to promote thyroid tumorigenesis (64, 86, 87).

TE D

The important roles played by lncRNAs in the origin and progression of various tumor types is slowly being unraveled (88-92). Three recent studies strongly suggest a role for various lncRNAs in thyroid carcinogenesis (93-95). Zheng et al. (2016) used quantitative reverse transcription polymerase chain reaction to demonstrate differential expression of three lncRNAs.

EP

Namely, (a) BRAF-activated lncRNA (BANCR), (b) PTC susceptibility candidate 3 (PTCSC3), and (c) lncRNA associated with MAPK pathway and cell cycle arrest (NAMA) in PTC (94).

AC C

BANCR, which exhibited the potential to regulate PTC proliferation through the cyclin D1 and TSHR pathways, showed upregulated expression while both of PTCSC3 and NAMA displayed downregulated expression in PTC (94). Downregulation of lncRNA NONHSAT037832 has been observed in PTC cell lines and was found to correlate with aggressive tumor characteristics in PTC, such as larger tumor size or lymph node metastasis (93). Plasmacytoma variant translocation 1 (PVT1), a recent addition to lncRNAs, was found upregulated in PTC, FTC, and ATC. Intriguingly, the same study implicated PVT1 as a promotor of thyroid cancer cell growth via EZH2 and TSHR (95). In addition to WDTCs, recent findings also clearly suggest important roles for epigenetic regulation in the initiation, promotion, and progression of PDTC and ATC

ACCEPTED MANUSCRIPT

(21-23, 96-100). The discussion below focuses on reviewing potential roles of various epigenetic candidates and mechanisms in promoting de novo dedifferentiation of thyroid follicular cells into PDTC’s and ATC’s, as well as continued dedifferentiation of WDTCs to PDTC’s and ATC’s.

RI PT

6. Characterization of PDTC and ATC epigenomes

Recent comprehensive genetic and epigenetic analyses of PTC and FTC helped to

generate a better understanding of their respective epigenomes (20-23, 30, 48, 49, 59, 66, 96-99, 101). Although next-generation genetic analysis of ATC revealed aberrations in multiple genes

SC

that could compromise the integrity of thyroid epigenome (46), a clear understanding of a defined and distinctive ATC epigenome is still missing. As a new entrant into the field of

M AN U

genetic/epigenetic analysis, epigenome information on PDTCs is also lacking. The PDTC/ATC epigenome analysis outlined below focuses on alterations identified in epigenetic candidates and mechanisms in comparison to normal thyroid follicle cells and other follicular cell-derived neoplasias including PTC and FTC. Alterations reported to have dominant roles in defining PDTC and ATC epigenomes are grouped into deregulated (a) genes, (b) miRNAs, and (c) lncRNAs (Table 3). The discussed alterations in this review—individually or in tandem—are

TE D

suggested to promote dedifferentiation de novo or to accentuate existing neoplasia-associated dedifferentiation depending on the context of the study reported (Figure 1). Dysregulated gene expressions associated with epigenetic alterations in PDTCs and ATCs are further grouped into (1) epigenetic modifiers, (2) epigenetic mediators, and (3) epigenetic modulators based on their

AC C

EP

implicated roles in the overall dedifferentiation process (66).

Table 3. Epigenetic alterations in PDTC and ATC Epigenetic modifiers

Poorly differentiated thyroid cancer

Anaplastic thyroid cancer

Histone modification: HMTs (KMT2A,



DNA methylation: MECP2

KMT2C, KMT2D, SETD2)



Histone modification: Aurora group, EZH2, HDAC1 & 2, HMTs (KMT2A, KMT2C, KMT2D, SETD2)



Chromatin remodeling: SWI/SNF complex, EP300

Epigenetic mediators

Maspin

RASSF1A, RASSF2, REC8, TTF-1, Maspin, MAP17

Epigenetic modulators

-

PTEN, p16, DAPK, UCHL1, TSHR, RASAL1, TCL1B, NOTCH4

ACCEPTED MANUSCRIPT

Downregulated

miR-23, miR-26a, miR-125b, miR-130b,

miR-1, miR-7, miR-15b, miR-17, miR-19b, miR19a-3p, miR-

miRNA

miR-139-5p, miR-150, miR-193a-5p, miR-

23b, miR-24, miR-25, miR-26a, miR-26b, miR-27b, miR-29

219-5p, miR-451, miR-455-3p, miR-886-3p,

family, miR-30 family, miR-34-b-3p, miR-98, miR -99a, miR-

let7c

99b, miR-100, miR-101, miR-106b, miR-107, miR-125a, miR125b, miR-126-3p, miR-129-3p, miR-130a, miR -135a-5p,

RI PT

miR-138, miR-141, miR-143, miR-144-3p, miR-145, miR148b, miR-151, miR-152, miR-181a, miR-191, miR-195, miR199 family, miR-200 family, miR-204, miR-206, miR-301a, miR-331-3p, miR-361, miR-486, miR-497, miR-513a-3p, miR -514-3p, miR-618, miR-708, let7 family miR-15a-3p, miR-125a-5p, miR-129, miR-

miR-17-92 cluster, miR-18a, miR-20a, miR-21, miR-92, miR-

miRNA

146b, miR-182, miR-183, miR-187, miR-221,

125a-3p, miR-130b, miR-137, miR-138-3p, miR-142, miR-

miR-222, miR-339

146a, miR-146b, miR-149-3p, miR-150, miR-205, miR-212,

SC

Upregulated

miR-221, miR-222, miR-223, miR-302c, miR-371, miR-422a,

M AN U

miR-487b, miR-491-3p, miR-494, miR-526b-3p, miR-584, miR-659-3p, miR-638, miR-665, miR-923, miR-4295, let7f-5p

Downregulated

MALAT1

MALAT1

lncRNA Upregulated lncRNA

-

MALAT1, LOC100507661, PVT1

6.1 Dysregulated genes

TE D

Bold miRNAs are the miRNAs that can be identified in both PDTC and ATC.

6.1.1 Epigenetic modifiers of PDTC and ATC 6.1.1.1 Aurora group

Contrary to the lack of expression in normal thyroid tissue, Aurora group members A, B,

EP

and C are found overexpressed in ATC (102, 103). The Aurora group acts as the regulators of mitotic events by controlling the histone H3 phosphorylation and chromatin remodeling process

AC C

(104, 105). Moreover, Aurora B silencing by RNA interference or inhibition of Aurora kinase activity has been shown to significantly reduce the growth of ATC cells in vivo, suggesting a tumor promoting role for Aurora proteins in ATC (103).

6.1.1.2 Histone lysine-methyltransferases Enhancer of Zeste homolog 2 (EZH2), a member of the polycomb family of proteins, is a histone lysine-methyltransferase that is overexpressed in ATC. In contrast, no significant change in expression was observed in WDTC compared to normal thyroid tissue (106). Silencing of EZH2 in ATC cell lines resulted in cell growth inhibition, loss of anchorage-independent growth

ACCEPTED MANUSCRIPT

capacity, cell migration, and invasion through Matrigel. Overexpression of EZH2 on the other hand, contributed to altered histone methylation, transcriptional silencing of thyroid-specific transcription factor paired-box gene 8 (PAX8), and ATC dedifferentiation (106).

RI PT

6.1.1.3 Histone deacetylases

Reduced acetylation of histones leads to altered expression of proteins involved in cell proliferation and cell cycle control. Specifically, dysregulation of both the ERK1/2-MEK1/2 and PI3K-Akt pathways were affected by HDAC’s overexpression (107). The overexpression of

6.1.1.4 Histone methyltransferases

M AN U

deregulation of cell cycle control observed in ATC’s (73).

SC

Histone deacetylases (HDACs) 1 and 2 has been reported in ATC and may contribute to the

KMT2D (Lysine Methyltransferase 2D), previously known as myeloid/ lymphoid or mixed-lineage leukemia protein 2 (MLL2), is a histone methyltransferase (HMT) that regulates gene transcription by modulating chromatin accessibility (108). Jeon et al. (2016) reported an association between KMT2D mutation status and shorter disease-specific survival in patients

TE D

with ATC (51). Recent transcriptome analysis of PDTC and ATC showed a greater increase in HMTs (KMT2A, KMT2C, KMT2D, and SETD2) expression in ATC than in PDTC. In addition, the rate of KMT2A, KMT2C, KMT2D, and SETD2 mutation has been found to be higher in ATCs (24%) than PDTCs (7%) (37). Whether the increased expression of HMTs is associated with the

EP

pattern of dedifferentiation observed in PDTC and ATC needs to be further investigated.

AC C

6.1.1.5 SWI/SNF chromatin remodeling complex Switch/Sucrose Non-Fermentable (SWI/SNF) is an important multi-subunit nucleosome-

remodeling complex found in both eukaryotes and prokaryotes. Landa et al. (2016) reported multiple mutations in SWI/SNF chromatin remodeling complex genes (ARID1A, ARID1B, ARID2, ARID5B, ATRX, SMARCB1, and PBRM1) in advanced thyroid tumors (37). SWI/SNF genes have a mutation rate in PDTC and ATC of 6% and 36% respectively (37), roughly aligning with their dedifferentiation status and potentially suggesting an active role in contributing to their aggressive behaviors.

ACCEPTED MANUSCRIPT

6.1.2 Epigenetic Mediators of PDTC and ATC 6.1.2.1 RAS association domain family members Epigenetic silencing of negative RAS effector, RASSF1A (Ras association domain family

RI PT

1 isoform A) has been detected in multiple thyroid cancer types including ATC and medullary thyroid carcinoma (MTC) (96, 109). Nakamura et al. (2005) reported methylation rates of

RASSF1A in PTC, MTC, and FTC as 32%, 40%, and 100% respectively (70). The RASSF1A methylation range reported in ATC was 33–78% (70, 96, 109). Analysis of the promoter

methylation status of RASSF2 (Ras Association (RalGDS/AF-6) Domain Family Member 2),

SC

another negative RAS effector, showed RASSF2 promoter methylation rates in MTC, PTC, FTC, and ATC as 0%, 54%, 80%, and 83% respectively (98). It remains to be seen whether the

M AN U

frequent RASSF family promoter methylation observed in ATCs deregulates the RAS pathway specifically or are part of global methylation changes prior to targeting RAS effectors as potential therapeutic candidates (98).

6.1.2.2 Meiosis-specific component of the cohesin complex

ATC samples have the highest methylation level of REC8 (REC8 Meiotic Recombination

TE D

Protein), one of the members of the cohesion complex, followed by FTC, PTC, and benign thyroid tumors. Hypermethylation of REC8 is associated with advanced disease stage and thyroid cancer-related mortality, potentially by abrogating the PI3K pathway as suggested by the

EP

experiments using ATC cell lines (100).

6.1.2.3 Thyroid transcription factor 1

AC C

Kondo et al. (2009) investigated DNA methylation status of thyroid transcription factor 1 (TTF-1) and found the lack of TTF-1 expression in ATC (6/10 cases) consistent with promoterhypermethylated TTF-1 gene. On the contrary, normal thyroid tissue and PTC displayed positive expression of TTF-1 with the nonmethylated TTF-1 promoter, leading to the conclusion that TTF-1 expression is regulated by an epigenetic promotor methylation process (97).

6.1.2.4 Serpin superfamily The methylation status of the promoter region of SERPINB5 (Serine (Or Cysteine) Proteinase Inhibitor, Clade B (Ovalbumin), Member 5), a mammary serine protease inhibitor

ACCEPTED MANUSCRIPT

(Maspin) was investigated in 6 human ATC cell lines, 17 PDTCs, and 13 ATCs by Ogasawara et al. (2004)(110). They found promoter hypomethylation in 41% of PDTCs and 62% of ATCs. The promotor hypomethylation of Maspin, causing Maspin overexpression is associated with

desirable therapeutic target for PDTC and ATC treatments.

6.1.2.5 Membrane-associated protein 17

RI PT

PDTC and ATC. With established metastatic promoting function, SERPINB5 could become a

Hypomethylation and overexpression of the membrane-associated protein 17 (MAP17), a

SC

reactive oxygen species dependent oncogene, was detected in 33% of ATCs. Overexpression of MAP17 promoted thyroid tumor cell growth in vitro and in vivo (111). It is currently unclear

M AN U

whether the MAP17 pathway is associated with the metabolic alterations observed in ATC.

6.1.3 Epigenetic modulators of PDTC and ATC 6.1.3.1 Phosphatase and tensin homolog

PTEN (phosphatase and tensin homolog) is frequently silenced via promoter methylation in ATC (112). Hou et al. (2008) suggest a positive correlation between PTEN hypermethylation

TE D

and progression from benign thyroid adenoma to FTC or ATC (112). This concept is consistent with the model of silencing PTEN by aberrant methylation and activating genetic aberrations of the PI3K/Akt pathways in multiple cancer types (112-114).

EP

6.1.3.2 Thyroid-stimulating hormone receptor

Hypermethylation of the thyroid-stimulating hormone receptor (TSHR) is frequently

AC C

observed in follicular adenoma, PTC, and ATC (109, 115). Hypermethylation and the resultant silencing of TSHR, the thyroidiodide-metabolizing gene, lead to the failure of iodine concentration, and consequent resistance to radioiodide therapy (68).

6.1.3.3 RAS protein activator like 1 The promoter sequence of RAS protein activator like 1 (RASAL1) gene coding for RASGTPase-activating protein was found hypermethylated in 33% of ATCs (116). Liu et al. (2013) concluded that RASAL1 silencing from promoter hypermethylation could influence ATC tumorigenesis via the RAS-coupled MAPK and PI3K pathways (116).

ACCEPTED MANUSCRIPT

6.1.3.4 T-cell leukemia/lymphoma 1B Dysregulated promoter methylation of T-cell leukemia/lymphoma 1B (TCL1B) oncogene, an established tumor driver gene of B-cell chronic lymphocytic leukemia, was found

RI PT

hypomethylated in 64% of ATCs (111). However, a definitive cancer driver role for TCL1B in thyroid tumorigenesis is yet to be established (117).

6.1.3.5 Neurogenic locus notch homolog protein 4

SC

The promoter of the neurogenic locus notch homolog protein 4 (NOTCH4) oncogene was found hypomethylated in 45% of ATC’s (111). Further, Geers et al. (2011) analyzed NOTCH4

M AN U

gene expression by immunohistochemistry, qRT-PCR, and Western-blot in normal thyroid tissue, benign nodules, and malignant thyroid cancer (PTC, FTC). This study determined that the overexpression of NOTCH4 in carcinoma has potential roles in thyroid dedifferentiation and angiogenesis (118).

Table 4. Genes epigenetically regulated in PDTC and ATC Full gene name

Type of

Mechanism

TE D

Gene

Regulation

Poorly differentiated thyroid cancer Maspin

Mammary serine

Histone

(KMT2

methyltransferase

SETD2)

Dysregulated

AC C

HMTs

family,

Upregulated

EP

protease inhibitor

Hypomethylation

Involved signaling

Outcome

References

pathway

Regulate the

Morphological

breakdown of proteins

dedifferentiation

Ogasawara (2004)

by inhibiting the catalytic activity of proteinases

Mutation

Histone

Deregulated gene

methyltransferase

expression

Landa (2016)

Dysregulated the

Morphological

methylation status

dedifferentiation

Histone

Important roles in

Impair of cell cycle

Sorrentino (2005),

modification

mitosis, Regulate

progression and

Wiseman (2007),

chromosome

maintenance of

Wunderlich (2011)

segregation and

genomic stability

Anaplastic thyroid cancer MECP2

Methyl CpG binding

Dysregulated

Gene fusion

protein 2

Aurora group

-

Upregulated

cytokinesis

Kasaian (2015)

ACCEPTED MANUSCRIPT

EZH2

Enhancer of Zeste

Upregulated

homolog 2

HDAC

Histone deacetylases

Upregulated

Histone

Transcriptional

Dysregulation of cell

Borbone (2011),

methylation

silencing of PAX8

proliferation and

Catalano (2012)

gene

differentiation

Histone

ERK1/2-MEK1/2 and

Condensation of

Borbone (2010),

acetylation

PI3K-Akt pathways

chromatin, preventing

Lin (2013)

transcription factor

RI PT

accessibility and function

HMTs

Histone

(KMT2

methyltransferase

Dysregulated

Mutation

Histone methyltransferase

family,

Switch/Sucrose Non-

complex

Fermentable

Dysregulated

Mutation

Chromatin structure

Dysregulated gene

Kasaian (2015),

modifier

expression

Kunstman (2015),

complex Histone

(EP300)

acetyltransferase

Dysregulated

RASSF1A

RAS association

Downregulated

Downregulated

Hypermethylation

-

Downregulated

Thyroid transcription

Downregulated

Mammary serine

Upregulated

Hypermethylation

Hypermethylation

Hypomethylation

AC C

protease inhibitor

MAP17

Cyclin D1 and MST1

Interact with the

Dysregulated cell

Membrane-

Upregulated

Landa (2016) Kasaian (2015)

growth and division Increased tumor cell

Schagdarsurengin

proliferation

(2002), Nakamura (2005), Schagdarsurengin (2006)

Reducing apoptosis

proapoptotic kinases

EP

Maspin

Hypermethylation

TE D

family 2

factor-1

Chromatin remodeling

deletion

family 1A

RAS association

Frame-shift

M AN U

HATs

TTF-1

Landa (2016),

SC

SWI/SNF

REC8

Kunstman (2015),

expression

Jeon (2016)

SETD2)

RASSF2

Dysregulated gene

Schagdarsurengin (2010)

MST1 and MST2 PI3K pathway

Associated with

Liu (2015)

advanced disease stages of thyroid cancer Regulate the

Dysregulating the

expression of TSH,

organogenesis and

NIS

thyroid function

Regulate the

Morphological

breakdown of proteins

dedifferentiation

Kondo (2009)

Ogasawara (2004)

by inhibiting the catalytic activity of proteinases Hypomethylation

PI3K/Akt pathway

associated protein 17

Promote thyroid tumor

Rodriguez-Rodero

growth in vitro and in

(2013)

vivo PTEN

Phosphatase and tensin homolog

Downregulated

Hypermethylation

Dephosphorylating

Reducing cell-cycle

Frisk (2002), Hou

PI(3,4,5)P3 to reduce

arrest and apoptosis

(2008)

the downstream activity of PKB/Akt kinase, PI3K/Akt

ACCEPTED MANUSCRIPT

pathway

TSHR

Thyroid-stimulating

Downregulated

Hypermethylation

TSHR pathway

hormone receptor

Tumor resistant to

Catalano (2012),

radioiodide therapy,

Schagdarsurengin

Dysregulation of

(2006)

RI PT

thyrocyte function and growth

RASAL1

RAS protein

Downregulated

Hypermethylation

activator like 1 TCL1B

T-cell

MAPK and PI3K pathways

Not associated

Hypomethylation

-

Rodriguez-Rodero

(2013)

Not associated

Hypomethylation

notch homolog

Notch pathway

Induce thyroid

Geers (2011),

carcinogenesis and

Rodriguez-Rodero

angiogenesis.

(2013)

M AN U

protein 4

SC

1B Neurogenic locus

Liu (2013)

development of ATC -

leukemia/lymphoma

NOTCH4

Promote the

6.2 MicroRNA deregulation in PDTC and ATC

MicroRNAs (miRNAs) are a group of small (19–25 nucleotide) RNA molecules that can negatively regulate their target gene expression by binding to the 3'- untranslated regions of the corresponding mRNAs and thereby inhibiting mRNA translation (119). They are involved in

TE D

regulating a variety of molecular processes including apoptosis, tumor dedifferentiation, and metastasis (99, 120). It is well established that improper epigenetic regulation by miRNAs contributes to tumor progression in multiple cancer types including PDTC and ATC (64, 86, 121-124). Up- or downregulation of miRNA can influence the tumorigenic outcome depending

EP

on the role(s) of the target genes on vital signaling processes (Table 5) (64, 86, 121-124).

6.2.1 Downregulated miRNAs and their targets in PDTC and ATC

AC C

While aberrant expression of hundreds of miRNAs has been identified in multiple types of thyroid cancers, very few were found to be exclusively dysregulated in either PDTC or ATC (64, 80, 86, 121-128). To date, approximately 10–15 miRNAs have been found to be downregulated in PDTC compared to 75–80 miRNAs in ATC. Three downregulated miRNAs (miR-26, miR-125, and let-7) were detected in both PDTC and ATC . There are some downregulated miRNAs in PDTC (for example; miR-23 and miR-150) and ATC (for example; miR-1, miR-26, miR-125, miR-141 and miR-618) that were not mentioned in this paragraph but we displayed them in (Table 3 and Table 5) (64, 121, 124-127, 129-132). The miR-30 family of miRNAs, composed of miR-30a, miR-30b, miR-30c, miR-30d, and miR-30e, is implicated to play tumor suppressor roles in multiple cancer types, and has been

ACCEPTED MANUSCRIPT

found to have reduced expression in ATC (64, 121, 125-127, 133, 134). Moreover, overexpression of miR-30a suppresses ATC migration, tumor spreading, and metastasis in vitro and in vivo, most likely mediated by modified LOX-ECM interactions (133).

RI PT

miR-138 potentially targets the human telomerase reverse transcriptase (hTERT) transcript and has been found significantly downregulated in ATC cell lines (130). This finding suggests an epigenetic regulatory mechanism for increasing hTERT expression in ATCs in addition to the noted TERT promoter mutations (54).

The inverse relationship observed between the expression levels of both miR-25 & miR-

SC

30d and EZH2 protein levels in ATC cell lines suggests a critical role for miR-25 and miR-30d in regulating EZH2 expression and potentially ATC carcinogenesis (135). Moreover, enforced

M AN U

upregulation of miR-25 expression in thyroid cells resulted in the reduction of two miR-25 protein targets, TNF-related apoptosis-inducing ligand (TRAIL) and mitogen-activated protein kinase kinase 4 (MEK4), resulting in aberrant cell adhesion and apoptosis, underscoring their potential roles in anaplastic dedifferentiation of the thyroid follicular cells (136). The miR-200 family consists of miR-200a, miR-200b, and miR-200c (124). Braun et al. (2010) demonstrated that downregulation of the miR-200 family can be useful in distinguishing

TE D

ATC from WDTCs (127). Examination of their potential role in regulating mesenchymalepithelial transition revealed not only the inhibition of TGFβ receptor 1 (TGFBR1) but also the upregulation of metastasis-promoting miR-21 in miR-200-silenced ATC cells. Zhang et al. (2012) further studied the molecular mechanism underlying miR-200 regulation of EGF/EGFR

EP

signaling in epithelial–mesenchymal transition (EMT) in ATC cells and showed Rho/ROCKmediated EMT signaling as potential targets for miR-200 in ATC (137).

AC C

A novel metastasis-specific role has been implicated for miR-206 in ATC. Zhang et al. (2015) demonstrated that miR-206 is downregulated in ATC leading to the upregulation of metastasis-related MRTF-A in metastatic ATC (138). Further, they showed that induced silencing of miR-206 resulted in promoting cellular migration and invasion of ATC cells in vitro. Let-7 family of miRNAs (let-7a, let-7b, let-7c, let-7d, let-7e, let-7f, let-7g, and let-7i) are

abundantly expressed in normal thyroid tissue (123). Let-7 displays the potential to overexpress the thyroid transcription factor-1 (TTF1/NKX2-1), which is an important component in regulating the sodium-iodine symporter gene expression. Multiple studies showed a marked decrease in the expression of let-7a, let-7c, let-7d, let-7f, let-7g, and let-7i in ATC (125-127). Incidentally,

ACCEPTED MANUSCRIPT

decreased expression of TTF-1 is frequently reported in PDTCs and almost universally reported in ATCs (15), suggesting a direct role for the let-7 family of miRNAs in the progression of

6.2.2 MicroRNA upregulation in PDTC and ATC

RI PT

thyroid cancer dedifferentiation.

Upregulation of miRNAs in PDTC and ATC are less frequent than downregulation. There are approximately 10–15 upregulated miRNAs in PDTC and 35–40 in ATC. Some

miRNAs, such as miR-146, miR-221, and miR-222, were found overexpressed in multiple

SC

thyroid cancer types including PTC, FTC, PDTC, and ATC (64). Although the target genes are not clearly understood, the most frequently upregulated miRNAs in PDTC are miR-129, miR-

M AN U

146b, miR-183 miR-187, miR-221, miR-222, and miR-339 (122, 128, 139) while ATC showed increased expression of miR-17-92 cluster, miR-137, miR-146 family, miR-205, miR-221/222 families, miR-302c, miR-584, and miR-4295 (64, 122, 124, 125, 140-142). Other upregulated miRNAs in PDTC (for example; miR-146b, miR-221 and miR-222) and ATC (for example; miR-20a) that were not mentioned in this paragraph, we demonstrated them in (Table 3 and Table 5) (64, 122, 128, 139, 143).

TE D

The miR-17-92 cluster transcribes a polycistron that yields seven different mature miRNA’s including miR-17-5p, miR-17-3p, miR-18a, miR-19a, miR-19b, miR-20a, and miR92a (124). Two of the seven cluster components, miR-17-3p and miR-17-5p, were found overexpressed in three out of the six ATC specimens tested. Inhibition of miR-17-5p, miR-17-

EP

3p, and miR-19a in vitro resulted in blocking of tumor proliferation and apoptosis, demonstrating a role for the miR-17-92 cluster in ATC progression (141).

AC C

The miR-146 family, including miR-146a and miR-146b, was found overexpressed in ATC, possibly regulated by the transcription factor NF- B (122, 144). Overexpression of miR146b promoted cell proliferation in ATC cells by inhibiting p21 (CDKN1A) cell cycle regulators (140). Upregulated expression of the miR-221/miR-222 cluster in ATC is associated with poor clinicopathologic features (122). Similar to miR-146b, miR-221/miR-222 also influenced cell proliferation in vitro by targeting the p27kip1 (CDKN1B) protein (83). Compared to PTC and normal thyroid samples, miR-584 is overexpressed in ATC (145). Induced expression of tumor suppressor candidate 2 (TUSC2), a target of miR-584, was shown to rescue the inhibition of apoptosis induced by miR-584 through a novel TWIST1/miR-584/TUSC2

ACCEPTED MANUSCRIPT

pathway in thyroid cancer cells, in vitro (145). In a similar fashion, overexpression of miR-4295 promoted ATC cell proliferation and invasion through direct targeting and inhibition of cyclindependent kinase inhibitor 1A (CDKN1A) (146). Damanakis et al. (2016) analyzed expression profiles of hsa-let7b-5p and hsa-let7f-5p

RI PT

and their predicted targets SLC5A5 (NIS) and high-mobility group AT-hook 2 (HMGA2) in

thyroid cancer and reported contrasting expression patterns in various subtypes of thyroid cancer (147). In ATC, hsa-let7f-5p was overexpressed compared to other types of thyroid cancers but

(147).

M AN U

6.3 Role of long non-coding RNAs in PDTC and ATC

SC

NIS was found stable or upregulated. There was no correlation between has-let7f-5p and NIS

lncRNAs are functionally defined as ‘non-protein coding transcripts longer than 200 nucleotides’ (65). Deregulation of lncRNA expression has been noted to regulate tumor proliferation, metastasis, and recurrence of breast, liver, lung, and thyroid cancer (65, 89, 95, 148-150). However, the limited numbers of studies addressing lncRNA’s regulation in thyroid cancer are mostly limited to PTC, PDTC, and ATC (Table 5) (93-95, 151, 152).

TE D

Zhang et al. (2017) studied the expression pattern of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a nuclear-abundant lncRNA, in 10 normal thyroid tissue, 28 PTC, 21 PDTC and 35 ATC samples using tissue microarrays and found slightly upregulation in PDTC and ATC if compared with normal thyroid tissue (152). In the same study,

EP

qRT-PCR analysis of MALAT1 expression demonstrated that PTC is the most upregulated among other thyroid cancer subtype (FTC, PDTC and ATC) and normal thyroid tissue (152).

AC C

MALAT1 and its suspected downstream target IQ Motif Containing GTPase Activating Protein 1 (IQGAP1), a scaffold protein involved in regulating the actin cytoskeleton, cellular adhesion, transcription, and migration, were found overexpressed in FTC samples (153). Additionally, the overexpression of MALAT1 was shown to induce ATC cell proliferation and invasion in vitro through the upregulation of IQGAP1 (153). LOC100507661 expression was analyzed in various thyroid cancer cell lines, that include PTC (TPC1, BCPAP), FTC (FTC133), and ATC (C643, 8505C) cells (151). PTC and ATC cells displayed LOC100507661 overexpression, with especially high levels in C643 and 8505C. However, LOC100507661 was undetectable in the FTC cell line FTC133. Moreover, elevated

ACCEPTED MANUSCRIPT

expression of LOC100507661 was found significantly correlated with lymph node metastasis (p = 0.035) and BRAF mutation status in PTCs. Additionally, stable overexpression of LOC100507661 induced proliferation, migration, and invasion in thyroid cancer cell lines in vitro, suggesting a potential role for LOC100507661 in promoting a more aggressive phenotype

RI PT

(151).

A comparative expression analysis of PVT1, a lncRNA encoded by the human PVT1 gene, was performed in 84 thyroid cancer cell lines including 66 PTC, 11 FTC, and 7 ATC cells, which showed universal overexpression of PVT1. Furthermore, silencing of PVT1 suppressed

SC

the expression of TSHR, the major regulator of thyroid proliferation, in the PTC, FTC, and ATC cell lines, supporting the proposed function of PVT1 in promoting thyroid cancer cell

M AN U

tumorigenesis including that of ATC’s via TSHR suppression (95).

Table 5. Dysregulated micro and long noncoding RNAs in PDTC and ATC ncRNA

Type of

Chromosomal

Regulation

Location

Poorly differentiated thyroid cancer Downregulated

miR-150

Downregulated

miR-146b

Upregulated

miR-221, miR-

Upregulated

222

19p13.12

19q13.33

Affected/Involved

Targets

signaling pathway

FGFR3

TP53

TE D

miR-23

Potential/Predicted

10q24.32

SMAD4

Xp11.3

PTEN, CDK1A,

11q13.1

IQGAP1

MAPK pathway

PI3K-Akt pathway

AC C

EP

Downregulated

References

Promote tumor

Dettmer (2014),

relapse

Zhang (2016)

Enhance cancer-

Dettmer (2014),

specific mortality

Zhang (2016)

TGF- β pathway,

Promote thyroid

Nikiforova (2008),

NF- B pathway

tumorigenesis

Geraldo (2012)

MAPK pathway

Induce the tumor

Nikiforova (2008),

angiogenesis

Dettmer (2014)

MAPK/ERK-

Dysregulated cell

Huang (2016),

Pathway

cycle, cell

Damanakis (2016),

proliferation, and

Zhang (2017)

CDK1B, TIMP3, KIT

MALAT1

Outcome

cell migration

Anaplastic thyroid cancer miR-1

miR-25

Downregulated

Downregulated

21q21.1

7q22.1

CXCR4 and SDF-1α

CXCR4/SDF-1α

Dysregulated

protein

pathway

cellular proliferation and migration

EZH2, TRAIL, BIM,

MAPK pathway

Dysregulated

Esposito (2012),

chromatin

Zhang (2016),

condensation

Aherne (2016)

NF- B pathway,

Slow the cell

Visone (2007),

TGF-β pathway

proliferation rate

Schwertheim (2009),

KLF4

miR-26a, miR26b

Downregulated

3p22.3, 2q35

Leone (2011)

HMGA

Braun (2010), Hebrant (2014),

ACCEPTED MANUSCRIPT

Zhang (2016)

miR-30 family

Downregulated

6q13, 8q24.22,

SMAD2, TGFBR1,

1q34.2

LOX, beclin 1

Autophagy pathway

Protect ATC cells

Zhu (2009),

from apoptosis and

Schwertheim (2009),

autophagy

Braun (2010), Zhang (2014), Hebrant

RI PT

(2014), Boufraqech (2015), Zhang (2016)

miR-125a, miR-

Downregulated

125b

19q13.41,

MMP1, HMGA2

p53-dependent

Promote tumor

Visone (2007),

11q24.1

LIN28A, p53

pathway

invasion, histone

Schwertheim (2009),

modification

Braun (2010), Furizawa (2014),

miR-141

Downregulated

miR-200 family

Downregulated

12p13.31

Downregulated

let7 family

Downregulated

Wnt pathway

TGFBR1, SMAD2

TGF-β pathway

Zhang (2016)

Increase with the

Mitomo (2008),

progression of

Zhang (2016)

histological dedifferentiation, and malignant behavior Increase the

Braun (2010)

invasive potential of tumor

ZEB1, ZEB2, EZH2,

TGF-β pathway,

Increase the

Braun (2010),

12p13.31

TGFBR1, SMAD2

EGF pathway

invasive potential of

Esposito (2012),

tumor

Zhang (2012),

6p12.2

12q21.31

AC C

miR-618

hTERT

1p36.33,

EP

miR-206

Downregulated

3p21.32

M AN U

Downregulated

TE D

miR-138

SC

Hebrant (2014),

9q22.32,

MRTF-A

XIAP

Hebrant (2014), Zhang (2016) MRTF pathway

Promote invasion,

Zhang (2015)

migration

PI3K/Akt pathway

Dysregulated cell

Cheng (2014), Yi

growth, cell cycle

(2015)

arrest

RAS pathways, Let-

Induce tumor

Johnson (2005),

19q13.41,

7-independent

proliferation,

Visone (2007),

3p21.2,

pathways

histone

Schwertheim (2009),

modification

Braun (2010),

12q14.1, 21q21.1

RAS, HMGA2, LIN28

Furizawa (2014), Hebrant (2014), Zhang (2016)

ACCEPTED MANUSCRIPT

Upregulated

miR-17-92

13q31.3

cluster

p21, TIMP3, PTEN,

PI3K growth

Promote tumor

Takakura (2008),

E2F1

pathway

growth and invasion

Smallridge (2009),

LIMK1

Rho pathway

Dysregulated

Xiong (2014), Zhang

cellular

(2016)

Furizawa (2014) miR-20a

Upregulated

13q31.3

proliferation and

miR‑146a, miR-

Upregulated

146b

miR-221, miR-

Upregulated

5q33.3,

p21, NF- B, THRB,

10q24.32

SMAD4

Xp11.3

p27, RECK, PTEN

222

RI PT

invasion

Dysregulated cell

Nikiforova (2008),

differentiation, and

Furizawa (2014),

invasion

Wang (2016)

TGF-β pathway,

Promote tumor

Visone (2007),

MAPK pathway

growth and invasion

Furizawa (2014),

NF- B pathway

Upregulated

5q32

TUSC2

SC

Zhang (2016)

miR-584

TWIST1/miR-

Induced resistance

584/TUSC2

to apoptosis

Orlandella (2016)

pathway Upregulated

10q25.2

CDKN1A

-

let7f-5p

Upregulated

9q22.3

SLC5A5 (NIS),

MAPK pathway

M AN U

miR-4295

HMGA2

Increased cell migration and invasion

Shao (2015)

Dysregulated cell

Marini (2011),

proliferation, and

Damanakis (2016)

differentiation

MALAT1

Downregulated

11q13.1

-

-

-

Zhang (2017)

MALAT1

Upregulated

11q13.1

IQGAP1

MAPK/ERK-

Dysregulated cell

Liu (2010), Huang

Pathway

cycle, cell

(2016)

LOC100507661

Upregulated

TE D

proliferation, and

3q26.2

Correlated with BRAF

cell migration -

mutation

8q24.21

EP

Upregulated

TSHR

Kim (2016)

proliferation, migration, and invasion Recruiting EZH2

Dysregulated tumor

Zhou (2016)

proliferation and arrested cell cycle at G0/G1 stage

AC C

PVT1

Promote tumor

7. Current status of epigenetic-targeted therapy in thyroid cancer The drugs targeting epigenetic alterations in tumors can be classified according to the

mechanism(s) that they target. HDAC inhibitors and DNA methylation inhibitors (DNMTi) are two main classes of epigenetic pharmaceuticals. To date, the US Food and Drug Administration (FDA) has approved four HDAC inhibitors including belinostat, panobinostat, romidepsin, and vorinostat for the treatment of hematologic malignancies (154-157). These drugs inhibit histone deacetylase, the enzyme that detaches acetyl group from an ε-N-acetyl lysine on histones, which

ACCEPTED MANUSCRIPT

regulates gene expression. Belinostat (PXD101) has shown effective clinical usage by improving the overall response rate of peripheral T-cell lymphoma patients (154, 158, 159). Panobinostat acts as a pan-HDAC inhibitor and showed clinical benefit for treating relapsed and refractory multiple myeloma patients (160). Panobinostat combined with bortezomib and dexamethasone

RI PT

showed enhanced tumor response rate compared to placebo (161). Romidepsin and vorinostat show clinical efficacy in the treatment of cutaneous T-cell lymphoma by increasing the treatment response rate (162-164).

Promoter hypermethylation leading to tumor suppressor silencing is a common epigenetic

SC

mechanism observed to drive a variety of tumor types. Consequently, multiple studies have shown improved treatment responses in multiple cancer types with methylation inhibitors. A

M AN U

widely used example of DNMTi is azacitidine or 5-azacytidine, which has been approved by both European Medicine Agency (EMA) and FDA for treatment of myelodysplastic syndrome (MDS) (165). Azacitidine displayed significantly prolonged overall median survival compared to the conventional care regimens (best supportive care, low-dose cytarabine, and intensive chemotherapy) in MDS patients (165, 166). Another DNMTi, decitabine (5-aza-2'deoxycytidine) efficiently inhibited DNA methyltransferase leading to a hypomethylation status

TE D

of target genes (168). Similar to azacitidine, decitabine also showed the potential to treat MDS by improving progression-free survival when compared to best supportive care (167).

8. Future prospects for epigenetic-targeted therapy in PDTC and ATC

EP

Currently, there are no thyroid-specific epigenetic medications available for the treatment of WDTC, PDTC, or ATC. However, multiple studies have recently demonstrated a progressive

AC C

understanding of the nature of the epigenetic alterations in PDTC and ATC, potentially paving the way to the development of targeted epigenetic pharmaceuticals in the near future (22-24, 37, 48, 59, 96, 97, 99, 142, 168). Aurora kinase inhibitors have also shown promise in laboratory tests. Wunderlich et al.

(2011) evaluated the functions of MLN8054, an Aurora serine ⁄ threonine kinases inhibitors, on ATC cells in vitro and in vivo. MLN8054 was found to be effective in inhibiting ATC cell proliferation and promoting apoptosis in vitro and tumor growth in vivo (169). Similarly, in vitro use of SNS-314 mesylate, a pan-inhibitor of the Aurora kinases, has also been shown to be effective in inhibiting ATC cell growth (170).

ACCEPTED MANUSCRIPT

BET inhibitors are immunosuppressive epigenetic anti-cancer drugs that bind to the extra-terminal motif (BET) proteins that prevent interaction between BET proteins and acetylated histones/transcription factor complexes (171). Mio et al. (2016) reported that BET inhibitors caused decreased viability in ATC cells by causing MCM5 silencing and blocking

RI PT

ATC cell proliferation (172). Epigenetic-driven usage of conventional chemotherapeutic agents alone has shown limited promise in improving the treatment of ATC. The use of cisplatin in a subset of ATCs that expressed miR-30 led to a decrease in tumor size. It was postulated that

cisplatin-activated autophagy response (134).

SC

beclin1, an autophagy gene, could be negatively regulated as it affects the repression of the

Both PDTC and ATC are very complex diseases driven by a multitude of independent

M AN U

and overlapping genetic and epigenetic events. This diversity also highlights the potential opportunity for unique therapeutic approaches including those that combine both genetic and epigenetic targets. Towards this aim, various epigenetic approaches are currently being explored. For instance, the combined use of HDAC and proteasome inhibitors in rat thyroid cell line in vitro resulted in an additive apoptotic response, potentially by interfering with the cell’s dominant survival and ubiquitin-dependent pathways (73). This early evidence supports the

TE D

notion that combined deacetylase-proteasome inhibitors would be potentially effective in treating ATCs where knock-down of multiple complimentary pathways supporting cell survival is likely to yield favorable therapeutic outcomes. In a similar approach, Catalano et al. (2012) used the non-selective HDAC inhibitor panobinostat to initiate ATC cell death both in cell culture and

EP

xenograft (78). Solitary use of PXD101, another HDAC inhibitor, showed inhibition of ATC cell proliferation but acted synergistically when used in combination with doxorubicin and paclitaxel

AC C

(107).

In summary, PDTC and ATC are aggressive and fatal malignancies characterized by

complex genetic and epigenetic modifications. A lack of efficient genetic targeting and the persistent failure of conventional therapeutic approaches make epigenetic targeting an attractive prospect for treating these lethal diseases. Although there are no epigenetic-driven thyroidtargeted therapies currently available, a detailed understanding of the signaling events that are compromised in PDTC’s and ATC’s via epigenetic modifications will dictate the development of precise epigenetic pharmaceuticals to repair the pathways that are being appropriated during the process of PDTC and ATC dedifferentiation.

ACCEPTED MANUSCRIPT

Acknowledgement: The authors thank Dr Ngoentra Tantranont for providing the histopathologic pictures of PDTC and ATC.

RI PT

Conflict of interest: The authors have no actual or potential conflicts of interest to declare.

References

AC C

EP

TE D

M AN U

SC

1. Horn-Ross PL, Lichtensztajn DY, Clarke CA, Dosiou C, Oakley-Girvan I, Reynolds P, et al. Continued rapid increase in thyroid cancer incidence in california: trends by patient, tumor, and neighborhood characteristics. Cancer Epidemiol Biomarkers Prev. 2014;23(6):1067-79. 2. La Vecchia C, Malvezzi M, Bosetti C, Garavello W, Bertuccio P, Levi F, et al. Thyroid cancer mortality and incidence: a global overview. International journal of cancer Journal international du cancer. 2015;136(9):2187-95. 3. Kitahara CM, Sosa JA. The changing incidence of thyroid cancer. Nat Rev Endocrinol. 2016;12(11):646-53. 4. Glaser SM, Mandish SF, Gill BS, Balasubramani GK, Clump DA, Beriwal S. Anaplastic thyroid cancer: Prognostic factors, patterns of care, and overall survival. Head & neck. 2016;38 Suppl 1:E208390. 5. Kebebew E, Greenspan FS, Clark OH, Woeber KA, McMillan A. Anaplastic thyroid carcinoma. Treatment outcome and prognostic factors. Cancer. 2005;103(7):1330-5. 6. Spitzweg C, Harrington KJ, Pinke LA, Vile RG, Morris JC. Clinical review 132: The sodium iodide symporter and its potential role in cancer therapy. J Clin Endocrinol Metab. 2001;86(7):3327-35. 7. O'Neill JP, Shaha AR. Anaplastic thyroid cancer. Oral oncology. 2013;49(7):702-6. 8. Setia N, Barletta JA. Poorly Differentiated Thyroid Carcinoma. Surg Pathol Clin. 2014;7(4):47589. 9. Sakamoto A, Kasai N, Sugano H. Poorly differentiated carcinoma of the thyroid. A clinicopathologic entity for a high-risk group of papillary and follicular carcinomas. Cancer. 1983;52(10):1849-55. 10. DeLellis RA LR, Heitz PU WHO classification of tumors, pathology and genetics-tumors of endocrine organs. IARC Press, Lyon. 2004. 11. Volante M, Collini P, Nikiforov YE, Sakamoto A, Kakudo K, Katoh R, et al. Poorly differentiated thyroid carcinoma: the Turin proposal for the use of uniform diagnostic criteria and an algorithmic diagnostic approach. The American journal of surgical pathology. 2007;31(8):1256-64. 12. Hiltzik D, Carlson DL, Tuttle RM, Chuai S, Ishill N, Shaha A, et al. Poorly differentiated thyroid carcinomas defined on the basis of mitosis and necrosis: a clinicopathologic study of 58 patients. Cancer. 2006;106(6):1286-95. 13. Asioli S, Erickson LA, Righi A, Jin L, Volante M, Jenkins S, et al. Poorly differentiated carcinoma of the thyroid: validation of the Turin proposal and analysis of IMP3 expression. Mod Pathol. 2010;23(9):1269-78. 14. Ragazzi M, Ciarrocchi A, Sancisi V, Gandolfi G, Bisagni A, Piana S. Update on anaplastic thyroid carcinoma: morphological, molecular, and genetic features of the most aggressive thyroid cancer. Int J Endocrinol. 2014;2014:790834. 15. Miettinen M, Franssila KO. Variable expression of keratins and nearly uniform lack of thyroid transcription factor 1 in thyroid anaplastic carcinoma. Hum Pathol. 2000;31(9):1139-45. 16. Patel KN, Shaha AR. Poorly differentiated and anaplastic thyroid cancer. Cancer control : journal of the Moffitt Cancer Center. 2006;13(2):119-28.

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

17. Wreesmann VB, Ghossein RA, Patel SG, Harris CP, Schnaser EA, Shaha AR, et al. Genomewide appraisal of thyroid cancer progression. Am J Pathol. 2002;161(5):1549-56. 18. Liu Z, Hou P, Ji M, Guan H, Studeman K, Jensen K, et al. Highly prevalent genetic alterations in receptor tyrosine kinases and phosphatidylinositol 3-kinase/akt and mitogen-activated protein kinase pathways in anaplastic and follicular thyroid cancers. J Clin Endocrinol Metab. 2008;93(8):3106-16. 19. Wang Y, Hou P, Yu H, Wang W, Ji M, Zhao S, et al. High prevalence and mutual exclusivity of genetic alterations in the phosphatidylinositol-3-kinase/akt pathway in thyroid tumors. J Clin Endocrinol Metab. 2007;92(6):2387-90. 20. Hou P, Liu D, Shan Y, Hu S, Studeman K, Condouris S, et al. Genetic alterations and their relationship in the phosphatidylinositol 3-kinase/Akt pathway in thyroid cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007;13(4):1161-70. 21. Eze OP, Starker LF, Carling T. The role of epigenetic alterations in papillary thyroid carcinogenesis. J Thyroid Res. 2011;2011:895470. 22. Vu-Phan D, Koenig RJ. Genetics and epigenetics of sporadic thyroid cancer. Mol Cell Endocrinol. 2014;386(1-2):55-66. 23. Faam B, Ghaffari MA, Ghadiri A, Azizi F. Epigenetic modifications in human thyroid cancer. Biomed Rep. 2015;3(1):3-8. 24. Xu B, Ghossein R. Genomic Landscape of poorly Differentiated and Anaplastic Thyroid Carcinoma. Endocr Pathol. 2016;27(3):205-12. 25. Soares P, Lima J, Preto A, Castro P, Vinagre J, Celestino R, et al. Genetic alterations in poorly differentiated and undifferentiated thyroid carcinomas. Curr Genomics. 2011;12(8):609-17. 26. Nikiforov YE. Genetic alterations involved in the transition from well-differentiated to poorly differentiated and anaplastic thyroid carcinomas. Endocr Pathol. 2004;15(4):319-27. 27. Santarpia L, El-Naggar AK, Cote GJ, Myers JN, Sherman SI. Phosphatidylinositol 3-kinase/akt and ras/raf-mitogen-activated protein kinase pathway mutations in anaplastic thyroid cancer. J Clin Endocrinol Metab. 2008;93(1):278-84. 28. Ricarte-Filho JC, Ryder M, Chitale DA, Rivera M, Heguy A, Ladanyi M, et al. Mutational profile of advanced primary and metastatic radioactive iodine-refractory thyroid cancers reveals distinct pathogenetic roles for BRAF, PIK3CA, and AKT1. Cancer research. 2009;69(11):4885-93. 29. Howell GM, Hodak SP, Yip L. RAS mutations in thyroid cancer. The oncologist. 2013;18(8):926-32. 30. Xing M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat Rev Cancer. 2013;13(3):184-99. 31. Alvarez-Nunez F, Bussaglia E, Mauricio D, Ybarra J, Vilar M, Lerma E, et al. PTEN promoter methylation in sporadic thyroid carcinomas. Thyroid : official journal of the American Thyroid Association. 2006;16(1):17-23. 32. Xing M. Identifying genetic alterations in poorly differentiated thyroid cancer: a rewarding pursuit. J Clin Endocrinol Metab. 2009;94(12):4661-4. 33. Garcia-Rostan G, Tallini G, Herrero A, D'Aquila TG, Carcangiu ML, Rimm DL. Frequent mutation and nuclear localization of beta-catenin in anaplastic thyroid carcinoma. Cancer research. 1999;59(8):1811-5. 34. Garcia-Rostan G, Camp RL, Herrero A, Carcangiu ML, Rimm DL, Tallini G. Beta-catenin dysregulation in thyroid neoplasms: down-regulation, aberrant nuclear expression, and CTNNB1 exon 3 mutations are markers for aggressive tumor phenotypes and poor prognosis. Am J Pathol. 2001;158(3):987-96. 35. Yu XM, 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. 36. Ferretti E, Tosi E, Po A, Scipioni A, Morisi R, Espinola MS, 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(10):4080-7.

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

37. Landa I, Ibrahimpasic T, Boucai L, Sinha R, Knauf JA, Shah RH, et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J Clin Invest. 2016;126(3):1052-66. 38. Montero-Conde C, Martin-Campos JM, Lerma E, Gimenez G, Martinez-Guitarte JL, Combalia N, et al. Molecular profiling related to poor prognosis in thyroid carcinoma. Combining gene expression data and biological information. Oncogene. 2008;27(11):1554-61. 39. Zhu W, Ou Y, Li Y, Xiao R, Shu M, Zhou Y, et al. A small-molecule triptolide suppresses angiogenesis and invasion of human anaplastic thyroid carcinoma cells via down-regulation of the nuclear factor-kappa B pathway. Mol Pharmacol. 2009;75(4):812-9. 40. Li X, Abdel-Mageed AB, Mondal D, Kandil E. The nuclear factor kappa-B signaling pathway as a therapeutic target against thyroid cancers. Thyroid : official journal of the American Thyroid Association. 2013;23(2):209-18. 41. Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009;361(11):1058-66. 42. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765-73. 43. Wang HY, Tang K, Liang TY, Zhang WZ, Li JY, Wang W, et al. The comparison of clinical and biological characteristics between IDH1 and IDH2 mutations in gliomas. J Exp Clin Cancer Res. 2016;35:86. 44. Smith N, Nucera C. Personalized therapy in patients with anaplastic thyroid cancer: targeting genetic and epigenetic alterations. J Clin Endocrinol Metab. 2015;100(1):35-42. 45. Guerra A, Di Crescenzo V, Garzi A, Cinelli M, Carlomagno C, Tonacchera M, et al. Genetic mutations in the treatment of anaplastic thyroid cancer: a systematic review. BMC surgery. 2013;13 Suppl 2:S44. 46. Murugan AK, Xing M. Anaplastic thyroid cancers harbor novel oncogenic mutations of the ALK gene. Cancer research. 2011;71(13):4403-11. 47. Fagin JA, Matsuo K, Karmakar A, Chen DL, Tang SH, Koeffler HP. High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas. J Clin Invest. 1993;91(1):179-84. 48. Kunstman JW, Juhlin CC, Goh G, Brown TC, Stenman A, Healy JM, et al. Characterization of the mutational landscape of anaplastic thyroid cancer via whole-exome sequencing. Hum Mol Genet. 2015;24(8):2318-29. 49. Xing M. Prognostic utility of BRAF mutation in papillary thyroid cancer. Mol Cell Endocrinol. 2010;321(1):86-93. 50. Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer. 2005;12(2):245-62. 51. Jeon MJ, Chun SM, Kim D, Kwon H, Jang EK, Kim TY, et al. Genomic Alterations of Anaplastic Thyroid Carcinoma Detected by Targeted Massive Parallel Sequencing in a BRAF(V600E) Mutation-Prevalent Area. Thyroid : official journal of the American Thyroid Association. 2016;26(5):683-90. 52. Sykorova V, Dvorakova S, Vcelak J, Vaclavikova E, Halkova T, Kodetova D, et al. Search for new genetic biomarkers in poorly differentiated and anaplastic thyroid carcinomas using next generation sequencing. Anticancer research. 2015;35(4):2029-36. 53. Garcia-Rostan G, Costa AM, Pereira-Castro I, Salvatore G, Hernandez R, Hermsem MJ, et al. Mutation of the PIK3CA gene in anaplastic thyroid cancer. Cancer research. 2005;65(22):10199-207. 54. Liu X, Bishop J, Shan Y, Pai S, Liu D, Murugan AK, et al. Highly prevalent TERT promoter mutations in aggressive thyroid cancers. Endocr Relat Cancer. 2013;20(4):603-10. 55. Hemmer S, Wasenius VM, Knuutila S, Franssila K, Joensuu H. DNA copy number changes in thyroid carcinoma. Am J Pathol. 1999;154(5):1539-47. 56. Lee J, Hwang JA, Lee EK. Recent progress of genome study for anaplastic thyroid cancer. Genomics Inform. 2013;11(2):68-75.

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

57. Marotta V, Guerra A, Sapio MR, Vitale M. RET/PTC rearrangement in benign and malignant thyroid diseases: a clinical standpoint. Eur J Endocrinol. 2011;165(4):499-507. 58. Kelly LM, Barila G, Liu P, Evdokimova VN, Trivedi S, Panebianco F, et al. Identification of the transforming STRN-ALK fusion as a potential therapeutic target in the aggressive forms of thyroid cancer. Proc Natl Acad Sci U S A. 2014;111(11):4233-8. 59. Rodriguez-Rodero S, Delgado-Alvarez E, Fernandez AF, Fernandez-Morera JL, Menendez-Torre E, Fraga MF. Epigenetic alterations in endocrine-related cancer. Endocr Relat Cancer. 2014;21(4):R31930. 60. Brien GL, Valerio DG, Armstrong SA. Exploiting the Epigenome to Control Cancer-Promoting Gene-Expression Programs. Cancer Cell. 2016;29(4):464-76. 61. Mazor T, Pankov A, Song JS, Costello JF. Intratumoral Heterogeneity of the Epigenome. Cancer Cell. 2016;29(4):440-51. 62. Stricker SH, Koferle A, Beck S. From profiles to function in epigenomics. Nat Rev Genet. 2017;18(1):51-66. 63. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128(4):683-92. 64. Zhang R, Hardin H, Chen J, Guo Z, Lloyd RV. Non-Coding RNAs in Thyroid Cancer. Endocr Pathol. 2016;27(1):12-20. 65. Schmitt AM, Chang HY. Long Noncoding RNAs in Cancer Pathways. Cancer Cell. 2016;29(4):452-63. 66. Feinberg AP, Koldobskiy MA, Gondor A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat Rev Genet. 2016;17(5):284-99. 67. Hu S, Liu D, Tufano RP, Carson KA, Rosenbaum E, Cohen Y, et al. Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. International journal of cancer Journal international du cancer. 2006;119(10):2322-9. 68. Xing M, Usadel H, Cohen Y, Tokumaru Y, Guo Z, Westra WB, et al. Methylation of the thyroidstimulating hormone receptor gene in epithelial thyroid tumors: a marker of malignancy and a cause of gene silencing. Cancer research. 2003;63(9):2316-21. 69. Xing M, Cohen Y, Mambo E, Tallini G, Udelsman R, Ladenson PW, et al. Early occurrence of RASSF1A hypermethylation and its mutual exclusion with BRAF mutation in thyroid tumorigenesis. Cancer research. 2004;64(5):1664-8. 70. Nakamura N, Carney JA, Jin L, Kajita S, Pallares J, Zhang H, et al. RASSF1A and NORE1A methylation and BRAFV600E mutations in thyroid tumors. Lab Invest. 2005;85(9):1065-75. 71. Kunstman JW, Korah R, Healy JM, Prasad M, Carling T. Quantitative assessment of RASSF1A methylation as a putative molecular marker in papillary thyroid carcinoma. Surgery. 2013;154(6):125561; discussion 61-2. 72. Mitsiades CS, Poulaki V, McMullan C, Negri J, Fanourakis G, Goudopoulou A, et al. Novel histone deacetylase inhibitors in the treatment of thyroid cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2005;11(10):3958-65. 73. Borbone E, Berlingieri MT, 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. 74. Furuya F, Shimura H, Suzuki H, Taki K, Ohta K, Haraguchi K, 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(6):2865-75. 75. Greenberg VL, Williams JM, Cogswell JP, Mendenhall M, Zimmer SG. Histone deacetylase inhibitors promote apoptosis and differential cell cycle arrest in anaplastic thyroid cancer cells. Thyroid : official journal of the American Thyroid Association. 2001;11(4):315-25. 76. 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 : official journal of the American Thyroid Association. 2013;23(7):838-46.

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

77. Sherman EJ, Su YB, Lyall A, Schoder H, Fury MG, Ghossein RA, et al. Evaluation of romidepsin for clinical activity and radioactive iodine reuptake in radioactive iodine-refractory thyroid carcinoma. Thyroid : official journal of the American Thyroid Association. 2013;23(5):593-9. 78. Catalano MG, Pugliese M, Gargantini E, Grange C, Bussolati B, Asioli S, et al. Cytotoxic activity of the histone deacetylase inhibitor panobinostat (LBH589) in anaplastic thyroid cancer in vitro and in vivo. International journal of cancer Journal international du cancer. 2012;130(3):694-704. 79. Noguchi H, Yamashita H, Murakami T, Hirai K, Noguchi Y, Maruta J, et al. Successful treatment of anaplastic thyroid carcinoma with a combination of oral valproic acid, chemotherapy, radiation and surgery. Endocr J. 2009;56(2):245-9. 80. Pallante P, Battista S, Pierantoni GM, Fusco A. Deregulation of microRNA expression in thyroid neoplasias. Nat Rev Endocrinol. 2014;10(2):88-101. 81. Sun Y, Yu S, Liu Y, Wang F, Liu Y, Xiao H. Expression of miRNAs in Papillary Thyroid Carcinomas Is Associated with BRAF Mutation and Clinicopathological Features in Chinese Patients. Int J Endocrinol. 2013;2013:128735. 82. Zhang X, Li D, Li M, Ye M, Ding L, Cai H, et al. MicroRNA-146a targets PRKCE to modulate papillary thyroid tumor development. International journal of cancer Journal international du cancer. 2014;134(2):257-67. 83. Visone R, Russo L, Pallante P, De Martino I, Ferraro A, Leone V, et al. MicroRNAs (miR)-221 and miR-222, both overexpressed in human thyroid papillary carcinomas, regulate p27Kip1 protein levels and cell cycle. Endocr Relat Cancer. 2007;14(3):791-8. 84. Jazdzewski K, Boguslawska J, Jendrzejewski J, Liyanarachchi S, Pachucki J, Wardyn KA, et al. Thyroid hormone receptor beta (THRB) is a major target gene for microRNAs deregulated in papillary thyroid carcinoma (PTC). J Clin Endocrinol Metab. 2011;96(3):E546-53. 85. Weber F, Teresi RE, Broelsch CE, Frilling A, Eng C. A limited set of human MicroRNA is deregulated in follicular thyroid carcinoma. J Clin Endocrinol Metab. 2006;91(9):3584-91. 86. Boufraqech M, Klubo-Gwiezdzinska J, Kebebew E. MicroRNAs in the thyroid. Best Pract Res Clin Endocrinol Metab. 2016;30(5):603-19. 87. Aragon Han P, Weng CH, Khawaja HT, Nagarajan N, Schneider EB, Umbricht CB, et al. MicroRNA Expression and Association with Clinicopathologic Features in Papillary Thyroid Cancer: A Systematic Review. Thyroid : official journal of the American Thyroid Association. 2015;25(12):1322-9. 88. Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, et al. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 2015;47(3):199-208. 89. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464(7291):1071-6. 90. Jendrzejewski J, He H, Radomska HS, Li W, Tomsic J, Liyanarachchi S, et al. The polymorphism rs944289 predisposes to papillary thyroid carcinoma through a large intergenic noncoding RNA gene of tumor suppressor type. Proc Natl Acad Sci U S A. 2012;109(22):8646-51. 91. Shao Y, Ye M, Jiang X, Sun W, Ding X, Liu Z, et al. Gastric juice long noncoding RNA used as a tumor marker for screening gastric cancer. Cancer. 2014;120(21):3320-8. 92. Hu X, Feng Y, Zhang D, Zhao SD, Hu Z, Greshock J, et al. A functional genomic approach identifies FAL1 as an oncogenic long noncoding RNA that associates with BMI1 and represses p21 expression in cancer. Cancer Cell. 2014;26(3):344-57. 93. Lan X, Sun W, Zhang P, He L, Dong W, Wang Z, et al. Downregulation of long noncoding RNA NONHSAT037832 in papillary thyroid carcinoma and its clinical significance. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2016;37(5):6117-23. 94. Zheng H, Wang M, Jiang L, Chu H, Hu J, Ning J, et al. BRAF-Activated Long Noncoding RNA Modulates Papillary Thyroid Carcinoma Cell Proliferation through Regulating Thyroid Stimulating Hormone Receptor. Cancer research and treatment : official journal of Korean Cancer Association. 2016;48(2):698-707. 95. Zhou Q, Chen J, Feng J, Wang J. Long noncoding RNA PVT1 modulates thyroid cancer cell proliferation by recruiting EZH2 and regulating thyroid-stimulating hormone receptor (TSHR). Tumour

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2016;37(3):3105-13. 96. Schagdarsurengin U, Gimm O, Hoang-Vu C, Dralle H, Pfeifer GP, Dammann R. Frequent epigenetic silencing of the CpG island promoter of RASSF1A in thyroid carcinoma. Cancer research. 2002;62(13):3698-701. 97. Kondo T, Nakazawa T, Ma D, Niu D, Mochizuki K, Kawasaki T, et al. Epigenetic silencing of TTF-1/NKX2-1 through DNA hypermethylation and histone H3 modulation in thyroid carcinomas. Lab Invest. 2009;89(7):791-9. 98. Schagdarsurengin U, Richter AM, Hornung J, Lange C, Steinmann K, Dammann RH. Frequent epigenetic inactivation of RASSF2 in thyroid cancer and functional consequences. Mol Cancer. 2010;9:264. 99. Catalano MG, Fortunati N, Boccuzzi G. Epigenetics modifications and therapeutic prospects in human thyroid cancer. Front Endocrinol (Lausanne). 2012;3:40. 100. Liu D, Shen X, Zhu G, Xing M. REC8 is a novel tumor suppressor gene epigenetically robustly targeted by the PI3K pathway in thyroid cancer. Oncotarget. 2015;6(36):39211-24. 101. Russo D, Damante G, Puxeddu E, Durante C, Filetti S. Epigenetics of thyroid cancer and novel therapeutic targets. J Mol Endocrinol. 2011;46(3):R73-81. 102. 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. Annals of surgical oncology. 2007;14(2):719-29. 103. Sorrentino R, Libertini S, Pallante PL, Troncone G, Palombini L, Bavetsias V, et al. Aurora B overexpression associates with the thyroid carcinoma undifferentiated phenotype and is required for thyroid carcinoma cell proliferation. J Clin Endocrinol Metab. 2005;90(2):928-35. 104. Wike CL, Graves HK, Hawkins R, Gibson MD, Ferdinand MB, Zhang T, et al. Aurora-A mediated histone H3 phosphorylation of threonine 118 controls condensin I and cohesin occupancy in mitosis. Elife. 2016;5:e11402. 105. Vader G, Lens SM. The Aurora kinase family in cell division and cancer. Biochim Biophys Acta. 2008;1786(1):60-72. 106. Borbone E, Troncone G, Ferraro A, Jasencakova Z, Stojic L, Esposito F, et al. Enhancer of zeste homolog 2 overexpression has a role in the development of anaplastic thyroid carcinomas. J Clin Endocrinol Metab. 2011;96(4):1029-38. 107. Lin SF, Lin JD, Chou TC, Huang YY, Wong RJ. Utility of a histone deacetylase inhibitor (PXD101) for thyroid cancer treatment. PloS one. 2013;8(10):e77684. 108. Guo C, Chen LH, Huang Y, Chang CC, Wang P, Pirozzi CJ, et al. KMT2D maintains neoplastic cell proliferation and global histone H3 lysine 4 monomethylation. Oncotarget. 2013;4(11):2144-53. 109. Schagdarsurengin U, Gimm O, Dralle H, Hoang-Vu C, Dammann R. CpG island methylation of tumor-related promoters occurs preferentially in undifferentiated carcinoma. Thyroid : official journal of the American Thyroid Association. 2006;16(7):633-42. 110. Ogasawara S, Maesawa C, Yamamoto M, Akiyama Y, Wada K, Fujisawa K, et al. Disruption of cell-type-specific methylation at the Maspin gene promoter is frequently involved in undifferentiated thyroid cancers. Oncogene. 2004;23(5):1117-24. 111. Rodriguez-Rodero S, Fernandez AF, Fernandez-Morera JL, Castro-Santos P, Bayon GF, Ferrero C, et al. DNA methylation signatures identify biologically distinct thyroid cancer subtypes. J Clin Endocrinol Metab. 2013;98(7):2811-21. 112. Hou P, Ji M, Xing M. Association of PTEN gene methylation with genetic alterations in the phosphatidylinositol 3-kinase/AKT signaling pathway in thyroid tumors. Cancer. 2008;113(9):2440-7. 113. Zhang X, Jia X, Mei L, Zheng M, Yu C, Ye M. Global DNA methylation and PTEN hypermethylation alterations in lung tissues from human silicosis. Journal of thoracic disease. 2016;8(8):2185-95.

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

114. Mueller S, Phillips J, Onar-Thomas A, Romero E, Zheng S, Wiencke JK, et al. PTEN promoter methylation and activation of the PI3K/Akt/mTOR pathway in pediatric gliomas and influence on clinical outcome. Neuro-oncology. 2012;14(9):1146-52. 115. Mohammadi-asl J, Larijani B, Khorgami Z, Tavangar SM, Haghpanah V, Kheirollahi M, et al. Qualitative and quantitative promoter hypermethylation patterns of the P16, TSHR, RASSF1A and RARbeta2 genes in papillary thyroid carcinoma. Med Oncol. 2011;28(4):1123-8. 116. Liu D, Yang C, Bojdani E, Murugan AK, Xing M. Identification of RASAL1 as a major tumor suppressor gene in thyroid cancer. J Natl Cancer Inst. 2013;105(21):1617-27. 117. Yuille MR, Condie A, Stone EM, Wilsher J, Bradshaw PS, Brooks L, et al. TCL1 is activated by chromosomal rearrangement or by hypomethylation. Genes Chromosomes Cancer. 2001;30(4):336-41. 118. Geers C, Colin IM, Gerard AC. Delta-like 4/Notch pathway is differentially regulated in benign and malignant thyroid tissues. Thyroid : official journal of the American Thyroid Association. 2011;21(12):1323-30. 119. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215-33. 120. Jafri MA, Al-Qahtani MH, Shay JW. Role of miRNAs in human cancer metastasis: Implications for therapeutic intervention. Semin Cancer Biol. 2017. 121. Hebrant A, Floor S, Saiselet M, Antoniou A, Desbuleux A, Snyers B, et al. miRNA expression in anaplastic thyroid carcinomas. PloS one. 2014;9(8):e103871. 122. Nikiforova MN, Tseng GC, Steward D, Diorio D, Nikiforov YE. MicroRNA expression profiling of thyroid tumors: biological significance and diagnostic utility. J Clin Endocrinol Metab. 2008;93(5):1600-8. 123. Marini F, Luzi E, Brandi ML. MicroRNA Role in Thyroid Cancer Development. J Thyroid Res. 2011;2011:407123. 124. Fuziwara CS, Kimura ET. MicroRNA Deregulation in Anaplastic Thyroid Cancer Biology. Int J Endocrinol. 2014;2014:743450. 125. Visone R, Pallante P, Vecchione A, Cirombella R, Ferracin M, Ferraro A, et al. Specific microRNAs are downregulated in human thyroid anaplastic carcinomas. Oncogene. 2007;26(54):7590-5. 126. Schwertheim S, Sheu SY, Worm K, Grabellus F, Schmid KW. Analysis of deregulated miRNAs is helpful to distinguish poorly differentiated thyroid carcinoma from papillary thyroid carcinoma. Horm Metab Res. 2009;41(6):475-81. 127. Braun J, Hoang-Vu C, Dralle H, Huttelmaier S. Downregulation of microRNAs directs the EMT and invasive potential of anaplastic thyroid carcinomas. Oncogene. 2010;29(29):4237-44. 128. Dettmer MS, Perren A, Moch H, Komminoth P, Nikiforov YE, Nikiforova MN. MicroRNA profile of poorly differentiated thyroid carcinomas: new diagnostic and prognostic insights. J Mol Endocrinol. 2014;52(2):181-9. 129. Leone V, D'Angelo D, Rubio I, de Freitas PM, Federico A, Colamaio M, et al. MiR-1 is a tumor suppressor in thyroid carcinogenesis targeting CCND2, CXCR4, and SDF-1alpha. J Clin Endocrinol Metab. 2011;96(9):E1388-98. 130. Mitomo S, Maesawa C, Ogasawara S, Iwaya T, Shibazaki M, Yashima-Abo A, et al. Downregulation of miR-138 is associated with overexpression of human telomerase reverse transcriptase protein in human anaplastic thyroid carcinoma cell lines. Cancer Sci. 2008;99(2):280-6. 131. Cheng Q, Zhang X, Xu X, Lu X. MiR-618 inhibits anaplastic thyroid cancer by repressing XIAP in one ATC cell line. Ann Endocrinol (Paris). 2014;75(4):187-93. 132. Yi L, Yuan Y. MicroRNA-618 modulates cell growth via targeting PI3K/Akt pathway in human thyroid carcinomas. Indian J Cancer. 2015;52 Suppl 3:E186-9. 133. Boufraqech M, Nilubol N, Zhang L, Gara SK, Sadowski SM, Mehta A, et al. miR30a inhibits LOX expression and anaplastic thyroid cancer progression. Cancer research. 2015;75(2):367-77. 134. Zhang Y, Yang WQ, Zhu H, Qian YY, Zhou L, Ren YJ, et al. Regulation of autophagy by miR30d impacts sensitivity of anaplastic thyroid carcinoma to cisplatin. Biochem Pharmacol. 2014;87(4):56270.

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

135. Esposito F, Tornincasa M, Pallante P, Federico A, Borbone E, Pierantoni GM, et al. Downregulation of the miR-25 and miR-30d contributes to the development of anaplastic thyroid carcinoma targeting the polycomb protein EZH2. J Clin Endocrinol Metab. 2012;97(5):E710-8. 136. Aherne ST, Smyth P, Freeley M, Smith L, Spillane C, O'Leary J, et al. Altered expression of mir222 and mir-25 influences diverse gene expression changes in transformed normal and anaplastic thyroid cells, and impacts on MEK and TRAIL protein expression. Int J Mol Med. 2016;38(2):433-45. 137. Zhang Z, Liu ZB, Ren WM, Ye XG, Zhang YY. The miR-200 family regulates the epithelialmesenchymal transition induced by EGF/EGFR in anaplastic thyroid cancer cells. Int J Mol Med. 2012;30(4):856-62. 138. Zhang WL, Lv W, Sun SZ, Wu XZ, Zhang JH. miR-206 inhibits metastasis-relevant traits by degrading MRTF-A in anaplastic thyroid cancer. International journal of oncology. 2015;47(1):133-42. 139. Geraldo MV, Yamashita AS, Kimura ET. MicroRNA miR-146b-5p regulates signal transduction of TGF-beta by repressing SMAD4 in thyroid cancer. Oncogene. 2012;31(15):1910-22. 140. Wang S, Chen Y, Bai Y. p21 participates in the regulation of anaplastic thyroid cancer cell proliferation by miR-146b. Oncol Lett. 2016;12(3):2018-22. 141. Takakura S, Mitsutake N, Nakashima M, Namba H, Saenko VA, Rogounovitch TI, et al. Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci. 2008;99(6):1147-54. 142. Smallridge RC, Marlow LA, Copland JA. Anaplastic thyroid cancer: molecular pathogenesis and emerging therapies. Endocr Relat Cancer. 2009;16(1):17-44. 143. Xiong Y, Zhang L, Kebebew E. MiR-20a is upregulated in anaplastic thyroid cancer and targets LIMK1. PloS one. 2014;9(5):e96103. 144. Pacifico F, Crescenzi E, Mellone S, Iannetti A, Porrino N, Liguoro D, et al. Nuclear factor{kappa}B contributes to anaplastic thyroid carcinomas through up-regulation of miR-146a. J Clin Endocrinol Metab. 2010;95(3):1421-30. 145. Orlandella FM, Di Maro G, Ugolini C, Basolo F, Salvatore G. TWIST1/miR-584/TUSC2 pathway induces resistance to apoptosis in thyroid cancer cells. Oncotarget. 2016. 146. Shao M, Geng Y, Lu P, Xi Y, Wei S, Wang L, et al. miR-4295 promotes cell proliferation and invasion in anaplastic thyroid carcinoma via CDKN1A. Biochem Biophys Res Commun. 2015;464(4):1309-13. 147. Damanakis AI, Eckhardt S, Wunderlich A, Roth S, Wissniowski TT, Bartsch DK, et al. MicroRNAs let7 expression in thyroid cancer: correlation with their deputed targets HMGA2 and SLC5A5. Journal of cancer research and clinical oncology. 2016;142(6):1213-20. 148. Lu Z, Xiao Z, Liu F, Cui M, Li W, Yang Z, et al. Long non-coding RNA HULC promotes tumor angiogenesis in liver cancer by up-regulating sphingosine kinase 1 (SPHK1). Oncotarget. 2016;7(1):24154. 149. Ji P, Diederichs S, Wang W, Boing S, Metzger R, Schneider PM, et al. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22(39):8031-41. 150. Prensner JR, Chinnaiyan AM. The emergence of lncRNAs in cancer biology. Cancer Discov. 2011;1(5):391-407. 151. Kim D, Lee WK, Jeong S, Seol MY, Kim H, Kim KS, et al. Upregulation of long noncoding RNA LOC100507661 promotes tumor aggressiveness in thyroid cancer. Mol Cell Endocrinol. 2016;431:36-45. 152. Zhang R, Hardin H, Huang W, Chen J, Asioli S, Righi A, et al. MALAT1 Long Non-coding RNA Expression in Thyroid Tissues: Analysis by In Situ Hybridization and Real-Time PCR. Endocr Pathol. 2017;28(1):7-12. 153. Huang JK, Ma L, Song WH, Lu BY, Huang YB, Dong HM, et al. MALAT1 promotes the proliferation and invasion of thyroid cancer cells via regulating the expression of IQGAP1. Biomed Pharmacother. 2016;83:1-7. 154. Lee HZ, Kwitkowski VE, Del Valle PL, Ricci MS, Saber H, Habtemariam BA, et al. FDA Approval: Belinostat for the Treatment of Patients with Relapsed or Refractory Peripheral T-cell

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

Lymphoma. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015;21(12):2666-70. 155. Lakshmaiah KC, Jacob LA, Aparna S, Lokanatha D, Saldanha SC. Epigenetic therapy of cancer with histone deacetylase inhibitors. Journal of cancer research and therapeutics. 2014;10(3):469-78. 156. De Souza C, Chatterji BP. HDAC Inhibitors as Novel Anti-Cancer Therapeutics. Recent Pat Anticancer Drug Discov. 2015;10(2):145-62. 157. Jones PA, Issa JP, Baylin S. Targeting the cancer epigenome for therapy. Nat Rev Genet. 2016;17(10):630-41. 158. Poole RM. Belinostat: first global approval. Drugs. 2014;74(13):1543-54. 159. Foss F, Advani R, Duvic M, Hymes KB, Intragumtornchai T, Lekhakula A, et al. A Phase II trial of Belinostat (PXD101) in patients with relapsed or refractory peripheral or cutaneous T-cell lymphoma. Br J Haematol. 2015;168(6):811-9. 160. Redic KA, Hough SM, Price EM. Clinical developments in the treatment of relapsed or relapsed and refractory multiple myeloma: impact of panobinostat, the first-in-class histone deacetylase inhibitor. Onco Targets Ther. 2016;9:2783-93. 161. Garnock-Jones KP. Panobinostat: first global approval. Drugs. 2015;75(6):695-704. 162. Duvic M, Olsen EA, Breneman D, Pacheco TR, Parker S, Vonderheid EC, et al. Evaluation of the long-term tolerability and clinical benefit of vorinostat in patients with advanced cutaneous T-cell lymphoma. Clin Lymphoma Myeloma. 2009;9(6):412-6. 163. Piekarz RL, Frye R, Turner M, Wright JJ, Allen SL, Kirschbaum MH, et al. Phase II multiinstitutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(32):5410-7. 164. Olsen EA, Kim YH, Kuzel TM, Pacheco TR, Foss FM, Parker S, et al. Phase IIb multicenter trial of vorinostat in patients with persistent, progressive, or treatment refractory cutaneous T-cell lymphoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25(21):3109-15. 165. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. The Lancet Oncology. 2009;10(3):223-32. 166. Prebet T, Sun Z, Ketterling RP, Zeidan A, Greenberg P, Herman J, et al. Azacitidine with or without Entinostat for the treatment of therapy-related myeloid neoplasm: further results of the E1905 North American Leukemia Intergroup study. Br J Haematol. 2016;172(3):384-91. 167. Lubbert M, Suciu S, Hagemeijer A, Ruter B, Platzbecker U, Giagounidis A, et al. Decitabine improves progression-free survival in older high-risk MDS patients with multiple autosomal monosomies: results of a subgroup analysis of the randomized phase III study 06011 of the EORTC Leukemia Cooperative Group and German MDS Study Group. Annals of hematology. 2016;95(2):191-9. 168. Kasaian K, Wiseman SM, Walker BA, Schein JE, Zhao Y, Hirst M, et al. The genomic and transcriptomic landscape of anaplastic thyroid cancer: implications for therapy. BMC cancer. 2015;15:984. 169. 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. 170. Baldini E, Sorrenti S, D'Armiento E, Guaitoli E, Morrone S, D'Andrea V, et al. Effects of the Aurora kinases pan-inhibitor SNS-314 mesylate on anaplastic thyroid cancer derived cell lines. Clin Ter. 2012;163(5):e307-13. 171. Shi J, Vakoc CR. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol Cell. 2014;54(5):728-36. 172. 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.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

HIGHLIGHTS

RI PT

SC M AN U TE D



EP



Epigenetic alterations may play potentially decisive roles in the origin and the agrressive clinical course of PDTC and ATC. Mechanisms of epigenetic deregulations in PDTC and ATC include DNA methylation, histone modification, chromatin remodeling, and ncRNA aberration. Understanding the underpinnings of PDTC and ATC epigenetic dysregulations may lead to the development of novel epigenetics-based therapeutic strategies.

AC C