Next Article in Journal
SERS-PLSR Analysis of Vaginal Microflora: Towards the Spectral Library of Microorganisms
Next Article in Special Issue
Immunomodulation of HDAC Inhibitor Entinostat Potentiates the Anticancer Effects of Radiation and PD-1 Blockade in the Murine Lewis Lung Carcinoma Model
Previous Article in Journal
Radical in the Peroxide-Produced F-Type Ferryl Form of Bovine Cytochrome c Oxidase
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Component and Function of Tumor Microenvironment in Thyroid Cancer

Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(20), 12578; https://doi.org/10.3390/ijms232012578
Submission received: 30 September 2022 / Accepted: 17 October 2022 / Published: 20 October 2022
(This article belongs to the Special Issue New Advances in Endocrine-Related Cancer 2022)

Abstract

:
Thyroid cancer is the most common cancer in the endocrine system. Most thyroid cancers have good prognosis, but some of them are resistant to treatment or show aggressive behavior. Like other tumors, thyroid cancers harbor tumor microenvironment (TME) composed of cancer associated fibroblasts (CAF) and immune cells. Autoimmune lymphocytic thyroiditis can occur in the thyroid, and it may be associated with cancer development. TME is involved in tumor progression through various mechanisms: (1) CAF is involved in tumor progression through cell proliferation and extracellular matrix (ECM) remodeling; and (2) immune cells are involved in tumor progression through cell proliferation, angiogenesis, epithelial mesenchymal transformation (EMT), and immune suppression. These events are activated by various cytokines, chemokines, and metabolites secreted from cells that comprise TME. This review is focused on how CAF and immune cells, two important cell components of thyroid cancer TME, are involved in tumor progression, and will explore their potential as therapeutic targets.

1. Introduction

Thyroid cancer is the most common cancer arising in the endocrine system [1], and its incidence rate is rapidly increasing [2]. Thyroid cancers mostly arise from follicular cells except for medullary carcinoma (MC) that arises from parafollicular c-cells. Thyroid cancers arising from follicular cells are classified as well-differentiated carcinoma, poorly differentiated carcinoma (PDC), and anaplastic (undifferentiated) carcinoma (ATC) according to histology and clinical features. Well-differentiated carcinomas, namely papillary thyroid carcinoma (PTC) and follicular thyroid carcinoma (FTC), comprise 90% of thyroid cancers [3]. Thyroid cancers show various clinical characteristics and treatment effects according to histologic subtypes [4], and PTC and FTC show relatively good prognosis. However, about 10% of PTC and FTC show resistance to radioactive iodine therapy, and ATC and MC, although rare in occurrence, have aggressive features with poor prognosis [5,6]. Treatment modalities for thyroid cancers include surgery, endocrine inhibition therapy, and radio-iodine therapy [7,8]. However, as these therapeutic options also have limitations and complications [9,10], research on new treatment targets is warranted.
Tumor microenvironment (TME) is the non-transformed area around tumor cells, mainly composed of cancer associated fibroblasts (CAF) and immune cells [11,12,13,14]. TME has an impact on tumor progression and treatment response through interactions with tumor cells. Like any other tumors, thyroid cancers have TME as well that affects various aspects of tumor behavior. This review is focused mainly on the cell components of TME in thyroid cancers and how they affect cancer biology. The possible potential of suppressing the crosstalk between TME and tumor cells as a new treatment option is also explored.

2. General Characteristics of Tumor Microenvironment

2.1. Cancer-Associated Fibroblasts (CAF)

TME is composed of immune cells, fibroblasts, myofibroblasts, endothelial cells, adipocytes, and extracellular matrix (ECM), and CAF are the main cellular component of TME [11,12,13,14]. CAF are larger in cell size, have more cytoplasmic branches, are more potent in proliferation and migration, and can produce more ECM than fibroblasts [11,13,15]. A number of markers, such as α-SMA, FSP-1, FAPα, NG2, PDGFR-β, and prolyl 4-hydroxase, have been explored to identify CAF; however, none of them are specific for CAF [12]. Resident tissue fibroblasts, mesenchymal stem cells, bone-marrow derived fibrocytes, adipocytes, endothelial cells, and epithelial cells are suggested as the origin of CAF, implying that various cells can differentiate into CAF [12,15]. Tumor cells and/or transformed epithelial cells that are destined to be tumor cells produce soluble factors and lipid-based particles, which induce fibroblasts to differentiate into CAF, and then recruit and activate CAF. Activated CAF are involved in tumor cell proliferation, expression of cancer cell stemness, and reprogramming of tumor metabolism through crosstalk such as cell to cell contact between tumor cells and receptor-ligands. CAF are also involved in ECM remodeling, immune modulation, and tumor cell migration and metastasis via EMT [16,17]. Growth factors such as HGF, VEGF, EGF, CTGF, IGF, and NGF, cytokines such as IL-6, IL-11, and IL-17, and chemokines such as CCL7, CCL5, CXCL12, and CXCL7 are produced by CAF in the process of interaction between tumor cells and CAF [18].

2.2. Tumor Infiltrating Immune Cells

Immune cells comprising TME can be subgrouped into tumor-associated myeloid cells (TAMC) and tumor-associated lymphoid cells (TALC). TAMC include tumor-associated macrophages (TAM), myeloid-derived suppressor cells (MDSC), tumor-associated neutrophils (TAN), tumor-associated dendritic cells (TADC), and tumor-associated mast cells (TMC). TALC include T cells and NK cells.

2.2.1. Tumor-Associated Myeloid Cells (TAMC)

TAM are the most common and large component among infiltrating immune cells and they are subdivided into M1 (classic) and M2 (alternative) [19,20]. M1 type TAM secrete cytokines such as TNF-α and IL-1, and thus they are associated with Th1 response, intracellular pathogen killing, and antitumor immunity [21,22]. M2 type TAM secrete IL-10 and IL-13, and they are associated with killing and encapsulation of parasites [23], Th2 activation [24], tissue remodeling [25], and tumor induction and growth [26]. Markers for M1 macrophages are CD64, IDO, SOCS1, and CXCL10. Markers for M2 macrophages are MRC1, TGM2, CD23, and CCL22 [27]. It is suggested that TAM are derived from tissue-resident macrophages (TRM) and bone-marrow-derived macrophages (BMDM), and the compositional ratio of TRM and BMDM in TME is different according to the tumor types [28]. The mechanisms by which TAM promote tumor progression are as follows: (1) TAM promote tumor invasion and metastasis by initiating EMT of tumor cells through secretion of signaling molecules such as EGF, SPARC, MMP, CCL2, and CCL18; (2) TAM induce angiogenesis within the tumor by secreting angiogenic factors such as VEGF, PDGF and bFGF; (3) TAM promote and maintain production of cancer stem cells by secreting cytokines such as TNF-α, IL-6, and EGF; and (4) TAM induce immunosuppression by suppressing CD8+ T-cells and NK cells and promoting proliferation of immunosuppressive Treg and MDSC through crosstalk between cytokines, metabolic enzymes and/or surface receptors such as IL-10, TGF-β, CCL2, CCL17, CCL20, CCL22, and PD-L1 [29].
MDSC are heterogeneous and immature myeloid cells, functioning as immune suppressor. The mechanism by which MDSC induce immune suppression is complex. Monocytic-MDSC produce nitric oxide (NO) through gene expression of inducible nitric oxide synthase (iNOS) gene. Granulocytic-MDSC produce ROS and arginase 1, which cause cell cycle arrest by amino acid l-arginine depletion and suppressing T cell receptor (TCR)–chain expression [30]. These NO and ROS induce TCR peroxynitration and T cell apoptosis [31]. MDSC activate Treg by secretion of IL-10, and they also activate Th17 cells by secretion of IL-6 and TGF-β [32]. In addition, when MDSC are activated, COX2 [33] and PD-L1 [34] are expressed, resulting in immunosuppression.
TAN, like TAM, can also differentiate into anti-tumor type (N1) and pro-tumor type (N2) [35,36,37]; N1 TAN have high levels of TNFα, CCL3, and ICAM-1 and low levels of arginase, whereas N2 TAN show high levels of CCL2, CCL3, CCL4, CCL8, CCL12, CCL17, CXCL1, CXCL2, IL-8/CXCL8, and CXCL16 [36,37]. The mechanisms by which N1 TAN show anti-tumor activities are antibody-dependent cytotoxicity by neutrophil elastase (NE), activation of innate and adaptive immune cells such as lymphocytes, NK cells and dendritic cells, and tumor cell suppression through ROS production by increasing NADPH oxidase activity [38,39]. The mechanisms by which N2 TAN induce tumor progression are tumor cell proliferation activated by NE, genetic alteration by ROS, CD8+ T cell suppression by arginase depletion, and immunosuppression by activating immunosuppressive Treg [36].
There are a few subsets of TADC comprising TME. Plasmacytoid dendritic cells (pDC) and pre-dendritic cells (pre-DC) are derived from bone marrow macrophage/dendritic cell (DC) progenitors (MDP), and CD11b + DCs and CD103 + DCs come from pre-DC. Inflammatory DCs (inf-DC) are derived from M-MDSC and monocytes. Therefore, TADC comprising TME are pDC, CD11b + DCs, CD103 + DCs, and Inf-DC [40]. DCs induce immune reaction through antigen presentation, and TADC interfere with the normal development of DCs, their activation and function through following mechanisms: (1) DCs are differentiated to tolerogenic DCs that have low expression of MHC molecule and secrete high level IL-10; (2) IL-10 that is produced by TAM suppresses IL-12 production from CD103 + DC, suppressing T cell activation; (3) accumulation of abnormal lipid within TADC lowers tumor antigen presenting capability of TADC; (4) tumor cell hypoxia, lactic acid accumulation, and low pH suppress TADC response to T cells; (5) IDO produced from pDC promote differentiation of immunosuppressive Treg; and (6) various soluble factors secreted from the tumor cells promote differentiation of inf-DC, which secrete tumor promoting IL-6 and immunosuppressive galectin-1 [40].
TMC play the dual role of tumor progression and tumor suppression. The mechanisms by which TMC play a role in tumor progression are as follows: (1) promotion of angiogenesis by heparin-like molecule, histamine, TNF-α, VEGF, platelet activating factor, IL-8, bFGF and prostaglandin that are secreted from TMC [41,42]; (2) promotion of tumor invasion and metastasis through extracellular matrix degradation by chymase, cathepsin G, carboxypeptidase, gelatinase A and B that are secreted from TMC [43,44]; and (3) immune suppression by secreting inhibitory cytokine IL-10 and maintaining Treg activation that is important in immune tolerance [45]. On the other hand, the mechanisms by which TMC play a role in tumor suppression are exocytosis of granules that contain serine protease and tumor cell cytotoxicity through receptor-ligand binding through TNF-α and FasL [46].

2.2.2. Tumor-Associated Lymphoid Cells (TALC)

T-cells and NK cells are two main types of TALC, and T-cells are classified into Treg, cytotoxic T cells, and helper T cells according to cell function. Treg can be found in TME, and they suppress anti-tumor immune reaction by recognizing TCR-antigen complex on MHC molecule [47]. The chemokine-chemokine receptors that are involved in recruiting Treg around tumor area are CCR4-CCL17/22, CCR10-CCL28, and CXCR4-CXCL12 [48]. Cytotoxic T cells are activated by recognizing tumor antigen presented by MHC class I molecule, bind to tumor cells and secrete granzyme B and perforin, and eventually induce tumor cell apoptosis [49]. Helper T cells are subdivided into Th1 and Th2. Th1 activate anti-tumor immune reaction by secreting IL-2 and IFN-γ whereas Th2 suppress anti-tumor immune reaction by secreting IL-4, suppressing NK cells, and lowering tumor antigen expression [50]. NK cells can be further classified into two subtypes, CD56dimCD16+ NK cells and CD56brightCD10−/low NK cells. CD56dimCD16+ NK cells clear tumor cells through cytotoxic activity, and CD56brightCD10−/low NK cells can suppress immune reaction by secreting IL-13 [51,52]. NK cells are activated by Fas ligand-Fas, secrete granzyme B and perforin, and induce tumor cell apoptosis, eventually inducing tumor cell necrosis by ADCC through antibody-FcR complex [53]. The mechanisms by which tumor cells interfere with NK cell function are as follows: (1) secretion of immunosuppressive factors such as TGF-β, IOD, and arginase-1; (2) decreased expression of MHC class I molecules on tumor cells; and (3) inhibition of ligand upregulation and activation of Treg [54].
One of the important mechanisms by which TALC promote tumor progression is immune check point modulation. Among immune check points, one that is important in tumor cells is inhibitory immune check points, namely CTLA-4 and PD-1. CTLA-4 is expressed on the surface of activated lymphocytes, binding to CD80 and CD86 on the surface of APC. This binding suppresses binding of CD80 and CD86 with CD28, suppressing T cell activation [55]. PD-1 is expressed on the surface of activated T cells, B cells, and NK cells, binding with PD-L1 and/or PD-L2 and suppressing CTL function [56].

3. Cell Component and Function of Tumor Microenvironment in Thyroid Cancer

TME in thyroid cancer, like TME in other tumors, are composed of CAF and immune cells and affect tumor biology of tumorigenesis, growth, and progression (Figure 1). First, thyroid cancer can have abundant fibrous stroma, especially in PTC [57,58,59] and ATC [60], suggesting the possible presence of abundant CAF. As the thyroid can often harbor chronic lymphocytic thyroiditis (CLT), many studies have reported that thyroid cancer, especially PTC, is related with CLT [61,62]. Warthin-like variant and diffuse sclerosing variant are histologic subtypes of PTC that are accompanied by an abundance of lymphoplasma cells [63,64]. PTC with CLT has good prognosis with limited disease progression [65,66]; however, PTC with tumor infiltrating lymphocytes is reported to be found at higher cancer stage and have more frequent lymph node metastases [67]. Lymphocyte density is correlated with lower tumor recurrence and longer OS in PTC [65]. In silico analysis study including 799 PTC and 194 normal thyroid samples has found higher levels and proportions of M2 macrophages, Tregs, monocytes, neutrophils, DCs, mast cells (MCs), and M0 macrophages in PTC than in normal thyroid tissues, and also in advanced PTC than in early PTC. It has also found more advanced stages, larger tumor sizes, greater lymph node metastases, higher tall-cell PTCs, lower follicular PTC proportions, more BRAF mutations, and fewer RAS mutations in high-immunity group [68], suggesting that thyroid cancer harbors an abundance of immune cells in TME that affects tumor biology.

3.1. CAF in Thyroid Cancer

A few studies on thyroid cancer tissues have found expression of CAF-related proteins in thyroid cancer stroma [69,70,71], and have revealed that certain CAF-related protein expression is related with cervical lymph node metastasis [69], histologic subtype, BRAF mutation, and prognosis [70]. Increased fibroblast infiltration around the tumor area has been found not only in human thyroid cancer tissues but also in mouse thyroid cancer models [72,73], both induced by oncogenic BRAF. Soluble factors produced by cancer cells are important in CAF generation and/or recruitment in thyroid cancer. It has been found in in vitro co-culture study that conditioned media by human ATC cells (8505c and KTC-2) are involved in reprogramming of thyroid fibroblast phenotype to CAF phenotype [74], and conditioned media by thyroid cancer cells that are induced by BRAF mutation promote CAF proliferation and migration in thyroid cancer mouse model [73]. Conditioned media by human ATC cells (8505c and KTC-2) activate Src and Akt pathway and accelerate human fibroblast proliferation, with increase in the expression of CAF-related markers, namely α-SMA and PDGFR-β [74]. Molecular candidates responsible for the reprogramming of fibroblast phenotype to CAF phenotype are IL-6, ROS, and PDGF [74]. The molecular mechanisms through which CAF affect tumor biology of thyroid cancer are as follows (Figure 2): (1) CAF promote thyroid cancer cell proliferation. Thyroid follicular cell (FRTL-Tc) injection in a mouse model did not develop thyroid cancer, but follicular cell (FRTL-Tc) injection with fibroblast caused tumor development [75]. Moreover, conditioned media by CAF in in vitro co-culture study promoted thyroid cancer cell proliferation, showing a high level of mitogen secretion, IL-6, in CAF [74]; (2) CAF promote migration and invasion in thyroid cancer cells through EMT. In in vitro co-culture study, conditioned media by CAF increased the expression of vimentin, an EMT marker, in thyroid cancer cells, and decreased the expression of E-cadherin, an anti-EMT marker, resulting in increased invasion activity [74]. Moreover, Sonic-Hedgehog ligand secreted by CAF induces ATC cell migration [76]; and (3) CAF induce metabolic reprogramming in thyroid cancer cells. One theory describing metabolic characteristics of cancer cells, the so-called Warburg effect, explains that cancer cells produce energy through glycolysis rather than mitochondrial oxidative phosphorylation [77]. However, the reverse-Warburg effect model describing metabolic crosstalk between cancer cells and TME explains that glycolysis in CAF produces lactate, ketone body, and pyruvate that are transported into cancer cells by lactate shuttle, resulting in energy production through mitochondrial oxidative phosphorylation [78,79,80]. An in vitro cell line study has found that CAF exposed to conditioned media by human ATC cells (8505c and KTC-2) have high levels of glycolysis related molecules, GLUT-1 and LDH-A, by HIF-1α modulation, and glucose uptake and glycolysis are increased as a result [74]. Additionally, an IHC study has confirmed that the expression of MCT4, a lactate shuttle, is increased in the stroma of ATC tissue [81]. CAF promote tumor progression in thyroid cancer through these various mechanisms, and they are related with dedifferentiation and aggressiveness. When mRNA-based CAF gene signature was investigated using thyroid cancer public data, high CAF score was correlated with anaplastic phenotype, poor prognosis, high genetic mutation, and oncogenic signaling pathway [82].

3.2. Immune Cells in Thyroid Cancer

Immune cells in TME in thyroid cancer affect cancer cell biology by various crosstalks (Figure 3). IHC has revealed the presence of CD68 positive TAM in thyroid cancer, and macrophage infiltration rate was significantly increased in PTC compared to in benign tumors and was correlated with lymph node metastasis and poor prognosis [83,84,85,86,87]. TAM density differs according to histologic subtypes; ATC has the highest TAM density and poor prognosis [84]. TAM density is correlated with invasiveness and metastasis in PTC, as a result of CXCL8 secreted from TAM binding with CXCR1/2 secreted from PTC [87]. As in PTC, TAM is related with capsular invasion and extrathyroidal extension in PDTC [85]. PTC employs CSF-1/CSF-1R signaling to recruit TAM as the tumor progresses [72]. TAM infiltration rate is higher in FTC than in follicular adenoma, and CCL15 is involved in this process [88]. TAM in BRAF V600E-related PTC are M2 phenotypes that show high levels of ARG1, CCL22, and IL-10 and a low level of IL-12 [87]. However, controversy exists regarding the association of TAM with prognosis, because there are reports that assert that the number of tumor-infiltrating macrophages is correlated with longer DFS in thyroid cancer patients [89]. The preoperative level of circulating MDSC was higher in thyroid cancer patients than in those with benign thyroid disease [90,91], and the circulating MDSC level was correlated with the aggressiveness of differentiated thyroid cancers [91]. There was no significant correlation between MDSC density and clinicopathologic factors in studies on MDSC in thyroid cancer tissues [89]. Studies on neutrophils in thyroid cancers mainly involved peripheral blood neutrophil-to-lymphocyte ratio (NLR). NLR was correlated with treatment response in thyroid cancers in that NLR was significantly decreased when treatment response was good and thus good prognosis, but NLR was significantly increased when treatment response was not good [92]. Increased NLR was correlated with poor treatment response in multivariate analysis, suggesting that the increased systemic inflammation after treatment incurred poor treatment response [92]. There are, however, reports that NLR is not correlated with benign or malignant tumor behavior and nor with disease prognosis [93]. TAN recruited by CXCL8/IL-8 secrete GM-CSF and promote tumor cell survival and tumor progression, evidenced by research involving human thyroid cancer tissues that have shown correlation between TAN density and tumor size [94]. PTC has increased TADC [95,96], especially CD1a positive TADC, that correlate with improved DFS [97]. TADC recruitment in PTC is due to MIP-1a secreted by HGF, and CCR6 expressing TADC clear damaged thyroid cells [98]. S100 positive TADC in PTC are not related with DFS [95], thus there are differences between TADC phenotypes. TADC are also involved in immune escape in PTC by transforming CD4 positive T cells to FOXP3 positive Treg [96].
Mast cells are one of the immune cells that have been researched more frequently in PTC than other immune cells. IHC for tryptase, a mast cell marker, in human thyroid cancer tissues has revealed that 95% of PTC show mast cells and the amount of TMC was correlated with extrathyroidal tumor extension [99]. TMC are also found in PDTC and ATC, in which TMC density was correlated with tumor invasiveness [100], and TMC density was significantly increased in FVPTC than in follicular adenoma [101]. Thyroid cancer cell line study has shown that VEGF-A is secreted from thyroid cancer cells and it promotes mast cell chemotaxis. Thus, it is suggested that VEGF pathway is involved in TMC recruitment in PTC [99]. TMC can be found in various thyroid cancer tissues and they are related with tumor aggressiveness, due to various soluble factors they produce, which are mostly non-IgE related. Histamine, IL-6, IL-1, TNF-α, and chemokines such as CXCL1/GRO-α, CXCL8/IL-8, and CXCL10/IP-10 are secreted from mast cells when thyroid cancer cells are activated [89]. First, TMC promote thyroid cancer cell proliferation by producing histamine that binds to H1 and H2 receptors in PTC tumor cells. A mouse xenograft study has suggested histamine, CXCL1/GRO-α and CXCL10/IP-10 as TMC secreting factors that are important in thyroid cancer cell proliferation [29]. Second, TMC initiate EMT, evidenced by studies using thyroid cancer cell lines (PTC, FTC, and ATC) that showed an EMT process of morphological change of thyroid cancer cells to spindle cells, increased EMT markers, and decreased epithelial markers when the cancer cells were exposed to conditioned media by activated mast cells [100]. TMC secreting factors that are essential in this EMT process are TNF, IL-6 and CXCL8/IL-8. CXCL8/IL-8 is especially important in the process [100]. Third, TMC induce cancer cell stemness, showing enhanced stemness features when thyroid cancer cells are exposed to conditioned media by mast cells or when exposed to recombinant CXCL8/IL-8 [100]. Such EMT and cancer cell stemness by TMC in thyroid cancer cells are activated by AKT/SLUG pathway. In IHC study of tryptase, a mast cell marker, and OCT-4, a stem cell marker, using human PTC tissues, there was a significant positive correlation between TMC density and OCT-4 expression. TMC density and OCT-4 expression were also correlated with higher T stage [100].
Treg, a subtype of T-cell, are found in PTC tissues in a significantly larger number than in benign goiter [96,102], and Treg in PTC are correlated with extrathyroidal extension and LN metastasis [102]. Additionally, Treg are found in abundance in metastatic lymph nodes of PTC, and they are correlated with recurrent PTC [103]. Treg are also related with the aggressiveness of papillary microcarcinoma, due to the immune suppression incurred by activation of Treg differentiation promoted by increased expression of IDO1 in tumor cells [104]. The mechanism by which Treg are recruited in PTC is transformation of CD4+ T cells into FoxP3+ICOS+Treg through inducible costimulatory (ICOS) ligand by plasmacytoid DC in PTC microenvironment [96]. In IHC study of FoxP3 involving thyroid cancer tissues (PTC and FTC) and benign thyroid tissue, FoxP3 expression was correlated with tumor aggressiveness, and tumor size was inversely correlated with FoxP3 expression [105]. New subtypes of T cells are suggested in thyroid cancer; (1) DN T cells (CD3+CD4-CD8-double negative T cells) comprise the main population of T cells in PTC TME, suppressing proliferation of activated T cells and hence decreased cytokine production [106]. CD4+IL17+ T cells (Th17) are more abundant in DTC than in benign thyroid tissue [89], and Th17 density is significantly higher in thyroid tumor than in normal thyroid tissue. Th17 density is also correlated with the serum level of IL-17 [107]. In contrast with Treg, CD8+ T cell concentration is lower in PTC patients, and CD8+ T cell/FoxP3+ Treg ratio is inversely correlated with tumor size [67]. It has been reported that CD8+ T cell infiltration in differentiated thyroid cancer is correlated with increased tumor recurrence [108], but there are reports of the contrary, that CD8+ T cell infiltration is correlated with improved DFS [89], because CD8+ T cell is in anergy without granzyme B [108]. In a gene informatics study, CLDN10 is related with CD8+ T cell infiltration in PTC and associated with good prognosis [109].
In a flow cytometry study, NK cells are significantly more abundant in PTC than in benign nodular goiter, but more abundant in early PTC than in advanced PTC [110]. NK cell dysfunction through PD-1/TIM-3 pathway in TME immune cells [111], and decreased expression of activated receptors for NK cells, such as NGK2D, by increased COX2 expression in tumor cells are two mechanisms by which tumor cells avoid cell lysis by NK cells in ATC [112]. ATC cells are sensitive to tumor cell lysis by ULBP2/5/6 and CXCR3-positive NK cells [112], and injection of NK cells in a mouse model of ATC pulmonary metastasis suppressed the growth of pulmonary metastatic tumor [113].

3.3. Immunologic Classification of Thyroid Cancer

Various immune cell infiltration in thyroid cancers is affected by various factors, but especially so by tumor genomic features. Thyroid cancers are classified according to histologic classification, and PTC can be further classified as BRAFV600E-like (BVL)-PTC and RAS-like (RS) PTC according to molecular classification on the basis of TCGA data [114]. BVL PTC and RS PTC have different genetic, epigenetic, and proteomic features, and development of BRAFV600E-RAS score (BRS) system can help elucidate molecular characteristics of PTC [115]. Certain genetic mutations in thyroid cancers can have various effects on immune cells. BRAF V600E mutation in PTC is associated with increased DC, TAM and mast cells, and also with elevated expression levels of CTLA-4, PD-L1 and PD-L2 [116]. It is also associated with Treg and immunosuppressive TAM [117]. TERT promotor methylation and BRAF V600E mutation are associated with PD-L1 expression in primary thyroid cancer [118]. RET/PTC3 genetic mutation promotes IDO1 expression through STAT1-IRF1 pathway [119]. IDO1 suppresses activated T cell proliferation and promotes Treg differentiation, controlling immune cell population phenotype in RET mutated thyroid cancer. Accordingly, such immunologic features can serve as classification criteria for thyroid cancers. A large scale immunogenomic analysis using TCGA data classified tumors into six immune subtypes (C1-C6) of wound healing, IFN-γ dominant, inflammatory, lymphocyte depleted, immunologically quiet, and TGF-β dominant [120]. These subtypes differ in lymphocyte/macrophage signature, Th1:Th2 ratio, intratumoral heterogeneity status, aneuploidy, neoantigen level, cell proliferation level, immunomodulatory gene expression, and prognosis. PTC is classified as inflammatory subtype, with balanced macrophage/lymphocyte ratio, low intratumoral heterogeneity, low aneuploidy level, low level of somatic copy number alteration, low cell proliferation, and high expression of genes representing Th17 differentiation. Another study using TCGA data and ESTIMATE datasets found 793 differentially expressed genes in thyroid cancer TME, and these genes were associated with immune score and stromal score [121]. Immune score and stromal score can be obtained from ESTIMATE website and they can predict immune cell and stromal cell infiltration in TME. Immunoscore has been developed in PTC, which can be obtained from the amount of CD3+ and CD8+ T cell population from the center of the tumor (CT) and from the invasive margin (IM). Immunoscore ranges from immunoscore 0 (I0) to immunoscore 4 (I4). [122]. I0 means low CD3+ and CD8+ T cell density in CT and IM, I4 means high CD3+ and CD8+ T cell density in CT and IM. In a study using TCGA data of PTC, immunoscore was negatively correlated with thyroid differentiation score (TDS), and high immunoscore was found in BRAFV600E mutated PTC [116]. An NGS study involving thyroid cancer tissues (25 PTC, 14 PDTC, and 13 ATC) and normal thyroid tissues (7 NT) has reported two clusters with immune-related genes [123]. The first cluster is composed of ATC and some of PTC showing high regulation of immune-related genes, and the second cluster is composed of PDTC and a part of PTC group showing low regulation of immune-related genes. According to the study results, the TAM and CD8+ T cell density in TME is highest in ATC, followed by PTC and PDTC. Another study has suggested four categories of thyroid cancer (hot, altered-immunosuppressed, altered-excluded, and cold) based on immune cell density and immunoscores [124]. Hot tumor is characterized by high T cell and CTL infiltration in CT and IM, high immunoscore, and suppressed T cell function due to activated immune checkpoints such as PD-1, CTLA-4, TIM3, and LAG3. Altered-immunosuppressed tumor shows an intermediate degree of T cell and CTL infiltration and immunoscore, T cell checkpoints (PD-1, CTLA-4, TIM3, and LAG3), immune suppressive cells (MDSC and Treg), and inhibitory cytokines (TGF-, IL-10 and VEGF). Altered-excluded tumor does not harbor T cell infiltration in CT, shows intermediate immunoscore in IM, and is associated with oncogenic activation, aberrant vasculature/stroma, and hypoxia. Cold tumor has the lowest immunoscore, no T cell infiltration in CT and IM, low tumor mutation burden, low antigen presentation, and T cell insensitivity. Another study has subgrouped thyroid cancers into ATC-like and PDTC-like phenotypes on the basis of NGS and immunoscore data with CD3+ cell density [125]. ATC-like tumor shows high levels of T cell infiltration, chemo-cytokines (CCL2, CCL3, CCL4, CCL5, CXCL9, and CXCL10), and immune checkpoints. PDTC-like tumor is characterized by low levels of T cell infiltration, chemo-cytokines and immune checkpoints. In this study, 50% of PTC were ATC-like and the rest were PDTC-like [125].

4. Future Targets for Thyroid Cancer Treatment

As has been described heretofore, many preclinical and clinical studies have been carried out on CAF and immune cells to explore their potential as treatment targets. CAF and immune cells are important components of TME in thyroid cancers and they play important roles in tumor biology.

4.1. Targeting CAF for Thyroid Cancer Treatment

CAF can be a good treatment target because they are genetically stable, they are important in maintaining ECM framework of cancer, and they are major barriers to cancer cells against anticancer drug. There are a few general strategies in using CAF as a cancer treatment target; (1) targeting biophysical stromal barrier in order to effectively transport drugs, (2) suppressing molecules secreted from CAF that activate cancer cells, (3) blocking ECM component in order to lower adhesion-induced signaling, and (4) suppressing CAF in order to suppress CAF downstream pathway. There are four strategic categories of CAF inhibitors (Table 1): (1) When the stromal barrier is targeted, the targets are TGF-β, MMPs, and Hedgehog which suppressed the tumorigenesis, migration, and invasion in ATC cells (8505C and/or SW1736) when TGF-β1 was inhibited [126,127]. BB94, an MMP inhibitor, suppressed migration and invasion of ATC cells [127], and when minocycline, an MMP inhibitor, was injected with manumycin and paclitaxel in ATC xenograft study, the tumor size was smaller than when single drug was administered [128]. When cyclopamine, a Hedgehog inhibitor, was administered, the expression of CSC-related transcription factors, B lymphoma Mo-MLV insertion region 1 homolog (BMI1) and SRY-Box Transcription Factor 2 (SOX2), were decreased and the growth of CSC-derived tumor xenografts was inhibited [129]. (2) When CAF secreted molecules are suppressed, the potential targets are CTGF, HGF-c-MET pathway, and CXCR4-CXCL12 axis. When PHA665752, a c-met inhibitor, was administered in PTC cells, HGF dependent cell growth, cell survival, cell invasion, and migration were suppressed by inhibition of c-MET phosphorylation [130]. c-MET inhibitors (tivantinib and crizotinib) suppressed cell growth in 50% of thyroid cancer cell lines [131]. AMD3100, a CXCR4 antagonist, inhibited tumor cell proliferation, invasion and xenograft tumor formation in PTC tumorigenic cell line (BHP10-3M) [132], and BAY11-7082, a CXCR4-CXCL12 axis inhibitor, suppressed CXCL12-CXCR4-induced migration, invasion, and EMT processes by inhibiting NF-κB signaling pathway in PTC cell line (B-CPAP) [133]. (3) When interaction with ECM is suppressed, the key targets are β-integrin and CD44. T315, an integrin-linked kinase inhibitor in thyroid cancer, inhibits thyroid cancer cell migration and shows cytotoxicity even in a very low level [134], and QLT0267, an integrin-linked kinase inhibitor, suppresses tumor cell growth and decreases xenograft tumor size in thyroid cancer cells, especially in ATC [135]. (4) When CAF itself and differentiation into CAF are suppressed, the key targets are FAPα, PDGFR kinase, and VDR ligand, but there has been no report on thyroid cancer.

4.2. Targeting Immune Cells for Thyroid Cancer Treatment

There are three strategies in targeting immune cells for cancer treatment in general: (1) tumor vaccines using specific tumor-associated antigen (TAA); (2) adoptive cell therapy using immunocompetent cells segregated from the tumor; and (3) immune checkpoint suppression that increases tumor suppressive ability of immune cells. Studies on immune therapy against thyroid cancer have mainly involved advanced cancer that do not respond to classic treatment or show treatment resistance (Table 2). NY-ESO-1 is a TAA discovered in thyroid cancer, especially in MTC [137,138]. MTC secretes carcinoembryonic antigen (CEA), and Yeast-CEA (GI-6207) vaccine was studied in a clinical trial on metastatic MTC [139]. Neo-antigen that can be used for tumor vaccine is mostly found in abundance in ATC that usually has a high level of mutation burden, and thus tumor vaccine may be more effective in ATC [115]. Oncolytic virus (OV) vaccine that initiates systemic antitumor immunity by tumor cell lysis has been tried in thyroid cancer, and dl922-947, an OV, not only suppressed tumor growth in ATC mouse model but also switched M2 TAM to M1 TAM [140,141,142]. Adoptive cell therapy for thyroid cancer involved injection of tumor-lysate-pulsed DC in advanced PTC, FTC, and MTC, and resulted in palliation of symptoms [143,144]. In a preclinical study of chimeric antigen receptor (CAR)-T cell therapy targeting ICAM-1 in PTC and ATC, tumor growth was suppressed effectively and patient survival improved [145,146]. Monoclonal antibodies for CTLA-4, PD-L1, and PD-1 are key immune checkpoint inhibitors, and when pembrolizumab, a PD-1 inhibitor, was administered in advanced PTC and FTC with PD-L1 expression, there was no major side effect and a few patients experienced symptom relief [147]. In addition, when pembrolizumab was administered with tyrosine kinase inhibitor in ATC patients showing PD-L1 expression, patient survival was extended [148]. The tumor size was decreased and survival period was extended in murine ATC model when anti-PD-1/PD-L1 antibody was administered with BRAF inhibitor [149]. PD-1 blockade together with CAR-T therapy for ICAM-1 in ATC showed tumor suppression and better survival in a mouse xenograft model than CAR-T therapy alone [150].

5. Conclusions

Like in other tumors, TME is important in tumorigenesis and tumor progression in thyroid cancer. CAF, one of the main cellular components of TME, are more abundant in fibrous tumor stroma of PTC and ATC among thyroid cancers. Types and functions of CAF have been studied in breast and pancreatic cancers, but not much in thyroid cancers. However, studies to date have reported that CAF can promote thyroid cancer proliferation and progression, and thus CAF suppression may be targeted for treatment in thyroid cancers, especially for refractory thyroid cancers and ATC that does not have effective treatment modality. Studies on CAF inhibitors in thyroid cancers are rather primitive yet and most are preclinical studies, so further studies are warranted. Effective suppression of CAF calls for understanding of specific CAF markers, but as aforementioned, CAF have various cell origins and harbor various markers that have plasticity in phenotypes. This CAF phenotype plasticity is a major barrier in applying CAF inhibitors in thyroid cancers, and thus further research is necessary.
Immune cells, another main cellular component in TME along with CAF, are also important in tumor biology and hence they may be possible treatment targets. A few points should be considered when immune cells are used for thyroid cancer treatment: (1) There are various types and differentiation status. Some studies have reported immune cells to be tumor progressor and some to be tumor suppressor. The role of immune cells in thyroid cancers according to immune cell type and differentiation status has to be studied further and single cell analysis would be important to meet this end. (2) Immune status of thyroid cancers should be considered. As aforementioned, when thyroid cancers are subclassified according to the genetic and immune features, types whose immune system is activated (ATC-like, hot, and altered-immunosuppressed type) and those whose immune system is not activated (PDTC-like, cold, and altered-excluded type), the former recover antitumor immune response by clearing away immunosuppressive signals or immune suppressing cells. The latter, on the other hand, are directed at tumor killing by immune cells through immune cell recruitment to around the tumor cells. (3) Biomarkers that are most effective for immunotherapy should be discovered. Possible biomarkers for cancer immunotherapy in general are PD-L1, tumor mutation burden (TMB), microsatellite instability (MSI), specific gene mutation, immunogenic neo-antigen, and gut microbiome [151], and possible application of these biomarkers for immunotherapy in thyroid cancers should be explored in further studies. PD-L1, for example, is an FDA-approved biomarker for immunotherapy, but its clonal types, interpretation method, and positive criteria are all differently established according to cancer types. Therefore, it is essential to discover effective biomarkers for targeted immune therapy that can be tailored for thyroid cancers.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors would like to thank Dong-Su Jang, MFA (Medical Illustrator), for his help with the illustrations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Carling, T.; Udelsman, R. Thyroid cancer. Annu. Rev. Med. 2014, 65, 125–137. [Google Scholar] [CrossRef]
  2. Davies, L.; Welch, H.G. Current thyroid cancer trends in the united states. JAMA Otolaryngol. Head Neck Surg. 2014, 140, 317–322. [Google Scholar] [CrossRef] [Green Version]
  3. Kitahara, C.M.; Sosa, J.A. The changing incidence of thyroid cancer. Nat. Rev. Endocrinol. 2016, 12, 646–653. [Google Scholar] [CrossRef]
  4. Cabanillas, M.E.; McFadden, D.G.; Durante, C. Thyroid cancer. Lancet 2016, 388, 2783–2795. [Google Scholar] [CrossRef]
  5. Nikiforova, M.N.; Nikiforov, Y.E. Molecular genetics of thyroid cancer: Implications for diagnosis, treatment and prognosis. Expert Rev. Mol. Diagn. 2008, 8, 83–95. [Google Scholar] [CrossRef]
  6. Molinaro, E.; Romei, C.; Biagini, A.; Sabini, E.; Agate, L.; Mazzeo, S.; Materazzi, G.; Sellari-Franceschini, S.; Ribechini, A.; Torregrossa, L.; et al. Anaplastic thyroid carcinoma: From clinicopathology to genetics and advanced therapies. Nat. Rev. Endocrinol. 2017, 13, 644–660. [Google Scholar] [CrossRef] [PubMed]
  7. Filetti, S.; Durante, C.; Hartl, D.; Leboulleux, S.; Locati, L.D.; Newbold, K.; Papotti, M.G.; Berruti, A. Thyroid cancer: Esmo clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 1856–1883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Grani, G.; Lamartina, L.; Durante, C.; Filetti, S.; Cooper, D.S. Follicular thyroid cancer and hürthle cell carcinoma: Challenges in diagnosis, treatment, and clinical management. Lancet Diabetes Endocrinol. 2018, 6, 500–514. [Google Scholar] [CrossRef]
  9. Tuttle, R.M. Controversial issues in thyroid cancer management. J. Nucl. Med. 2018, 59, 1187–1194. [Google Scholar] [CrossRef] [Green Version]
  10. Raue, F.; Frank-Raue, K. Thyroid cancer: Risk-stratified management and individualized therapy. Clin. Cancer Res. 2016, 22, 5012–5021. [Google Scholar]
  11. Cirri, P.; Chiarugi, P. Cancer associated fibroblasts: The dark side of the coin. Am. J. Cancer Res. 2011, 1, 482–497. [Google Scholar]
  12. Öhlund, D.; Elyada, E.; Tuveson, D. Fibroblast heterogeneity in the cancer wound. J. Exp. Med. 2014, 211, 1503–1523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef] [PubMed]
  14. Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.M.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
  15. LeBleu, V.S.; Kalluri, R. A peek into cancer-associated fibroblasts: Origins, functions and translational impact. Dis. Model. Mech. 2018, 11, dmm029447. [Google Scholar] [CrossRef] [Green Version]
  16. Chen, Y.; McAndrews, K.M.; Kalluri, R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat. Rev. Clin. Oncol. 2021, 18, 792–804. [Google Scholar] [CrossRef]
  17. Geng, X.; Chen, H.; Zhao, L.; Hu, J.; Yang, W.; Li, G.; Cheng, C.; Zhao, Z.; Zhang, T.; Li, L.; et al. Cancer-associated fibroblast (caf) heterogeneity and targeting therapy of cafs in pancreatic cancer. Front. Cell Dev. Biol. 2021, 9, 655152. [Google Scholar] [CrossRef]
  18. Mhaidly, R.; Mechta-Grigoriou, F. Role of cancer-associated fibroblast subpopulations in immune infiltration, as a new means of treatment in cancer. Immunol. Rev. 2021, 302, 259–272. [Google Scholar] [CrossRef]
  19. Martinez, F.O.; Sica, A.; Mantovani, A.; Locati, M. Macrophage activation and polarization. Front. Biosci. 2008, 13, 453–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Cassetta, L.; Cassol, E.; Poli, G. Macrophage polarization in health and disease. ScientificWorldJournal 2011, 11, 2391–2402. [Google Scholar] [CrossRef] [Green Version]
  21. Montes, V.N.; Turner, M.S.; Subramanian, S.; Ding, Y.; Hayden-Ledbetter, M.; Slater, S.; Goodspeed, L.; Wang, S.; Omer, M.; Den Hartigh, L.J.; et al. T cell activation inhibitors reduce cd8+ t cell and pro-inflammatory macrophage accumulation in adipose tissue of obese mice. PLoS ONE 2013, 8, e67709. [Google Scholar] [CrossRef]
  22. Xu, Y.; Romero, R.; Miller, D.; Kadam, L.; Mial, T.N.; Plazyo, O.; Garcia-Flores, V.; Hassan, S.S.; Xu, Z.; Tarca, A.L.; et al. An m1-like macrophage polarization in decidual tissue during spontaneous preterm labor that is attenuated by rosiglitazone treatment. J. Immunol. 2016, 196, 2476–2491. [Google Scholar] [CrossRef]
  23. Nelson, M.P.; Christmann, B.S.; Dunaway, C.W.; Morris, A.; Steele, C. Experimental pneumocystis lung infection promotes m2a alveolar macrophage-derived mmp12 production. Am. J. Physiol. Lung Cell Mol. Physiol. 2012, 303, L469–L475. [Google Scholar] [CrossRef] [Green Version]
  24. Ohama, H.; Asai, A.; Ito, I.; Suzuki, S.; Kobayashi, M.; Higuchi, K.; Suzuki, F. M2b macrophage elimination and improved resistance of mice with chronic alcohol consumption to opportunistic infections. Am. J. Pathol. 2015, 185, 420–431. [Google Scholar] [CrossRef] [PubMed]
  25. Lu, J.; Cao, Q.; Zheng, D.; Sun, Y.; Wang, C.; Yu, X.; Wang, Y.; Lee, V.W.; Zheng, G.; Tan, T.K.; et al. Discrete functions of m2a and m2c macrophage subsets determine their relative efficacy in treating chronic kidney disease. Kidney Int. 2013, 84, 745–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Cao, W.; Peters, J.H.; Nieman, D.; Sharma, M.; Watson, T.; Yu, J. Macrophage subtype predicts lymph node metastasis in oesophageal adenocarcinoma and promotes cancer cell invasion in vitro. Br. J. Cancer 2015, 113, 738–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Martinez, F.O.; Gordon, S. The m1 and m2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 2014, 6, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Laviron, M.; Boissonnas, A. Ontogeny of tumor-associated macrophages. Front. Immunol. 2019, 10, 1799. [Google Scholar] [CrossRef] [Green Version]
  29. Petty, A.J.; Yang, Y. Tumor-associated macrophages: Implications in cancer immunotherapy. Immunotherapy 2017, 9, 289–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Weber, R.; Fleming, V.; Hu, X.; Nagibin, V.; Groth, C.; Altevogt, P.; Utikal, J.; Umansky, V. Myeloid-derived suppressor cells hinder the anti-cancer activity of immune checkpoint inhibitors. Front. Immunol. 2018, 9, 1310. [Google Scholar] [CrossRef] [Green Version]
  31. Nagaraj, S.; Gupta, K.; Pisarev, V.; Kinarsky, L.; Sherman, S.; Kang, L.; Herber, D.L.; Schneck, J.; Gabrilovich, D.I. Altered recognition of antigen is a mechanism of cd8+ t cell tolerance in cancer. Nat. Med. 2007, 13, 828–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Pan, P.Y.; Ma, G.; Weber, K.J.; Ozao-Choy, J.; Wang, G.; Yin, B.; Divino, C.M.; Chen, S.H. Immune stimulatory receptor cd40 is required for t-cell suppression and t regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. 2010, 70, 99–108. [Google Scholar] [CrossRef]
  33. Chen, J.; Cai, S.; Li, R.; Xie, J.; Yang, F.; Liu, T. Blockade of cycloxygenase-2 ameliorates sepsis induced immune-suppression by regulating myeloid-derived suppressor cells. Int. Immunopharmacol. 2022, 104, 108506. [Google Scholar] [CrossRef]
  34. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. Pd-l1 is a novel direct target of hif-1α, and its blockade under hypoxia enhanced mdsc-mediated t cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [Green Version]
  35. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by tgf-beta: “N1” versus “n2” tan. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [Green Version]
  36. Masucci, M.T.; Minopoli, M.; Carriero, M.V. Tumor associated neutrophils. Their role in tumorigenesis, metastasis, prognosis and therapy. Front. Oncol. 2019, 9, 1146. [Google Scholar] [CrossRef] [Green Version]
  37. Fridlender, Z.G.; Albelda, S.M. Tumor-associated neutrophils: Friend or foe? Carcinogenesis 2012, 33, 949–955. [Google Scholar] [CrossRef] [Green Version]
  38. Mantovani, A.; Cassatella, M.A.; Costantini, C.; Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 2011, 11, 519–531. [Google Scholar] [CrossRef]
  39. Granot, Z.; Henke, E.; Comen, E.A.; King, T.A.; Norton, L.; Benezra, R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell 2011, 20, 300–314. [Google Scholar] [CrossRef] [Green Version]
  40. Veglia, F.; Gabrilovich, D.I. Dendritic cells in cancer: The role revisited. Curr. Opin. Immunol. 2017, 45, 43–51. [Google Scholar] [CrossRef] [Green Version]
  41. Coussens, L.M.; Raymond, W.W.; Bergers, G.; Laig-Webster, M.; Behrendtsen, O.; Werb, Z.; Caughey, G.H.; Hanahan, D. Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis. Genes Dev. 1999, 13, 1382–1397. [Google Scholar] [CrossRef]
  42. Norrby, K. Mast cells and de novo angiogenesis: Angiogenic capability of individual mast-cell mediators such as histamine, tnf, il-8 and bfgf. Inflamm. Res. 1997, 46, 7–8. [Google Scholar] [CrossRef]
  43. Ch’ng, S.; Wallis, R.A.; Yuan, L.; Davis, P.F.; Tan, S.T. Mast cells and cutaneous malignancies. Mod. Pathol. 2006, 19, 149–159. [Google Scholar] [CrossRef] [Green Version]
  44. Rojas, I.G.; Spencer, M.L.; Martínez, A.; Maurelia, M.A.; Rudolph, M.I. Characterization of mast cell subpopulations in lip cancer. J. Oral Pathol. Med. 2005, 34, 268–273. [Google Scholar] [CrossRef]
  45. Huang, B.; Lei, Z.; Zhang, G.M.; Li, D.; Song, C.; Li, B.; Liu, Y.; Yuan, Y.; Unkeless, J.; Xiong, H.; et al. Scf-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment. Blood 2008, 112, 1269–1279. [Google Scholar] [CrossRef]
  46. Ozdemir, O. Immunosurveillance function of human mast cell? World J. Gastroenterol. 2005, 11, 7054–7056. [Google Scholar] [CrossRef] [Green Version]
  47. Whiteside, T.L. Foxp3+ treg as a therapeutic target for promoting anti-tumor immunity. Expert Opin. Ther. Targets 2018, 22, 353–363. [Google Scholar] [CrossRef]
  48. Yin, H.; Tang, Y.; Guo, Y.; Wen, S. Immune microenvironment of thyroid cancer. J. Cancer 2020, 11, 4884–4896. [Google Scholar] [CrossRef]
  49. Mahmoud, S.M.; Paish, E.C.; Powe, D.G.; Macmillan, R.D.; Grainge, M.J.; Lee, A.H.; Ellis, I.O.; Green, A.R. Tumor-infiltrating cd8+ lymphocytes predict clinical outcome in breast cancer. J. Clin. Oncol. 2011, 29, 1949–1955. [Google Scholar] [CrossRef]
  50. Akdis, M.; Burgler, S.; Crameri, R.; Eiwegger, T.; Fujita, H.; Gomez, E.; Klunker, S.; Meyer, N.; O’Mahony, L.; Palomares, O.; et al. Interleukins, from 1 to 37, and interferon-γ: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2011, 127, e701–e770. [Google Scholar] [CrossRef]
  51. Robertson, F.C.; Berzofsky, J.A.; Terabe, M. Nkt cell networks in the regulation of tumor immunity. Front. Immunol. 2014, 5, 543. [Google Scholar] [CrossRef] [Green Version]
  52. Galdiero, M.R.; Varricchi, G.; Marone, G. The immune network in thyroid cancer. Oncoimmunology 2016, 5, e1168556. [Google Scholar] [CrossRef]
  53. Trapani, J.A.; Smyth, M.J. Functional significance of the perforin/granzyme cell death pathway. Nat. Rev. Immunol. 2002, 2, 735–747. [Google Scholar] [CrossRef]
  54. Bi, J.; Tian, Z. Nk cell dysfunction and checkpoint immunotherapy. Front. Immunol. 2019, 10, 1999. [Google Scholar] [CrossRef]
  55. Krummel, M.F.; Allison, J.P. Cd28 and ctla-4 have opposing effects on the response of t cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [Green Version]
  56. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. Pd-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [Green Version]
  57. Naganuma, H.; Iwama, N.; Nakamura, Y.; Ohtani, N.; Ohtani, H.; Takaya, K.; Sakai, N. Papillary carcinoma of the thyroid gland forming a myofibroblastic nodular tumor: Report of two cases and review of the literature. Pathol. Int. 2002, 52, 54–58. [Google Scholar] [CrossRef]
  58. Suster, D.; Michal, M.; Nishino, M.; Piana, S.; Bongiovanni, M.; Blatnik, O.; Hájková, V.; Ptáková, N.; Michal, M.; Suster, S. Papillary thyroid carcinoma with prominent myofibroblastic stromal component: Clinicopathologic, immunohistochemical and next-generation sequencing study of seven cases. Mod. Pathol. 2020, 33, 1702–1711. [Google Scholar] [CrossRef]
  59. Takada, N.; Hirokawa, M.; Ito, M.; Ito, A.; Suzuki, A.; Higuchi, M.; Kuma, S.; Hayashi, T.; Kishikawa, M.; Horikawa, S.; et al. Papillary thyroid carcinoma with desmoid-type fibromatosis: A clinical, pathological, and immunohistochemical study of 14 cases. Endocr. J. 2017, 64, 1017–1023. [Google Scholar] [CrossRef] [Green Version]
  60. Dahlman, T.; Lammerts, E.; Wik, M.; Bergström, D.; Grimelius, L.; Westermark, K.; Rubin, K.; Heldin, N.E. Fibrosis in undifferentiated (anaplastic) thyroid carcinomas: Evidence for a dual action of tumour cells in collagen type i synthesis. J. Pathol. 2000, 191, 376–386. [Google Scholar] [CrossRef]
  61. Di Pasquale, M.; Rothstein, J.L.; Palazzo, J.P. Pathologic features of hashimoto’s-associated papillary thyroid carcinomas. Hum. Pathol. 2001, 32, 24–30. [Google Scholar] [CrossRef]
  62. Ott, R.A.; McCall, A.R.; McHenry, C.; Jarosz, H.; Armin, A.; Lawrence, A.M.; Paloyan, E. The incidence of thyroid carcinoma in hashimoto’s thyroiditis. Am. Surg. 1987, 53, 442–445. [Google Scholar]
  63. Yeo, M.K.; Bae, J.S.; Lee, S.; Kim, M.H.; Lim, D.J.; Lee, Y.S.; Jung, C.K. The warthin-like variant of papillary thyroid carcinoma: A comparison with classic type in the patients with coexisting hashimoto’s thyroiditis. Int. J. Endocrinol. 2015, 2015, 456027. [Google Scholar] [CrossRef]
  64. Wang, Q.; Chang, Q.; Zhang, R.; Sun, C.; Li, L.; Wang, S.; Wang, Q.; Li, Z.; Niu, L. Diffuse sclerosing variant of papillary thyroid carcinoma: Ultrasonographic and clinicopathological features in children/adolescents and adults. Clin. Radiol. 2022, 77, e356–e362. [Google Scholar] [CrossRef]
  65. Matsubayashi, S.; Kawai, K.; Matsumoto, Y.; Mukuta, T.; Morita, T.; Hirai, K.; Matsuzuka, F.; Kakudoh, K.; Kuma, K.; Tamai, H. The correlation between papillary thyroid carcinoma and lymphocytic infiltration in the thyroid gland. J. Clin. Endocrinol. Metab. 1995, 80, 3421–3424. [Google Scholar]
  66. Fiumara, A.; Belfiore, A.; Russo, G.; Salomone, E.; Santonocito, G.M.; Ippolito, O.; Vigneri, R.; Gangemi, P. In situ evidence of neoplastic cell phagocytosis by macrophages in papillary thyroid cancer. J. Clin. Endocrinol. Metab. 1997, 82, 1615–1620. [Google Scholar] [CrossRef]
  67. French, J.D.; Weber, Z.J.; Fretwell, D.L.; Said, S.; Klopper, J.P.; Haugen, B.R. Tumor-associated lymphocytes and increased foxp3+ regulatory t cell frequency correlate with more aggressive papillary thyroid cancer. J. Clin. Endocrinol. Metab. 2010, 95, 2325–2333. [Google Scholar] [CrossRef]
  68. Xie, Z.; Li, X.; He, Y.; Wu, S.; Wang, S.; Sun, J.; He, Y.; Lun, Y.; Zhang, J. Immune cell confrontation in the papillary thyroid carcinoma microenvironment. Front. Endocrinol. 2020, 11, 570604. [Google Scholar] [CrossRef]
  69. Cho, J.G.; Byeon, H.K.; Oh, K.H.; Baek, S.K.; Kwon, S.Y.; Jung, K.Y.; Woo, J.S. Clinicopathological significance of cancer-associated fibroblasts in papillary thyroid carcinoma: A predictive marker of cervical lymph node metastasis. Eur. Arch. Otorhinolaryngol. 2018, 275, 2355–2361. [Google Scholar] [CrossRef]
  70. Sun, W.Y.; Jung, W.H.; Koo, J.S. Expression of cancer-associated fibroblast-related proteins in thyroid papillary carcinoma. Tumour. Biol. 2016, 37, 8197–8207. [Google Scholar] [CrossRef]
  71. Minna, E.; Brich, S.; Todoerti, K.; Pilotti, S.; Collini, P.; Bonaldi, E.; Romeo, P.; Cecco, L.; Dugo, M.; Perrone, F.; et al. Cancer associated fibroblasts and senescent thyroid cells in the invasive front of thyroid carcinoma. Cancers 2020, 12, 112. [Google Scholar] [CrossRef] [Green Version]
  72. Ryder, M.; Gild, M.; Hohl, T.M.; Pamer, E.; Knauf, J.; Ghossein, R.; Joyce, J.A.; Fagin, J.A. Genetic and pharmacological targeting of csf-1/csf-1r inhibits tumor-associated macrophages and impairs braf-induced thyroid cancer progression. PLoS ONE 2013, 8, e54302. [Google Scholar]
  73. Jolly, L.A.; Novitskiy, S.; Owens, P.; Massoll, N.; Cheng, N.; Fang, W.; Moses, H.L.; Franco, A.T. Fibroblast-mediated collagen remodeling within the tumor microenvironment facilitates progression of thyroid cancers driven by brafv600e and pten loss. Cancer Res. 2016, 76, 1804–1813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Fozzatti, L.; Alamino, V.A.; Park, S.; Giusiano, L.; Volpini, X.; Zhao, L.; Stempin, C.C.; Donadio, A.C.; Cheng, S.Y.; Pellizas, C.G. Interplay of fibroblasts with anaplastic tumor cells promotes follicular thyroid cancer progression. Sci. Rep. 2019, 9, 8028. [Google Scholar] [CrossRef] [Green Version]
  75. Saitoh, O.; Mitsutake, N.; Nakayama, T.; Nagayama, Y. Fibroblast-mediated in vivo and in vitro growth promotion of tumorigenic rat thyroid carcinoma cells but not normal fisher rat thyroid follicular cells. Thyroid 2009, 19, 735–742. [Google Scholar] [CrossRef] [PubMed]
  76. Parascandolo, A.; Laukkanen, M.O.; De Rosa, N.; Ugolini, C.; Cantisani, M.C.; Cirafici, A.M.; Basolo, F.; Santoro, M.; Castellone, M.D. A dual mechanism of activation of the sonic hedgehog pathway in anaplastic thyroid cancer: Crosstalk with ras-braf-mek pathway and ligand secretion by tumor stroma. Oncotarget 2018, 9, 4496–4510. [Google Scholar] [CrossRef] [Green Version]
  77. Schwartz, L.; Supuran, C.T.; Alfarouk, K.O. The warburg effect and the hallmarks of cancer. Anticancer Agents Med. Chem. 2017, 17, 164–170. [Google Scholar] [CrossRef] [PubMed]
  78. Fu, Y.; Liu, S.; Yin, S.; Niu, W.; Xiong, W.; Tan, M.; Li, G.; Zhou, M. The reverse warburg effect is likely to be an achilles’ heel of cancer that can be exploited for cancer therapy. Oncotarget 2017, 8, 57813–57825. [Google Scholar] [CrossRef] [Green Version]
  79. Pavlides, S.; Whitaker-Menezes, D.; Castello-Cros, R.; Flomenberg, N.; Witkiewicz, A.K.; Frank, P.G.; Casimiro, M.C.; Wang, C.; Fortina, P.; Addya, S.; et al. The reverse warburg effect: Aerobic glycolysis in cancer associated fibroblasts and the tumor stroma. Cell Cycle 2009, 8, 3984–4001. [Google Scholar] [CrossRef] [Green Version]
  80. Wilde, L.; Roche, M.; Domingo-Vidal, M.; Tanson, K.; Philp, N.; Curry, J.; Martinez-Outschoorn, U. Metabolic coupling and the reverse warburg effect in cancer: Implications for novel biomarker and anticancer agent development. Semin. Oncol. 2017, 44, 198–203. [Google Scholar] [CrossRef]
  81. Nahm, J.H.; Kim, H.M.; Koo, J.S. Glycolysis-related protein expression in thyroid cancer. Tumour. Biol. 2017, 39, 1010428317695922. [Google Scholar] [CrossRef] [Green Version]
  82. Wen, S.; Qu, N.; Ma, B.; Wang, X.; Luo, Y.; Xu, W.; Jiang, H.; Zhang, Y.; Wang, Y.; Ji, Q. Cancer-associated fibroblasts positively correlate with dedifferentiation and aggressiveness of thyroid cancer. Onco. Targets Ther. 2021, 14, 1205–1217. [Google Scholar] [CrossRef]
  83. Qing, W.; Fang, W.Y.; Ye, L.; Shen, L.Y.; Zhang, X.F.; Fei, X.C.; Chen, X.; Wang, W.Q.; Li, X.Y.; Xiao, J.C.; et al. Density of tumor-associated macrophages correlates with lymph node metastasis in papillary thyroid carcinoma. Thyroid 2012, 22, 905–910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Jung, K.Y.; Cho, S.W.; Kim, Y.A.; Kim, D.; Oh, B.C.; Park, D.J.; Park, Y.J. Cancers with higher density of tumor-associated macrophages were associated with poor survival rates. J. Pathol. Transl. Med. 2015, 49, 318–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Ryder, M.; Ghossein, R.A.; Ricarte-Filho, J.C.; Knauf, J.A.; Fagin, J.A. Increased density of tumor-associated macrophages is associated with decreased survival in advanced thyroid cancer. Endocr. Relat. Cancer 2008, 15, 1069–1074. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, S.; Cho, S.W.; Min, H.S.; Kim, K.M.; Yeom, G.J.; Kim, E.Y.; Lee, K.E.; Yun, Y.G.; Park, D.J.; Park, Y.J. The expression of tumor-associated macrophages in papillary thyroid carcinoma. Endocrinol. Metab. 2013, 28, 192–198. [Google Scholar] [CrossRef]
  87. Fang, W.; Ye, L.; Shen, L.; Cai, J.; Huang, F.; Wei, Q.; Fei, X.; Chen, X.; Guan, H.; Wang, W.; et al. Tumor-associated macrophages promote the metastatic potential of thyroid papillary cancer by releasing cxcl8. Carcinogenesis 2014, 35, 1780–1787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Huang, F.J.; Zhou, X.Y.; Ye, L.; Fei, X.C.; Wang, S.; Wang, W.; Ning, G. Follicular thyroid carcinoma but not adenoma recruits tumor-associated macrophages by releasing ccl15. BMC Cancer 2016, 16, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Cunha, L.L.; Morari, E.C.; Guihen, A.C.; Razolli, D.; Gerhard, R.; Nonogaki, S.; Soares, F.A.; Vassallo, J.; Ward, L.S. Infiltration of a mixture of immune cells may be related to good prognosis in patients with differentiated thyroid carcinoma. Clin. Endocrinol. 2012, 77, 918–925. [Google Scholar] [CrossRef] [PubMed]
  90. Suzuki, S.; Shibata, M.; Gonda, K.; Kanke, Y.; Ashizawa, M.; Ujiie, D.; Suzushino, S.; Nakano, K.; Fukushima, T.; Sakurai, K.; et al. Immunosuppression involving increased myeloid-derived suppressor cell levels, systemic inflammation and hypoalbuminemia are present in patients with anaplastic thyroid cancer. Mol. Clin. Oncol. 2013, 1, 959–964. [Google Scholar] [CrossRef] [Green Version]
  91. Angell, T.E.; Lechner, M.G.; Smith, A.M.; Martin, S.E.; Groshen, S.G.; Maceri, D.R.; Singer, P.A.; Epstein, A.L. Circulating myeloid-derived suppressor cells predict differentiated thyroid cancer diagnosis and extent. Thyroid 2016, 26, 381–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lee, F.; Yang, P.S.; Chien, M.N.; Lee, J.J.; Leung, C.H.; Cheng, S.P. An increased neutrophil-to-lymphocyte ratio predicts incomplete response to therapy in differentiated thyroid cancer. Int. J. Med. Sci. 2018, 15, 1757–1763. [Google Scholar] [CrossRef] [Green Version]
  93. Lang, B.H.; Ng, C.P.; Au, K.B.; Wong, K.P.; Wong, K.K.; Wan, K.Y. Does preoperative neutrophil lymphocyte ratio predict risk of recurrence and occult central nodal metastasis in papillary thyroid carcinoma? World J. Surg. 2014, 38, 2605–2612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Galdiero, M.R.; Varricchi, G.; Loffredo, S.; Bellevicine, C.; Lansione, T.; Ferrara, A.L.; Iannone, R.; di Somma, S.; Borriello, F.; Clery, E.; et al. Potential involvement of neutrophils in human thyroid cancer. PLoS ONE 2018, 13, e0199740. [Google Scholar] [CrossRef]
  95. Hilly, O.; Koren, R.; Raz, R.; Rath-Wolfson, L.; Mizrachi, A.; Hamzany, Y.; Bachar, G.; Shpitzer, T. The role of s100-positive dendritic cells in the prognosis of papillary thyroid carcinoma. Am. J. Clin. Pathol. 2013, 139, 87–92. [Google Scholar] [CrossRef]
  96. Yu, H.; Huang, X.; Liu, X.; Jin, H.; Zhang, G.; Zhang, Q.; Yu, J. Regulatory t cells and plasmacytoid dendritic cells contribute to the immune escape of papillary thyroid cancer coexisting with multinodular non-toxic goiter. Endocrine 2013, 44, 172–181. [Google Scholar] [CrossRef] [PubMed]
  97. Hilly, O.; Rath-Wolfson, L.; Koren, R.; Mizrachi, A.; Hamzany, Y.; Bachar, G.; Shpitzer, T. Cd1a-positive dendritic cell density predicts disease-free survival in papillary thyroid carcinoma. Pathol. Res. Pract. 2015, 211, 652–656. [Google Scholar] [CrossRef] [PubMed]
  98. Scarpino, S.; Stoppacciaro, A.; Ballerini, F.; Marchesi, M.; Prat, M.; Stella, M.C.; Sozzani, S.; Allavena, P.; Mantovani, A.; Ruco, L.P. Papillary carcinoma of the thyroid: Hepatocyte growth factor (hgf) stimulates tumor cells to release chemokines active in recruiting dendritic cells. Am. J. Pathol. 2000, 156, 831–837. [Google Scholar] [CrossRef]
  99. Melillo, R.M.; Guarino, V.; Avilla, E.; Galdiero, M.R.; Liotti, F.; Prevete, N.; Rossi, F.W.; Basolo, F.; Ugolini, C.; de Paulis, A.; et al. Mast cells have a protumorigenic role in human thyroid cancer. Oncogene 2010, 29, 6203–6215. [Google Scholar] [CrossRef] [Green Version]
  100. Visciano, C.; Liotti, F.; Prevete, N.; Cali, G.; Franco, R.; Collina, F.; de Paulis, A.; Marone, G.; Santoro, M.; Melillo, R.M. Mast cells induce epithelial-to-mesenchymal transition and stem cell features in human thyroid cancer cells through an il-8-akt-slug pathway. Oncogene 2015, 34, 5175–5186. [Google Scholar] [CrossRef]
  101. Proietti, A.; Ugolini, C.; Melillo, R.M.; Crisman, G.; Elisei, R.; Santoro, M.; Minuto, M.; Vitti, P.; Miccoli, P.; Basolo, F. Higher intratumoral expression of cd1a, tryptase, and cd68 in a follicular variant of papillary thyroid carcinoma compared to adenomas: Correlation with clinical and pathological parameters. Thyroid 2011, 21, 1209–1215. [Google Scholar] [CrossRef]
  102. Liu, Y.; Yun, X.; Gao, M.; Yu, Y.; Li, X. Analysis of regulatory t cells frequency in peripheral blood and tumor tissues in papillary thyroid carcinoma with and without hashimoto’s thyroiditis. Clin. Transl. Oncol. 2015, 17, 274–280. [Google Scholar] [CrossRef]
  103. French, J.D.; Kotnis, G.R.; Said, S.; Raeburn, C.D.; McIntyre, R.C., Jr.; Klopper, J.P.; Haugen, B.R. Programmed death-1+ t cells and regulatory t cells are enriched in tumor-involved lymph nodes and associated with aggressive features in papillary thyroid cancer. J. Clin. Endocrinol. Metab. 2012, 97, E934–E943. [Google Scholar] [CrossRef] [PubMed]
  104. Ryu, H.S.; Park, Y.S.; Park, H.J.; Chung, Y.R.; Yom, C.K.; Ahn, S.H.; Park, Y.J.; Park, S.H.; Park, S.Y. Expression of indoleamine 2,3-dioxygenase and infiltration of foxp3+ regulatory t cells are associated with aggressive features of papillary thyroid microcarcinoma. Thyroid 2014, 24, 1232–1240. [Google Scholar] [CrossRef] [PubMed]
  105. Cunha, L.L.; Morari, E.C.; Nonogaki, S.; Soares, F.A.; Vassallo, J.; Ward, L.S. Foxp3 expression is associated with aggressiveness in differentiated thyroid carcinomas. Clinics 2012, 67, 483–488. [Google Scholar] [CrossRef]
  106. Imam, S.; Paparodis, R.; Sharma, D.; Jaume, J.C. Lymphocytic profiling in thyroid cancer provides clues for failure of tumor immunity. Endocr. Relat. Cancer 2014, 21, 505–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Jiang, G.; Ma, S.; Wei, Y.; Wu, Y.; Yu, X.; Liu, H. The prevalence and distribution of th17 and tc17 cells in patients with thyroid tumor. Immunol. Lett. 2014, 162, 68–73. [Google Scholar] [CrossRef]
  108. Cunha, L.L.; Marcello, M.A.; Nonogaki, S.; Morari, E.C.; Soares, F.A.; Vassallo, J.; Ward, L.S. Cd8+ tumour-infiltrating lymphocytes and cox2 expression may predict relapse in differentiated thyroid cancer. Clin. Endocrinol. 2015, 83, 246–253. [Google Scholar] [CrossRef] [PubMed]
  109. Xiang, Z.; Zhong, C.; Chang, A.; Ling, J.; Zhao, H.; Zhou, W.; Zhuo, X. Immune-related key gene cldn10 correlates with lymph node metastasis but predicts favorable prognosis in papillary thyroid carcinoma. Aging 2020, 12, 2825–2839. [Google Scholar] [CrossRef] [PubMed]
  110. Gogali, F.; Paterakis, G.; Rassidakis, G.Z.; Kaltsas, G.; Liakou, C.I.; Gousis, P.; Neonakis, E.; Manoussakis, M.N.; Liapi, C. Phenotypical analysis of lymphocytes with suppressive and regulatory properties (tregs) and nk cells in the papillary carcinoma of thyroid. J. Clin. Endocrinol. Metab. 2012, 97, 1474–1482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Yin, M.; Di, G.; Bian, M. Dysfunction of natural killer cells mediated by pd-1 and tim-3 pathway in anaplastic thyroid cancer. Int. Immunopharmacol. 2018, 64, 333–339. [Google Scholar] [CrossRef]
  112. Wennerberg, E.; Pfefferle, A.; Ekblad, L.; Yoshimoto, Y.; Kremer, V.; Kaminskyy, V.O.; Juhlin, C.C.; Höög, A.; Bodin, I.; Svjatoha, V.; et al. Human anaplastic thyroid carcinoma cells are sensitive to nk cell-mediated lysis via ulbp2/5/6 and chemoattract nk cells. Clin. Cancer Res. 2014, 20, 5733–5744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhu, L.; Li, X.J.; Kalimuthu, S.; Gangadaran, P.; Lee, H.W.; Oh, J.M.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; et al. Natural killer cell (nk-92mi)-based therapy for pulmonary metastasis of anaplastic thyroid cancer in a nude mouse model. Front. Immunol. 2017, 8, 816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [CrossRef] [Green Version]
  115. Landa, I.; Ibrahimpasic, T.; Boucai, L.; Sinha, R.; Knauf, J.A.; Shah, R.H.; Dogan, S.; Ricarte-Filho, J.C.; Krishnamoorthy, G.P.; Xu, B.; et al. Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers. J. Clin. Investig. 2016, 126, 1052–1066. [Google Scholar] [CrossRef] [PubMed]
  116. Na, K.J.; Choi, H. Immune landscape of papillary thyroid cancer and immunotherapeutic implications. Endocr. Relat. Cancer 2018, 25, 523–531. [Google Scholar] [CrossRef] [Green Version]
  117. Angell, T.E.; Lechner, M.G.; Jang, J.K.; Correa, A.J.; LoPresti, J.S.; Epstein, A.L. Braf v600e in papillary thyroid carcinoma is associated with increased programmed death ligand 1 expression and suppressive immune cell infiltration. Thyroid 2014, 24, 1385–1393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Ahn, S.; Kim, T.H.; Kim, S.W.; Ki, C.S.; Jang, H.W.; Kim, J.S.; Kim, J.H.; Choe, J.H.; Shin, J.H.; Hahn, S.Y.; et al. Comprehensive screening for pd-l1 expression in thyroid cancer. Endocr. Relat. Cancer 2017, 24, 97–106. [Google Scholar] [CrossRef] [Green Version]
  119. Moretti, S.; Menicali, E.; Nucci, N.; Voce, P.; Colella, R.; Melillo, R.M.; Liotti, F.; Morelli, S.; Fallarino, F.; Macchiarulo, A.; et al. Signal transducer and activator of transcription 1 plays a pivotal role in ret/ptc3 oncogene-induced expression of indoleamine 2,3-dioxygenase 1. J. Biol. Chem. 2017, 292, 1785–1797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Thorsson, V.; Gibbs, D.L.; Brown, S.D.; Wolf, D.; Bortone, D.S.; Ou Yang, T.H.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The immune landscape of cancer. Immunity 2018, 48, 812–830.e814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Zhao, K.; Yang, H.; Kang, H.; Wu, A. Identification of key genes in thyroid cancer microenvironment. Med. Sci. Monit. 2019, 25, 9602–9608. [Google Scholar] [CrossRef] [PubMed]
  122. Galon, J.; Mlecnik, B.; Bindea, G.; Angell, H.K.; Berger, A.; Lagorce, C.; Lugli, A.; Zlobec, I.; Hartmann, A.; Bifulco, C.; et al. Towards the introduction of the ‘immunoscore’ in the classification of malignant tumours. J. Pathol. 2014, 232, 199–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Cesano, A. Ncounter(®) pancancer immune profiling panel (Nanostring Technologies, Inc., Seattle, WA). J. Immunother. Cancer 2015, 3, 42. [Google Scholar] [CrossRef] [Green Version]
  124. Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
  125. Giannini, R.; Moretti, S.; Ugolini, C.; Macerola, E.; Menicali, E.; Nucci, N.; Morelli, S.; Colella, R.; Mandarano, M.; Sidoni, A.; et al. Immune profiling of thyroid carcinomas suggests the existence of two major phenotypes: An atc-like and a pdtc-like. J. Clin. Endocrinol. Metab. 2019, 104, 3557–3575. [Google Scholar] [CrossRef] [PubMed]
  126. Sun, W.; Xu, Y.; Zhao, C.; Hao, F.; Chen, D.; Guan, J.; Zhang, K. Targeting tgf-β1 suppresses survival of and invasion by anaplastic thyroid carcinoma cells. Am. J. Transl. Res. 2017, 9, 1418–1425. [Google Scholar] [PubMed]
  127. Zhang, K.; Liu, X.; Hao, F.; Dong, A.; Chen, D. Targeting tgf-β1 inhibits invasion of anaplastic thyroid carcinoma cell through smad2-dependent s100a4-mmp-2/9 signalling. Am. J. Transl. Res. 2016, 8, 2196–2209. [Google Scholar] [PubMed]
  128. She, M.; Jim Yeung, S.C. Combining a matrix metalloproteinase inhibitor, a farnesyltransferase inhibitor, and a taxane improves survival in an anaplastic thyroid cancer model. Cancer Lett. 2006, 238, 197–201. [Google Scholar] [CrossRef] [PubMed]
  129. Lu, Y.; Zhu, Y.; Deng, S.; Chen, Y.; Li, W.; Sun, J.; Xu, X. Targeting the sonic hedgehog pathway to suppress the expression of the cancer stem cell (csc)-related transcription factors and csc-driven thyroid tumor growth. Cancers 2021, 13, 418. [Google Scholar] [CrossRef]
  130. Chattopadhyay, C.; El-Naggar, A.K.; Williams, M.D.; Clayman, G.L. Small molecule c-met inhibitor pha665752: Effect on cell growth and motility in papillary thyroid carcinoma. Head. Neck. 2008, 30, 991–1000. [Google Scholar] [CrossRef]
  131. Zhou, Y.; Zhao, C.; Gery, S.; Braunstein, G.D.; Okamoto, R.; Alvarez, R.; Miles, S.A.; Doan, N.B.; Said, J.W.; Gu, J.; et al. Off-target effects of c-met inhibitors on thyroid cancer cells. Mol. Cancer Ther. 2014, 13, 134–143. [Google Scholar] [CrossRef] [Green Version]
  132. Jung, Y.H.; Lee, D.Y.; Cha, W.; Kim, B.H.; Sung, M.W.; Kim, K.H.; Ahn, S.H. Antitumor effect of cxcr4 antagonist amd3100 on the tumorigenic cell line of bhp10-3 papillary thyroid cancer cells. Head. Neck. 2016, 38, 1479–1486. [Google Scholar] [CrossRef] [PubMed]
  133. Lin, Y.; Ma, Q.; Li, L.; Wang, H. The cxcl12-cxcr4 axis promotes migration, invasiveness, and emt in human papillary thyroid carcinoma b-cpap cells via nf-κb signaling. Biochem. Cell Biol. 2018, 96, 619–626. [Google Scholar] [CrossRef] [PubMed]
  134. Shirley, L.A.; McCarty, S.; Yang, M.C.; Saji, M.; Zhang, X.; Phay, J.; Ringel, M.D.; Chen, C.S. Integrin-linked kinase affects signaling pathways and migration in thyroid cancer cells and is a potential therapeutic target. Surgery 2016, 159, 163–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Younes, M.N.; Kim, S.; Yigitbasi, O.G.; Mandal, M.; Jasser, S.A.; Dakak Yazici, Y.; Schiff, B.A.; El-Naggar, A.; Bekele, B.N.; Mills, G.B.; et al. Integrin-linked kinase is a potential therapeutic target for anaplastic thyroid cancer. Mol. Cancer Ther. 2005, 4, 1146–1156. [Google Scholar] [CrossRef] [PubMed]
  136. Yin, Q.; Liu, S.; Dong, A.; Mi, X.; Hao, F.; Zhang, K. Targeting transforming growth factor-beta1 (tgf-β1) inhibits tumorigenesis of anaplastic thyroid carcinoma cells through erk1/2-nfκkb-puma signaling. Med. Sci. Monit. 2016, 22, 2267–2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Maio, M.; Coral, S.; Sigalotti, L.; Elisei, R.; Romei, C.; Rossi, G.; Cortini, E.; Colizzi, F.; Fenzi, G.; Altomonte, M.; et al. Analysis of cancer/testis antigens in sporadic medullary thyroid carcinoma: Expression and humoral response to ny-eso-1. J. Clin. Endocrinol. Metab. 2003, 88, 748–754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Gunda, V.; Frederick, D.T.; Bernasconi, M.J.; Wargo, J.A.; Parangi, S. A potential role for immunotherapy in thyroid cancer by enhancing ny-eso-1 cancer antigen expression. Thyroid 2014, 24, 1241–1250. [Google Scholar] [CrossRef] [Green Version]
  139. Del Rivero, J.; Donahue, R.N.; Marté, J.L.; Gramza, A.W.; Bilusic, M.; Rauckhorst, M.; Cordes, L.; Merino, M.J.; Dahut, W.L.; Schlom, J.; et al. A case report of sequential use of a yeast-cea therapeutic cancer vaccine and anti-pd-l1 inhibitor in metastatic medullary thyroid cancer. Front. Endocrinol. 2020, 11, 490. [Google Scholar] [CrossRef] [PubMed]
  140. Jiang, K.; Song, C.; Kong, L.; Hu, L.; Lin, G.; Ye, T.; Yao, G.; Wang, Y.; Chen, H.; Cheng, W.; et al. Recombinant oncolytic newcastle disease virus displays antitumor activities in anaplastic thyroid cancer cells. BMC Cancer 2018, 18, 746. [Google Scholar] [CrossRef] [PubMed]
  141. Mundi, N.; Um, S.; Yoo, J.; Rizzo, G.; Black, M.; Pinto, N.; Palma, D.A.; Fung, K.; MacNeil, D.; Mymryk, J.S.; et al. The control of anaplastic thyroid carcinoma cell lines by oncolytic poxviruses. Virus. Res. 2014, 190, 53–59. [Google Scholar] [CrossRef] [PubMed]
  142. Passaro, C.; Borriello, F.; Vastolo, V.; Di Somma, S.; Scamardella, E.; Gigantino, V.; Franco, R.; Marone, G.; Portella, G. The oncolytic virus dl922-947 reduces il-8/cxcl8 and mcp-1/ccl2 expression and impairs angiogenesis and macrophage infiltration in anaplastic thyroid carcinoma. Oncotarget 2016, 7, 1500–1515. [Google Scholar] [CrossRef] [Green Version]
  143. Kuwabara, K.; Nishishita, T.; Morishita, M.; Oyaizu, N.; Yamashita, S.; Kanematsu, T.; Obara, T.; Mimura, Y.; Inoue, Y.; Kaminishi, M.; et al. Results of a phase i clinical study using dendritic cell vaccinations for thyroid cancer. Thyroid 2007, 17, 53–58. [Google Scholar] [CrossRef] [PubMed]
  144. Stift, A.; Sachet, M.; Yagubian, R.; Bittermann, C.; Dubsky, P.; Brostjan, C.; Pfragner, R.; Niederle, B.; Jakesz, R.; Gnant, M.; et al. Dendritic cell vaccination in medullary thyroid carcinoma. Clin. Cancer Res. 2004, 10, 2944–2953. [Google Scholar] [CrossRef] [Green Version]
  145. Min, I.M.; Shevlin, E.; Vedvyas, Y.; Zaman, M.; Wyrwas, B.; Scognamiglio, T.; Moore, M.D.; Wang, W.; Park, S.; Park, S.; et al. Car t therapy targeting icam-1 eliminates advanced human thyroid tumors. Clin. Cancer Res. 2017, 23, 7569–7583. [Google Scholar] [CrossRef]
  146. Vedvyas, Y.; McCloskey, J.E.; Yang, Y.; Min, I.M.; Fahey, T.J.; Zarnegar, R.; Hsu, Y.S.; Hsu, J.M.; Van Besien, K.; Gaudet, I.; et al. Manufacturing and preclinical validation of car t cells targeting icam-1 for advanced thyroid cancer therapy. Sci. Rep. 2019, 9, 10634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Mehnert, J.M.; Varga, A.; Brose, M.S.; Aggarwal, R.R.; Lin, C.C.; Prawira, A.; de Braud, F.; Tamura, K.; Doi, T.; Piha-Paul, S.A.; et al. Safety and antitumor activity of the anti-pd-1 antibody pembrolizumab in patients with advanced, pd-l1-positive papillary or follicular thyroid cancer. BMC Cancer 2019, 19, 196. [Google Scholar] [CrossRef] [PubMed]
  148. Iyer, P.C.; Dadu, R.; Gule-Monroe, M.; Busaidy, N.L.; Ferrarotto, R.; Habra, M.A.; Zafereo, M.; Williams, M.D.; Gunn, G.B.; Grosu, H.; et al. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J. Immunother. Cancer 2018, 6, 68. [Google Scholar] [CrossRef] [Green Version]
  149. Gunda, V.; Gigliotti, B.; Ndishabandi, D.; Ashry, T.; McCarthy, M.; Zhou, Z.; Amin, S.; Freeman, G.J.; Alessandrini, A.; Parangi, S. Combinations of braf inhibitor and anti-pd-1/pd-l1 antibody improve survival and tumour immunity in an immunocompetent model of orthotopic murine anaplastic thyroid cancer. Br. J. Cancer 2018, 119, 1223–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Gray, K.D.; McCloskey, J.E.; Vedvyas, Y.; Kalloo, O.R.; Eshaky, S.E.; Yang, Y.; Shevlin, E.; Zaman, M.; Ullmann, T.M.; Liang, H.; et al. Pd1 blockade enhances icam1-directed car t therapeutic efficacy in advanced thyroid cancer. Clin. Cancer Res. 2020, 26, 6003–6016. [Google Scholar] [CrossRef]
  151. Otoshi, T.; Nagano, T.; Tachihara, M.; Nishimura, Y. Possible biomarkers for cancer immunotherapy. Cancers 2019, 11, 935. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Tumor stroma in thyroid cancer. Papillary thyroid carcinoma (PTC) shows desmoplastic tumor stroma mainly composed of caner-associated fibroblasts (CAF) (×100) (a). Prominent immune cells are noted in PTC subtype such as Warthin-like variant (×100) (b). Fibrous stroma composed of CAF is noted in anaplastic thyroid carcinoma (×100) (c). Immune cells stroma is present in poorly differentiated thyroid carcinoma (×100) (d).
Figure 1. Tumor stroma in thyroid cancer. Papillary thyroid carcinoma (PTC) shows desmoplastic tumor stroma mainly composed of caner-associated fibroblasts (CAF) (×100) (a). Prominent immune cells are noted in PTC subtype such as Warthin-like variant (×100) (b). Fibrous stroma composed of CAF is noted in anaplastic thyroid carcinoma (×100) (c). Immune cells stroma is present in poorly differentiated thyroid carcinoma (×100) (d).
Ijms 23 12578 g001
Figure 2. Interaction between cancer cells and caner-associated fibroblasts (CAF) in thyroid cancer. The expressions of CAF markers, α-SMA and PDGFR-β, are increased by SRC and AKT pathway activated by IL-6, ROS, PDGF that are secreted by thyroid cancer cells. SRC and AKT pathway differentiates and activates CAF. IL-6 secreted by CAF promotes tumor cell proliferation, and Sonic-Hedgehog ligand secreted by CAF promotes cancer cell invasion and metastasis through EMT process. In this EMT process, the expression of vimentin is increased and the expression of E-cadherin is attenuated. Moreover, lactate and ketone produced by glycolysis in CAF are transported to tumor cells and used in energy production by TCA cycle. This metabolic reprogramming is called the reverse-Warburg effect.
Figure 2. Interaction between cancer cells and caner-associated fibroblasts (CAF) in thyroid cancer. The expressions of CAF markers, α-SMA and PDGFR-β, are increased by SRC and AKT pathway activated by IL-6, ROS, PDGF that are secreted by thyroid cancer cells. SRC and AKT pathway differentiates and activates CAF. IL-6 secreted by CAF promotes tumor cell proliferation, and Sonic-Hedgehog ligand secreted by CAF promotes cancer cell invasion and metastasis through EMT process. In this EMT process, the expression of vimentin is increased and the expression of E-cadherin is attenuated. Moreover, lactate and ketone produced by glycolysis in CAF are transported to tumor cells and used in energy production by TCA cycle. This metabolic reprogramming is called the reverse-Warburg effect.
Ijms 23 12578 g002
Figure 3. Interaction between cancer cells and immune cells in thyroid cancer. CSF-1 and CCL15 produced by thyroid cancer cells recruit TAM, which produce CXCL8 that binds to CXCR1/2 and promotes tumor progression. TAM in thyroid cancers are M2 types that express ARG1, CCL22, IL10, and IL12. CXCL8 and IL-8 recruit TAN, which produce GM-CSF that is involved in tumor progression. VEGF-A recruits TMC, which produce histamine, CXCL1/GRO-α, and CXCL10/IP-10 that promote cancer cell proliferation. TMC also produce TNF, IL-6, and CXCL8/IL-8 that are involved in EMT. HGF and MIP-1a recruit TADC, which recruit Treg by ICOS, and PD-1 and JIM-3 recruit NK cells. TADC in thyroid cancers express CD1a, CCR6 and S100, and thyroid cancer cells produce COX-2 to recruit NK cells.
Figure 3. Interaction between cancer cells and immune cells in thyroid cancer. CSF-1 and CCL15 produced by thyroid cancer cells recruit TAM, which produce CXCL8 that binds to CXCR1/2 and promotes tumor progression. TAM in thyroid cancers are M2 types that express ARG1, CCL22, IL10, and IL12. CXCL8 and IL-8 recruit TAN, which produce GM-CSF that is involved in tumor progression. VEGF-A recruits TMC, which produce histamine, CXCL1/GRO-α, and CXCL10/IP-10 that promote cancer cell proliferation. TMC also produce TNF, IL-6, and CXCL8/IL-8 that are involved in EMT. HGF and MIP-1a recruit TADC, which recruit Treg by ICOS, and PD-1 and JIM-3 recruit NK cells. TADC in thyroid cancers express CD1a, CCR6 and S100, and thyroid cancer cells produce COX-2 to recruit NK cells.
Ijms 23 12578 g003
Table 1. CAF inhibitors in thyroid cancer.
Table 1. CAF inhibitors in thyroid cancer.
DrugTargetCancer TypeStudy GroupEffect Reference
Stromal barrier target agents
TGFβ inhibitorTGFβATCCell linetumorigenesis↓
migration and invasion ↓
[136]
[126,127]
BB94MMPATCCell linemigration and invasion ↓[127]
monocyclineMMPATCMouse xenograftxenograft tumor size ↓[128]
cyclopamineHedgehogATCCell line, mouse xenograftxenograft tumor size ↓[129]
CAF secreting factor inhibitors
PHA665752c-metPTCCell linecell growth, cell survival, cell invasion, and migration ↓[130]
tivantinib and crizotinibc-met Cell linecell proliferation ↓[131]
AMD3100CXCR4PTC Cell line (BHP10-3M), mouse xenograftcell proliferation, invasion and xenograft tumor formation ↓[132]
BAY11-7082CXCR4-CXCL12 axisPTCCell line (B-CPAP)migration, invasion, and EMT processes ↓[133]
CAF-ECM cross-talk inhibitors
T315integrin-linked kinase Cell linecell migration ↓, cell death ↑[134]
QLT0267integrin-linked kinaseATCCell line, mouse xenograftcell proliferation, and xenograft tumor formation ↓[135]
Table 2. Immune cell modulator in thyroid cancer.
Table 2. Immune cell modulator in thyroid cancer.
Drug Target Cancer TypeStudy GroupEffectReference
Cancer vaccine
GI-6207Yeast-CEAMTCMetastatic MCTOngoing clinical trial[139]
dl922-947oncolytic virus (OV)ATCMouse modelCell growth ↓,
M2 TAM → M1 TAM
[140,141,142]
Adoptive cell therapy
tumor lysate-pulsed DC PTC, FTC, and MTCAdvanced PTC, FTC, and MTCSymptom relieved [143,144]
CAR-T cellICAM-1PTC and ATC [145,146]
Immune checkpoint inhibitors
pembrolizumabPD-1PTC and FTCadvanced PTC and FTCPatient survival time ↑[147,148]
anti-PD-1/PD-L1 antibodyPD-1/PD-L1ATCMouse modelTumor size ↓, survival time ↑[149,150]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shin, E.; Koo, J.S. Cell Component and Function of Tumor Microenvironment in Thyroid Cancer. Int. J. Mol. Sci. 2022, 23, 12578. https://doi.org/10.3390/ijms232012578

AMA Style

Shin E, Koo JS. Cell Component and Function of Tumor Microenvironment in Thyroid Cancer. International Journal of Molecular Sciences. 2022; 23(20):12578. https://doi.org/10.3390/ijms232012578

Chicago/Turabian Style

Shin, Eunah, and Ja Seung Koo. 2022. "Cell Component and Function of Tumor Microenvironment in Thyroid Cancer" International Journal of Molecular Sciences 23, no. 20: 12578. https://doi.org/10.3390/ijms232012578

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop