Next Article in Journal
Melatonin Modulation of Radiation and Chemotherapeutics-induced Changes on Differentiation of Breast Fibroblasts
Next Article in Special Issue
Toll-Like Receptors-2 and -4 in Graves’ Disease—Key Players or Bystanders?
Previous Article in Journal
Streptococcal Pyrogenic Exotoxin A-Stimulated Monocytes Mediate Regulatory T-Cell Accumulation through PD-L1 and Kynurenine
Previous Article in Special Issue
Identification of Human Enzymes Oxidizing the Anti-Thyroid-Cancer Drug Vandetanib and Explanation of the High Efficiency of Cytochrome P450 3A4 in its Oxidation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Immune Landscape of Thyroid Cancer in the Context of Immune Checkpoint Inhibition

by
Gilda Varricchi
1,2,3,†,
Stefania Loffredo
1,2,3,†,
Giancarlo Marone
4,
Luca Modestino
1,2,
Poupak Fallahi
5,
Silvia Martina Ferrari
5,
Amato de Paulis
1,2,3,
Alessandro Antonelli
5 and
Maria Rosaria Galdiero
1,2,3,*
1
Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80131 Naples, Italy
2
Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80131 Naples, Italy
3
WAO Center of Excellence, 80131 Naples, Italy
4
Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
5
Department of Clinical and Experimental Medicine, University of Pisa, School of Medicine, 56126 Pisa, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2019, 20(16), 3934; https://doi.org/10.3390/ijms20163934
Submission received: 11 June 2019 / Revised: 1 August 2019 / Accepted: 8 August 2019 / Published: 13 August 2019
(This article belongs to the Special Issue Cell and Molecular Biology of Thyroid Disorders 2.0)

Abstract

:
Immune cells play critical roles in tumor prevention as well as initiation and progression. However, immune-resistant cancer cells can evade the immune system and proceed to form tumors. The normal microenvironment (immune cells, fibroblasts, blood and lymphatic vessels, and interstitial extracellular matrix (ECM)) maintains tissue homeostasis and prevents tumor initiation. Inflammatory mediators, reactive oxygen species, cytokines, and chemokines from an altered microenvironment promote tumor growth. During the last decade, thyroid cancer, the most frequent cancer of the endocrine system, has emerged as the fifth most incident cancer in the United States (USA), and its incidence is steadily growing. Inflammation has long been associated with thyroid cancer, raising critical questions about the role of immune cells in its pathogenesis. A plethora of immune cells and their mediators are present in the thyroid cancer ecosystem. Monoclonal antibodies (mAbs) targeting immune checkpoints, such as mAbs anti-cytotoxic T lymphocyte antigen 4 (anti-CTLA-4) and anti-programmed cell death protein-1/programmed cell death ligand-1 (anti-PD-1/PD-L1), have revolutionized the treatment of many malignancies, but they induce thyroid dysfunction in up to 10% of patients, presumably by enhancing autoimmunity. Combination strategies involving immune checkpoint inhibitors (ICIs) with tyrosine kinase (TK) or serine/threonine protein kinase B-raf (BRAF) inhibitors are showing considerable promise in the treatment of advanced thyroid cancer. This review illustrates how different immune cells contribute to thyroid cancer development and the rationale for the antitumor effects of ICIs in combination with BRAF/TK inhibitors.

1. Introduction

Thyroid cancer (TC) is the most frequent type of cancer of the endocrine system [1], accounting for approximately 90% of the endocrine malignancies and for 70% of deaths due to endocrine cancers [2]. During the past two decades, TC incidence increased [3,4]. Risk factors for TC include being female, having a history of goiter or thyroid nodule, a family history of TC, a low-iodine diet, radiation exposure, and obesity [5]. TC is three to four times more likely to develop in women, but it is more aggressive in men, who present with more advanced disease and have lower survival rates [6]. Differentiated TC (DTC) includes papillary thyroid carcinoma (PTC) (≈80%) and follicular thyroid carcinoma (FTC) (≈2%) and medullary thyroid cancer (MTC) (≈4%). Poorly differentiated TC (PDTC) and anaplastic thyroid carcinoma (ATC) account for approximately 5–10% of all TCs [7], but the majority of deaths for TC are attributable to this disease [8,9,10]. Calcitonin-producing parafollicular cells within the thyroid can give rise to MTC [11]. The association between smoldering inflammation and TC has long been recognized [12,13,14]. Indeed, a mixture of immune cells and proinflammatory mediators has been associated with TC initiation and progression [13]. In this review, we discuss recent findings investigating how the cells of the innate and adaptive immune system are involved in TC development and progression. We also discuss the rationale and preliminary results of treatment with monoclonal antibodies (mAbs), targeting immune checkpoints in patients with aggressive TCs.

2. Molecular and Genetic Factors

Genetic studies of PTCs reported a high frequency of activating somatic alterations of genes encoding effectors in the mitogen-activated protein kinase (MAPK) signaling pathway, including point mutations of BRAF (≈60%) and the GTPase RAS family genes (≈13%) [15,16,17], as well as fusions involving the REarranged during Transfection (RET) [18] and neutrotrophic receptor tyrosine kinase 1 (NTRK1) [19]. The most prevalent (≈60%) mutation in PTC is the activating BRAFV600E mutation [9,17,20]. The second most prevalent genetic event in PTC is paracentric inversions of the long arm of chromosome 10, causing the fusion of the REarranged during Transfection (RET) intracellular domain and leading to ligand-independent RET kinase activation [21]. RET fusions are found in approximately 7% of sporadic PTC [1] and in about 30% of young patients [22]. Mutations in the tumor suppressor TP53 gene are the most frequent genetic alterations in PDTC [23]. RAS mutations have also been found in PDTC [24]. Apart from the above-mentioned genetic alterations, several other genes have been found mutated in FTC, including PI3CA, PTEN, DICER1, EXH1, and SPOP [25,26]. MTC are commonly associated with RET mutations [21]. In a minority of PTCs, EWSR1 rearrangements have been identified [27,28].
The TElomerase Reverse Transcriptase (TERT) [29] promoter mutations chr5, 1,295,228 C>T (C228T) and 1,295,250 C>T (C250T) are present, on average, 0% (benign thyroid tumor), 11.3% (PTC), 17.1% (FTC), 43.2% (PDTC) and 40.1% (ATC) in thyroid tumors, displaying an association with aggressive types of TC. Coexisting BRAFV600E and TERT promoter mutations have a robust synergistic impact on the aggressiveness of PTC [30].
ATC is characterized by the accumulation of several different genetic alterations [9]. ATCs and to a lesser extent PDTC are a challenge for genomic studies due to their extensive infiltration of macrophages [31,32]. Three recent studies using a next-generation sequencing [33] approach performed extensive cancer gene exome sequencing in PDTC and ATC [34,35,36]. Profound genomic differences between the two advanced forms of the disease have been found. ATCs harbored a higher number of oncogenic alterations than PDTCs, and the mutations in PDTCs were increased compared to PTC. BRAFV600E mutations were present in 33% of PDTC and 45% of ATC, whereas mutations in NRAS, HRAS, or KRAS occurred in 28% and 24% of PDTCs and ATCs, respectively. TERT promoter mutations, which are important in tumorigenesis, were highly prevalent in ATC compared to PDTC (73% versus 8%). Mutations in PIK3CA and PTEN were prevalent in ATC (18% and 53%, respectively). Mutations in EIF1AX were reported in 11% of PDTC and 9% of ATC. Mutations of genes encoding the phosphoinositide-3-kinase–protein kinase B/Akt-mammalian target of rapamycin (PI3K-PKB/Akt-mTOR) pathway were more frequent in ATC than PDTC (39% versus 11%). Anaplastic lymphoma kinase (ALK) gene rearrangements have been found in ATC [37] and in PTC [38]. GLIS3 rearrangements are prevalent in hyalinizing trabecular tumor (HTT), which is a rare TC with a characteristic trabecular growth pattern and hyalinization [39]. Collectively, these results have identified several genetic lesions that distinguish PDTC from ATC.

3. Cytokines

Cytokines, produced by thyroid-infiltrating immune cells and by follicular cells, play a role in the pathogenesis of autoimmune thyroid disease [14] and contribute to several aspects of TC initiation and growth [13]. Interleukin (IL)-1 stimulated thyroid cell proliferation and the production of IL-8 [40]. IL-1β serum concentrations were increased in patients with atrophic thyroiditis and decreased in those with PTC [41].
IL-4 and IL-13 produced by human T helper 2 (Th2) cells, basophils, and T follicular helper cells (Tfh) [42,43] induce alternative (M2) activation of macrophages and tumor-associated macrophages [44]. Macrophages in TC displayed an M2-like phenotype [45]. The exposure of thyreocytes to ionizing radiation (IR) caused IL-13 production, which stimulated reactive oxygen species (ROS) increase, and was presumably responsible for genetic instability and the emergence of neoplastic clones in TC [46].
TC-derived conditioned media (CM) induced the up-regulation of IL-6 in human mast cells [47] and contributed to epithelial-to-mesenchymal transition (EMT) and stemness, which are features of TC [48]. Studies comparing levels of circulating IL-6 among TC patients and controls have provided inconsistent results [49,50,51].
Tumor-associated macrophages (TAMs) [44] and tumor cells themselves produce IL-10 [52]. IL-10 circulating levels were higher in PTC associated with multinodular goiter (MNG) compared to MNG alone [53]. Malignant epithelial cells isolated from PTC and ATC [7] produced in vitro IL-4 and IL-10, which increased the expression of the anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2), B-cell lymphoma-extra large (Bcl-xL), cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein (cFLIP), and Phosphoprotein enriched in diabetes/Astrocytic phosphoprotein (PED/PEA-15) in TC cells [54,55]. IL-4 and IL-10 serum concentrations were higher in PTC patients compared to Hashimoto’s thyroiditis [56]. Moreover, IL-10 expression was correlated with thyroid tumor size and malignancy [57]. Interestingly, B cells from TC patients produced higher levels of IL-10 compared to healthy controls [58].
IL-12, a proinflammatory cytokine with antitumor activity [59], was effective against ATC in a mouse xenograft model [60], and its administration inhibited tumor growth and prolonged survival by activating both CD8+ T and natural killer (NK) cells in a genetically engineered BRAFV600E mouse model [61].
IL-17A is produced mainly by human CD4+ Th17 cells and to a lesser extent by CD8+ T cells (Tc17 cells), and was associated with both protumor or antitumor responses [62,63]. The serum concentration of IL-17 was increased in TC patients compared to thyroid adenoma [64] and correlated with the percent of circulating Th17 cells [62]. IL17RA polymorphisms negatively correlated with the development of PTC [65]. The serum levels of IL-17 and the percentage of Th17, Tc17, and T regulatory cells (Tregs) were increased in patients with DTCs compared to healthy controls [66]. IL-17 and IL-23 expression in DTCs and MTCs were increased compared to thyroid adenoma, and IL-17 expression was associated with recurrence and mortality [67].
IL-21 is highly expressed by both Tfh and Th9 cells [68]. In a large cohort of a Chinese population, IL-21 polymorphism was associated with an increased risk of TC development [69].
IL-24 was induced by RET/PTC3 expression in the thyrocytes of RET/PTC3 transgenic mice, and it was involved in autocrine growth/survival of RET/PTC3-expressing thyroid cells supporting its role in early cellular transformation. Its expression decreased in poorly differentiated mouse tumors, paralleling the loss of RET/PTC3 expression along with tumor progression, thus supporting a role for IL-24 as a tumor suppressor factor [70].
In a murine model of transgenic BRAF, mouse thyrocytes and TAMs produced TGF-β, which was responsible for the acquisition of EMT features and the invasiveness of TC cells [71]. In human PTCs, TGF-β overexpression correlated with tumor invasiveness, regardless of BRAF mutations [72]. The expression of TGF-β in PTC correlated with CD68+ cell infiltration in PTC [73] compared to benign thyroid nodules [74]. These findings led to suggest that targeting transforming growth factor beta β1 (TGF-β1) could inhibit ATC tumorigenesis [75].
All three classes of interferons (IFNs), type I (IFN-α/β), type II (IFN-γ), and type III (IFN-λ/s), induce the apoptosis of tumor cells and modulate cancer immunosurveillance [76]. In TC, type I and type II IFNs induced the expression of Major Histocompatibility Complex (MHC)-I molecules on human TC cell lines, thus limiting TC immunoevasion and potentiating TC susceptibility to immune destruction [77]. IFN-γ inhibited the migration of PTC cells in vitro and induced EMT [78]. Moreover, IFN-γ inhibited CXCL8 secretion from BRAFV600E mutated thyroid cell lines [79].

4. Chemokines

Chemokines are functionally related small molecules with chemoattractant and cytokine-like functions that also modulate angiogenesis and lymphangiogenesis [80]. Chemokines can be produced by TC cells, following the activation of the MAPK pathways by the RET/PTC, RAS, and BRAF oncogenic drivers [81,82]. Thyroid cells produce a wide spectrum of CXC chemokines (i.e., CXCL1, CXCL8, CXCL9, CXCL10, and CXCL11) in basal conditions and/or under the influence of specific stimuli [83,84]. In thyroid tissue, recruited T helper 1 (Th1) lymphocytes can enhance IFN-γ and tumor necrosis factor α (TNF-α) production, which in turn stimulate the chemokine secretion from the thyroid cells, therefore creating an amplification feedback loop, initiating and perpetuating the autoimmune process. PTC and ATC cell lines release CXCL8 in basal conditions as well as under inflammatory stimuli, such as IL-1 and TNF-α [40,85]. The exogenous induction of RET/PTC1 oncogene in primary normal human thyrocytes induced the expression of CCL2, CCL20, and CXCL8 genes. CCL20 and CXCL8 were up-regulated in clinical samples of PTC [82]. PTCs displayed the highest expression of CCL20 and CXCL8 compared to normal tissues and thyroiditis, regardless of the genetic lesion beard by the tumor [86]. In a model of orthotopic TC xenograft in nude mice, CXCL8 was involved in tumor growth and progression [87]. Also, the tumor-infiltrating immune cells can be a source of chemokines. Indeed, mast cell-derived CXCL8 in vitro induced EMT features and stemness in human TC cells [48]. TAMs purified from PTC patients released CXCL8 [88].
PTC and ATC cell lines produce CXCL1/GRO-α and CXCL10/IP10 in basal conditions and under inflammatory stimuli [81,83,89]. Moreover, mast cell-derived CXCL1/GRO-α (growth-regulated oncogene α) and CXCL10/IP10 increased TC cell proliferation through the engagement of CXCR2 and CXCR3 on TC cells [47,81]. CXCL12/SDF-1 and its receptor CXCR7 have been found in PTC [90]. The CXCL12/SDF-1-CXCR7 axis promoted in vitro TC cell line proliferation and invasion [91]. Also, CCL20 promoted TC cell invasion and migration in vitro [92]. Two splice variants of CXCR3 (A and B), the receptor on follicular thyroid cells activated by several chemokines (CXCL4, CXCL9, CXCL10, and CXCL11), have been found [93]. A progressive increase in CXCR3A expression over the CXCR2B isoform was found from benign tumors to PTC.
We have found that the oncolytic adenovirus dl922-947 reduced CXCL8 and CCL2 production by ATC cell lines. dl922-947 treatment impaired angiogenesis and favored the switch of tumor macrophages toward an M1 phenotype. These results indicate that dl922-947 treatment, along with its role in inducing TC cell death, has an impact on the ATC immune microenvironment.

5. Angiogenic and Lymphangiogenic Factors

The formation of blood and lymphatic vessels is a complex process, requiring a finely tuned balance between stimulatory and inhibitory signals such as vascular endothelial growth factors (VEGFs), angiopoietins (ANGPTs), chemokines, and many others [94,95,96]. Several TC-infiltrating immune cells can impact tumor angiogenesis and lymphangiogenesis. Figure 1 shows that a plethora of cytokines, chemokines, angiogenic factors, and lymphangiogenic factors derived from immune cells enrich the complexity of the inflammatory tumor microenvironment in TC. For instance, mast cells (MCs) are a major source of proangiogenic (VEGF-A and VEGF-B) and lymphangiogenic (VEGF-C and VEGF-D) factors [97]. Thyroid MCs modulated angiogenesis through the release of CXCL8 [48]. Human macrophages are also a major source of both angiogenic and lymphangiogenic factors [98], as well as of several proangiogenic enzymes such as matrix metallopeptidase 9 (MMP-9), cyclooxygenase -2 (Cox-2), andinducible nitric oxide synthase (iNOS) [44]. Tumor-associated NK cells produce VEGF-A and CXCL8 [99]. Tumor-associated DCs release VEGF-A, CXCL8, and osteopontin [100]. Human eosinophils promote angiogenesis [101,102], and eosinophilia can be observed in ATC patients [103]. Tregs play a role in maintaining tolerance in TC and drive tumor angiogenesis through the release of VEGF-A [104]. Myeloid-derived suppressor cells (MDSCs) promote tumor angiogenesis through the release of VEGF-A and MMP-9 [105]. Human Th2 cells and neutrophils are sources of angiogenic factors [94,106,107].

6. The Immune Landscape in TC

The tumor microenvironment (i.e., immune cells, fibroblasts, blood and lymphatic vessels, endothelial cell progenitors, and extracellular matrix components (ECM)) plays a central role in tumor initiation and progression. The normal tissue microenvironment can suppress malignancy, while certain pathogenetic tissue features can induce tumor progression [111,112].

6.1. Tumor-Associated Macrophages (TAM)

Macrophages are key components of the tumor microenvironment and are highly plastic cells [113,114,115]. Under the influence of IFN-γ, macrophages undergo M1 polarization, which is characterized by an immunostimulatory phenotype. In contrast, IL-4 or IL-13 induce the M2 phenotype, which promotes tumor angiogenesis and suppresses immune responses [44]. This model distinguishing between classically polarized antitumor M1 and alternatively polarized M2 subtypes incompletely accounts for the extraordinary phenotypic diversity of macrophages in vivo [116,117]. High-resolution analysis of TAMCs led to the identification of 17 major phenotypes of human macrophages [116].
In PTCs, TAMs correlated with lymph node metastasis [88], larger tumor size [118], and reduced survival. In PDTC, TAM density correlated with capsular invasion and extrathyroid extension [32]. TAMs represent more than 50% of immune cells in ATCs, forming a “microglia-like” in close contact with cancer cells [31]. In the diffuse sclerosing variant of PTC, M2-like macrophages can be found in lymphatic emboli and correlated with tumor cell lymphatic invasion [119].
Macrophages are present in the immune landscape of PTC, especially in the BRAFV600E+ group [120]. The histologic grading of CD68+ TAM increased in more aggressive thyroid cancers (i.e., PDTC and ATC) compared to PTC [121]. In a murine model of transgenic BRAFV600E-induced thyroid carcinogenesis, tumors displayed a high TAM infiltration due to the increased expression of Csf-1r and Ccr2 by tumor cells. In this model, TAMs displayed an M2-like phenotype. In addition, Csf-1 −/− BRAF transgenic mice displayed a reduction in tumor growth [45]. TAMs purified from human PTC displayed a higher expression of IL-10 and CD206 compared to peripheral blood monocytes [122], and promoted the invasiveness of TC cell lines in vitro through the production of CXCL8 [88]. TAMs are present in ATC to variable degrees, ranging from 22% to 95% [31,32,123]. ATCs and to a lesser extent PDTCs have an extensive infiltration of macrophages [31,32], which make an interconnected network that envelops the tumor cells throughout the cancer specimen. A total of 68 genes that were overexpressed in M2 macrophages were examined in PDTCs and ATCs, and M2 signatures clearly differentiated the two TCs [34]. The role of macrophages in the formation of lung metastasis has been evaluated in an experimental model of ATC [124]. Macrophage depletion in mice injected with clodronate reduced the lung metastasis of ATC cell lines. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a lung non-coding RNA that is up-regulated in several cancers, including TC [125]. MALAT1 is up-regulated in TC tissues and cells, and induced the expression of basic fibroblast growth factor (FGF2) and IL-10 in TAM, which promoted FTC133 proliferation and angiogenesis [126].
Soluble factors released by TC cell lines (TPC1, BC-PAP, and FTC133) induced an inflammatory phenotype of peripheral blood monocytes [127]. Zhang et al. examined the protumorigenic role of testosterone using the ThrbPV/PV transgenic mouse model, which mimics human FTC development. They found that testosterone reduced the expression of the immune regulatory genes Glipr1 and Sfrp1, the M1 macrophage, and CD8+ T cell infiltration in thyroid samples. These immunosuppressive events resulted in TC progression in ThrbPV/PV mice [128]. These interesting findings were corroborated by the analysis of the database of the National and Cancer Institute’s Surveillance Epidemiology End Results (NIH, Bethesda, USA), which found that men had a higher rate of large primary or locally advanced FTC than women. Furthermore, there was higher FTC-associated mortality in men than in women in the 40-to-60-year age group.
High-dimensional analysis, particularly single-cell RNA-seq, will be necessary in order to better characterize the role of macrophages in thyroid tumorigenesis.

6.2. Dendritic Cells (DCs)

Tumor-infiltrating dendritic cells (DCs) display an immature phenotype, with impaired antigen presentation ability [129]. S100+ (mature and immature), CD1a+ (immature), and CD83+ (mature) DCs are increased in human PTC compared to normal thyroid tissue [130]. Conditioned media from the primary cultures of PTC induced the chemotaxis of peripheral blood monocyte-derived DCs. Hepatocyte growth factor (HGF) enhanced this chemotactic activity through the engagement of the receptor Met on TC cells [131]. PTCs expressed the DC chemotactic molecule macrophage inflammatory protein 1α (MIP-1α), and DCs expressed CCR6, thus suggesting a role for TC cells in recruiting DCs [132]. PTC displayed a higher CD1a+ DC infiltration compared to FTC and adenomas [132]. In contrast, a reduced or absent DC infiltration in PDTC and ATC was described in comparison to DTC [133]. DCs are present in the immune landscape of PTC, especially in the BRAFV600E+group [120].

6.3. Tumor-Associated Mast Cells (TAMCs)

Mast cells are ubiquitous in nearly all tissue and in close proximity to epithelia, fibroblasts, blood vessels, lymphatic vessels, and nerves [95,134]. Mast cells can play a role in angiogenesis [134], lymphangiogenesis [97], and tumor initiation and progression [110,135,136]. Mast cells are present in the microenvironment of several solid [47,137,138,139,140,141,142,143] and hematologic tumors [144]). The contribution of mast cells in cancer varies according to the stages of tumorigenesis and to their microlocalization [142].
Melillo et al. were the first to investigate the contribution of mast cells in thyroid cancer. Mast cell density was very low in normal thyroid tissue, whereas in PTC samples, there was a mast cell infiltration that correlated with tumor extrathyroid extension [47]. These findings were extended to a limited number of PDTCs and ATCs [48]. The presence of mast cells in the immune landscape of PTC has been recently confirmed [120].
A higher mast cell density was also found in FTC compared with adenomas and correlated with extracapsular extension [145]. In vitro studies demonstrated that TC cell line conditioned media induced mast cell chemotaxis through the release of VEGF-A [47], which activated the VEGFRs on human mast cells [97,146]. Moreover, TC cells activated mast cells to release cytokines (IL-6, TNF-α, granulocyte-macrophage colony-stimulating factor—GM-CSF) and chemokines (CXCL10/IP10 and CXCL1/Gro-α). In turn, mast cells promoted TC cell proliferation through CXCL1/GROα and CXCL10/IP10. In an in vivo model of a TC xenograft, mast cells were recruited at the tumor site and accelerated tumor growth, enhancing tumor vascularization and cell proliferation of the xenograft [47]. These results indicate that mast cells are present in human TCs and play a protumorigenic role in TC.
Epithelial-to-mesenchymal transition (EMT) is important in tumor progression, and is in part responsible for the acquisition of the invasive properties of cancer cells [147]. A mast cell-conditioned medium induced the EMT of human TC cell lines mainly through the release of CXCL8 [48]. Interestingly, the blockade of CXCL8 receptors (i.e., CXCR1 and CXCR2) with blocking antibodies markedly reduced the sphere-forming ability of TC cells [48]. Collectively, these findings support the hypothesis that mast cell-derived CXCL8 favors the acquisition of stem-like features of TC cells.
Recent evidence indicates that mast cells, similar to macrophages [116,148] and neutrophils [107], comprise several subsets of cells [149,150]. Single-cell RNA-seq analysis will be necessary to better understand the role(s) of mast cells subsets in TC development and the formation of metastases.

6.4. Tumor-Associated Neutrophils (TANs)

Neutrophils participate in the early phases of inflammation and resistance against extracellular pathogens [107,151], and play a role in cancer initiation and growth [152]. In humans, the ratio between peripheral blood neutrophil and lymphocyte count (neutrophil-to-lymphocyte ratio—NLR) has been proposed as an index of systemic inflammation, and has been found to be associated with tumor development [153]. A higher NLR was associated with a larger tumor size and higher risk of recurrence in TC patients, but failed to distinguish patients with benign or malignant nodules [154]. In contrast, an increased NLR has been found in TC compared with benign lesions and healthy controls [155]. No correlation was found with patient disease-free survival or risk of occult metastasis [156]. In a large cohort of PTC, PDTC, and ATC, NLR was increased in ATC and to a lesser extent in PDTC [157]. In a meta-analysis based on seven prospective cohorts comprising 7349 patients, no difference in NLR was found between DTC and patients with benign nodules [158]. Preoperative NLR was not associated with clinicopathological characteristics in PTC patients [159,160]. By contrast, other studies reported that the preoperative NRL was correlated with the size and lymph node metastasis of PTC patients [161,162,163,164]. In conclusion, the diagnostic and prognostic significance of NLR in different types of TC remains uncertain.
Maria Rosaria Galdiero et al. elegantly investigated the potential involvement of neutrophils in human TC [165]. Conditioned media from TC cell lines TPC1 and 8505 (TC-CM) promoted human neutrophils chemotaxis and survival. Neutrophil chemotaxis was mediated at least in part by CXCL8, and survival was mediated by GM-CSF. TC-CM induced morphological changes and the activation of neutrophils (i.e., CD11b and CD66 overexpression and CD62L shedding). Moreover, TC-CM induced the production of ROS and angiogenic mediators (i.e., VEGF-A and CXCL8). Importantly, the density of tumor-infiltrating neutrophils correlated with TC size. This study was the first to reveal the possible involvement of neutrophils in human TC.
Due to their relatively low transcriptional activity and short lifespan [166], neutrophils are generally believed to be terminally differentiated homogeneous cells once they leave the bone marrow. This view is rapidly changing, and various subsets of neutrophils that are mostly defined by surface markers [167] and density [168] have been described. Studies in murine models of cancer have identified two functionally antagonistic populations of neutrophils, which are referred to as N1 and N2 to mirror the nomenclature of M1 and M2 macrophages with similar activity [152]. The lack of definitive cell markers of human N1 and N2 subsets has been so far a major obstacle to establish their role in TC.

6.5. Myeloid-Derived Suppressor Cells (MDSCs)

Myeloid-derived suppressor cells (MDSCs) are largely immature myeloid cells that are characterized by a state of activation and display potent immune suppressive activity [169]. Two major subsets of MDSCs have been identified so far: monocytic (M-MDSCs) and polymorphonuclear (PMN-MDSCs) [170]. M-MDSCs share phenotypic and morphologic features with monocytes, whereas PMN-MDSCs are similar to neutrophils. MDSCs have been implicated in tumor immune responses, tumor initiation and progression, angiogenesis, and the formation of pre-metastatic niches [169,170]. MDSCs inhibit anti-cancer immune responses by releasing cytokines (IL-1 and TGF-β), ROS, and reactive nitrogen species (RNS) that condition the tumor microenvironment and by stimulating Foxp3+ Treg cells and M2 tumor-associated macrophages (TAM) [171]. Moreover, MDSCs promote angiogenesis and condition the pre-metastatic niches [170]. M-MDSCs and PMN-MDSCs differentiate from normal progenitors of monocytes and neutrophils, respectively [172]. Recently, PMN-MDSCs were distinguished from human neutrophils by the expression of the lectin-type oxidized LDL receptor 1 (LOX-1). LOX-1+ neutrophils were potent suppressor of T cells, while LOX-1 neutrophils, presumably classical neutrophils, were not [173,174].
Peripheral blood MDSC levels were increased in patients with ATC compared to healthy controls and correlated with the serum level of IL-10, suggesting a correlation between MDSCs and systemic immunosuppression [175]. MDSCs are attracted to tumor sites in response to various cytokines (CCL2, CCL5, and CSF1 for M-MDSCs, and CXCL1, CXCL5, CXCL6, CXCL8, and CXCL12 for PMN-MDSCs) [176]. In tumors, the incoming cells dive into a fairly hostile microenvironment characterized by hypoxia, low pH, and high concentrations of cytokines, lactate, adenosine, and oxidative agents (ROS, NO) [176,177]. These conditions heavily affect MDSC survival, functions, and differentiation. It is generally thought that PMN-MDSCs in tissues are short-living cells, and that monocytic cells in the tumor microenvironment most likely represent bona fide M-MDSCs. In a wide cohort of TC patients (253 PTCs and 13 FTCs), the intratumoral MDSCs count, which were identified as CD11b+ and CD33+ cells, did not correlate with patient clinicopathological features [178]. Two studies did not find increased peripheral blood MDSCs, which were defined as CD11b+ CD33+ cells, in PTC patients compared to control, but showed accumulation in patients with ATC [175,179]. More recently, circulating CD11b+ HLA-DRlow (presumably PMN-MDSCs), but not CD33+ HLA-DRlow (presumably M-MDSCs), were increased in DTC, mostly PTC, compared to benign nodules [180]. The lack of definite cell markers for human MDSC subsets has been so far a major obstacle to establish their clinical significance in TC patients. Single-cell RNA-seq will be needed to identify subsets of M-MDSCs and PMN-MDSCs in different types of human TCs.
A mouse model HrasG12V/Pten/TPO-Cre developed multifocal FTC, which displayed many of the classical hallmarks of high-grade human FTC and PDTC, including an extrathyroidal extension and lung metastasis [181]. These tumors were heavily infiltrated by macrophages, MDSCs, and double-positive CD4+ CD25+ T cells (Treg cells), and contained arginase-1+ cells. These findings are suggestive of an immunosuppressive tumor microenvironment (TME) of TC.

6.6. Natural Killer (NK) Cells

Natural killer (NK) cells are a family of innate immune cells that play crucial roles in protective immunity against tumors and viral infections [182]. Subtypes of human NK cells have been identified on the basis of the relative surface expression of CD16 and CD56. CD56dim CD16+ NK cells display a higher cytotoxic activity, whereas CD56bright CD16−/low NK cells are more efficient in cytokine production [183]. Tumor-infiltrating NK cells were increased in PTCs compared to goiters and healthy thyroids, whereas no differences were found in peripheral blood NK cells [184,185]. In PTC patients, NK cell infiltration negatively correlated with disease stage [186]. An increased infiltration of the immunoregulatory subset of NK cells CD56bright was found in PTC samples compared to MNG. CD56bright NK cells inversely correlated with the disease stage, whereas cytotoxic NK cells positively correlated with the disease stage in PTC patients. These results indicate that the TC microenvironment modulates the phenotype of NK cells [187].
NK cells mediated the lysis of ATC cell lines through the expression of the activating receptor NKG2D on NK cells and its ligands UL16 binding proteins (ULBP2/5/6) on ATC cells [188]. ATC cell lines and TC derived from human fine-needle aspiration [189] samples from ATC patients expressed ULB2/5/6, whereas non-malignant thyroid tissue did not. ATC cell lines induced NK cell migration through the activation of the CXCL10–CXCR3 axis. NK cells from FNA and the peripheral blood of ATC patients displayed a suppressed phenotype, which was characterized by a lower percentage of CD56dim cells and CXCR3+ cells and a reduced NKG2D expression, compared to peripheral blood NK cells. PGE2 produced by thyroid cancer cells [190] can be responsible for ATC-mediated NK cell suppression [188]. A recent study found a decrease frequency of peripheral blood cytotoxic NK cells (i.e., CD56lo CD16hi) in ATC patients compared to controls [191]. By contrast, NK cells CD56hi CD16lo/hi (cytokine producing) were increased in ATC compared to non-ATC patients and healthy controls.
In a knock-in mouse model, BRAF oncogene was expressed under the control of the thyroid peroxidase (TPO) promoter (LSL-BRAFV600E/TPO Cre mice). IL-12 gene therapy and recombinant IL-12 reduced tumor growth, restored the follicular architecture of the gland, and improved the survival of tumor-bearing mice. Interestingly, IL-12 improved the cytotoxicity of CD8+ T cells and NK cells, and increased the infiltration of M1 macrophages within the tumors [61]. NK cells inhibited the growth of metastasis in an in vivo mouse model of ATC pulmonary metastasis [192].

6.7. Natural Killer T Cells (NKT)

NKT cells are a heterogeneous lymphoid population that recognizes the lipid antigens presented by CD1d and plays an important role in tumor immunosurveillance [193]. Two NKT subsets have been identified: type I induce the lysis of tumor cells directly via a perforin/granzyme B-mediated mechanism or indirectly via the activation of NK cells and DCs; type II show immunosuppressive activity through the production of IL-13 [194]. NKT subsets have not been characterized in TC.

6.8. γδ T Cells

γδ T lymphocytes, expressing a γδ T cell receptor (TCR), are not MHC-restricted and do not recognize peptide antigens [195]. The contribution of γδ T cells in tumor immunosurveillance is controversial. Zitvogel et al. demonstrated that IL-17-producing γδ T cells play a prominent role in chemotherapy-induced anti-cancer immune responses [196]. The role of tumor-infiltrating γδ T cells in TC is still unknown.

6.9. Innate Lymphoid Cells (ILCs)

Innate lymphoid cells (ILCs) lack TCR, but functionally resemble effector T cells [197]. ILCs include three subsets of cytokine-producing helper cells: group 1 ILCs produce IFN-γ and include conventional NK cells; group 2 produces Th2-type cytokines (e.g., IL-4, IL-5, and IL-13); group 3 ILCs comprises several distinct cell subsets [198]. ILCs have a critical role in the development of lymphoid structures, the maintenance of immune homeostasis, tissue remodeling, and the maintenance of epithelial integrity [199]. ILCs have also been implicated in the control and suppression of tumors [200]. There is evidence that the tumor microenvironment dictates the fate of the tumor-suppressive functions of ILC2. Moreover, ILC2 can modulate T cell-to-MDSC balance in cancer [201]. Studies are required to understand the role of ILC subsets in different TCs.

6.10. CD8+ Cytotoxic T Cells

CD8+ cytotoxic T lymphocytes (CTLs) recognize and attack tumor cells expressing tumor antigens [202]. In a wide immunohistochemical characterization of the immune network in patients with chronic lymphocytic thyroiditis concurrent with DTC, a high CD8+ T lymphocyte infiltration was associated with improved disease-free survival [178]. In a study conducted in a wide cohort of DTC patients, including papillary and follicular subtypes, immunohistochemical analysis of tumor samples revealed that the combined enrichment of CD8+ cells and Cox-2 overexpression correlated with the highest risk of disease relapse. In the majority of the tumor samples analyzed (68%), CD8+ cells were granzyme B negative, reflecting a state of anergy [203].
A low intratumoral CD8+/Foxp3+ ratio was found in human BRAFV600E PTC, which was associated with an increased expression of the immunosuppressive molecules arginase-1, indoleamine 2,3-dioxygenase (IDO), and programmed death-ligand 1 (PD-L1). The latter findings suggest a BRAF-driven tumor-promoting microenvironment [204]. A recent study evaluated the CXCR5+ CD8+ T cell subset in peripheral blood, tumor-draining lymph nodes (TDLNs), and tumors from TC patients [205]. Although CXCR5+ CD8+ T cells expressed higher PD-1, T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) markers than CXCR5 CD8+ T cells, these cells displayed a higher expression of cytotoxic molecules (e.g., granzymes and perforin).

6.11. CD4+ Cells

The role of different subsets of CD4+ T cells in tumor immunity remains underappreciated. Th1-mediated immunity is generally considered as antitumoral [206], while polarized Th2 and/or Treg activity is believed to be protumorigenic [207]. This simplistic view is complicated by the plasticity of Th differentiation, which can be extensively modulated by the tumor microenvironment [115,208]. In TC, the extent of tumor-infiltrating CD4+ cells does not appear to predict patient outcome [203]. No differences were found between PTC and MNG patients, with respect to tissue or peripheral blood CD4+ cell frequencies [53]. Interestingly, a double negative CD4 CD8 lymphocyte population was the dominant cell type in PTC, and was more abundant in PTC than in thyroid autoimmunity and ex vivo released IFN-γ and IL-17 [209].

6.12. Treg Cells

Tregs shut down antitumor immune response via the production of IL-10, the expression of immunosuppressive molecules (e.g., CTLA-4 and PD-1), and the stimulation of angiogenesis [210]. Moreover, increased PD-1+ T and Treg cells in metastatic lymph nodes correlated with a more aggressive TC [211]. Foxp3+ Tregs and VEGF were found in PTC samples, and Treg infiltration correlated with disease stage and lymph node metastasis [212]. A higher Treg density was also observed in PTC samples compared with nodular goiter, and positively correlated to the stage of the disease [186]. Accordingly, a high infiltration of Foxp3+ Treg cells was associated with aggressive features of PTC [213]. Moreover, increased Foxp3+ and reduced CD3+ tumor-infiltrating lymphocytes correlated with IDO1 expression. Similarly to PD-L1, IDO1 is overexpressed in different tumors, and is associated with the activation of Foxp3+ Tregs and the down-regulation of cytotoxic cellular immunity in the tumor microenvironment [214].
A higher percentage of Foxp3+ T cells and Inducible T-cell COStimulator (ICOS) + Treg cells were found in tissues, but not in the peripheral blood of PTC patients with MNG compared to MNG alone [53]. In PTC plus MNG, tissue ICOS+ Foxp3+ T cells were increased in advanced stages and metastatic tumors. Tissue ICOS+ Foxp3+ T cell numbers correlated with tissue plasmocytoid DCs, which favor an immunosuppressive microenvironment [53].

6.13. IL-17+ Cells

CD4+ IL-17+ T cells (Th17) cells can exert protumor or antitumor functions, depending on the tissue microenvironment [215]. Only one study examined the prevalence and distribution of Th17 cells in TC samples. In peripheral blood and tissue samples of PTC patients, increased Th17 levels were found compared to healthy controls, whereas the percentage of CD8+IL-17+ T cells (Tc17) in the peripheral blood was reduced. The frequency of peripheral blood Th17 cells was positively correlated with the IL-17 serum level, while no correlation between the serum level of IL-17 and Tc17 cells was found. Peripheral blood Th17 cells inversely correlated with tumor size [62].

6.14. T Follicular Helper Cells (Tfh)

T follicular helper cells (Tfh) were discovered based on their expression of the essential transcription factor BCL6 [42,216]. This led to the recognition of Tfh cells as an independent CD4+ subset specialized in helping B cells in lymph nodes [42,217]. No single marker or combination of markers reliably identifies Tfh cells as a discrete immune cell population. In fact, Tfh are part of the CD4+ T cell differentiation spectrum. Several surface molecules (CCR5, PD-1, BCL6, BTLA4, and ICOS) vary in expression and represent plastic features of Tfh heterogeneity. A fraction of CD4+ T cells in human blood express CXCR5 or PD-1 and are called circulating Tfh (cTfh) cells. The latter cells are closely related to tissue Tfh cells. Human cTfh are heterogeneous [42], and are divided into three major functional subtypes (Tfh1, Tfh2, and Tfh17) [218]. IL-21 is a B-cell helper cytokine produced by Tfh cells [219].
Overreactive Tfh cells have been reported in several human systemic autoimmune diseases [220]. An increased frequency of cTfh cells has been reported in patients with autoimmune thyroid disease, and Tfh cells were also detected in the thyroid tissue of Hashimoto’s thyroiditis patients [221]. CXCR5+ CD4+ T cells were increased in the thyroid tissue of patients with Graves’ disease compared to control subjects [222]. Furthermore, CD4+ IL-21R+ T cells and CD19+ IL-21R+ B cells were also observed in Graves’ disease tissues.
Recent studies suggested that Tfh cell infiltrates may influence the growth and survival of certain tumors [223,224]. To our knowledge, the characterization of different subtypes of circulating and intratumor Tfh in different types of TC has not been reported yet. Future studies should investigate the presence and functions of Tfh subsets in different types of TC.

6.15. Th9 Cells

IL-9-producing CD4+ helper T cells (Th9 cells) are a subset of CD4+ helper T cells with proinflammatory functions and anti-cancer properties in vivo [225]. The release of IL-9 has been proposed to account for anti-cancer efficacy. Moreover, Th9 cells release IL-21, which promotes the production of IFN-γ and tumor elimination by CD8+ T cells and NK cells [226]. The Th9 antitumor efficacy has been attributed to the production of IL-9 [227]. Studies investigating the relevance of Th9 cells in thyroid oncogenesis are urgently needed. Figure 2 schematically illustrates a hypothetical immune landscape of TC.

7. Immune Checkpoint Inhibitors in TC

Immune checkpoints physiologically prevent excessive immune responses and the development of autoimmunity [230,231,232]. Monoclonal antibodies (mAbs) targeting immune checkpoints (immune checkpoint inhibitors: ICIs) have revolutionized the treatment of malignancies characterized by DNA microsatellite instability [233]. Two major classes of ICIs have clinically emerged: those targeting cytotoxic T lymphocyte antigen 4 (CTLA-4) (i.e., ipilimumab and tremelimumab), and those targeting programmed cell death protein-1 (PD-1) (i.e., nivolumab, pembrolizumab, spartalizumab) or its ligand, programmed cell death ligand-1 (PD-L1) (i.e., avelumab, atezolizumab, and durvalumab). CTLA-4 and PD-1 regulate different stages of the immune response. For instance, CTLA-4 modulates immune response primarily in draining lymph nodes, whereas the primary site of action of PD-1 and its ligands (PD-L1/PD-L2) is in the tumor microenvironment (TME). PD-1 expressed on activated T cells interact with PD-L1 or PD-L2 expressed on the surface of cancer cells or tumor-infiltrating immune cells, thus inhibiting the cytotoxic action of T cells [234]. The up-regulation of PD-L1 by tumor cells leads to increased T-cell exhaustion, and is thought to be a means of cancer cell immune evasion [235]. When T cells are primed by exposure to antigens presented by APCs in draining lymph nodes, they become activated and overexpress CTLA-4 on their surfaces, which competes with CD28 for binding to CD80 or CD86 on the surface of APCs. This competition between CTLA-4 and CD28 attenuates the early activation of CD4+ and CD8+ T cells, and improves the immunosuppressive functions of Treg cells [236]. Figure 2A schematically illustrates the two interactions between CTLA-4 on T lymphocytes and CD80/CD86 on APCs and between PD-1 on T cells and tumor cells expressing PD-L1 or PD-L2. Both interactions result in inhibitory signals that lead to T-cell exhaustion, decreased T-cell cytotoxicity, and cancer cell proliferation.
The PD-1/PD-L1 pathway is expressed in ATC and DTC [237,238]. BRAFV600E cells express higher expression of PD-L1 compared to BRAFWT cells [228]. The low rate of mutations—and therefore, of neoantigens in DTC—might suggest that this would be a poor target for monotherapy with ICIs [239]. In fact, the results of a preliminary trial of the PD-1 inhibitor pembrolizumab in patients with PD-L1+ PTC or FTC confirmed that hypothesis [240]. More recently, higher numbers of mutation load and genetic alterations have been identified in poorly DTCs and ATC than in DTCs [34]. To increase the response rate of ICIs, a major focus is to find combinations that act synergistically. For example, combination strategies that target multiple aspects of the immune response (e.g., PD-1/PD-L1 and CTLA-4 pathways) or tumor cells themselves (e.g., tyrosine kinase inhibitors, BRAF inhibitors, radiotherapy) could provide benefits for patients with advanced thyroid disease.
Brauner et al. found that combining the BRAF inhibitor and anti-PD-L1 antibody markedly improved tumor immunity (e.g., CD8+ cell infiltration and CD8+: Treg ratio) and tumor regression in an immunocompetent murine model of ATC. These findings were extended in an immunocompetent model of orthotopic murine ATC by showing that the combination of the BRAF inhibitor (i.e., PLX4720) and anti-PD-1/PD-L1 mAbs reduced tumor volume and improved survival [241]. Moreover, the combination of lenvatinib—a multi-tyrosine kinase (TK) inhibitor—with anti-PD-1/PD-L1 mAb reduced tumor volume and improved survival in immunocompetent mice with orthotopic ATC [229]. This combination treatment was associated with an increase in tumor-infiltrating CD8+ T cells and granzyme B staining without changes in NK cells. Preliminary studies indicate that the combination of a mAb anti-PD1 (i.e., pembrolizumab) with a TK inhibitor may be an effective salvage therapy for the treatment of ATC [242]. Several ongoing clinical trials are evaluating the strategies that target PD-1 alone or in combination with CTLA-4 inhibitor (ipilimumab), chemotherapy, radiation, or TKI (lenvatinib) in advanced TCs (Table 1). Other clinical trials are evaluating the effects of PD-L1 inhibitors (avelumab, atezolizumab, and durvalumab) in combination with CTLA-4 inhibitor (tremelimumab), chemotherapy, or radiotherapy in advanced TCs (Table 2). Figure 2B schematically illustrates the rationale of combinatorial therapies of advanced TC involving ICIs, BRAFi, multi-targeted TKIs, chemotherapies, or radiotherapies.

8. Thyroid Disorders Induced by ICIs

CTLA-4-knockout mice often develop autoimmune diseases, such as pancreatitis and myocarditis [243]. Similar to CTLA-4, mouse models lacking expression of PD-1 have distinct autoimmune phenotypes, such as lupus-like syndromes or dilated cardiomyopathy [244,245]. In humans, inactivating CTLA-4 (i.e., ipilimumab) is linked with a wide array of autoimmune disorders including thyroiditis [246,247,248,249,250,251]. Immune-related adverse events (irAEs) associated with inhibitors of PD-1/PD-L1 axis tend to be more limited in severity and incidence, but have an earlier onset than the adverse effects of CTLA-4 inhibition associated with these agents, which are sometimes severe and can occur during treatment or even long after treatment cessation [230]). Hypothyroidism is more common with anti-PD-1 antibodies than ipilimumab (4–10% versus 2–4% respectively), is rarely severe [252,253,254,255,256,257,258], and occurs commonly after subclinical hyperthyroidism [259,260,261].

9. Outstanding Questions and Conclusions

During the last years, the incidence of TC has increased, and it now represents approximately 90% of all endocrine malignancies and 70% of deaths due to endocrine cancers [2]. While the prognosis of DTC is favorable, PDTC and ATC are among the most lethal human malignancies. Nearly all immune cells are present in the TC microenvironment, and in some cases are associated with patient outcome [32,47,121,123,124]. Studies assessing the functional role of TAMs, TAMCs, and TANs have provided evidence for their protumorigenic role [45,47,165]. The majority of these studies were merely quantitative and qualitative analyses of immune cells in the TME of TC. Unfortunately, the presence and functional role(s) of several subsets of immune cells (e.g., NKT and γδ T cells, Tfh, Th9, Th17, and Tc17), which are known to be relevant for tumor initiation and growth, have not yet been investigated in the microenvironment of different types of TC.
Macrophages are key components of the TC microenvironment, and TAM and M2 macrophages are associated with more aggressive cancers (PDTC and ATC) [121], larger tumor size [118], lymph nodes [88], and lung metastases [124]. However, increasing evidence indicates that TAMs comprise more than two (M1 and M2) subsets of cells [116,148]. Mast cell density in human PTC and FTC correlated with tumor extrathyroid extension [47,145]. Mast cells, similar to macrophages, also comprise several subsets of cells [149,150]. Therefore, single-cell RNA-seq will be necessary in order to understand the role of different subsets of macrophages and mast cells in thyroid tumorigenesis and as biomarkers of response to ICIs in patients with advanced disease.
Increasing evidence indicates that neutrophils, which were originally believed to be homogeneous and terminally differentiated cells, are involved in tumor immunity [107,152]. Several studies have simplistically suggested that the NLR could be associated with clinicopathological characteristics of TC patients. The significance of NLR in different types of TC remains uncertain. Galdiero et al. have demonstrated that neutrophils are present in human TC, and TC-CM promoted their activation [165]. Further studies are urgently needed to explore the role of subsets (e.g., low-density, high-density, N1, N2) of neutrophils in different types of TC.
MDSC, similar to macrophages, mast cells, and neutrophils, comprise at least two subsets of cells (i.e., PMN-MDSC and M-MDSC) [170]. PMN-MDSCs in tissues are considered short-living cells, whereas MDSCs in the tumor microenvironment likely represent bona fide M-MDSCs. Unfortunately, the lack of definitive markers for subsets of human MDSCs has so far prevented establishing their exact role in different types of TC. NK cells play a central role in immune surveillance against tumors, and comprise at least two subsets (i.e., CD56hi CD16lo/hi and CD56lo CD16hi). In a mouse model of ATC, it was shown that NK cell-based immunotherapy is an effective therapy of pulmonary metastases [192].
TK inhibitors targeting RET or BRAF can induce either stable disease or partial responses in PTC and FTC metastatic patients, but are much less effective in ATC [9,262]. Therefore, alternative therapeutic approaches for these TC histotypes are needed. A possible approach is represented by drugs targeting the interactions between immune cells and cancer cells and/or tumor stromal cells, including immune cells. Various immunologic approaches are under evaluation in preclinical studies or in early phase clinical trials for the treatment of ATC. Preclinical and preliminary clinical studies have reported encouraging results on the efficacy of mAbs targeting the PD-1/PD-L1 network [228]. The rationale of this immunologic approach is based on the expression of the PD-1/PD-L1 pathway in DTC and ATC [237,238]. A promising strategy is the immunotherapy combined or sequenced with targeted therapy in the treatment of tumors [263,264]. Preliminary results suggest that combining BRAF inhibitor and anti-PD-1 mAb can improve immunity in a murine model of ATC [229]. Similarly, the combination of a TK inhibitor with anti-PD-1/PD-L1 mAbs improved survival in a murine model of ATC [229]. Figure 2 schematically illustrates the rationale for the antitumor effect of BRAF or TK inhibitor in combination with ICI.
Several other immune checkpoint receptors such as TIM-3, Lymphocyte-activation gene 3 (LAG-3), T Cell Immunoreceptor With Ig And ITIM Domains (TIGIT), B- and T-lymphocyte attenuator (BTLA), V-domain Ig suppressor of T cell activation (VISTA), sialic acid-binding immunoglobulin-type lectins (SIGLEC) 9 and 7, and P-selectin glycoprotein ligand (PSGL)-1 have been identified to be potential therapeutic targets in the immunotherapy of tumors [265]. mAbs targeting the above immune checkpoints are under evaluation in preclinical and/or clinical studies, and should be considered also for the treatment of ATC.
TC cells and several immune cells are a major source of protumorigenic and pro-angiogenic cytokines and chemokines [97,98]. Anti-angiogenic agents, eventually in combination with ICIs, could be exploited to block TC growth, since this strategy has been already developed for other tumors [266]. Moreover, blocking immunosuppressive molecules (TGF-β, IL-10) expressed either by cancer cells or by tumor-infiltrating immune cells [267] could represent another therapeutic strategy for the treatment of TC.
Oncolytic viruses (OVs) are non-pathogenetic viral stains or viral mutants that selectively replicate in and kill tumor cells without causing damage to normal cells [268]. The OV dl922-947 reduced CXCL8 and CCL2/MCP-1 expression and inhibited angiogenesis and macrophage infiltration in ATC [269]. The Food and Drug Administration (FDA) has approved the first OV to treat patients with advanced melanoma [270]. Preclinical and clinical studies appear necessary for evaluating ATC virotherapy in the context of TC immunotherapy.
Most of the in vivo experimental studies of TC have been performed with athymic nude mice models. Studies conducted with these models have demonstrated a protumorigenic role of mast cells in human TC [47,48]. The protumorigenic role of macrophages in TC has been established in mice in which macrophage depletion has been obtained by either pharmacological or genetic tools [45,124]. Genetically modified mouse models of TC should be employed to better characterize the role of different immune cell subsets in different stages of tumorigenesis [271].
Extraordinary progress has been made in recent years in the characterization of several, but not all, cells of the immune system in the tumor microenvironment of different TCs. Moreover, it is becoming clear that different subtypes of immune cells play a protumorigenic role, whereas other types play a protective role in TC. Single-cell analysis of peritumoral and intratumoral immune cells could help to elucidate the functions of subsets of cells in different types of TC. The result that will emerge from these studies will contribute to elaborate targeted immunotherapy strategies for the treatment of advanced thyroid cancer.

Author Contributions

G.V., M.R.G., and G.M. wrote the initial draft. S.L., L.M., P.F., S.M.F., A.d.P., and A.A. made substantial contributions and discussed the content. All the authors reviewed the manuscript prior submission.

Funding

This work was supported in part by grants from the CISI-Lab Project (University of Naples Federico II), the CRèME Project, and the TIMING Project (Regione Campania) (G.M.) and from MIUR-PRIN 2017M8YMR8_005 (M.R.G.).

Acknowledgments

The authors apologize to the many researchers who have contributed importantly to this field and whose work has not been cited due to space and citation restrictions. The authors thank Gjada Criscuolo for the critical reading of the manuscript, scientists from CISI Laboratory not listed as authors for invaluable collaborations, medical graphic artist Fabrizio Fiorbianco for preparing the figures, and the administrative staff (Roberto Bifulco and Anna Ferraro), without whom we could not function as an integrated team.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

ALKanaplastic lymphoma kinase
ANGPTangiopoietin
ATCanaplastic thyroid carcinoma
CTLcytotoxic T lymphocyte
CTLA-4cytotoxic T lymphocyte antigen 4
DCsdendritic cell
DTCdifferentiated thyroid carcinoma
ECMextracellular matrix
EMTepithelial-to-mesenchymal transition
FNAfine-needle aspiration
FTCfollicular thyroid cancer
GM-CSFgranulocyte-macrophage colony-stimulating factor
HGFhepatocyte growth factor
HTThyalinizing trabecular tumor
ICIimmune checkpoint inhibitor
IDO1indoleamine 2,3-dioxygenase 1
IFNinterferon
ILCinnate lymphoid cell
INFinterferon
mAbmonoclonal antibody
MDSCmyeloid-derived suppressor cell
MMPmetalloproteinase
MNGmultinodular goiter
MTCmedullary thyroid cancer
NGSnext-generation sequencing
NKnatural killer
NKTnatural killer T cell
NLRneutrophil-to-lymphocyte ratio
NSCLCnon-small-cell lung cancer
OSoverall survival
PD-1programmed cell death protein-1
PD-L1programmed cell death ligand-1
PDTCpoorly differentiated thyroid cancer
PMNpolymorphonuclear cell
PTCpapillary thyroid cancer
RETREarranged during Transfection
RNSreactive nitrogen species
ROSreactive oxygen species
SCFstem cell factor
TAMtumor-associated macrophage
TAMCtumor-associated mast cell
TANtumor-associated neutrophil
TCthyroid cancer
TCRT cell receptor
TDLNtumor-draining lymph node
TfhT follicular helper cell
ThT helper cell
TKtyrosine kinase
TKItyrosine kinase inhibitor
TMEtumor microenvironment
TNF-αtumor necrosis factor-α
Tregregulatory T cell
VEGFvascular endothelial growth factor
VEGFRvascular endothelial growth factor receptor

References

  1. Fagin, J.A.; Wells, S.A., Jr. Biologic and Clinical Perspectives on Thyroid Cancer. N. Engl. J. Med. 2016, 375, 1054–1067. [Google Scholar] [CrossRef] [PubMed]
  2. Lim, H.; Devesa, S.S.; Sosa, J.A.; Check, D.; Kitahara, C.M. Trends in Thyroid Cancer Incidence and Mortality in the United States, 1974–2013. JAMA 2017, 317, 1338–1348. [Google Scholar] [CrossRef] [PubMed]
  3. Kilfoy, B.A.; Zheng, T.; Holford, T.R.; Han, X.; Ward, M.H.; Sjodin, A.; Zhang, Y.; Bai, Y.; Zhu, C.; Guo, G.L.; et al. International patterns and trends in thyroid cancer incidence, 1973–2002. Cancer Causes Control 2009, 20, 525–531. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, A.Y.; Jemal, A.; Ward, E.M. Increasing incidence of differentiated thyroid cancer in the United States, 1988–2005. Cancer 2009, 115, 3801–3807. [Google Scholar] [CrossRef] [PubMed]
  5. Carling, T.; Udelsman, R. Thyroid cancer. Ann. Rev. Med. 2014, 65, 125–137. [Google Scholar] [CrossRef] [PubMed]
  6. Nilubol, N.; Zhang, L.; Kebebew, E. Multivariate analysis of the relationship between male sex, disease-specific survival, and features of tumor aggressiveness in thyroid cancer of follicular cell origin. Thyroid 2013, 23, 695–702. [Google Scholar] [CrossRef] [PubMed]
  7. Butts, C.A.; Brady, J.J., III; Wilhelm, S.; Castor, L.; Sherwood, A.; McCall, A.; Patch, J.; Jones, P.; Cortes, V.; Ong, A.W. Do simple beside lung function tests predict morbidity after rib fractures? Am. J. Surg. 2017, 213, 473–477. [Google Scholar] [CrossRef] [PubMed]
  8. Besic, N.; Auersperg, M.; Us-Krasovec, M.; Golouh, R.; Frkovic-Grazio, S.; Vodnik, A. Effect of primary treatment on survival in anaplastic thyroid carcinoma. Eur J. Surg Oncol. 2001, 27, 260–264. [Google Scholar] [CrossRef]
  9. 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]
  10. Smallridge, R.C.; Marlow, L.A.; Copland, J.A. Anaplastic thyroid cancer: Molecular pathogenesis and emerging therapies. Endocr. Relat. Cancer 2009, 16, 17–44. [Google Scholar] [CrossRef]
  11. Hadoux, J.; Pacini, F.; Tuttle, R.M.; Schlumberger, M. Management of advanced medullary thyroid cancer. Lancet Diabetes Endocrinol. 2016, 4, 64–71. [Google Scholar] [CrossRef]
  12. Fridman, W.H.; Zitvogel, L.; Sautes-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
  13. Cunha, L.L.; Marcello, M.A.; Ward, L.S. The role of the inflammatory microenvironment in thyroid carcinogenesis. Endocr. Relat. Cancer 2014, 21, 85–103. [Google Scholar] [CrossRef]
  14. Antonelli, A.; Ferrari, S.M.; Corrado, A.; Di Domenicantonio, A.; Fallahi, P. Autoimmune thyroid disorders. Autoimmun. Rev. 2015, 14, 174–180. [Google Scholar] [CrossRef]
  15. Suarez, H.G.; Du Villard, J.A.; Caillou, B.; Schlumberger, M.; Tubiana, M.; Parmentier, C.; Monier, R. Detection of activated ras oncogenes in human thyroid carcinomas. Oncogene 1988, 2, 403–406. [Google Scholar]
  16. Lemoine, N.R.; Mayall, E.S.; Wyllie, F.S.; Farr, C.J.; Hughes, D.; Padua, R.A.; Thurston, V.; Williams, E.D.; Wynford-Thomas, D. Activated ras oncogenes in human thyroid cancers. Cancer Res. 1988, 48, 4459–4463. [Google Scholar]
  17. Agrawal, N.; Akbani, R.; Aksoy, B.A.; Ally, A.; Arachchi, H.; Asa, S.L.; Auman, J.T.; Balasundaram, M.; Balu, S.; Baylin, S.B.; et al. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690. [Google Scholar] [CrossRef]
  18. Grieco, M.; Santoro, M.; Berlingieri, M.T.; Melillo, R.M.; Donghi, R.; Bongarzone, I.; Pierotti, M.A.; Della Porta, G.; Fusco, A.; Vecchio, G. PTC is a novel rearranged form of the ret proto-oncogene and is frequently detected in vivo in human thyroid papillary carcinomas. Cell 1990, 60, 557–563. [Google Scholar] [CrossRef]
  19. Pierotti, M.A.; Bongarzone, I.; Borrello, M.G.; Mariani, C.; Miranda, C.; Sozzi, G.; Greco, A. Rearrangements of TRK proto-oncogene in papillary thyroid carcinomas. J. Endocrinol. Investig. 1995, 18, 130–133. [Google Scholar] [CrossRef]
  20. Xing, M. BRAF mutation in thyroid cancer. Endocr.-Relat. Cancer 2005, 12, 245–262. [Google Scholar] [CrossRef] [Green Version]
  21. Mulligan, L.M. RET revisited: Expanding the oncogenic portfolio. Nat. Rev. Cancer 2014, 14, 173–186. [Google Scholar] [CrossRef]
  22. Vanden Borre, P.; Schrock, A.B.; Anderson, P.M.; Morris, J.C., III; Heilmann, A.M.; Holmes, O.; Wang, K.; Johnson, A.; Waguespack, S.G.; Ou, S.I.; et al. Pediatric, Adolescent, and Young Adult Thyroid Carcinoma Harbors Frequent and Diverse Targetable Genomic Alterations, Including Kinase Fusions. Oncologist 2017, 22, 255–263. [Google Scholar] [CrossRef] [Green Version]
  23. Rodrigues, R.F.; Roque, L.; Krug, T.; Leite, V. Poorly differentiated and anaplastic thyroid carcinomas: Chromosomal and oligo-array profile of five new cell lines. Br. J. Cancer 2007, 96, 1237–1245. [Google Scholar] [CrossRef]
  24. Basolo, F.; Pisaturo, F.; Pollina, L.E.; Fontanini, G.; Elisei, R.; Molinaro, E.; Iacconi, P.; Miccoli, P.; Pacini, F. N-ras mutation in poorly differentiated thyroid carcinomas: Correlation with bone metastases and inverse correlation to thyroglobulin expression. Thyroid 2000, 10, 19–23. [Google Scholar] [CrossRef]
  25. Jung, S.H.; Kim, M.S.; Jung, C.K.; Park, H.C.; Kim, S.Y.; Liu, J.; Bae, J.S.; Lee, S.H.; Kim, T.M.; Chung, Y.J. Mutational burdens and evolutionary ages of thyroid follicular adenoma are comparable to those of follicular carcinoma. Oncotarget 2016, 7, 69638–69648. [Google Scholar] [CrossRef]
  26. Yoo, S.K.; Lee, S.; Kim, S.J.; Jee, H.G.; Kim, B.A.; Cho, H.; Song, Y.S.; Cho, S.W.; Won, J.K.; Shin, J.Y.; et al. Comprehensive Analysis of the Transcriptional and Mutational Landscape of Follicular and Papillary Thyroid Cancers. PLoS Genet. 2016, 12, e1006239. [Google Scholar] [CrossRef]
  27. Oliveira, G.; Polonia, A.; Cameselle-Teijeiro, J.M.; Leitao, D.; Sapia, S.; Sobrinho-Simoes, M.; Eloy, C. EWSR1 rearrangement is a frequent event in papillary thyroid carcinoma and in carcinoma of the thyroid with Ewing family tumor elements (CEFTE). Virchows Arch. 2017, 470, 517–525. [Google Scholar] [CrossRef]
  28. Kabata, P.; Kaniuka-Jakubowska, S.; Kabata, W.; Lakomy, J.; Biernat, W.; Sworczak, K.; Jaskiewicz, J.; Swierblewski, M. Primary Ewing Sarcoma of the Thyroid-Eight Cases in a Decade: A Case Report and Literature Review. Front. Endocrinol. Lausanne 2017, 8, 257. [Google Scholar] [CrossRef]
  29. Gane, E.J.; Pianko, S.; Roberts, S.K.; Thompson, A.J.; Zeuzem, S.; Zuckerman, E.; Ben-Ari, Z.; Foster, G.R.; Agarwal, K.; Laursen, A.L.; et al. Safety and efficacy of an 8-week regimen of grazoprevir plus ruzasvir plus uprifosbuvir compared with grazoprevir plus elbasvir plus uprifosbuvir in participants without cirrhosis infected with hepatitis C virus genotypes 1, 2, or 3 (C-CREST-1 and C-CREST-2, part A): Two randomised, phase 2, open-label trials. Lancet Gastroenterol. Hepatol. 2017, 2, 805–813. [Google Scholar]
  30. Liu, R.; Xing, M. TERT promoter mutations in thyroid cancer. Endocr. Relat. Cancer 2016, 23, 143–155. [Google Scholar] [CrossRef]
  31. Caillou, B.; Talbot, M.; Weyemi, U.; Pioche-Durieu, C.; Al Ghuzlan, A.; Bidart, J.M.; Chouaib, S.; Schlumberger, M.; Dupuy, C. Tumor-associated macrophages (TAMs) form an interconnected cellular supportive network in anaplastic thyroid carcinoma. PLoS ONE 2011, 6, 22567. [Google Scholar] [CrossRef]
  32. 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] [Green Version]
  33. Matricardi, P.M.; Dramburg, S.; Alvarez-Perea, A.; Antolin-Amerigo, D.; Apfelbacher, C.; Atanaskovic-Markovic, M.; Berger, U.; Blaiss, M.S.; Blank, S.; Boni, E.; et al. The Role of Mobile Health Technologies in Allergy Care: An EAACI Position Paper. Allergy 2019. [Google Scholar] [CrossRef]
  34. 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] [Green Version]
  35. Kunstman, J.W.; Juhlin, C.C.; Goh, G.; Brown, T.C.; Stenman, A.; Healy, J.M.; Rubinstein, J.C.; Choi, M.; Kiss, N.; Nelson-Williams, C. Characterization of the mutational landscape of anaplastic thyroid cancer via whole-exome sequencing. Hum. Mol. Genet. 2015, 24, 2318–2329. [Google Scholar] [CrossRef] [Green Version]
  36. Jeon, M.J.; Chun, S.M.; Kim, D.; Kwon, H.; Jang, E.K.; Kim, T.Y.; Kim, W.B.; Shong, Y.K.; Jang, S.J.; Song, D.E.; et al. Genomic Alterations of Anaplastic Thyroid Carcinoma Detected by Targeted Massive Parallel Sequencing in a BRAF(V600E) Mutation-Prevalent Area. Thyroid 2016, 26, 683–690. [Google Scholar] [CrossRef]
  37. Kelly, L.M.; Barila, G.; Liu, P.; Evdokimova, V.N.; Trivedi, S.; Panebianco, F.; Gandhi, M.; Carty, S.E.; Hodak, S.P.; Luo, J.; 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. USA 2014, 111, 4233–4238. [Google Scholar] [CrossRef]
  38. Arndt, A.; Steinestel, K.; Rump, A.; Sroya, M.; Bogdanova, T.; Kovgan, L.; Port, M.; Abend, M.; Eder, S. Anaplastic lymphoma kinase (ALK) gene rearrangements in radiation-related human papillary thyroid carcinoma after the Chernobyl accident. J. Pathol. Clin. Res. 2018, 4, 175–183. [Google Scholar] [CrossRef]
  39. Nikiforova, M.N.; Nikitski, A.V.; Panebianco, F.; Kaya, C.; Yip, L.; Williams, M.; Chiosea, S.I.; Seethala, R.R.; Roy, S.; Condello, V.; et al. GLIS Rearrangement is a Genomic Hallmark of Hyalinizing Trabecular Tumor of the Thyroid Gland. Thyroid 2019, 29, 161–173. [Google Scholar] [CrossRef]
  40. Weetman, A.P.; Bennett, G.L.; Wong, W.L. Thyroid follicular cells produce interleukin-8. J. Clin. Endocrinol. Metab. 1992, 75, 328–330. [Google Scholar]
  41. Kammoun-Krichen, M.; Bougacha-Elleuch, N.; Mnif, M.; Bougacha, F.; Charffedine, I.; Rebuffat, S.; Rebai, A.; Glasson, E.; Abid, M.; Ayadi, F.; et al. IL-1beta a potential factor for discriminating between thyroid carcinoma and atrophic thyroiditis. Eur. Cytokine Netw. 2012, 23, 101–106. [Google Scholar]
  42. Varricchi, G.; Harker, J.; Borriello, F.; Marone, G.; Durham, S.R.; Shamji, M.H. T follicular helper (Tfh) cells in normal immune responses and in allergic disorders. Allergy 2016, 71, 1086–1094. [Google Scholar] [CrossRef]
  43. Bacher, P.; Scheffold, A. The effect of regulatory T cells on tolerance to airborne allergens and allergen immunotherapy. J. Allergy Clin. Immunol. 2018, 142, 1697–1709. [Google Scholar] [CrossRef] [Green Version]
  44. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef]
  45. 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] [CrossRef]
  46. Ameziane-El-Hassani, R.; Talbot, M.; de Souza Dos Santos, M.C.; Al Ghuzlan, A.; Hartl, D.; Bidart, J.M.; De Deken, X.; Miot, F.; Diallo, I.; de Vathaire, F.; et al. NADPH oxidase DUOX1 promotes long-term persistence of oxidative stress after an exposure to irradiation. Proc. Natl. Acad. Sci. USA 2015, 112, 5051–5056. [Google Scholar] [CrossRef] [Green Version]
  47. 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]
  48. 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]
  49. Martins, M.B.; Marcello, M.A.; Batista, F.A.; Peres, K.C.; Meneghetti, M.; Ward, M.A.L.; Etchebehere, E.; da Assumpcao, L.V.M.; Ward, L.S. Serum interleukin measurement may help identify thyroid cancer patients with active disease. Clin. Biochem. 2018, 52, 1–7. [Google Scholar] [CrossRef]
  50. Warakomski, J.; Romuk, E.; Jarzab, B.; Krajewska, J.; Sieminska, L. Concentrations of Selected Adipokines, Interleukin-6, and Vitamin D in Patients with Papillary Thyroid Carcinoma in Respect to Thyroid Cancer Stages. Int. J. Endocrinol. 2018, 2018, 4921803. [Google Scholar] [CrossRef]
  51. Sloot, Y.J.E.; Rabold, K.; Netea, M.G.; Smit, J.W.A.; Hoogerbrugge, N.; Netea-Maier, R.T. Effect of PTEN inactivating germline mutations on innate immune cell function and thyroid cancer-induced macrophages in patients with PTEN hamartoma tumor syndrome. Oncogene 2019, 38, 3743. [Google Scholar] [CrossRef]
  52. Li, C.; Li, H.; Jiang, K.; Li, J.; Gai, X. TLR4 signaling pathway in mouse Lewis lung cancer cells promotes the expression of TGF-beta1 and IL-10 and tumor cells migration. Biomed. Mater. Eng. 2014, 24, 869–875. [Google Scholar]
  53. 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]
  54. Todaro, M.; Zerilli, M.; Ricci-Vitiani, L.; Bini, M.; Perez Alea, M.; Maria Florena, A.; Miceli, L.; Condorelli, G.; Bonventre, S.; Di Gesu, G.; et al. Autocrine production of interleukin-4 and interleukin-10 is required for survival and growth of thyroid cancer cells. Cancer Res. 2006, 66, 1491–1499. [Google Scholar] [CrossRef]
  55. Stassi, G.; Todaro, M.; Zerilli, M.; Ricci-Vitiani, L.; Di Liberto, D.; Patti, M.; Florena, A.; Di Gaudio, F.; Di Gesu, G.; De Maria, R. Thyroid cancer resistance to chemotherapeutic drugs via autocrine production of interleukin-4 and interleukin-10. Cancer Res. 2003, 63, 6784–6790. [Google Scholar]
  56. Stanciu, A.E.; Serdarevic, N.; Hurduc, A.E.; Stanciu, M.M. IL-4, IL-10 and high sensitivity-CRP as potential serum biomarkers of persistent/recurrent disease in papillary thyroid carcinoma with/without Hashimoto’s thyroiditis. Scand. J. Clin. Lab. Investig. 2015, 75, 539–548. [Google Scholar] [CrossRef]
  57. Cunha, L.L.; Morari, E.C.; Nonogaki, S.; Marcello, M.A.; Soares, F.A.; Vassallo, J.; Ward, L.S. Interleukin 10 expression is related to aggressiveness and poor prognosis of patients with thyroid cancer. Cancer Immunol. Immunother. 2017, 66, 141–148. [Google Scholar] [CrossRef]
  58. Wang, X.; Li, J.; Lu, C.; Wang, G.; Wang, Z.; Liu, X.; Liu, B.; Zhang, Q.; Yang, Q. IL-10-producing B cells in differentiated thyroid cancer suppress the effector function of T cells but improve their survival upon activation. Exp. Cell Res. 2019, 376, 192–197. [Google Scholar] [CrossRef]
  59. Lu, X. Impact of IL-12 in Cancer. Curr. Cancer Drug Targets 2017, 17, 682–697. [Google Scholar] [CrossRef]
  60. Shi, Y.; Parhar, R.S.; Zou, M.; Baitei, E.; Kessie, G.; Farid, N.R.; Alzahrani, A.; Al-Mohanna, F.A. Gene therapy of anaplastic thyroid carcinoma with a single-chain interleukin-12 fusion protein. Hum. Gene Ther. 2003, 14, 1741–1751. [Google Scholar] [CrossRef]
  61. Parhar, R.S.; Zou, M.; Al-Mohanna, F.A.; Baitei, E.Y.; Assiri, A.M.; Meyer, B.F.; Shi, Y. IL-12 immunotherapy of Braf(V600E)-induced papillary thyroid cancer in a mouse model. Lab. Investig. 2016, 96, 89–97. [Google Scholar] [CrossRef]
  62. 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]
  63. Bailey, S.R.; Nelson, M.H.; Himes, R.A.; Li, Z.; Mehrotra, S.; Paulos, C.M. Th17 cells in cancer: The ultimate identity crisis. Front. Immunol. 2014, 5, 276. [Google Scholar] [CrossRef]
  64. Lu, Y.; Yuan, Y. Serum level of interleukin-17 and interleukin-35 as a biomarker for diagnosis of thyroid cancer. J. Cancer Res. Ther. 2015, 11, 209–211. [Google Scholar] [CrossRef]
  65. Lee, Y.C.; Chung, J.H.; Kim, S.K.; Rhee, S.Y.; Chon, S.; Oh, S.J.; Hong, I.K.; Eun, Y.G. Association between interleukin 17/interleukin 17 receptor gene polymorphisms and papillary thyroid cancer in Korean population. Cytokine 2015, 71, 283–288. [Google Scholar] [CrossRef]
  66. Zhang, L.; Chen, J.; Xu, C.; Qi, L.; Ren, Y. Effects of iodine-131 radiotherapy on Th17/Tc17 and Treg/Th17 cells of patients with differentiated thyroid carcinoma. Exp. Ther. Med. 2018, 15, 2661–2666. [Google Scholar] [CrossRef]
  67. Carvalho, D.F.G.; Zanetti, B.R.; Miranda, L.; Hassumi-Fukasawa, M.K.; Miranda-Camargo, F.; Crispim, J.C.O.; Soares, E.G. High IL-17 expression is associated with an unfavorable prognosis in thyroid cancer. Oncol. Lett. 2017, 13, 1925–1931. [Google Scholar] [CrossRef] [Green Version]
  68. Crotty, S. A brief history of T cell help to B cells. Nat. Rev. Immunol. 2015, 15, 185–189. [Google Scholar] [CrossRef]
  69. Xiao, M.; Hu, S.; Tang, J.; Zhang, L.; Jiang, H. Interleukin (IL)-21 promoter polymorphism increases the risk of thyroid cancer in Chinese population. Gene 2014, 537, 15–19. [Google Scholar] [CrossRef]
  70. Shinohara, S.; Rothstein, J.L. Interleukin 24 is induced by the RET/PTC3 oncoprotein and is an autocrine growth factor for epithelial cells. Oncogene 2004, 23, 7571–7579. [Google Scholar] [CrossRef] [Green Version]
  71. Knauf, J.A.; Sartor, M.A.; Medvedovic, M.; Lundsmith, E.; Ryder, M.; Salzano, M.; Nikiforov, Y.E.; Giordano, T.J.; Ghossein, R.A.; Fagin, J.A. Progression of BRAF-induced thyroid cancer is associated with epithelial-mesenchymal transition requiring concomitant MAP kinase and TGFbeta signaling. Oncogene 2011, 30, 3153–3162. [Google Scholar] [CrossRef]
  72. Eloy, C.; Santos, J.; Cameselle-Teijeiro, J.; Soares, P.; Sobrinho-Simoes, M. TGF-beta/Smad pathway and BRAF mutation play different roles in circumscribed and infiltrative papillary thyroid carcinoma. Virchows Arch. 2012, 460, 587–600. [Google Scholar] [CrossRef]
  73. Ivanova, K.; Manolova, I.; Ignatova, M.M.; Gulubova, M. Immunohistochemical Expression of TGF-Beta1, SMAD4, SMAD7, TGFbetaRII and CD68-Positive TAM Densities in Papillary Thyroid Cancer. Open Access Maced. J. Med. Sci. 2018, 6, 435–441. [Google Scholar] [CrossRef]
  74. Brace, M.D.; Wang, J.; Petten, M.; Bullock, M.J.; Makki, F.; Trites, J.; Taylor, S.M.; Hart, R.D. Differential expression of transforming growth factor-beta in benign vs. papillary thyroid cancer nodules; a potential diagnostic tool? J. Otolaryngol. Head Neck Surg. 2014, 43, 22. [Google Scholar] [CrossRef]
  75. Zhang, K.; Liu, X.; Hao, F.; Dong, A.; Chen, D. Targeting TGF-beta1 inhibits invasion of anaplastic thyroid carcinoma cell through SMAD2-dependent S100A4-MMP-2/9 signalling. Am. J. Transl. Res. 2016, 8, 2196. [Google Scholar]
  76. Zitvogel, L.; Galluzzi, L.; Kepp, O.; Smyth, M.J.; Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 2015, 15, 405–414. [Google Scholar] [CrossRef]
  77. 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]
  78. Lv, N.; Gao, Y.; Guan, H.; Wu, D.; Ding, S.; Teng, W.; Shan, Z. Inflammatory mediators, tumor necrosis factor-alpha and interferon-gamma, induce EMT in human PTC cell lines. Oncol. Lett. 2015, 10, 2591–2597. [Google Scholar] [CrossRef]
  79. Rotondi, M.; Coperchini, F.; Awwad, O.; Pignatti, P.; Di Buduo, C.A.; Abbonante, V.; Magri, F.; Balduini, A.; Chiovato, L. Effect of Interferon-gamma on the Basal and the TNFalpha-Stimulated Secretion of CXCL8 in Thyroid Cancer Cell Lines Bearing Either the RET/PTC Rearrangement Or the BRAF V600e Mutation. Mediat. Inflamm. 2016, 2016, 8512417. [Google Scholar] [CrossRef]
  80. Bosisio, D.; Ronca, R.; Salvi, V.; Presta, M.; Sozzani, S. Dendritic cells in inflammatory angiogenesis and lymphangiogenesis. Curr. Opin. Immunol. 2018, 53, 180–186. [Google Scholar] [CrossRef]
  81. Melillo, R.M.; Castellone, M.D.; Guarino, V.; De Falco, V.; Cirafici, A.M.; Salvatore, G.; Caiazzo, F.; Basolo, F.; Giannini, R.; Kruhoffer, M.; et al. The RET/PTC-RAS-BRAF linear signaling cascade mediates the motile and mitogenic phenotype of thyroid cancer cells. J. Clin. Investig. 2005, 115, 1068–1081. [Google Scholar] [CrossRef] [Green Version]
  82. Borrello, M.G.; Alberti, L.; Fischer, A.; Degl’innocenti, D.; Ferrario, C.; Gariboldi, M.; Marchesi, F.; Allavena, P.; Greco, A.; Collini, P.; et al. Induction of a proinflammatory program in normal human thyrocytes by the RET/PTC1 oncogene. Proc. Natl. Acad. Sci. USA 2005, 102, 14825–14830. [Google Scholar] [CrossRef]
  83. Antonelli, A.; Ferrari, S.M.; Fallahi, P.; Piaggi, S.; Di Domenicantonio, A.; Galleri, D.; Santarpia, L.; Basolo, F.; Ferrannini, E.; Miccoli, P. Variable modulation by cytokines and thiazolidinediones of the prototype Th1 chemokine CXCL10 in anaplastic thyroid cancer. Cytokine 2012, 59, 218–222. [Google Scholar] [CrossRef]
  84. Rotondi, M.; Coperchini, F.; Latrofa, F.; Chiovato, L. Role of Chemokines in Thyroid Cancer Microenvironment: Is CXCL8 the Main Player? Front. Endocrinol. Lausanne 2018, 9, 314. [Google Scholar] [CrossRef]
  85. Rotondi, M.; Coperchini, F.; Pignatti, P.; Magri, F.; Chiovato, L. Metformin reverts the secretion of CXCL8 induced by TNF-alpha in primary cultures of human thyroid cells: An additional indirect anti-tumor effect of the drug. J. Clin. Endocrinol. Metab. 2015, 100, 427–432. [Google Scholar] [CrossRef]
  86. Muzza, M.; Degl’Innocenti, D.; Colombo, C.; Perrino, M.; Ravasi, E.; Rossi, S.; Cirello, V.; Beck-Peccoz, P.; Borrello, M.G.; Fugazzola, L. The tight relationship between papillary thyroid cancer, autoimmunity and inflammation: Clinical and molecular studies. Clin. Endocrinol. Oxf. 2010, 72, 702–708. [Google Scholar] [CrossRef]
  87. Bauerle, K.T.; Schweppe, R.E.; Lund, G.; Kotnis, G.; Deep, G.; Agarwal, R.; Pozdeyev, N.; Wood, W.M.; Haugen, B.R. Nuclear factor kappaB-dependent regulation of angiogenesis, and metastasis in an in vivo model of thyroid cancer is associated with secreted interleukin-8. J. Clin. Endocrinol. Metab. 2014, 99, 1436–1444. [Google Scholar] [CrossRef]
  88. 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] [Green Version]
  89. Antonelli, A.; Ferrari, S.M.; Fallahi, P.; Frascerra, S.; Piaggi, S.; Gelmini, S.; Lupi, C.; Minuto, M.; Berti, P.; Benvenga, S.; et al. Dysregulation of secretion of CXC alpha-chemokine CXCL10 in papillary thyroid cancer: Modulation by peroxisome proliferator-activated receptor-gamma agonists. Endocr. Relat. Cancer 2009, 16, 1299–1311. [Google Scholar] [CrossRef]
  90. Liu, Z.; Sun, D.X.; Teng, X.Y.; Xu, W.X.; Meng, X.P.; Wang, B.S. Expression of stromal cell-derived factor 1 and CXCR7 in papillary thyroid carcinoma. Endocr. Pathol. 2012, 23, 247–253. [Google Scholar] [CrossRef]
  91. Liu, Z.; Yang, L.; Teng, X.; Zhang, H.; Guan, H. The involvement of CXCR7 in modulating the progression of papillary thyroid carcinoma. J. Surg. Res. 2014, 191, 379–388. [Google Scholar] [CrossRef]
  92. Zeng, W.; Chang, H.; Ma, M.; Li, Y. CCL20/CCR6 promotes the invasion and migration of thyroid cancer cells via NF-kappa B signaling-induced MMP-3 production. Exp. Mol. Pathol. 2014, 97, 184–190. [Google Scholar] [CrossRef]
  93. Urra, S.; Fischer, M.C.; Martinez, J.R.; Veliz, L.; Orellana, P.; Solar, A.; Bohmwald, K.; Kalergis, A.; Riedel, C.; Corvalan, A.H.; et al. Differential expression profile of CXCR3 splicing variants is associated with thyroid neoplasia. Potential role in papillary thyroid carcinoma oncogenesis? Oncotarget 2018, 9, 2445–2467. [Google Scholar] [CrossRef]
  94. Loffredo, S.; Staiano, R.I.; Granata, F.; Genovese, A.; Marone, G. Immune cells as a source and target of angiogenic and lymphangiogenic factors. Chem. Immunol. Allergy 2014, 99, 15–36. [Google Scholar]
  95. Varricchi, G.; Raap, U.; Rivellese, F.; Marone, G.; Gibbs, B.F. Human mast cells and basophils-How are they similar how are they different? Immunol. Rev. 2018, 282, 8–34. [Google Scholar] [CrossRef]
  96. Varricchi, G.; Loffredo, S.; Galdiero, M.R.; Marone, G.; Cristinziano, L.; Granata, F. Innate effector cells in angiogenesis and lymphangiogenesis. Curr. Opin. Immunol. 2018, 53, 152–160. [Google Scholar] [CrossRef]
  97. Detoraki, A.; Staiano, R.I.; Granata, F.; Giannattasio, G.; Prevete, N.; de Paulis, A.; Ribatti, D.; Genovese, A.; Triggiani, M.; Marone, G. Vascular endothelial growth factors synthesized by human lung mast cells exert angiogenic effects. J. Allergy Clin. Immunol. 2009, 123, 1142–1149. [Google Scholar] [CrossRef]
  98. Granata, F.; Frattini, A.; Loffredo, S.; Staiano, R.I.; Petraroli, A.; Ribatti, D.; Oslund, R.; Gelb, M.H.; Lambeau, G.; Marone, G. Production of vascular endothelial growth factors from human lung macrophages induced by group IIA and group X secreted phospholipases A2. J. Immunol. 2010, 184, 5232–5241. [Google Scholar] [CrossRef]
  99. Bruno, A.; Focaccetti, C.; Pagani, A.; Imperatori, A.S.; Spagnoletti, M.; Rotolo, N.; Cantelmo, A.R.; Franzi, F.; Capella, C.; Ferlazzo, G.; et al. The proangiogenic phenotype of natural killer cells in patients with non-small cell lung cancer. Neoplasia 2013, 15, 133–142. [Google Scholar] [CrossRef]
  100. Curiel, T.J.; Cheng, P.; Mottram, P.; Alvarez, X.; Moons, L.; Evdemon-Hogan, M.; Wei, S.; Zou, L.; Kryczek, I.; Hoyle, G.; et al. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004, 64, 5535–5538. [Google Scholar] [CrossRef]
  101. Nissim Ben Efraim, A.H.; Levi-Schaffer, F. Roles of eosinophils in the modulation of angiogenesis. Chem. Immunol. Allergy 2014, 99, 138–154. [Google Scholar]
  102. Varricchi, G.; Galdiero, M.R.; Loffredo, S.; Lucarini, V.; Marone, G.; Mattei, F.; Schiavoni, G. Eosinophils: The unsung heroes in cancer? Oncoimmunology 2018, 7, e1393134. [Google Scholar] [CrossRef]
  103. Shiraishi, J.; Koyama, H.; Seki, M.; Hatayama, M.; Naka, M.; Kurajoh, M.; Okazaki, H.; Shoji, T.; Moriwaki, Y.; Yamamoto, T.; et al. Anaplastic thyroid carcinoma accompanied by uncontrollable eosinophilia. Int. Med. 2015, 54, 611–616. [Google Scholar] [CrossRef]
  104. Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef]
  105. Murdoch, C.; Muthana, M.; Coffelt, S.B.; Lewis, C.E. The role of myeloid cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer 2008, 8, 618–631. [Google Scholar] [CrossRef]
  106. Loffredo, S.; Borriello, F.; Iannone, R.; Ferrara, A.L.; Galdiero, M.R.; Gigantino, V.; Esposito, P.; Varricchi, G.; Lambeau, G.; Cassatella, M.A.; et al. Group V Secreted Phospholipase A2 Induces the Release of Proangiogenic and Antiangiogenic Factors by Human Neutrophils. Front. Immunol. 2017, 8, 443. [Google Scholar] [CrossRef] [Green Version]
  107. Galdiero, M.R.; Varricchi, G.; Loffredo, S.; Mantovani, A.; Marone, G. Roles of neutrophils in cancer growth and progression. J. Leukoc. Biol. 2018, 103, 457–464. [Google Scholar] [CrossRef]
  108. Carretero, R.; Sektioglu, I.M.; Garbi, N.; Salgado, O.C.; Beckhove, P.; Hammerling, G.J. Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8+ T cells. Nat. Immunol. 2015, 16, 609–617. [Google Scholar] [CrossRef]
  109. Lucarini, V.; Ziccheddu, G.; Macchia, I.; La Sorsa, V.; Peschiaroli, F.; Buccione, C.; Sistigu, A.; Sanchez, M.; Andreone, S.; D’Urso, M.T.; et al. IL-33 restricts tumor growth and inhibits pulmonary metastasis in melanoma-bearing mice through eosinophils. Oncoimmunology 2017, 6, e1317420. [Google Scholar] [CrossRef]
  110. Galdiero, M.R.; Varricchi, G.; Marone, G. The immune network in thyroid cancer. Oncoimmunology 2016, 5, e1168556. [Google Scholar] [CrossRef] [Green Version]
  111. Bissell, M.J.; Hines, W.C. Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat. Med. 2011, 17, 320–329. [Google Scholar] [CrossRef]
  112. Coussens, L.M.; Zitvogel, L.; Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: A magic bullet? Science 2013, 339, 286–291. [Google Scholar] [CrossRef]
  113. Galdiero, M.R.; Garlanda, C.; Jaillon, S.; Marone, G.; Mantovani, A. Tumor associated macrophages and neutrophils in tumor progression. J. Cell Physiol. 2013, 228, 1404–1412. [Google Scholar] [CrossRef]
  114. Flavell, R.A.; Sanjabi, S.; Wrzesinski, S.H.; Licona-Limon, P. The polarization of immune cells in the tumour environment by TGFbeta. Nat. Rev. Immunol. 2010, 10, 554–567. [Google Scholar] [CrossRef]
  115. Schiavoni, G.; Gabriele, L.; Mattei, F. The tumor microenvironment: A pitch for multiple players. Front. Oncol. 2013, 3, 90. [Google Scholar] [CrossRef]
  116. Chevrier, S.; Levine, J.H.; Zanotelli, V.R.T.; Silina, K.; Schulz, D.; Bacac, M.; Ries, C.H.; Ailles, L.; Jewett, M.A.S.; Moch, H.; et al. An Immune Atlas of Clear Cell Renal Cell Carcinoma. Cell 2017, 169, 736–749. [Google Scholar] [CrossRef]
  117. Dick, S.A.; Macklin, J.A.; Nejat, S.; Momen, A.; Clemente-Casares, X.; Althagafi, M.G.; Chen, J.; Kantores, C.; Hosseinzadeh, S.; Aronoff, L.; et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat. Immunol. 2019, 20, 29–39. [Google Scholar] [CrossRef]
  118. 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. Seoul 2013, 28, 192–198. [Google Scholar] [CrossRef]
  119. Chang, W.C.; Chen, J.Y.; Lee, C.H.; Yang, A.H. Expression of decoy receptor 3 in diffuse sclerosing variant of papillary thyroid carcinoma: Correlation with M2 macrophage differentiation and lymphatic invasion. Thyroid 2013, 23, 720–726. [Google Scholar] [CrossRef]
  120. 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]
  121. Cho, J.W.; Kim, W.W.; Lee, Y.M.; Jeon, M.J.; Kim, W.G.; Song, D.E.; Park, Y.; Chung, K.W.; Hong, S.J.; Sung, T.Y. Impact of tumor-associated macrophages and BRAF(V600E) mutation on clinical outcomes in patients with various thyroid cancers. Head Neck 2019, 41, 686–691. [Google Scholar] [CrossRef]
  122. 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]
  123. 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] [Green Version]
  124. Li, X.J.; Gangadaran, P.; Kalimuthu, S.; Oh, J.M.; Zhu, L.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Role of pulmonary macrophages in initiation of lung metastasis in anaplastic thyroid cancer. Int. J. Cancer 2016, 139, 2583–2592. [Google Scholar] [CrossRef]
  125. Zhang, R.; Hardin, H.; Chen, J.; Guo, Z.; Lloyd, R.V. Non-Coding RNAs in Thyroid Cancer. Endocr. Pathol. 2016, 27, 12–20. [Google Scholar] [CrossRef]
  126. Huang, J.K.; Ma, L.; Song, W.H.; Lu, B.Y.; Huang, Y.B.; Dong, H.M.; Ma, X.K.; Zhu, Z.Z.; Zhou, R. LncRNA-MALAT1 Promotes Angiogenesis of Thyroid Cancer by Modulating Tumor-Associated Macrophage FGF2 Protein Secretion. J. Cell Biochem. 2017, 118, 4821–4830. [Google Scholar] [CrossRef]
  127. Arts, R.J.; Plantinga, T.S.; Tuit, S.; Ulas, T.; Heinhuis, B.; Tesselaar, M.; Sloot, Y.; Adema, G.J.; Joosten, L.A.; Smit, J.W.; et al. Transcriptional and metabolic reprogramming induce an inflammatory phenotype in non-medullary thyroid carcinoma-induced macrophages. Oncoimmunology 2016, 5, e1229725. [Google Scholar] [CrossRef]
  128. Zhang, L.J.; Xiong, Y.; Nilubol, N.; He, M.; Bommareddi, S.; Zhu, X.; Jia, L.; Xiao, Z.; Park, J.W.; Xu, X. Testosterone regulates thyroid cancer progression by modifying tumor suppressor genes and tumor immunity. Carcinogenesis 2015, 36, 420–428. [Google Scholar] [CrossRef] [Green Version]
  129. Dudek, A.M.; Martin, S.; Garg, A.D.; Agostinis, P. Immature, Semi-Mature, and Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments Anticancer Immunity. Front. Immunol. 2013, 4, 438. [Google Scholar] [CrossRef] [Green Version]
  130. 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]
  131. 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]
  132. Tsuge, K.; Takeda, H.; Kawada, S.; Maeda, K.; Yamakawa, M. Characterization of dendritic cells in differentiated thyroid cancer. J. Pathol. 2005, 205, 565–576. [Google Scholar] [CrossRef]
  133. Ugolini, C.; Basolo, F.; Proietti, A.; Vitti, P.; Elisei, R.; Miccoli, P.; Toniolo, A. Lymphocyte and immature dendritic cell infiltrates in differentiated, poorly differentiated, and undifferentiated thyroid carcinoma. Thyroid 2007, 17, 389–393. [Google Scholar] [CrossRef]
  134. Marone, G.; Varricchi, G.; Loffredo, S.; Granata, F. Mast cells and basophils in inflammatory and tumor angiogenesis and lymphangiogenesis. Eur J. Pharmacol. 2016, 778, 146–151. [Google Scholar] [CrossRef]
  135. Varricchi, G.; Galdiero, M.R.; Loffredo, S.; Marone, G.; Iannone, R.; Granata, F. Are Mast Cells MASTers in Cancer? Front. Immunol. 2017, 8, 424. [Google Scholar] [CrossRef] [Green Version]
  136. Varricchi, G.; Galdiero, M.R.; Marone, G.; Granata, F.; Borriello, F. Controversial role of mast cells in skin cancers. Exp. Dermatol. 2017, 26, 11–17. [Google Scholar] [CrossRef]
  137. Ribatti, D.; Vacca, A.; Ria, R.; Marzullo, A.; Nico, B.; Filotico, R.; Roncali, L.; Dammacco, F. Neovascularisation, expression of fibroblast growth factor-2, and mast cells with tryptase activity increase simultaneously with pathological progression in human malignant melanoma. Eur. J. Cancer 2003, 39, 666–674. [Google Scholar] [CrossRef]
  138. Johnson, C.; Huynh, V.; Hargrove, L.; Kennedy, L.; Graf-Eaton, A.; Owens, J.; Trzeciakowski, J.P.; Hodges, K.; DeMorrow, S.; Han, Y.; et al. Inhibition of Mast Cell-Derived Histamine Decreases Human Cholangiocarcinoma Growth and Differentiation via c-Kit/Stem Cell Factor-Dependent Signaling. Am. J. Pathol. 2016, 186, 123–133. [Google Scholar] [CrossRef]
  139. Pittoni, P.; Tripodo, C.; Piconese, S.; Mauri, G.; Parenza, M.; Rigoni, A.; Sangaletti, S.; Colombo, M.P. Mast cell targeting hampers prostate adenocarcinoma development but promotes the occurrence of highly malignant neuroendocrine cancers. Cancer Res. 2011, 71, 5987–5997. [Google Scholar] [CrossRef]
  140. Giannou, A.D.; Marazioti, A.; Spella, M.; Kanellakis, N.I.; Apostolopoulou, H.; Psallidas, I.; Prijovich, Z.M.; Vreka, M.; Zazara, D.E.; Lilis, I.; et al. Mast cells mediate malignant pleural effusion formation. J. Clin. Investig. 2015, 125, 2317–2334. [Google Scholar] [CrossRef]
  141. Siiskonen, H.; Poukka, M.; Bykachev, A.; Tyynela-Korhonen, K.; Sironen, R.; Pasonen-Seppanen, S.; Harvima, I.T. Low numbers of tryptase+ and chymase+ mast cells associated with reduced survival and advanced tumor stage in melanoma. Melanoma Res. 2015, 25, 479–485. [Google Scholar] [CrossRef]
  142. Sammarco, G.; Varricchi, G.; Ferraro, V.; Ammendola, M.; De Fazio, M.; Altomare, D.F.; Luposella, M.; Maltese, L.; Curro, G.; Marone, G.; et al. Mast Cells, Angiogenesis and Lymphangiogenesis in Human Gastric Cancer. Int. J. Mol. Sci. 2019, 20, 2106. [Google Scholar] [CrossRef]
  143. Varricchi, G.; Pecoraro, A.; Marone, G.; Criscuolo, G.; Spadaro, G.; Genovese, A. Thymic Stromal Lymphopoietin Isoforms, Inflammatory Disorders, and Cancer. Front. Immunol. 2018, 9, 1595. [Google Scholar] [CrossRef] [Green Version]
  144. Ribatti, D.; Vacca, A.; Nico, B.; Quondamatteo, F.; Ria, R.; Minischetti, M.; Marzullo, A.; Herken, R.; Roncali, L.; Dammacco, F. Bone marrow angiogenesis and mast cell density increase simultaneously with progression of human multiple myeloma. Br. J. Cancer 1999, 79, 451–455. [Google Scholar] [CrossRef] [Green Version]
  145. 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]
  146. Detoraki, A.; Granata, F.; Staibano, S.; Rossi, F.W.; Marone, G.; Genovese, A. Angiogenesis and lymphangiogenesis in bronchial asthma. Allergy 2010, 65, 946–958. [Google Scholar] [CrossRef]
  147. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
  148. Aran, D.; Looney, A.P.; Liu, L.; Wu, E.; Fong, V.; Hsu, A.; Chak, S.; Naikawadi, R.P.; Wolters, P.J.; Abate, A.R.; et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 2019, 20, 163–172. [Google Scholar] [CrossRef]
  149. Gentek, R.; Ghigo, C.; Hoeffel, G.; Bulle, M.J.; Msallam, R.; Gautier, G.; Launay, P.; Chen, J.; Ginhoux, F.; Bajenoff, M. Hemogenic Endothelial Fate Mapping Reveals Dual Developmental Origin of Mast Cells. Immunity 2018, 48, 1160–1171. [Google Scholar] [CrossRef]
  150. Li, Z.; Liu, S.; Xu, J.; Zhang, X.; Han, D.; Liu, J.; Xia, M.; Yi, L.; Shen, Q.; Xu, S. Adult Connective Tissue-Resident Mast Cells Originate from Late Erythro-Myeloid Progenitors. Immunity 2018, 49, 640–653. [Google Scholar] [CrossRef]
  151. Jaillon, S.; Galdiero, M.R.; Del Prete, D.; Cassatella, M.A.; Garlanda, C.; Mantovani, A. Neutrophils in innate and adaptive immunity. Semin. Immunopathol. 2013, 35, 377–394. [Google Scholar] [CrossRef]
  152. Sionov, R.V.; Fridlender, Z.G.; Granot, Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron. 2015, 8, 125–158. [Google Scholar] [CrossRef]
  153. Bhatti, I.; Peacock, O.; Lloyd, G.; Larvin, M.; Hall, R.I. Preoperative hematologic markers as independent predictors of prognosis in resected pancreatic ductal adenocarcinoma: Neutrophil-lymphocyte versus platelet-lymphocyte ratio. Am. J. Surg. 2010, 200, 197–203. [Google Scholar] [CrossRef]
  154. Liu, C.L.; Lee, J.J.; Liu, T.P.; Chang, Y.C.; Hsu, Y.C.; Cheng, S.P. Blood neutrophil-to-lymphocyte ratio correlates with tumor size in patients with differentiated thyroid cancer. J. Surg Oncol. 2013, 107, 493–497. [Google Scholar] [CrossRef]
  155. Seretis, C.; Gourgiotis, S.; Gemenetzis, G.; Seretis, F.; Lagoudianakis, E.; Dimitrakopoulos, G. The significance of neutrophil/lymphocyte ratio as a possible marker of underlying papillary microcarcinomas in thyroidal goiters: A pilot study. Am. J. Surg. 2013, 205, 691–696. [Google Scholar] [CrossRef]
  156. 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]
  157. Cho, M.J.; Park, K.S.; Yoo, Y.B.; Yang, J.H. A comparative analysis of endoscopic thyroidectomy versus conventional thyroidectomy in clinically lymph node negative thyroid cancer. Ann. Surg Treat. Res. 2015, 88, 69–76. [Google Scholar] [CrossRef] [Green Version]
  158. Liu, J.F.; Ba, L.; Lv, H.; Lv, D.; Du, J.T.; Jing, X.M.; Yang, N.J.; Wang, S.X.; Li, C.; Li, X.X. Association between neutrophil-to-lymphocyte ratio and differentiated thyroid cancer: A meta-analysis. Sci. Rep. 2016, 6, 38551. [Google Scholar] [CrossRef]
  159. Kim, S.M.; Kim, E.H.; Kim, B.H.; Kim, J.H.; Park, S.B.; Nam, Y.J.; Ahn, K.H.; Oh, M.Y.; Kim, W.J.; Jeon, Y.K.; et al. Association of the Preoperative Neutrophil-to-ymphocyte Count Ratio and Platelet-to-Lymphocyte Count Ratio with Clinicopathological Characteristics in Patients with Papillary Thyroid Cancer. Endocrinol. Metab. Seoul 2015, 30, 494–501. [Google Scholar] [CrossRef]
  160. Machairas, N.; Kostakis, I.D.; Prodromidou, A.; Stamopoulos, P.; Feretis, T.; Garoufalia, Z.; Damaskos, C.; Tsourouflis, G.; Kouraklis, G. Trends in white blood cell and platelet indices in a comparison of patients with papillary thyroid carcinoma and multinodular goiter do not permit differentiation between the conditions. Endocr. Res. 2017, 42, 311–317. [Google Scholar] [CrossRef]
  161. Gong, W.; Yang, S.; Yang, X.; Guo, F. Blood preoperative neutrophil-to-lymphocyte ratio is correlated with TNM stage in patients with papillary thyroid cancer. Clin. Sao Paulo 2016, 71, 311–314. [Google Scholar] [CrossRef]
  162. Ozmen, S.; Timur, O.; Calik, I.; Altinkaynak, K.; Simsek, E.; Gozcu, H.; Arslan, A.; Carlioglu, A. Neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) may be superior to C-reactive protein (CRP) for predicting the occurrence of differentiated thyroid cancer. Endocr. Regul. 2017, 51, 131–136. [Google Scholar] [CrossRef] [Green Version]
  163. 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]
  164. Xu, N.; Jian, Y.; Wang, Y.; Tian, W. Evaluation of neutrophil-to-lymphocyte ratio and calcitonin concentration for predicting lymph node metastasis and distant metastasis in patients with medullary thyroid cancer. Mol. Clin. Oncol. 2018, 9, 629–634. [Google Scholar] [CrossRef]
  165. 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]
  166. Adrover, J.M.; Nicolas-Avila, J.A.; Hidalgo, A. Aging: A Temporal Dimension for Neutrophils. Trends Immunol. 2016, 37, 334–345. [Google Scholar] [CrossRef]
  167. Silvestre-Roig, C.; Hidalgo, A.; Soehnlein, O. Neutrophil heterogeneity: Implications for homeostasis and pathogenesis. Blood 2016, 127, 2173–2181. [Google Scholar] [CrossRef]
  168. Sagiv, J.Y.; Michaeli, J.; Assi, S.; Mishalian, I.; Kisos, H.; Levy, L.; Damti, P.; Lumbroso, D.; Polyansky, L.; Sionov, R.V.; et al. Phenotypic diversity and plasticity in circulating neutrophil subpopulations in cancer. Cell Rep. 2015, 10, 562–573. [Google Scholar] [CrossRef]
  169. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer. 2019, 120, 16–25. [Google Scholar] [CrossRef]
  170. Tcyganov, E.; Mastio, J.; Chen, E.; Gabrilovich, D.I. Plasticity of myeloid-derived suppressor cells in cancer. Curr. Opin. Immunol. 2018, 51, 76–82. [Google Scholar] [CrossRef]
  171. Senovilla, L.; Aranda, F.; Galluzzi, L.; Kroemer, G. Impact of myeloid cells on the efficacy of anticancer chemotherapy. Curr. Opin. Immunol. 2014, 30, 24–31. [Google Scholar] [CrossRef]
  172. Millrud, C.R.; Bergenfelz, C.; Leandersson, K. On the origin of myeloid-derived suppressor cells. Oncotarget 2017, 8, 3649–3665. [Google Scholar] [CrossRef]
  173. Kumar, V.; Cheng, P.; Condamine, T.; Mony, S.; Languino, L.R.; McCaffrey, J.C.; Hockstein, N.; Guarino, M.; Masters, G.; Penman, E.; et al. CD45 Phosphatase Inhibits STAT3 Transcription Factor Activity in Myeloid Cells and Promotes Tumor-Associated Macrophage Differentiation. Immunity 2016, 44, 303–315. [Google Scholar] [CrossRef] [Green Version]
  174. Condamine, T.; Dominguez, G.A.; Youn, J.I.; Kossenkov, A.V.; Mony, S.; Alicea-Torres, K.; Tcyganov, E.; Hashimoto, A.; Nefedova, Y.; Lin, C.; et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. 2016, 1. [Google Scholar] [CrossRef]
  175. 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]
  176. Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef]
  177. Chiu, D.K.; Tse, A.P.; Xu, I.M.; Di Cui, J.; Lai, R.K.; Li, L.L.; Koh, H.Y.; Tsang, F.H.; Wei, L.L.; Wong, C.M.; et al. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat. Commun. 2017, 8, 517. [Google Scholar] [CrossRef]
  178. 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. Oxf. 2012, 77, 918–925. [Google Scholar] [CrossRef]
  179. Ohki, S.; Shibata, M.; Gonda, K.; Machida, T.; Shimura, T.; Nakamura, I.; Ohtake, T.; Koyama, Y.; Suzuki, S.; Ohto, H. Circulating myeloid-derived suppressor cells are increased and correlate to immune suppression, inflammation and hypoproteinemia in patients with cancer. Oncol. Rep. 2012, 28, 453–458. [Google Scholar] [CrossRef]
  180. 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] [Green Version]
  181. Jolly, L.A.; Massoll, N.; Franco, A.T. Immune Suppression Mediated by Myeloid and Lymphoid Derived Immune Cells in the Tumor Microenvironment Facilitates Progression of Thyroid Cancers Driven by Hras(G12V) and Pten Loss. J. Clin. Cell Immunol. 2016, 7. [Google Scholar] [CrossRef]
  182. Michelet, X.; Dyck, L.; Hogan, A.; Loftus, R.M.; Duquette, D.; Wei, K.; Beyaz, S.; Tavakkoli, A.; Foley, C.; Donnelly, R. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat. Immunol. 2018, 19, 1330–1340. [Google Scholar] [CrossRef]
  183. Wang, F.; Tian, Z.; Wei, H. Genomic expression profiling of NK cells in health and disease. Eur. J. Immunol. 2015, 45, 661–678. [Google Scholar] [CrossRef]
  184. Modi, J.; Patel, A.; Terrell, R.; Tuttle, R.M.; Francis, G.L. Papillary thyroid carcinomas from young adults and children contain a mixture of lymphocytes. J. Clin. Endocrinol. Metab. 2003, 88, 4418–4425. [Google Scholar] [CrossRef]
  185. Xu, X.; Rao, G.; Gaffud, M.J.; Ding, H.G.; Maki, G.; Klingemann, H.G.; Groh, V.; Spies, T.; Caillat-Zucman, S.; Gattuso, P. Clinicopathological significance of major histocompatibility complex class I-related chain a and B expression in thyroid cancer. J. Clin. Endocrinol. Metab. 2006, 91, 2704–2712. [Google Scholar] [CrossRef]
  186. 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]
  187. Gogali, F.; Paterakis, G.; Rassidakis, G.Z.; Liakou, C.I.; Liapi, C. CD3(-)CD16(-)CD56(bright) immunoregulatory NK cells are increased in the tumor microenvironment and inversely correlate with advanced stages in patients with papillary thyroid cancer. Thyroid 2013, 23, 1561–1568. [Google Scholar] [CrossRef]
  188. Wennerberg, E.; Pfefferle, A.; Ekblad, L.; Yoshimoto, Y.; Kremer, V.; Kaminskyy, V.O.; Juhlin, C.C.; Hoog, 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]
  189. Azad, P.; Zhao, H.W.; Cabrales, P.J.; Ronen, R.; Zhou, D.; Poulsen, O.; Appenzeller, O.; Hsiao, Y.H.; Bafna, V.; Haddad, G.G. Senp1 drives hypoxia-induced polycythemia via GATA1 and Bcl-xL in subjects with Monge’s disease. J. Exp. Med. 2016, 213, 2729–2744. [Google Scholar] [CrossRef]
  190. Park, A.; Lee, Y.; Kim, M.S.; Kang, Y.J.; Park, Y.J.; Jung, H.; Kim, T.D.; Lee, H.G.; Choi, I.; Yoon, S.R. Prostaglandin E2 Secreted by Thyroid Cancer Cells Contributes to Immune Escape Through the Suppression of Natural Killer (NK) Cell Cytotoxicity and NK Cell Differentiation. Front. Immunol. 2018, 9, 1859. [Google Scholar] [CrossRef]
  191. 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]
  192. 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] [Green Version]
  193. Crosby, C.M.; Kronenberg, M. Tissue-specific functions of invariant natural killer T cells. Nat. Rev. Immunol. 2018, 18, 559–574. [Google Scholar] [CrossRef]
  194. 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]
  195. Morrow, E.S.; Roseweir, A.; Edwards, J. The role of gamma delta T lymphocytes in breast cancer: A review. Transl Res. 2019, 203, 88–96. [Google Scholar] [CrossRef]
  196. Ma, Y.; Aymeric, L.; Locher, C.; Mattarollo, S.R.; Delahaye, N.F.; Pereira, P.; Boucontet, L.; Apetoh, L.; Ghiringhelli, F.; Casares, N.; et al. Contribution of IL-17-producing gamma delta T cells to the efficacy of anticancer chemotherapy. J. Exp. Med. 2011, 208, 491–503. [Google Scholar] [CrossRef]
  197. Krabbendam, L.; Bal, S.M.; Spits, H.; Golebski, K. New insights into the function, development, and plasticity of type 2 innate lymphoid cells. Immunol. Rev. 2018, 286, 74–85. [Google Scholar] [CrossRef]
  198. Artis, D.; Spits, H. The biology of innate lymphoid cells. Nature 2015, 517, 293–301. [Google Scholar] [CrossRef]
  199. McHedlidze, T.; Waldner, M.; Zopf, S.; Walker, J.; Rankin, A.L.; Schuchmann, M.; Voehringer, D.; McKenzie, A.N.; Neurath, M.F.; Pflanz, S.; et al. Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis. Immunity 2013, 39, 357–371. [Google Scholar] [CrossRef]
  200. Crome, S.Q.; Nguyen, L.T.; Lopez-Verges, S.; Yang, S.Y.; Martin, B.; Yam, J.Y.; Johnson, D.J.; Nie, J.; Pniak, M.; Yen, P.H. A distinct innate lymphoid cell population regulates tumor-associated T cells. Nat. Med. 2017, 23, 368–375. [Google Scholar] [CrossRef]
  201. Chevalier, M.F.; Trabanelli, S.; Racle, J.; Salome, B.; Cesson, V.; Gharbi, D.; Bohner, P.; Domingos-Pereira, S.; Dartiguenave, F.; Fritschi, A.S.; et al. ILC2-modulated T cell-to-MDSC balance is associated with bladder cancer recurrence. J. Clin. Investig. 2017, 127, 2916–2929. [Google Scholar] [CrossRef]
  202. Russell, J.H.; Ley, T.J. Lymphocyte-mediated cytotoxicity. Ann. Rev. Immunol. 2002, 20, 323–370. [Google Scholar] [CrossRef]
  203. 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. Oxf. 2015, 83, 246–253. [Google Scholar] [CrossRef]
  204. Angell, T.E.; Lechner, M.G.; Jang, J.K.; LoPresti, J.S.; Epstein, A.L. MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in vitro. Clin. Cancer Res. 2014, 20, 6034–6044. [Google Scholar] [CrossRef]
  205. Zhou, Y.; Guo, L.; Sun, H.; Xu, J.; Ba, T. CXCR5+ CD8 T cells displayed higher activation potential despite high PD-1 expression, in tumor-involved lymph nodes from patients with thyroid cancer. Int. Immunopharmacol. 2018, 62, 114–119. [Google Scholar] [CrossRef]
  206. Quezada, S.A.; Simpson, T.R.; Peggs, K.S.; Merghoub, T.; Vider, J.; Fan, X.; Blasberg, R.; Yagita, H.; Muranski, P.; Antony, P.A.; et al. Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J. Exp. Med. 2010, 207, 637–650. [Google Scholar] [CrossRef]
  207. Ruffell, B.; DeNardo, D.G.; Affara, N.I.; Coussens, L.M. Lymphocytes in cancer development: Polarization towards pro-tumor immunity. Cytokine Growth Factor Rev. 2010, 21, 3–10. [Google Scholar] [CrossRef] [Green Version]
  208. Speiser, D.E.; Ho, P.C.; Verdeil, G. Regulatory circuits of T cell function in cancer. Nat. Rev. Immunol. 2016, 16, 599–611. [Google Scholar] [CrossRef]
  209. 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]
  210. Wolf, D.; Sopper, S.; Pircher, A.; Gastl, G.; Wolf, A.M. Treg(s) in Cancer: Friends or Foe? J. Cell Physiol. 2015, 230, 2598–2605. [Google Scholar] [CrossRef]
  211. French, J.D.; Kotnis, G.R.; Said, S.; Raeburn, C.D.; McIntyre, R.C.; Klopper, J.P., Jr.; 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, 934–943. [Google Scholar] [CrossRef]
  212. 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]
  213. 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]
  214. 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] [Green Version]
  215. 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-gamma: Receptors, functions, and roles in diseases. J. Allergy Clin. Immunol. 2011, 127, 701–721. [Google Scholar] [CrossRef]
  216. Johnston, R.J.; Poholek, A.C.; DiToro, D.; Yusuf, I.; Eto, D.; Barnett, B.; Dent, A.L.; Craft, J.; Crotty, S. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 2009, 325, 1006–1010. [Google Scholar] [CrossRef]
  217. Vinuesa, C.G.; Linterman, M.A.; Yu, D.; MacLennan, I.C. Follicular Helper T Cells. Ann. Rev. Immunol. 2016, 34, 335–368. [Google Scholar] [CrossRef]
  218. Ueno, H.; Banchereau, J.; Vinuesa, C.G. Pathophysiology of T follicular helper cells in humans and mice. Nat. Immunol. 2015, 16, 142–152. [Google Scholar] [CrossRef] [Green Version]
  219. Suan, D.; Nguyen, A.; Moran, I.; Bourne, K.; Hermes, J.R.; Arshi, M.; Hampton, H.R.; Tomura, M.; Miwa, Y.; Kelleher, A.D.; et al. T follicular helper cells have distinct modes of migration and molecular signatures in naive and memory immune responses. Immunity 2015, 42, 704–718. [Google Scholar] [CrossRef]
  220. Craft, J.E. Follicular helper T cells in immunity and systemic autoimmunity. Nat. Rev. Rheumatol. 2012, 8, 337–347. [Google Scholar] [CrossRef]
  221. Zhu, C.; Ma, J.; Liu, Y.; Tong, J.; Tian, J.; Chen, J.; Tang, X.; Xu, H.; Lu, L.; Wang, S. Increased frequency of follicular helper T cells in patients with autoimmune thyroid disease. J. Clin. Endocrinol. Metab. 2012, 97, 943–950. [Google Scholar] [CrossRef]
  222. Zhang, J.; Ren, M.; Zeng, H.; Guo, Y.; Zhuang, Z.; Feng, Z.; Yan, H.; Xu, M.; Liang, W.; Yang, C. Elevated follicular helper T cells and expression of IL-21 in thyroid tissues are involved in the pathogenesis of Graves’ disease. Immunol. Res. 2015, 62, 163–174. [Google Scholar] [CrossRef]
  223. Gu-Trantien, C.; Loi, S.; Garaud, S.; Equeter, C.; Libin, M.; de Wind, A.; Ravoet, M.; Le Buanec, H.; Sibille, C.; Manfouo-Foutsop, G.; et al. CD4+ follicular helper T cell infiltration predicts breast cancer survival. J. Clin. Investig. 2013, 123, 2873–2892. [Google Scholar] [CrossRef]
  224. Couillault, C.; Germain, C.; Dubois, B.; Kaplon, H. Identification of Tertiary Lymphoid Structure-Associated Follicular Helper T Cells in Human Tumors and Tissues. Methods Mol. Biol. 2018, 1845, 205–222. [Google Scholar]
  225. Zhao, Y.; Chu, X.; Chen, J.; Wang, Y.; Gao, S.; Jiang, Y.; Zhu, X.; Tan, G.; Zhao, W.; Yi, H.; et al. Dectin-1-activated dendritic cells trigger potent antitumour immunity through the induction of Th9 cells. Nat. Commun. 2016, 7, 12368. [Google Scholar] [CrossRef]
  226. Vegran, F.; Berger, H.; Boidot, R.; Mignot, G.; Bruchard, M.; Dosset, M.; Chalmin, F.; Rebe, C.; Derangere, V.; Ryffel, B.; et al. The transcription factor IRF1 dictates the IL-21-dependent anticancer functions of TH9 cells. Nat. Immunol. 2014, 15, 758–766. [Google Scholar] [CrossRef]
  227. Lu, Y.; Wang, Q.; Xue, G.; Bi, E.; Ma, X.; Wang, A.; Qian, J.; Dong, C.; Yi, Q. Th9 Cells Represent a Unique Subset of CD4+ T Cells Endowed with the Ability to Eradicate Advanced Tumors. Cancer Cell. 2018, 33, 1048–1060. [Google Scholar] [CrossRef]
  228. Brauner, E.; Gunda, V.; Vanden Borre, P.; Zurakowski, D.; Kim, Y.S.; Dennett, K.V.; Amin, S.; Freeman, G.J.; Parangi, S. Combining BRAF inhibitor and anti PD-L1 antibody dramatically improves tumor regression and anti tumor immunity in an immunocompetent murine model of anaplastic thyroid cancer. Oncotarget 2016, 7, 17194–17211. [Google Scholar] [CrossRef]
  229. Gunda, V.; Gigliotti, B.; Ashry, T.; Ndishabandi, D.; McCarthy, M.; Zhou, Z.; Amin, S.; Lee, K.E.; Stork, T.; Wirth, L.; et al. Anti-PD-1/PD-L1 therapy augments lenvatinib’s efficacy by favorably altering the immune microenvironment of murine anaplastic thyroid cancer. Int. J. Cancer 2018, 144, 2266–2278. [Google Scholar] [CrossRef]
  230. Varricchi, G.; Galdiero, M.R.; Tocchetti, C.G. Cardiac Toxicity of Immune Checkpoint Inhibitors: Cardio-Oncology Meets Immunology. Circulation 2017, 136, 1989–1992. [Google Scholar] [CrossRef]
  231. Tocchetti, C.G.; Galdiero, M.R.; Varricchi, G. Cardiac Toxicity in Patients Treated with Immune Checkpoint Inhibitors: It Is Now Time for Cardio-Immuno-Oncology. J. Am. Coll. Cardiol. 2018, 71, 1765–1767. [Google Scholar] [CrossRef]
  232. Varricchi, G.; Galdiero, M.R.; Mercurio, V.; Bonaduce, D.; Marone, G.; Tocchetti, C.G. Pharmacovigilating cardiotoxicity of immune checkpoint inhibitors. Lancet Oncol. 2018, 19, 1545–1546. [Google Scholar] [CrossRef]
  233. Fritz, J.M.; Lenardo, M.J. Development of immune checkpoint therapy for cancer. J. Exp. Med. 2019, 216, 1244–1254. [Google Scholar] [CrossRef]
  234. Okazaki, T.; Chikuma, S.; Iwai, Y.; Fagarasan, S.; Honjo, T. A rheostat for immune responses: The unique properties of PD-1 and their advantages for clinical application. Nat. Immunol. 2013, 14, 1212–1218. [Google Scholar] [CrossRef]
  235. Kataoka, K.; Shiraishi, Y.; Takeda, Y.; Sakata, S.; Matsumoto, M.; Nagano, S.; Maeda, T.; Nagata, Y.; Kitanaka, A.; Mizuno, S.; et al. Aberrant PD-L1 expression through 3’-UTR disruption in multiple cancers. Nature 2016, 534, 402–406. [Google Scholar] [CrossRef]
  236. Peggs, K.S.; Quezada, S.A.; Chambers, C.A.; Korman, A.J.; Allison, J.P. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J. Exp. Med. 2009, 206, 1717–1725. [Google Scholar] [CrossRef]
  237. Bastman, J.J.; Serracino, H.S.; Zhu, Y.; Koenig, M.R.; Mateescu, V.; Sams, S.B.; Davies, K.D.; Raeburn, C.D.; McIntyre, R.C.; Haugen, B.R., Jr.; et al. Tumor-Infiltrating T Cells and the PD-1 Checkpoint Pathway in Advanced Differentiated and Anaplastic Thyroid Cancer. J. Clin. Endocrinol. Metab. 2016, 101, 2863–2873. [Google Scholar] [CrossRef]
  238. Chintakuntlawar, A.V.; Rumilla, K.M.; Smith, C.Y.; Jenkins, S.M.; Foote, R.L.; Kasperbauer, J.L.; Morris, J.C.; Ryder, M.; Alsidawi, S.; Hilger, C. Expression of PD-1 and PD-L1 in Anaplastic Thyroid Cancer Patients Treated with Multimodal Therapy: Results From a Retrospective Study. J. Clin. Endocrinol. Metab. 2017, 102, 1943–1950. [Google Scholar] [CrossRef]
  239. Colli, L.M.; Machiela, M.J.; Myers, T.A.; Jessop, L.; Yu, K.; Chanock, S.J. Burden of Nonsynonymous Mutations among TCGA Cancers and Candidate Immune Checkpoint Inhibitor Responses. Cancer Res. 2016, 76, 3767–3772. [Google Scholar] [CrossRef]
  240. 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]
  241. 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] [Green Version]
  242. 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]
  243. Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef]
  244. Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef]
  245. Nishimura, H.; Okazaki, T.; Tanaka, Y.; Nakatani, K.; Hara, M.; Matsumori, A.; Sasayama, S.; Mizoguchi, A.; Hiai, H.; Minato, N.; et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001, 291, 319–322. [Google Scholar] [CrossRef]
  246. Vaidya, B.; Pearce, S.H.; Charlton, S.; Marshall, N.; Rowan, A.D.; Griffiths, I.D.; Kendall-Taylor, P.; Cawston, T.E.; Young-Min, S. An association between the CTLA4 exon 1 polymorphism and early rheumatoid arthritis with autoimmune endocrinopathies. Rheumatology 2002, 41, 180–183. [Google Scholar] [CrossRef] [Green Version]
  247. Hudson, L.L.; Rocca, K.; Song, Y.W.; Pandey, J.P. CTLA-4 gene polymorphisms in systemic lupus erythematosus: A highly significant association with a determinant in the promoter region. Hum. Genet. 2002, 111, 452–455. [Google Scholar] [CrossRef]
  248. Ueda, H.; Howson, J.M.; Esposito, L.; Heward, J.; Snook, H.; Chamberlain, G.; Rainbow, D.B.; Hunter, K.M.; Smith, A.N.; Di Genova, G.; et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature 2003, 423, 506–511. [Google Scholar] [CrossRef]
  249. Blomhoff, A.; Lie, B.A.; Myhre, A.G.; Kemp, E.H.; Weetman, A.P.; Akselsen, H.E.; Huseby, E.S.; Undlien, D.E. Polymorphisms in the cytotoxic T lymphocyte antigen-4 gene region confer susceptibility to Addison’s disease. J. Clin. Endocrinol. Metab. 2004, 89, 3474–3476. [Google Scholar] [CrossRef]
  250. Zhernakova, A.; Eerligh, P.; Barrera, P.; Wesoly, J.Z.; Huizinga, T.W.; Roep, B.O.; Wijmenga, C.; Koeleman, B.P. CTLA4 is differentially associated with autoimmune diseases in the Dutch population. Hum. Genet. 2005, 118, 58–66. [Google Scholar] [CrossRef]
  251. Fernandez-Mestre, M.; Sanchez, K.; Balbas, O.; Gendzekhzadze, K.; Ogando, V.; Cabrera, M.; Layrisse, Z. Influence of CTLA-4 gene polymorphism in autoimmune and infectious diseases. Hum. Immunol. 2009, 70, 532–535. [Google Scholar] [CrossRef]
  252. Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef]
  253. Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H.; Lao, C.D., Jr.; et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015, 16, 375–384. [Google Scholar] [CrossRef]
  254. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef]
  255. De Filette, J.; Jansen, Y.; Schreuer, M.; Everaert, H.; Velkeniers, B.; Neyns, B.; Bravenboer, B. Incidence of Thyroid-Related Adverse Events in Melanoma Patients Treated With Pembrolizumab. J. Clin. Endocrinol. Metab. 2016, 101, 4431–4439. [Google Scholar] [CrossRef]
  256. Delivanis, D.A.; Gustafson, M.P.; Bornschlegl, S.; Merten, M.M.; Kottschade, L.; Withers, S.; Dietz, A.B.; Ryder, M. Pembrolizumab-Induced Thyroiditis: Comprehensive Clinical Review and Insights Into Underlying Involved Mechanisms. J. Clin. Endocrinol. Metab. 2017, 102, 2770–2780. [Google Scholar] [CrossRef]
  257. Byun, D.J.; Wolchok, J.D.; Rosenberg, L.M.; Girotra, M. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat. Rev. Endocrinol. 2017, 13, 195–207. [Google Scholar] [CrossRef]
  258. Alhusseini, M.; Samantray, J. Hypothyroidism in Cancer Patients on Immune Checkpoint Inhibitors with anti-PD1 Agents: Insights on Underlying Mechanisms. Exp. Clin. Endocrinol. Diabetes. 2017, 125, 267–269. [Google Scholar] [CrossRef]
  259. Eggermont, A.M.; Chiarion-Sileni, V.; Grob, J.J.; Dummer, R.; Wolchok, J.D.; Schmidt, H.; Hamid, O.; Robert, C.; Ascierto, P.A.; Richards, J.M.; et al. Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): A randomised, double-blind, phase 3 trial. Lancet Oncol. 2015, 16, 522–530. [Google Scholar] [CrossRef]
  260. Ferrari, S.M.; Fallahi, P.; Galetta, F.; Citi, E.; Benvenga, S.; Antonelli, A. Thyroid disorders induced by checkpoint inhibitors. Rev. Endocr. Metab. Disord. 2018, 19, 325–333. [Google Scholar] [CrossRef]
  261. Antonelli, A.; Ferrari, S.M.; Fallahi, P. Current and future immunotherapies for thyroid cancer. Expert Rev. Anticancer Ther. 2018, 18, 149–159. [Google Scholar] [CrossRef]
  262. Ferrari, S.M.; Fallahi, P.; Politti, U.; Materazzi, G.; Baldini, E.; Ulisse, S.; Miccoli, P.; Antonelli, A. Molecular Targeted Therapies of Aggressive Thyroid Cancer. Front. Endocrinol. Lausanne 2015, 6, 176. [Google Scholar] [CrossRef] [Green Version]
  263. Atkins, M.B.; Larkin, J. Immunotherapy Combined or Sequenced With Targeted Therapy in the Treatment of Solid Tumors: Current Perspectives. J. Natl. Cancer Inst. 2016, 108, 414. [Google Scholar] [CrossRef]
  264. Palmer, A.C.; Sorger, P.K. Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy. Cell 2017, 171, 1678–1691. [Google Scholar] [CrossRef]
  265. Lee, C.S.; Cragg, M.; Glennie, M.; Johnson, P. Novel antibodies targeting immune regulatory checkpoints for cancer therapy. Br. J. Clin. Pharmacol. 2013, 76, 233–247. [Google Scholar] [CrossRef]
  266. Khan, K.A.; Kerbel, R.S. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat. Rev. Clin. Oncol. 2018, 15, 310–324. [Google Scholar] [CrossRef]
  267. Nguyen, L.T.; Ohashi, P.S. Clinical blockade of PD1 and LAG3–potential mechanisms of action. Nat. Rev. Immunol. 2015, 15, 45–56. [Google Scholar] [CrossRef]
  268. Russell, S.J.; Peng, K.W.; Bell, J.C. Oncolytic virotherapy. Nat. Biotechnol. 2012, 30, 658–670. [Google Scholar] [CrossRef] [Green Version]
  269. 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]
  270. Martinez-Quintanilla, J.; Seah, I.; Chua, M.; Shah, K. Oncolytic viruses: Overcoming translational challenges. J. Clin. Investig. 2019, 130. [Google Scholar] [CrossRef]
  271. Kirschner, L.S.; Qamri, Z.; Kari, S.; Ashtekar, A. Mouse models of thyroid cancer: A 2015 update. Mol. Cell Endocrinol. 2016, 421, 18–27. [Google Scholar] [CrossRef]
Figure 1. Hypothetical scheme of immune contexture of thyroid cancer (TC). The immune network in thyroid cancer is a complex and dynamic system characterized by multiple interactions between tumor cells and nearly all immune cells. Tumor-associated macrophages (TAM), M2 macrophages, tumor-associated mast cells, monocytes, polymorphonuclear-myeloid-derived suppressor cells (PMN-MDSCs), monocyte-derived suppressor cells (M-MDSCs), T regulatory cells (Treg) and T helper 2 (Th2) cells, tumor-associated neutrophils (TAN), and immature DCs (iDCs) and their mediators play protumorigenic roles in thyroid cancer. M1 macrophages, cytotoxic CD8+ T cells, natural killer (NK) cells, Th1 cells, mature DCs (mDCs), and their mediators play an antitumorigenic role. There is increasing evidence that eosinophils play an antitumorigenic role in different cancers [102,108,109]. VEGF-A and CXCL8 produced by thyroid cancer cells activate tumor angiogenesis. Mast cells and macrophages are major producers of lymphangiogenic factors (VEGF-C and VEGF-D). The antitumorigenic role of γδ T cells, Th9 cells, and type I natural killer T (NKT) cells (grey and dashed lines) have been demonstrated in several other human cancers. The protumorigenic role of Tfh cells and of type II NKT cells has been shown in several other human tumors (grey and dashed lines). Protumor or antitumor activities of Th17 and Tc17 cells are context-dependent (grey and dashed line). Modified with permission from Galdiero et al. [110].
Figure 1. Hypothetical scheme of immune contexture of thyroid cancer (TC). The immune network in thyroid cancer is a complex and dynamic system characterized by multiple interactions between tumor cells and nearly all immune cells. Tumor-associated macrophages (TAM), M2 macrophages, tumor-associated mast cells, monocytes, polymorphonuclear-myeloid-derived suppressor cells (PMN-MDSCs), monocyte-derived suppressor cells (M-MDSCs), T regulatory cells (Treg) and T helper 2 (Th2) cells, tumor-associated neutrophils (TAN), and immature DCs (iDCs) and their mediators play protumorigenic roles in thyroid cancer. M1 macrophages, cytotoxic CD8+ T cells, natural killer (NK) cells, Th1 cells, mature DCs (mDCs), and their mediators play an antitumorigenic role. There is increasing evidence that eosinophils play an antitumorigenic role in different cancers [102,108,109]. VEGF-A and CXCL8 produced by thyroid cancer cells activate tumor angiogenesis. Mast cells and macrophages are major producers of lymphangiogenic factors (VEGF-C and VEGF-D). The antitumorigenic role of γδ T cells, Th9 cells, and type I natural killer T (NKT) cells (grey and dashed lines) have been demonstrated in several other human cancers. The protumorigenic role of Tfh cells and of type II NKT cells has been shown in several other human tumors (grey and dashed lines). Protumor or antitumor activities of Th17 and Tc17 cells are context-dependent (grey and dashed line). Modified with permission from Galdiero et al. [110].
Ijms 20 03934 g001
Figure 2. Schematic representation of the rationale developing combinatorial therapies of advanced thyroid cancer involving immune checkpoint inhibitors (monoclonal antibodies (mAbs) anti-cytotoxic T lymphocyte antigen 4 (anti-CTLA-4), anti-programmed cell death protein-1 (anti-PD-1), or anti-programmed cell death ligand-1 (anti-PD-L1)), BRAF inhibitors (BRAFi), multi-targeted tyrosine kinase inhibitors (TKIs) (e.g., lenvatinib), chemotherapies, or radiotherapies. (A) Cancer cells release neoantigens (dots of different colors) that are captured by antigen-presenting cells (APCs). These cells present peptides in the context of MHC I molecule/T cell receptor (TCR) on the surface of CD8+ cytotoxic T cells. APCs can also present peptides bound to MHC II molecules on CD4+ T helper cells. T-cell activation requires costimulatory signals transmitted via CD28, which is activated by binding to CD80 and/or CD86 on APCs. Tumor cells up-regulate CTLA-4 on T cells, which competes with CD28 for binding to CD80/CD86 on APCs. The interaction of CTLA-4 with CD80/CD86 results in inhibitory signaling in T cells, which favors thyroid cancer cell proliferation. The immunosuppressive activity of CTLA-4 is mediated by the down-regulation of Th cells and the enhancement of Treg cells. Moreover, tumor cells express high levels of PD-L1 and/or PD-L2, which binds to PD-1 on T cells, resulting in inhibitory signals that decrease cytotoxicity and lead to T-cell exhaustion. (B) mAbs blocking CTLA-4 (e.g., ipilimumab, tremelimumab), PD-1 (nivolumab, pembrolizumab, spartalizumab), or PD-L1 (avelumab, atezolizumab, durvalumab) inhibit the interactions of CTLA-4/CD80/86 and PD-1/PD-L1, respectively, and activate T-cell cytotoxicity. BRAF inhibitors (BRAFi), TKIs (e.g., lenvatinib), chemotherapies, and radiotherapies can induce thyroid cancer cell death, increasing the release of tumor neoantigens in the tumor microenvironment. Combining an anti-PD-L1 antibody with BRAFi [228,229] or with lenvatinib [229] improved survival and tumor immunity in a immunocompetent murine model of ATC. Several combination strategies involving immune checkpoint inhibitors (ICIs) are under evaluation in patients with advanced TC (see Table 1 and Table 2).
Figure 2. Schematic representation of the rationale developing combinatorial therapies of advanced thyroid cancer involving immune checkpoint inhibitors (monoclonal antibodies (mAbs) anti-cytotoxic T lymphocyte antigen 4 (anti-CTLA-4), anti-programmed cell death protein-1 (anti-PD-1), or anti-programmed cell death ligand-1 (anti-PD-L1)), BRAF inhibitors (BRAFi), multi-targeted tyrosine kinase inhibitors (TKIs) (e.g., lenvatinib), chemotherapies, or radiotherapies. (A) Cancer cells release neoantigens (dots of different colors) that are captured by antigen-presenting cells (APCs). These cells present peptides in the context of MHC I molecule/T cell receptor (TCR) on the surface of CD8+ cytotoxic T cells. APCs can also present peptides bound to MHC II molecules on CD4+ T helper cells. T-cell activation requires costimulatory signals transmitted via CD28, which is activated by binding to CD80 and/or CD86 on APCs. Tumor cells up-regulate CTLA-4 on T cells, which competes with CD28 for binding to CD80/CD86 on APCs. The interaction of CTLA-4 with CD80/CD86 results in inhibitory signaling in T cells, which favors thyroid cancer cell proliferation. The immunosuppressive activity of CTLA-4 is mediated by the down-regulation of Th cells and the enhancement of Treg cells. Moreover, tumor cells express high levels of PD-L1 and/or PD-L2, which binds to PD-1 on T cells, resulting in inhibitory signals that decrease cytotoxicity and lead to T-cell exhaustion. (B) mAbs blocking CTLA-4 (e.g., ipilimumab, tremelimumab), PD-1 (nivolumab, pembrolizumab, spartalizumab), or PD-L1 (avelumab, atezolizumab, durvalumab) inhibit the interactions of CTLA-4/CD80/86 and PD-1/PD-L1, respectively, and activate T-cell cytotoxicity. BRAF inhibitors (BRAFi), TKIs (e.g., lenvatinib), chemotherapies, and radiotherapies can induce thyroid cancer cell death, increasing the release of tumor neoantigens in the tumor microenvironment. Combining an anti-PD-L1 antibody with BRAFi [228,229] or with lenvatinib [229] improved survival and tumor immunity in a immunocompetent murine model of ATC. Several combination strategies involving immune checkpoint inhibitors (ICIs) are under evaluation in patients with advanced TC (see Table 1 and Table 2).
Ijms 20 03934 g002
Table 1. Clinical Trials Evaluating the Effects of PD-1 Inhibitors in Thyroid Cancer.
Table 1. Clinical Trials Evaluating the Effects of PD-1 Inhibitors in Thyroid Cancer.
Clinical Trial Registry NCT NumberPD-1 InhibitorCombinationStudy Phase
NCT03246958NivolumabNivolumab + IpilimumabPhase 2
NCT02834013NivolumabNivolumab + IpilimumabPhase 2
NCT03274258NivolumabNivolumab + IpilimumabPhase 2
NCT03866382NivolumabNivolumab + IpilumabPhase 2
NCT02688608PembrolizumabPembrolizumabPhase 2
NCT03072160PembrolizumabPembrolizumabPhase 2
NCT03360890PembrolizumabPembrolizumab + ChemotherapyPhase 2
NCT03211117PembrolizumabPembrolizumab + Chemotherapy + RadiationPhase 2
NCT02973997PembrolizumabPembrolizumab + LenvatinibPhase 2
NCT03012620PembrolizumabPembrolizumabPhase 2
NCT03435952PembrolizumabPembrolizumab + Clostridium Novyi-NTPhase 1
NCT02628067PembrolizumabPembrolizumabPhase 2
Table 2. Clinical Trials Evaluating the Effects of PD-L1 Inhibitors in Thyroid Cancer.
Table 2. Clinical Trials Evaluating the Effects of PD-L1 Inhibitors in Thyroid Cancer.
NTC NumberPD-L1 InhibitorCombinationStudy Phase
NCT03181100AtezolizumabAtezolizumab + ChemotherapyPhase 2
NCT03170960AtezolizumabAtezolizumab + CabozantinibPhase 1 and 2
NCT03217747AvelumabAvelumab + ChemotherapyPhase 1 and 2
NCT03753919DurvalumabDurvalumab + TremelimumabPhase 2
NCT03215095DurvalumabDurvalumab + RadioiodinePhase 1
NCT03122496DurvalumabDurvalumab + RadiotherapyPhase 1

Share and Cite

MDPI and ACS Style

Varricchi, G.; Loffredo, S.; Marone, G.; Modestino, L.; Fallahi, P.; Ferrari, S.M.; de Paulis, A.; Antonelli, A.; Galdiero, M.R. The Immune Landscape of Thyroid Cancer in the Context of Immune Checkpoint Inhibition. Int. J. Mol. Sci. 2019, 20, 3934. https://doi.org/10.3390/ijms20163934

AMA Style

Varricchi G, Loffredo S, Marone G, Modestino L, Fallahi P, Ferrari SM, de Paulis A, Antonelli A, Galdiero MR. The Immune Landscape of Thyroid Cancer in the Context of Immune Checkpoint Inhibition. International Journal of Molecular Sciences. 2019; 20(16):3934. https://doi.org/10.3390/ijms20163934

Chicago/Turabian Style

Varricchi, Gilda, Stefania Loffredo, Giancarlo Marone, Luca Modestino, Poupak Fallahi, Silvia Martina Ferrari, Amato de Paulis, Alessandro Antonelli, and Maria Rosaria Galdiero. 2019. "The Immune Landscape of Thyroid Cancer in the Context of Immune Checkpoint Inhibition" International Journal of Molecular Sciences 20, no. 16: 3934. https://doi.org/10.3390/ijms20163934

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