Open access peer-reviewed chapter

Papillary Thyroid Carcinoma Intertwined with Hashimoto’s Thyroiditis: An Intriguing Correlation

Written By

Maria V. Deligiorgi and Dimitrios T. Trafalis

Submitted: 02 December 2018 Reviewed: 12 February 2019 Published: 10 April 2019

DOI: 10.5772/intechopen.85128

From the Edited Volume

Knowledges on Thyroid Cancer

Edited by Omer Engin

Chapter metrics overview

1,353 Chapter Downloads

View Full Metrics

Abstract

Illustrating the ancient link connecting inflammation with cancer, the correlation of papillary thyroid carcinoma (PTC) with Hashimoto’s thyroiditis (HT) has long been pursued as intersection of autoimmunity-induced chronic inflammation and tumor-induced immunity. The dramatic rise of the incidence of PTC οver the last decades—the main culprit for “thyroid cancer (TC) epidemic”—parallels the increasing incidence of HT, potentially reflecting a pathogenetic link that could be harnessed in diagnostics and therapeutics. Prompted by this perspective, in the present chapter, we dissect the hitherto elusive interrelationship of PTC with HT, focusing on four issues: firstly, an unresolved conundrum is whether PTC emerges due to or notwithstanding immune response or mirrors the “tumor defense-induced autoimmunity.” Secondly, the interrelationship of HT with PTC may be merely epiphenomenon of selection bias inherent in thyroidectomy series. Thirdly, the impact of HT on coexistent PTC is equivocal—host protective versus tumor protective. Fourthly, translating serum concentrations of thyroid autoantibodies and thyroid-stimulating hormone (TSH) into predictive and prognostic PTC biomarkers dichotomizes, till now, the researchers. In the era of precision medicine, illuminating whether HT precipitates PTC or vice versa is awaited with anticipation in order to refine the preventive and therapeutic policy counteracting “TC epidemic.”

Keywords

  • papillary thyroid carcinoma
  • hashimoto’s thyroiditis
  • anti-thyroglobulin autoantibodies
  • anti-thyroperoxidase autoantibodies
  • thyroid-stimulating hormone

1. Introduction

Initially reported by Dailey et al. in 1955, the correlation of papillary thyroid carcinoma (PTC)—the most common thyroid cancer (TC) histotype—with Hashimoto’s thyroiditis (HT) [1] has long been pursued, rekindling the ancient link between inflammation and cancer [2]. Bearing in mind the rising incidence of PTC over the last decades [3], establishing causality between PTC and HT—an issue highly contested—could lay the groundwork for a preventive policy. Moreover, harnessing the interrelationship of PTC with HT could refine therapeutics with respect to PTC. The present chapter dissects the correlation of PTC with HT, delving into a strongly insinuated immunological link. A comprehensive review of current literature emphasizes on the bewildering clinical significance of the interrelationship of PTC with HT. The intriguing predictive and prognostic value of serum concentrations of thyroid autoantibodies and thyroid-stimulating hormone (TSH) in the context of PTC coexistent with HT paints a more nuanced and sophisticated picture.

Advertisement

2. Tailoring the treatment of PTC: where does coexistent HT stand?

Thyroid cancer (TC) is the most common endocrine malignancy [4], though comprising only 2.1% of global cancer burden [5]. It is estimated that 52,070 new TC cases will occur in 2019 in the United States, while 2170 patients will die of this cancer type [6]. Derived from follicular epithelial cells, PTC constitutes the most common TC subtype in iodine sufficient areas, accounting for 85% of differentiated TC (DTC) [7] and 70–80% of TC [8]. In light of the interface between “TC epidemic” and “epidemic of diagnosis,” a true increase of the incidence of PTC due to environmental, hormonal, and lifestyle risk factors appears to be merged with overdiagnosis of subclinical disease owing to meticulous screening [3, 9, 10, 11].

The indolent nature of PTC imposes a paradigm shift from ameliorating 10-year survival rates exceeding 90% to eliminating the recurrence incidence that hovers at 15–30% [12]. Individualization of therapeutic approach is deemed to confront the emerging challenges [13]. Seminal studies [4, 14, 15] recently illuminated the “dark matter” of the previously unidentified driver genetic events in 96% of PTC [4], being translated into molecular-based risk-adapted therapeutic strategies [7]. Although surgery is the cornerstone of treatment of PTC, a tailored approach with respect to the extent of thyroidectomy and lymph node dissection, the radioiodine ablation, and the management of radioiodine-refractory recurrent/metastatic disease has been endorsed [7].

Provided that the clinical relevance of the increasingly reported interrelationship of PTC with HT is clarified, the incorporation thereof in current PTC risk stratification systems may empower a personalized treatment. This perspective is anticipated to build on accomplishing a fine-tuned balance in terms of decision-making concerning PTC, precluding both overestimating an innocent disease and ignoring a metastatic potential.

Advertisement

3. HT at a glance

HT, originally designated as “struma lymphomatosa” by Dr. Hakaru Hashimoto in 1912 [16], is the most common autoimmune thyroid disease and the most common cause of hypothyroidism in iodine sufficient areas, showing a worldwide annual incidence varying from 0.3 to 1.5 cases per 1000 individuals [17]. An insightful approach concerning the multifactorial etiology of HT has been proposed by Weetman et al.: aligned in a way reminiscent of the wholes of the Swiss cheese are genetic factors acting as susceptibility loci—major histocompatibility human leukocyte antigen (HLA) genes, immunoregulatory genes, thyroid specific genes-environmental factors—excess iodine intake, viral infections, stress, endocrine disruptors—as well as non-modifiable intrinsic factors—female sex, parity, age. Traversed by a hypothetical arrow, this conceivable line translates in a catastrophic event [18].

The histopathologically confirmed HT is characterized by diffuse lymphocytic infiltrate, formation of lymphoid follicles with germinal centers within normal thyroid tissue [19], and, potentially, atrophy of parenchymal tissue gradually replaced by fibrous tissue [20]. The identification of the autoantibodies hallmark of HT in 1936 [21] paved the way for Rose and Witebsky to designate HT as the archetype of autoimmune destructive disorders [22]. Whereas the pathogenesis of HT is unclear, crucial is considered the imbalance between T-helper (Th)2 cells—Th CD (cluster of differentiation)4+ cells credited with stimulation of B cells, which in turn produce thyroid autoantibodies- and Th1 cells-cytotoxic Th CD4+ cells directly attacking the thyroid follicular cells. This concept has been refined by the imbalance between Th17 cells and Th cells producing mainly IL-17, involved also in carcinomas- and T regulatory (Treg) cells-Th CD4+ cells deemed to halt the immune response [23]. Especially, an increased TH17/Treg ratio ascribed to both enhancement of TH17 expression and decrease of Treg is involved in the pathogenesis of HT [24]. Incriminated for the depletion of thyrocytes in HT is principally the autocrine/paracrine Fas-/Fas ligand (FasL)-induced extrinsic apoptotic pathway [24, 25].

Advertisement

4. Rationality in the investigation of the interrelationship of PTC with HT

Apart from the well-established connection of HT with thyroid lymphoma [26], which is beyond the scope of the present chapter, the association of TC with HT concerns almost exclusively the PTC [27], alluding to a discriminating, though unknown, pathogenetic link.

Since PTC is conceived as the main culprit for the explosive rise of TC incidence [3], the hypothesis that the increasing incidence of HT hastens the “TC epidemic” is appealing. Considering that the inflammation has been envisaged as the “seventh hallmark of cancer” [28], the autoimmunity-induced inflammatory milieu [29] merits further interrogation as the missing piece in the puzzle of the interrelationship of PTC with HT.

An alternative explanation that cannot be ruled out is that third extraneous variables actually cause the coexistence of PTC with HT. Indeed, both PTC and HT are precipitated by an interplay among genetic factors and environmental influences most of which are shared by the two entities. Emphasis is placed on risk factors implicated in the pathogenesis of both PTC and HT, such as female predominance, excess iodine intake, and exposure to radiation [30, 31, 32, 33, 34], implying a spurious correlation.

Nonetheless, a common origin of PTC and HT from cancer stem cells expressing p63 proteins—homologs of p53 proteins postulated to regulate squamous stem cell commitment—has been suggested. In fact, the cancer stem cells constitute pluripotent cells deemed to remain undifferentiated or undergo benign squamoid and glandural maturation or be differentiated to follicular epithelial cells, harboring the potential to elicit both PTC and HT [35, 36].

The interrelationship of PTC with HT spurs a realm of intense research, principally in four respects. Firstly, the pathogenetic link between HT and PTC remains elusive; however, accumulative evidence suggests that these two entities are immunologically linked [29]. Secondly, some authors argue that this interrelationship is merely epiphenomenon of selection bias inherent in studies encompassing surgical series [37, 38]. Thirdly, equivocal—favorable versus unfavorable—is the impact of HT on the prognosis of concurrent PTC [39, 40, 41, 42, 43, 44, 45, 46, 47, 48]. Finally, the translation of the serum concentrations of thyroid autoantibodies [49, 50, 51, 52, 53, 54, 55, 56] and thyroid-stimulating hormone (TSH) [55, 57, 58, 59, 60, 61, 62, 63] into predictive and prognostic PTC biomarkers incites a perpetual conflict.

Advertisement

5. Exploring the immunological link between PTC and HT

Compelling evidence insinuate that the PTC and the HT represent two extremes in the continuum of immune response. In cancer, dominant is an anti-inflammatory response dictated by cancer cells per se, counteracting the antitumor immune surveillance. Quite the contrary, an overactivated inflammatory response owing to breakage of self-tolerance attacks host tissue cells, resulting in tissue damage in the context of autoimmune diseases. Despite the fundamental differences between the tumor microenvironment and the autoimmune milieu, certain parallel aspects of these two landscapes have been recognized [64]. For instance, the macrophages (M) and the neutrophils (N)—cells of myeloid origin—are encountered in both cancer and autoimmunity that act as well-coordinated partners to orchestrate the innate immune attack. Showing plasticity, these cells transition from proinflammatory M1/N1 polarization, devoted to kill pathogens or cancer cells, to anti-inflammatory M2/N2 polarization, dedicated to repair tissue damage and promote angiogenesis. A shift toward M2 macrophage polarization is a core component of tumor microenvironment, observed in autoimmune milieu as well, providing a hint to the interface thereof. Furthermore, supportive of the tumor-promoting M2 macrophage polarization is the local hypoxic milieu inherent in both autoimmune and cancerous diseases [64].

The elucidation of the continuum of immune response could provide insights into the pathogenetic background of the coexistence of PTC with HT. Given that the macrophage phenotype M2 is considered tumor-promoting contrary to the antitumor effect of M1 phenotype, an appealing hypothesis connecting PTC with HT is derived from the intrathyroidal immune profiling of euthyroid HT conducted very recently by Imam et al. [65]. The immune infiltrate in euthyroid HT proved to contain low count of natural killer (NK) cells, facilitating the differentiation of the macrophage phenotype M0 to the M2 phenotype, which in concert with the observed low count of M1 macrophages may interpret the higher risk of PTC inherent in euthyroid HT [65].

Interestingly, overexpression of Toll-like receptors (TLR)—cell surface receptors credited with recognition of pathogen-related molecules, crucial for activation of innate and adaptive immunity—is detected immunohistochemically in human thyrocytes surrounded by immune cells in all patients with HT. The high basal TLR3 mRNA levels observed in PTC, reinforcing the shared immunological landscape, are consistent [66, 67].

Dissecting the interface of HT with PTC is expected to unveil novel targets for immunomodulation. For instance, triggering the innate immunity via the TLR5 agonist flagellin, being already in clinical trials as inducer of NK activation [68], could be interrogated as a modality to reverse the M2 macrophage phenotype in PTC coexistent with HT.

In pursuit of the immunological link connecting PTC with HT, three hypotheses, rather interrelated, shape a conceptual framework outlined below.

5.1. Thyroid malignancy develops despite immune response in the context of HT

Manifold mechanisms have been proposed to underlie the escape of PTC cells from immune response in the context of autoimmunity: (i) the ability of PTC cells to manipulate the expression of immune-regulatory cytokines, editing the immune response; (ii) the enhancement of Treg known to suppress the NK cell effector functions, mainly the cytotoxicity; and (iii) the promotion of expression of specific surface molecules facilitating tumor development and growth, such as the membrane-bound transforming growth factor b (TGFb), histocompatibility antigen, class 1, G (HLA-G), FasL, and B7 homolog 1(B7H1) [28].

Moreover, the interrelationship of PTC with HT may empower the escape of cancer cells from immune surveillance, consolidating the dogma that “cancer is a wound that never heals since tumor cells hijack the wound healing machinery for their own gain” [69]. In fact, a recently discovered “unexpected player”, the T cell double negative (DN) CD4(−) CD8(−), expressed both in PTC and in thyroid autoimmunity, downregulates the proliferation of activated T effector cells and the cytokine production, fostering an immunosuppressive microenvironment [70]. Favoring immune tolerance, the FOXP3+ Treg cells—crucial players of thyroid autoimmunity [71]—are encountered also in PTC [70]. The dendritic cells (DCs), beyond governing the autoimmune milieu, are also expressed in PTC, being responsible for the expansion of FOXP3+ Treg cells, allowing the tumor immune evasion and, thus, enabling the PTC progression [72].

5.2. Thyroid malignancy develops owing to thyroid autoimmunity

The first detection of lymphocytes in neoplastic tissues by Virchow in 1863 [73] paved the way for the endorsement of chronic inflammation as a precipitating factor for certain cancer types. In that respect, thyroid gland could be conceived as an intersection of HT-induced chronic inflammation and cancer; however, a causal relationship is yet to be defined. In light of the cancer-related inflammation (CRI), the concurrence of PTC with HT might reflect either the malignant transformation ascribed to an autoimmunity-induced chronic inflammatory milieu (extrinsic pathway) or the inflammatory response to tumor (intrinsic pathway) [74].

The perpetually overactive immune response in the context of HT initiates an inflammatory vicious cycle with the potential to gear the journey of normal cells toward malignancy, rendering the interrelationship of PTC with HT the epitome of the extrinsic pathway of CRI [74].

Central in the extrinsic pathway is the “smoldering inflammation,” an ungoverned inflammatory milieu orchestrated by immune/inflammatory cells, involving macrophages, immature DCs, and mast cells, expressing a myriad of cytokines, chemokines, and growth factors, such as interleukin (IL)-1b, tumor necrosis factor a (TNFa), IL-6, (C-C motif) ligand 2 (CCL2)/monocyte chemoattractant protein 1 (MCP-1), CXC chemokine ligand (CXCL8)/IL-8, vascular endothelial growth factor (VEGF), as well as reactive oxygen species (ROS) and reactive nitrogen species (RNS), spurring tissue damage, neo-angiogenesis, and tissue remodeling [75, 76]. Implicated in this milieu is the hypoxic microenvironment, inherent in both PTC and HT, favoring the progression of tumor, reinforcing, among others, the neo-angiogenesis and the shift of metabolism toward anaerobic glycolysis [64].

Overexpression of cyclooxygenase-2 (COX-2)—an enzyme involved in initiation [77] and progression of thyroid tumors [78]—and inducible nitric oxide synthases (iNOS), key elements of CRI, has been observed in epithelial cells of lymphocytic thyroiditis, follicular adenoma, and PTC contrary to the absence or the limited expression thereof in normal thyroid epithelium, potentially linking carcinogenesis to autoimmunity [79].

Intertwined with the extrinsic pathway is the intrinsic pathway: genetic alterations caused by DNA damage induced by the “smoldering inflammation” [80] trigger a proinflammatory transcriptional program [74]. For instance, the oncogene RAS is involved in the induction of chemokine CXCL8 [75], an inflammatory mediator of both cancer [75] and autoimmunity [64]. Moreover, phosphatase and tensin homolog (PTEN) mutation, a key element of the oncogenic phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, leads to upregulation of hypoxia-inducible factor-1 (HIF1), which, in turn, upregulates the CXC chemokine receptor 4 (CXCR4) [75], well-recognized player of autoimmunity [81]. Accordingly, the observed activation of the PI3K/AKT pathway in HT, PTC, and HT coexistent with PTC contrary to the absence of activation thereof in normal follicles is rational [82].

Illustrating the common molecular background shared by PTC and HT, the rearranged during transfection (RET)/PTC rearrangements—landmarks of PTC—are detected in 95% of HT [83]. Moreover, the RET/PTC1 rearrangement has been detected more frequently in PTC coexistent with autoimmunity than PTC alone (31% versus 13%, respectively) [84]. The inflammatory milieu fosters the genesis of RET/PTC rearrangements either via secreting ROS and RNS [85]—the main culprit for mutagenic-mediated DNA damage [2]—or sustaining the survival of thyroid cells that harbor RET/PTC rearrangements.

The oncogenic RET/PTC-RAS-BRAF-mitogen-activated protein kinase (MAPK) cascade [74] may connect the oxyphil cell metaplasia of HT with PTC, considering the enhancement of the expression of RET, nuclear RAS, and extracellular signal-regulated kinases (ERKs)—core components of MAPK cascade—not only in PTC but also in oxyphil cells in the context of HT [86].

Further, experimental data unravel that the RET/PTC1 exogenously expressed on normal human thyroid cells induces an inflammatory milieu involving crucial chemokines and their receptors, promoting functions vital for tumor progression, such as proliferation and survival of cancer cells [e.g., CXCR4/CXCL1] as well as neo-angiogenesis (e.g., CXL1, 2, 3, 5, 6, and 8) [87].

Additionally, a constellation of RET/PTC1-induced molecules fosters the genesis and evolution of cancer, including (i) matrix metalloproteinases (MMPs) and dipeptidyl peptidase IV (DPP IV), molecules crucial for tissue remodeling, tumor invasiveness, and neo-angiogenesis [87]; (ii) urokinase-type plasminogen activator (UPA) and urokinase-type plasminogen activator receptor (UPAR), involved in cancer progression and metastasis [87]; (iii) l-selectin [87], an adhesion molecule facilitating metastasis [88]; and (iv) osteopontin (OPN) and CD44, implicated in proliferation and invasion of transformed PCCl 3 cells, rat thyroid follicular cells [89].

An intriguing RET/PTC3-induced mechanism pivotal for tumor progression is the recruitment of CD11b+Gr1+ myeloid-derived suppressor cells, providing cancer cells with the advantage of evading immune surveillance [90, 91].

However, skepticism raise the technical limitations of the applied PCR techniques and the lack of reproducibility of the results of studies detecting the RET/PTC rearrangements in HT [92]. Furthermore, the equivocal nature of RET/PTC-induced transcriptional program—tumor-promoting versus antitumor—should be considered [87].

5.3. The immune attack against PTC triggers thyroid autoimmunity

The association of PTC with HT seems more intricate than initially conceived in view of a seminal cyclic model governed by the overactive immune response, acting as a driving force for carcinogenesis, while being also a marker of tumor immunity [93]. An assumption that merits further exploration is whether the cross reaction of antitumor immunity with normal thyrocytes may precipitate HT in PTC patients genetically predisposed to thyroid autoimmunity, consolidating the hypothesis of “tumor defense-induced autoimmunity” [29]. With the advent of the era of cancer immunotherapy, new light on the coexistence of HT with PTC is shed by the increasingly reported development of HT as an adverse event of the monoclonal antibodies blocking programmed cell death (PD) protein 1 (PD-1) and PD ligand 1 (PD-L1). This revolutionary anticancer treatment unleashes the antitumor immunity at the expense of abrogating the self-tolerance, exemplifying the “tumor defense-induced” immunity [94]. For instance, a loss of circulatory PD1+ CD4+ and CD8+ T cells, an increase in peripheral CD56+CD16+ NK cells and an increase in activated monocytes have been implicated in pembrolizumab (anti-PD1 monoclonal antibody)-induced thyroiditis [94].

Advertisement

6. Does the coexistence of HT with PTC really exist?

An issue of major concern is whether the coexistence of PTC with HT is real or a myth nurtured by methodological pitfalls implicit in studies addressing this issue.

The great variety of the incidence of the coexistence of PTC with HT ranging from 0.5 to 38% [95] or, alternatively, from 5 to 85% is noticeable [96]. The results of the meta-analyses addressing the coexistence of PTC with HT are highly divergent [40, 48, 96, 97], as depicted in Table 1. The broad array of the mean rate of PTC among patients with HT extending from 1.1 to 40.1% blurs the landscape [97]. Nevertheless, according to a systematic review, the correlation of PTC with HT is statistically significant with a relative risk (RR) of HT among PTC equal to 2.36 and a RR of PTC among HT equal to 1.40 [98].

Reference Results
Moon et al. [48] PTC coexistent with HT is negatively associated with ETE (OR: 0.74, 95% CI, 0.68–0.81), LNM (OR: 0.82, 95% CI, 0.72–0.94), distant metastasis (OR: 0.49, 95% CI, 0.32–0.76), and recurrence (RR: 0.50, 95% CI, 0.41–0.61)
Lee et al. [96] Frequency of HT in PTC: ≈23%
2.8 times higher occurrence rate of HT in PTC than in benign thyroid diseases (p < 0.001)
2.4 times higher incidence of HT in PTC than in other TC (p < 0.001)
Significant association of PTC concurrent with HT with female sex (OR: 2.7; p < 0.001), multifocality (OR: 1.5, p = 0.010), absence of ETE (OR: 1.3, p = 0.002) and LNM (OR: 1.3, p = 0.041), long recurrence-free survival (HR: 0.6, p = 0.001)
Lai et al. [97] Range of mean rate of PTC among patients with HT: 1.12–40.11%
Overall pooled OR of PTC risk for HT (HT versus non-HT): 2.12 (95% CI, 1.78–2.52)
Singh et al. [40] 2.77 times elevated rate of PTC in patients with HT compared with control population (OR: 2.77, 95% CI, 1.24–6.21)
1.89 times higher rate of HT in patients with PTC compared with other TC types (OR: 1.89, 95% CI, 1.02–3.50)
Increased PTC-free survival in patients with coexistent HT (r: 0.08, 95% CI, 0.05–0.12)
Increased overall survival in PTC patients with coexistent HT (r: 0.11; 95% CI, 0.07–0.14)

Table 1.

Meta-analyses addressing the correlation of PTC with HT.

Abbreviations: CI, confidence interval; ETE, extrathyroidal extension; HR, hazard ratio; HT, Hashimoto’s thyroiditis, LNM, lymph node metastasis; OR, odds ratio; PTC, papillary thyroid carcinoma, RR, risk ratio; TC, thyroid cancer.

In an attempt to annotate the diverse epidemiological profile of the coexistence of PTC with HT, attention should be paid to the discrepancy among pertinent studies concerning the design, the enrolled populations, and the histopathologic definitions of HT [99]. Moreover, certain caveats hamper hitherto the interpretation of the lymphocytic infiltration and the positivity of thyroid autoantibodies. Firstly, thyroid lymphocytic infiltration confirmed on histology has been significantly associated with PTC even in the absence of thyroid autoantibodies [100]. Secondly, the pattern of Tg recognition by anti-thyroglobulin autoantibodies (TgAbs) differs between autoimmune and non-autoimmune thyroid disorders, being more restricted in autoimmune disorders as compared with nodular goiter and PTC harboring no thyroid lymphocytic infiltration [101]. However, in PTC correlated with histopathologically confirmed HT, the pattern of Tg recognition does not differ from that observed in HT [101]. Thirdly, it should be mentioned that the thyroid autoantibodies may be detected in healthy individuals [102]. Finally, the discordance among available TgAbs assays should be considered [103].

Another hurdle in evaluating the coexistence of PTC with HT is the selection bias inherent in data derived from surgical specimens wherein the prevalence of PTC is a priori higher than that in fine needle aspiration biopsy (FNAB) studies. Jankovic et al. showed that the average prevalence rate of PTC in HT patients differed significantly between FNAB and thyroidectomy studies: 1.20 and 27.56%, respectively. Likewise, the relative risk of PTC in HT patients extended from 0.39 to 1.00 in the FNAB studies, significantly lower than that observed in the thyroidectomy studies (1.15–4.16) [37]. In that respect, Castagna et al. demonstrated absence of association of nodular HT with TC based on cytology. The same authors observed a significantly higher prevalence of DTC in nodular HT compared to nodular Graves’ disease, nodular goiter with either negative or positive thyroid autoantibodies, according to surgical series. This result raised the possibility of selection bias ascribed to the fact that 60.7% of patients with nodular HT underwent surgery due to cytological data suspicious of thyroid malignancy [38]. The FNAB data from 10,508 patients revealing no statistically significant relationship between PTC and HT are consistent [104].

Nevertheless, the fear of the selection bias was abolished by the recent demonstration of a significant association of PTC with HT based on either pathological examination of surgical specimens or FNAB studies [60].

Advertisement

7. Effect of HT on coexistent PTC: host protective or tumor protective?

Irrespectively of whether HT is etiologically linked to PTC or merely judged “guilty by association,” the importance of this coexistence lies on its clinical significance. Since a complex immune network has been considered a core component of PTC microenvironment, it is rational to assume that HT—the epitome of aberrant immune reaction—influences the progression of coexistent PTC [105]. In that respect, the positive association of a favorable outcome of coexistent PTC with HT, tumor-associated macrophage infiltration, and CD8+ lymphocytes highlights the antitumor potential of the immunological landscape intrinsic in HT [106]. The recently reported negative correlation of RORγt—a nuclear transcription protein of Th17—with lymph node metastases in PTC concurrent with HT is consistent. In fact, RORγt is positively associated with the upregulation of caveolin 1, a tumor suppressor gene [107]. Another plausible mechanism underlying the host-protective effect of HT coexistent with PTC could be the lower frequency of BRAF V600E mutation—a genetic alteration associated with aggressive PTC phenotype—in PTC concurrent with HT compared with PTC alone [41, 42].

A rich repertoire of features indicative of auspicious PTC prognosis are significantly associated with coexistent HT, including increased relapse-free and overall survival [39], increased survival rate [96, 108], decreased risk of recurrence [108], lower rate [108] or absence of extrathyroidal extension [96], and lower rate [41] or absence of lymph node metastases [96], observed in PTC coexistent with HT compared with PTC alone. The results of a recent meta-analysis including 71 published studies with 44,034 participants revealing that PTC coexistent with HT significantly correlated with reduced incidence of extrathyroidal extension, lymph node, and distant metastasis and increased recurrence-free survival duration compared with PTC alone are seminal [47]. Noticeably, the coexistence of HT with PTC has been proven an independent indicator of favorable prognosis of PTC [105], irrespectively of the extent of lymph node dissection [46], though inconsistently [108]. On the other hand, the reported absence of host-protective effect of HT on coexistent PTC [42, 43, 44, 45] hampers the endorsement of HT as a prognostic PTC biomarker.

In fact, the inflammatory cell infiltration of tumor microenvironment plays an equivocal role, tumor-promoting versus antitumor, posing a “Dr. Jekyll or Mr. Hyde” enigma [109]. Challenging is the illumination of the precise factors that define the fate of cancer cells in the context of the interface of PTC with HT.

Advertisement

8. Thyroid autoantibodies in the context of HT coexistent with PTC: predictive and/or prognostic PTC biomarkers or not?

Whether HT constitutes the driving force for PTC or vice versa remains elusive; nevertheless, the thyroid autoantibodies, the landmark of HT, and especially the anti-thyroperoxidase autoantibodies (TPOAbs)—a more sensitive marker of HT than the TgAbs—merit interrogation as potential hallmarks of the interrelationship of PTC with HT.

A great body of evidence sustains that the positivity of thyroid autoantibodies translates into predictive and prognostic knowledge. In particular, the positivity of TPOAbs [49, 52, 53], TgAbs [51, 53, 110], as well as TPOAbs coexistent with TgAbs [52], has been shown to harbor a predictive value. Moreover, the positivity of TPOAbs [52, 57] and TgAbs [50, 51, 52, 53, 110] has been designated as an independent predictive factor for thyroid malignancy in nodular goiter. Interestingly, the coexistence of TgAbs and TPOAbs is associated with a PTC risk greater than that connected with isolated positivity of either TgAbs or TPOAbs [52]. A host-protective role of TPOAbs in the context of coexistent PTC has been demonstrated [27, 54, 55], rationalized by the speculation that the TPOAbs exert a cytotoxic effect [110].

However, skepticism imposes a multivariate analysis failing to consolidate the host-protective effect of thyroid autoantibodies in the case of coexistent PTC [55]. Importantly, awareness raises the correlation of the positivity of thyroid autoantibodies with features indicative of ominous PTC prognosis, such as advanced disease stage [52]. As a potential link between positive thyroid autoantibodies and aggressive phenotype of PTC could be suggested the excess iodine intake that unmasks a cryptic epitope on Tg, triggering the development of TgAbs [33, 34], while exerting stimulative effect on the genesis of BRAF V600E mutation as well [111]. However, this hypothesis is debunked by the observation that the BRAF V600E mutation in DTC is inversely correlated with coexistent HT [42]. In the light of the foregoing, the designation of thyroid autoantibodies as predictive and/or prognostic biomarkers of PTC is not yet feasible.

Advertisement

9. Elevated TSH levels in HT coexistent with PTC: the mediator of the effect of HT on PTC?

Considering that TSH constitutes a growth factor for thyrocytes [58], rational is the designation of increased, even within the normal range, serum TSH levels, in the case of PTC concurrent with HT, as a predictor of PTC risk [49, 52, 58] and a harbinger of aggressive tumor behavior [55, 59].

A strong argument in favor of the role of TSH in thyroid tumorigenesis is the detection of activating mutations of TSH receptors (TSH-R) in DTC [112]. Moreover, the cross-talk between the TSH-R/protein kinase A (PKA) signaling transduction and the well-recognized oncogenic pathways involving Wingless/int-1 (Wnt), PI3K, and MAPK has been implicated in initiation and progression of TC [113]. However, many arguments against the pathogenetic role of TSH in TC have been raised [114, 115, 116, 117].

Nevertheless, the demonstration of HT as a risk factor for PTC in univariate analysis while being a host-protective factor in multivariate analysis after controlling TSH levels should be mentioned [61]. Similarly, multivariate analysis showed that increased TSH levels were an independent risk factor of malignancy in most FNAB studies, albeit not consistently related to HT [60]. Consequently, the subclinical or overt hypothyroidism due to autoimmune destruction of thyroid—and not HT per se—could be the real culprit for the increased PTC risk in the context of HT.

Experimental data derived from mouse models suggest the TSH-induced signaling mediated via cyclic adenosine monophosphate (cAMP) as a prerequisite for the BRAF V600E-stimulated PTC genesis, providing a plausible explanation for the implication of elevated TSH levels in PTC [62]. Furthermore, a protein kinase C (PKC)-mediated pathway has been demonstrated in vitro to transduce the TSH-induced signaling, dictating the invasiveness and the growth of human follicular TC cell lines [118].

However, the reported association of subclinical hypothyroidism with a less aggressive PTC phenotype compared with euthyroidism cannot be ignored [119]. Consistent is the higher risk of DTC enclosed in HT requiring low levothyroxine (LT4) replacement doses as compared with HT-induced hypothyroidism requiring higher LT4 replacement doses [27]. A hypothesis mandating further exploration is that the toxic effect of TSH mediated by H2O2—an element essential for thyroid hormone synthesis being simultaneously a mitogenic and mutagenic factor—concerns the residual functioning thyroid tissue, while sparing the completely destructed thyroid [27].

Intriguingly, according to the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort, low TSH levels may induce DTC [63], likely forming a less differentiated epithelium susceptible to malignant transformation [27]. Interestingly, two genetic variants predisposing to PTC located on 9q22.23 and 14q13.3 have been also associated with low TSH levels [120]. Consequently, the perplexing role of TSH in PTC fuels a contention regarding the endorsement of TSH levels as predictive and/or prognostic biomarker of PTC.

Advertisement

10. Conclusions

Despite the major strides toward the elucidation of the correlation of PTC with HT, integrating coexistent HT per se, as well as thyroid autoantibodies and TSH levels into PTC risk stratification systems, awaits further consolidation. Translating the coexistence of PTC with HT into the therapeutic approach of PTC is currently uncertain. A burning question is whether the broad clinical spectrum of HT, mirroring the wide array of HT histopathology, defines the trajectory of the coexistence of PTC with HT. The designation of HT as a premalignant lesion or PTC as a precipitating factor for HT is thwarted by the blurred, till now, pathogenetic landscape. Illuminating the temporal precedence, a parameter sine qua non for the embracement of a causal relationship between PTC and TC, is daunting. Nevertheless, harnessing the immunological link between PTC and HT should guide future efforts in clinical research, aiming to widen the horizons of immunotherapy.

In the interim, active surveillance of HT cannot be undermined, since it yields a tangible perspective of a prompt therapeutic intervention in the case of coexistent PTC.

Nonetheless, striking is, to date, the dearth of solid evidence to guide clinical decision-making on surveillance of HT based on the presumptive correlation thereof with PTC; in fact, a patient-oriented standard of care of HT should be applauded. Although thyroid ultrasonography (US) is not required for diagnosing and monitoring the majority of HT, an individualized approach should be endorsed in clinical settings. Bearing in mind the negativity of TPOAbs and/or TgAbs in 10% of HT patients [121] and approximately 20% of patients with subclinical hypothyroidism [122], identifying a hypoechoic or an inhomogeneous US thyroid pattern will provide invaluable information as regards the diagnosis of HT. Even though a thyroid/neck US is not routinely recommended unless a palpable thyroid lesion is detected [7], averting underdiagnosis of a PTC smaller than 1 centimeter (cm) in greatest dimension—the so-called papillary thyroid microcarcinoma—raises awareness. In that respect, US could unravel a nodular variant of HT that merits further evaluation. The management of nodules in the context of HT is governed by the rules applied for any thyroid nodule irrespectively of HT, based on US-guided stratification of risk of malignancy [7]. FNAB is indicated in (i) nodules equal to or larger than 1 cm in greatest dimension presenting sonographic features of high or intermediate suspicion for PTC, (ii) nodules equal to or greater than 1.5 cm in greatest dimension presenting sonographic features of low suspicion for PTC, and (iii) nodules equal to or greater than 2 cm in greatest dimension presenting features of very low suspicion for PTC. Lower size cutoffs are embraced in the presence of clinical risk factors for PTC [7]. Pending the illumination of the clinical significance of the correlation of PTC with HT, clinicians should rely on their discretion and judgment, implementing the principle “primum non nocere.”

Conflict of interest

The authors declare no conflicts of interest.

AKTprotein kinase B
B7H1B7 Homolog 1
cAMPcyclic adenosine monophosphate
CCLchemokine (C-C motif) ligand
CDcluster of differentiation
cmcentimeter
COX-2cyclooxygenase-2
CRIcancer-related inflammation
CXCLCXC chemokine ligand
CXCR4CXC chemokine receptor 4
DCsdendritic cells
DNdouble negative
DPP IVdipeptidyl peptidase IV
DTCdifferentiated thyroid cancer
ERKsextracellular signal-regulated kinases
FasLFas ligand
FNABfine needle aspiration biopsy
HIF-1hypoxia-inducible factor-1
HLAhuman leukocyte antigen
HLA-Ghistocompatibility antigen, class 1, G, known also as human leukocyte antigen G
HTHashimoto’s thyroiditis
ILinterleukin
iNOSinducible nitric oxide synthases
LT4levothyroxine
Mmacrophages
MAPKmitogen-activated protein kinase
MCP-1monocyte chemoattractant protein 1
MMPsmatrix metalloproteinases
Nneutrophils
NKnatural killer
OPNosteopontin
PDprogrammed cell death
PD-1PD protein 1
PD-L1programmed cell death ligand 1
PI3Kphosphoinositide 3-kinase
PKAprotein kinase A
PKCprotein kinase C
PTCpapillary thyroid carcinoma
PTENphosphatase and tensin homolog
RETrearranged during transfection
RNSreactive nitrogen species
ROSreactive oxygen species
TCthyroid cancer
TgAbsanti-thyroglobulin autoantibodies
TGFbtransforming growth factor b
ThT-helper
TLRToll-like receptors
TNFatumor necrosis factor a
TPOAbsanti-thyroperoxidase autoantibodies
TregT regulatory cells
TSHthyroid-stimulating hormone
TSH-RTSH receptor
UPAurokinase-type plasminogen activator
UPARurokinase-type plasminogen activator receptor
USultrasonography
VEGFvascular endothelial growth factor
WntWingless/int-1

References

  1. 1. Dailey ME, Lindsay S, Skahen R. Relation of thyroid neoplasms to Hashimoto disease of the thyroid gland. AMA Archives of Surgery. 1955;70:291-297. DOI: 10.1001/archsurg.1955.01270080137023
  2. 2. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860-867. DOI: 10.1038/nature01322
  3. 3. Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R. Worldwide increasing incidence of thyroid cancer: Update on epidemiology and risk factors. Journal of Cancer Epidemiology. 2013;2013:965212. DOI: 10.1155/2013/965212
  4. 4. Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014;159:676-690. DOI: 10.1016/j.cell.2014.09.050
  5. 5. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer. 2015;136:E359-E386. DOI: 10.1002/ijc.29210
  6. 6. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA: A Cancer Journal for Clinicians. 2019;69:7-34. DOI: 10.3322/caac.21551
  7. 7. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid. 2016;26:1-133. DOI: 10.1089/thy.2015.0020
  8. 8. Nguyen QT, Lee EJ, Huang MG, Park YI, Khullar A, Plodkowski RA. Diagnosis and treatment of patients with thyroid cancer. American Health & Drug Benefits. 2015;8:30-40
  9. 9. Vigneri R, Malandrino P, Vigneri P. The changing epidemiology of thyroid cancer: Why is incidence increasing? Current Opinion in Oncology. 2015;27:1-7. DOI: 10.1097/CCO.0000000000000148
  10. 10. Udelsman R, Zhang Y. The epidemic of thyroid cancer in the United States: The role of endocrinologists and ultrasounds. Thyroid. 2014;24:472-479. DOI: 10.1089/thy.2013.0257
  11. 11. Morris LG, Myssiorek D. Improved detection does not fully explain the rising incidence of well-differentiated thyroid cancer: A population-based analysis. American Journal of Surgery. 2010;200:454-461. DOI: 10.1016/j.amjsurg.2009.11.008
  12. 12. Wong KP, Lang BHH. The role of prophylactic central neck dissection in differentiated thyroid carcinoma: Issues and controversies. Journal of Oncology. 2011;2011:127929. DOI: 10.1155/2011/127929
  13. 13. Shaha AR. Recurrent differentiated thyroid cancer. Endocrine Practice. 2012;18:600-603. DOI: 10.4158/EP12047.CO
  14. 14. Qiu WH, Chen GY, Cui L, Zhang TM, Wei F, Yang Y. Identification of differential pathways in papillary thyroid carcinoma utilizing pathway co-expression analysis. Journal of BUON. 2016;21:1501-1509
  15. 15. Zhao M, Wang KJ, Tan Z, Zheng CM, Liang Z, Zhao JQ. Identification of potential therapeutic targets for papillary thyroid carcinoma by bioinformatics analysis. Oncology Letters. 2016;11:51-58
  16. 16. Hashimoto H. Zur Kenntis der lymphomatosen Veranderung der Schilddruse (Struma lymphomatosa). Langenbecks Archiv für klinische Chirurgie … vereinigt mit Deutsche Zeitschrift für Chirurgie. 1912;97:219-248
  17. 17. Ahmed R, Al-Shaikh S, Akhtar M. Hashimoto thyroiditis: A century later. Advances in Anatomic Pathology. 2012;19:181-186. DOI: 10.1097/PAP.0b013e3182534868
  18. 18. Weetman AP. The immunopathogenesis of chronic autoimmune thyroiditis one century after Hashimoto. European Thyroid Journal. 2013;1:243-250. DOI: 10.1159/000343834
  19. 19. Liu X, Zhu L, Cui D, Wang Z, Chen H, Duan Y, et al. Coexistence of histologically confirmed Hashimoto’s thyroiditis with different stages of papillary thyroid carcinoma in a consecutive Chinese cohort. International Journal of Endocrinology. 2014;2014:769294. DOI: 10.1155/2014/769294
  20. 20. Ward LS. Immune response in thyroid cancer: Widening the boundaries. Scientifica (Cairo). 2014;2014:125450. DOI: 10.1155/2014/125450
  21. 21. Picado C, Rotter W. Precipitines seriques antithyroidiennes chez le goitreux. Comptes rendus des séances de la Société de biologie et de ses filiales. 1936;123:1111
  22. 22. Rose NR, Witebsky E. Studies in organ specificity. V. Changes in the thyroid glands of rabbits following active immunization with rabbit thyroid extracts. Journal of Immunology. 1956;76:417-427
  23. 23. Pyzik A, Grywalska E, Matyjaszek-Matuszek B, Roliński J. Immune disorders in Hashimoto’s thyroiditis: What do we know so far? Journal of Immunology Research. 2015;2015:979167. DOI: 10.1155/2015/979167
  24. 24. Li C, Yuan J, Zhu YF, Yang XJ, Wang Q , Xu J, et al. Imbalance of Th17/Treg in different subtypes of autoimmune thyroid diseases. Cellular Physiology and Biochemistry. 2016;40:245-252. DOI: 10.1159/000452541
  25. 25. Stassi G, Todaro M, Bucchieri F, Stoppacciaro A, Farina F, Zummo G, et al. Fas/Fas ligand-driven T cell apoptosis as a consequence of ineffective thyroid immunoprivilege in Hashimoto’s thyroiditis. Journal of Immunology. 1999;162:263-267
  26. 26. Foppiani L, Secondo V, Arlandini A, Quilici P, Cabria M, Del Monte P. Thyroid lymphoma: A rare tumor requiring combined management. Hormones (Athens, Greece). 2009;8:214-218
  27. 27. Paparodis R, Imam S, Todorova-Koteva K, Staii A, Jaume JC. Hashimoto’s thyroiditis pathology and risk for thyroid cancer. Thyroid. 2014;24:1107-1114. DOI: 10.1089/thy.2013.0588
  28. 28. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144:646-741. DOI: 10.1016/j.cell.2011.02.013
  29. 29. Ehlers M, Schott M. Hashimoto’s thyroiditis and papillary thyroid cancer: Are they immunologically linked? Trends in Endocrinology and Metabolism. 2014;25:656-664. DOI: 10.1016/j.tem.2014.09.001
  30. 30. Dong LQ , Sun XM, Xiang CF, Wu J, Yu P. Hashimoto’s thyroiditis and papillary carcinoma in an adolescent girl: A case report. Molecular and Clinical Oncology. 2016;5:129-131. DOI: 10.3892/mco.2016.895
  31. 31. Effraimidis G, Wiersinga WM. Mechanisms in endocrinology: Autoimmune thyroid disease: Old and new players. European Journal of Endocrinology. 2014;170:R241-R252. DOI: 10.1530/EJE-14-0047
  32. 32. Farahat SA, Mansour N, Sheta MM, Alramlawy SA, Ramadan M. Autoimmune thyroiditis among ionizing radiation exposed workers in cardiac catheterization units. Egyptian Journal of Occupational Medicine. 2017;41:127-1241. DOI: 10.21608/EJOM.2017.966
  33. 33. Li Y, Teng D, Shan Z, Teng X, Guan H, Yu X, et al. Antithyroperoxidase and antithyroglobulin antibodies in a five-year follow-up survey of populations with different iodine intakes. The Journal of Clinical Endocrinology and Metabolism. 2008;93:1751-1757. DOI: 10.1210/jc.2007-2368
  34. 34. Zimmermann MB, Valeria GV. Iodine intake as a risk factor for thyroid cancer: A comprehensive review of animal and human studies. Thyroid Research. 2015;8:8. DOI: 10.1186/s13044-015-0020-8
  35. 35. Burstein DE, Nagi C, Wang BY, Unger P. Immunohistochemical detection of p53 homolog p63 in solid cell nests, papillary thyroid carcinoma, and Hashimoto’s thyroiditis: A stem cell hypothesis of papillary carcinoma oncogenesis. Human Pathology. 2004;35:465-473. DOI: 10.1016/j.humpath.2003.10.027
  36. 36. Unger P, Ewart M, Wang BY, Gan L, Kohtz DS, Burstein DE. Expression of p63 in papillary thyroid carcinoma and in Hashimoto’s thyroiditis: A pathobiologic link? Human Pathology. 2003;34:764-769. DOI: 10.1016/S0046-8177(03)00239-9
  37. 37. Jankovic B, Le KT, Hershman JM. Clinical review: Hashimoto's thyroiditis and papillary thyroid carcinoma: Is there a correlation? The Journal of Clinical Endocrinology and Metabolism. 2013;98:474-482. DOI: 10.1210/jc.2012-2978
  38. 38. Castagna MG, Belardini V, Memmo S, Maino F, Di Santo A, Toti P, et al. Nodules in autoimmune thyroiditis are associated with increased risk of thyroid cancer in surgical series but not in cytological series: Evidence for selection bias. The Journal of Clinical Endocrinology and Metabolism. 2014;99:3193-3198. DOI: 10.1210/jc.2014-1302
  39. 39. Kashima K, Yokoyama S, Noguchi S, Murakami N, Yamashita H, Watanabe S, et al. Chronic thyroiditis as a favorable prognostic factor in papillary thyroid carcinoma. Thyroid. 1998;8:197-202. DOI: 10.1089/thy.1998.8.197
  40. 40. Singh B, Shaha AR, Trivedi H, Carew JF, Poluri A, Shah JP. Coexistent Hashimoto's thyroiditis with papillary thyroid carcinoma: Impact on presentation, management, and outcome. Surgery. 1999;126:1070-1076. DOI: 10.1067/msy.2099.101431
  41. 41. Ma H, Li L, Li K, Wang T, Zhang Y, Zhang C, et al. Hashimoto's thyroiditis, nodular goiter or follicular adenoma combined with papillary thyroid carcinoma play protective role in patients. Neoplasma. 2018;65:436-440. DOI: 10.4149/neo_2018_170428N317
  42. 42. Kim SK, Song KH, Lim SOD, Lim YC, Yoo YB, Kim JS, et al. Clinical and pathological features and the BRAF mutation in patients with papillary thyroid carcinoma with and without concurrent Hashimoto thyroiditis. Thyroid. 2009;19:137-141. DOI: 10.1089/thy.2008.0144
  43. 43. Ye Z, Gu D, Hu H, Zhou Y, Hu X, Zhang X. Hashimoto’s Thyroiditis, microcalcification and raised thyrotropin levels within normal range are associated with thyroid cancer. World Journal of Surgical Oncology. 2013;11:56. DOI: 10.1186/1477-7819-11-56
  44. 44. Del Rio P, Cataldo S, Sommaruga L, Concione L, Arcuri MF, Sianesi M. The association between papillary carcinoma and chronic lymphocytic thyroiditis: Does it modify the prognosis of cancer? Minerva Endocrinologica. 2008;33:1-5
  45. 45. Kebebew E, Treseler PA, Ituarte PH, Clark OH. Coexisting chronic lymphocytic thyroiditis and papillary thyroid cancer revisited. World Journal of Surgery. 2001;25:632-637. DOI: 10.1007/s002680020165
  46. 46. Song E, Jeon MJ, Park S, Kim M, Oh HS, Song DE, et al. Influence of coexistent Hashimoto's thyroiditis on the extent of cervical lymph node dissection and prognosis in papillary thyroid carcinoma. Clinical Endocrinology. 2018;88:123-128. DOI: 10.1111/cen.13475
  47. 47. Kim SS, Lee BJ, Lee JC, Kim SJ, Jeon YK, Kim MR, et al. Coexistence of Hashimoto’s thyroiditis with papillary thyroid carcinoma: The influence of central lymph node metastasis. Head & Neck. 2011;33:1272-1277. DOI: 10.1002/hed.21594
  48. 48. Moon S, Chung HS, Yu JM, Yoo HJ, Park JH, Kim DS, et al. Associations between Hashimoto thyroiditis and clinical outcomes of papillary thyroid cancer: A meta-analysis of observational studies. Endocrinology and Metabolism (Seoul, Korea). 2018;33:473-484. DOI: 10.3803/EnM.2018.33.4.473
  49. 49. Cho YA, Kong S-Y, Shin A, Lee J, Lee EK, Lee YJ, et al. Biomarkers of thyroid function and autoimmunity for predicting high-risk groups of thyroid cancer: A nested case–control study. BMC Cancer. 2014;14:873. DOI: 10.1186/1471-2407-14-873
  50. 50. Zhang X, Zhang X, Chang Z, Wu C, Guo H. Correlation analyses of thyroid-stimulating hormone and thyroid autoantibodies with differentiated thyroid cancer. JBUON. 2018;23:1467-1471
  51. 51. Kim ES, Lim DJ, Baek KH, Lee JM, Kim MK, Kwon HS, et al. Thyroglobulin antibody is associated with increased cancer risk in thyroid nodules. Thyroid. 2010;20:885-891. DOI: 10.1089/thy.2009.0384
  52. 52. Wu X, Lun Y, Jiang H, Gang Q , Xin S, Duan Z, et al. Coexistence of thyroglobulin antibodies and thyroid peroxidase antibodies correlates with elevated thyroid-stimulating hormone level and advanced tumor stage of papillary thyroid cancer. Endocrine. 2013;46:554-560. DOI: 10.1007/s12020-013-0121-x
  53. 53. Qin J, Yu Z, Guan H, Shi L, Liu Y, Zhao N, et al. High thyroglobulin antibody levels increase the risk of differentiated thyroid carcinoma. Disease Markers. 2015;2015:648670. DOI: 10.1155/2015/648670
  54. 54. Souza SL, da Assumpção LVM, Ward LS. Impact of previous thyroid autoimmune diseases on prognosis of patients with well-differentiated thyroid cancer. Thyroid. 2003;13:491-495. DOI: 10.1089/105072503322021160
  55. 55. McLeod DS, Cooper DS, Ladenson PW, Ain KB, Brierley JD, Fein HG, et al. The national thyroid cancer treatment cooperative study group. Prognosis of differentiated thyroid cancer in relation to serum thyrotropin and thyroglobulin antibody status at time of diagnosis. Thyroid. 2014;24:35-42. DOI: 10.1089/thy.2013.0062
  56. 56. Morand GB, da Silva SD, Mlynarek AM, Black MJ, Payne RJ, Hier MP. Clinicopathological relevance of antithyroglobulin antibodies in low-risk papillary thyroid cancer. Clinical Otolaryngology. 2017;42:1130-1134. DOI: 10.1111/coa.12835
  57. 57. Krátký J, Ježková J, Kosák M, Vítková H, Bartáková J, Mráz M, et al. Positive antithyroid antibodies and nonsuppressed TSH are associated with thyroid cancer: A retrospective cross-sectional study. International Journal of Endocrinology. 2018;2018:9793850. DOI: 10.1155/2018/9793850
  58. 58. McLeod DS, Watters KF, Carpenter AD, Ladenson PW, Cooper DS, Ding EL. Thyrotropin and thyroid cancer diagnosis: A systematic review and dose-response meta-analysis. The Journal of Clinical Endocrinology and Metabolism. 2012;97:2682-2692. DOI: 10.1210/jc.2012-1083. Epub 2012 May 23
  59. 59. Haymart MR, Repplinger DJ, Leverson GE, Elson DF, Sippel RS, Jaume JC, et al. Higher serum thyroid stimulating hormone level in thyroid nodule patients is associated with greater risks of differentiated thyroid cancer and advanced tumor stage. The Journal of Clinical Endocrinology and Metabolism. 2008;93:809-814. DOI: 10.1210/jc.2007-2215
  60. 60. Boi F, Pani F, Mariotti S. Thyroid autoimmunity and thyroid cancer: Review focused on cytological studies. European Thyroid Journal. 2017;6:1781-1786. DOI: 10.1159/000468928
  61. 61. Zhang L, Li H, Ji QH, Zhu YX, Wang ZY, Wang Y, et al. The clinical features of papillary thyroid cancer in Hashimoto’s thyroiditis patients from an area with a high prevalence of Hashimoto’s disease. BMC Cancer. 2012;12:610. DOI: 10.1186/1471-2407-12-610
  62. 62. Franco AT, Malaguarnera R, Refetoff S, Liao XH, Lundsmith E, Kimura S, et al. Thyrotrophin receptor signaling dependence of Braf-induced thyroid tumor initiation in mice. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:1615-1620. DOI: 10.1073/pnas.1015557108
  63. 63. Rinaldi S, Plummer M, Biessy C, Tsilidis KK, Østergaard JN, Overvad K, et al. Thyroid-stimulating hormone, thyroglobulin, and thyroid hormones and risk of differentiated thyroid carcinoma: The EPIC study. Journal of the National Cancer Institute. 2014;106:dju097. DOI: 10.1093/jnci/dju097
  64. 64. Rahat MA, Shakya J. Parallel aspects of the microenvironment in cancer and autoimmune disease. Mediators of Inflammation. 2016;2016:1-17. DOI: 10.1155/2016/4375120
  65. 65. Imam S, Dar P, Paparodis R, Almotah K, Al-Khudhair A, Hasan SA, et al. Nature of coexisting thyroid autoimmune disease determines success or failure of tumor immunity in thyroid cancer. Journal for ImmunoTherapy of Cancer. 2019;7:3. DOI: 10.1186/s40425-018-0483-y
  66. 66. Harii N, Lewis CJ, Vasko V, McCall K, Benavides-Peralta U, Sun X, et al. Thyrocytes express a functional toll-like receptor 3: Overexpression can be induced by viral infection and reversed by phenylmethimazole and is associated with Hashimoto’s autoimmune thyroiditis. Molecular Endocrinology. 2005;19:1231-1250. DOI: 10.1210/me.2004-0100
  67. 67. McCall KD, Harii N, Lewis CJ, Malgor R, Kim WB, Saji M, et al. High basal levels of functional toll-like receptor 3 (TLR3) and noncanonical Wnt5a are expressed in papillary thyroid cancer and are coordinately decreased by phenylmethimazole together with cell proliferation and migration. Endocrinology. 2007;148:4226-4237. DOI: 10.1210/en.2007-0459
  68. 68. Hajam IA, Dar PA, Shahnawaz I, Jaume JC, Lee JH. Bacterial flagellin-a potent immunomodulatory agent. Experimental & Molecular Medicine. 2017;49:e373. DOI: 10.1038/emm.2017.172
  69. 69. Dvorak HF. Tumors: Wounds that do not heal-redux. Cancer Immunology Research. 2015;3:1-11. DOI: 10.1158/2326-6066.CIR-14-0209
  70. 70. Imam S, Paparodis R, Sharma D, Jaume JC. Lymphocytic profiling in thyroid cancer provides clues for failure of tumor immunity. Endocrine-Related Cancer. 2014;21:505-516. DOI: 10.1530/ERC-13-0436
  71. 71. Ban Y, Tozaki T, Tobe T, Ban Y, Jacobson EM, Concepcion ES, et al. The regulatory T cell gene FOXP3 and genetic susceptibility to thyroid autoimmunity: An association analysis in Caucasian and Japanese cohorts. Journal of Autoimmunity. 2007;28:201-207. DOI: 10.1016/j.jaut.2007.02.016
  72. 72. Lewinski A, Sliwka PW, Stasiolek M. Dendritic cells in autoimmune disorders and cancer of the thyroid. Folia Histochemica et Cytobiologica. 2014;52:18-28. DOI: 10.5603/FHC.2014.0002
  73. 73. Virchow R. Standpoints in scientific medicine, 1877. Bulletin of the History of Medicine. 1956;30:537-543
  74. 74. Liotti F, Visciano C, Melillo RM. Inflammation in thyroid oncogenesis. American Journal of Cancer Research. 2012;2:286-297
  75. 75. Del Prete A, Allavena P, Santoro G, Fumarulo R, Corsi MM, Mantovani A. Molecular pathways in cancer-related inflammation. Biochemia Medica. 2011;21:264-275
  76. 76. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005;7:211-217. DOI: 10.1016/j.ccr.2005.02.013
  77. 77. Kajita S, Ruebel KH, Casey MB, Nakamura N, Lloyd RV. Role of COX-2, thromboxane A2 synthase, and prostaglandin I2 synthase in papillary thyroid carcinoma growth. Modern Pathology. 2005;18:221-227. DOI: 10.1038/modpathol.3800285
  78. 78. Scarpino S, Duranti E, Giglio S, Di Napoli A, Galafate D, Del Bufalo D, et al. Papillary carcinoma of the thyroid: High expression of COX-2 and low expression of KAI-1/CD82 are associated with increased tumor invasiveness. Thyroid. 2013;23:1127-1137. DOI: 10.1089/thy.2011.0421
  79. 79. Nose F, Ichikawa T, Fujiwara M, Okayasu I. Up-regulation of Cyclooxygenase-2 expression in lymphocytic thyroiditis and thyroid tumors. American Journal of Clinical Pathology. 2002;117:546-551. DOI: 10.1309/9CCJ-XQ8P-PMFM-M65K
  80. 80. Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis. 2009;30:1073-1081. DOI: 10.1016/j.gde.2008.01.003
  81. 81. Pozzobon T, Goldon G, Viola A, Molon B. CXCR4 signaling in health and disease. Immunology Letters. 2016;177:6-15. DOI: 10.1016/j.imlet.2016.06.006
  82. 82. Larson SD, Jackson LN, Riall TS, Uchida T, Thomas RP, Qiu S, et al. Increased incidence of well-differentiated thyroid cancer associated with Hashimoto thyroiditis and the role of the PI3k/Akt pathway. Journal of the American College of Surgeons. 2007;204:764-773. DOI: 10.1016/j.jamcollsurg.2006.12.037
  83. 83. Wirtschafter A, Schmidt R, Rosen D, Kundu N, Santoro M, Fusco A, et al. Expression of the RET/PTC fusion gene as a marker for papillary carcinoma in Hashimoto’s thyroiditis. The Laryngoscope. 1997;107:95-100. DOI: 10.1097/00005537-199701000-00019
  84. 84. Muzza M, Degl'Innocenti D, Colombo C, Perrino M, Ravasi E, Rossi S, et al. The tight relationship between papillary thyroid cancer, autoimmunity and inflammation: Clinical and molecular studies. Clinical Endocrinology. 2010;72:702-708. DOI: 10.1111/j.1365-2265.2009.03699.x
  85. 85. Guarino V, Castellone MD, Avilla E, Melillo RM. Thyroid cancer and inflammation. Molecular and Cellular Endocrinology. 2010;321:94-102. DOI: 10.1016/j.mce.2009.10.003
  86. 86. Kang DY, Kim KH, Kim JM, Kim SH, Kim JY, Baik HW, et al. High prevalence of RET, RAS, and ERK expression in Hashimoto's thyroiditis and in papillary thyroid carcinoma in the Korean population. Thyroid. 2007;17:1031-1038. DOI: 10.1089/thy.2007.0035
  87. 87. Borrello MG, Alberti L, Fischer A, Degl'innocenti D, Ferrario C, Gariboldi M, et al. Induction of a proinflammatory program in normal human thyrocytes by the RET/PTC1 oncogene. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:14825-14830. DOI: 10.1073/pnas.0503039102
  88. 88. Qian F, Hanahan D, Weissman IL. L-selectin can facilitate metastasis to lymph nodes in a transgenic mouse model of carcinogenesis. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:3976-3981. DOI: 10.1073/pnas.061633698
  89. 89. Castellone MD, Celetti A, Guarino V, Cirafici AM, Basolo F, Giannini R, et al. Autocrine stimulation by osteopontin plays a pivotal role in the expression of the mitogenic and invasive phenotype of RET/PTC-transformed thyroid cells. Oncogene. 2004;23:2188-2196. DOI: 10.1038/sj.onc.1207322
  90. 90. Pufnock JS, Rothstein JL. Oncoprotein signaling mediates tumor-specific inflammation and enhances tumor progression. Journal of Immunology. 2009;182:5498-5506. DOI: 10.4049/jimmunol.0801284
  91. 91. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nature Reviews. Immunology. 2009;9:162-174. DOI: 10.1038/nri2506
  92. 92. Nikiforov YE. RET/PTC Rearrangement—A link between Hashimoto’s thyroiditis and thyroid cancer …or not. The Journal of Clinical Endocrinology and Metabolism. 2006;91:2040-2042. DOI: 10.1210/jc.2006-0791
  93. 93. Franks AL, Slansky JE. Multiple associations between a broad spectrum of autoimmune diseases, chronic inflammatory diseases and cancer. Anticancer Research. 2012;32:1119-1136
  94. 94. Girotra M, Hansen A, Farooki A, Byun DJ, Min L, Creelan BC, et al. The current understanding of the endocrine effects from immune checkpoint inhibitors and recommendations for management. JNCI Cancer Spectrum. 2018;2:pky021. DOI: 10.1093/jncics/pky021
  95. 95. Zhang Y, Ma XP, Deng FS, Liu ZR, Wei HQ , Wang XH, et al. The effect of chronic lymphocytic thyroiditis on patients with thyroid cancer. World Journal of Surgical Oncology. 2014;12:277. DOI: 10.1186/1477-7819-12-277
  96. 96. Lee JH, Kim Y, Choi JW, Kim YS. The association between papillary thyroid carcinoma and histologically proven Hashimoto’s thyroiditis: A meta-analysis. European Journal of Endocrinology. 2013;15(168):343-349. DOI: 10.1530/EJE-12-0903
  97. 97. Lai X, Xia Y, Zhang B, Li J, Jiang Y. A meta-analysis of Hashimoto’s thyroiditis and papillary thyroid carcinoma risk. Oncotarget. 2017;8:62414-62424. DOI: 10.18632/oncotarget.18620
  98. 98. Resende de Paiva C, Grønhøj C, Feldt-Rasmussen U, von Buchwald C. Association between Hashimoto’s thyroiditis and thyroid cancer in 64,628 patients. Frontiers in Oncology. 2017;7:53. DOI: 10.3389/fonc.2017.00053
  99. 99. McLeod DSA, Cooper DS. The incidence and prevalence of thyroid autoimmunity. Endocrine Journal. 2012;42:252-265
  100. 100. Fiore E, Rago T, Scutari M, Ugolini C, Proietti A, Di Coscio G, et al. Papillary thyroid cancer, although strongly associated with lymphocytic infiltration on histology, is only weakly predicted by serum thyroid auto-antibodies in patients with nodular thyroid diseases. Journal of Endocrinological Investigation. 2009;32:344-335. DOI: 10.1007/BF03345725
  101. 101. Fiore E, Latrofa F, Vitti P. Iodine, thyroid autoimmunity and cancer. European Thyroid Journal. 2015;4:26-35. DOI: 10.1159/000371741
  102. 102. Kohno Y, Yamaguchi F, Saito K, Niimi H, Nishikawa T, Hosoya T. Anti-thyroid peroxidase antibodies in sera from healthy subjects and from patients with chronic thyroiditis: Differences in the ability to inhibit thyroid peroxidase activities. Clinical and Experimental Immunology. 1991;85:459-463
  103. 103. Latrofa F, Ricci D, Montanelli L, Rocchi R, Piaggi P, Sisti E, et al. Thyroglobulin autoantibodies in patients with papillary thyroid carcinoma: Comparison of different assays and evaluation of causes of discrepancies. The Journal of Clinical Endocrinology and Metabolism. 2012;97:3974-3982. DOI: 10.1210/jc.2012-2406
  104. 104. Matesa-Anic D, Matesa N, Dabelic N, Kusic Z. Coexistence of papillary carcinoma and Hashimoto’s thyroiditis. Acta Clinica Croatica. 2009;48:9-12
  105. 105. Marotta V, Sciammarella C, Chiofalo MG, Gambardella C, Bellevicine C, Grasso M, et al. Hashimoto’s thyroiditis predicts outcome in intrathyroidal papillary thyroid cancer. Endocrine-Related Cancer. 2017;24:485-493. DOI: 10.1530/ERC-17-0085
  106. 106. Cunha LL, Morari EC, Guihen AC, Razolli D, Gerhard R, Nonogaki S, et al. Infiltration of a mixture of immune cells may be related to good prognosis in patients with differentiated thyroid carcinoma. Clinical Endocrinology. 2012;77:918-925. DOI: 10.1111/j.1365-2265.2012.04482.x
  107. 107. Zeng R, Lyu Y, Zhang G, et al. Positive effect of RORγt on the prognosis of thyroid papillary carcinoma patients combined with Hashimoto’s thyroiditis. American Journal of Translational Research. 2018;10:3011-3024
  108. 108. Jeong JS, Kim HK, Lee CR, Park S, Park JH, Kang SW, et al. Coexistence of chronic lymphocytic thyroiditis with papillary thyroid carcinoma: Clinical manifestation and prognostic outcome. Journal of Korean Medical Science. 2012;27:883-889. DOI: 10.3346/jkms.2012.27.8.883
  109. 109. Talmadge JE, Donkor M, Scholar E. Inflammatory cell infiltration of tumors: Jekyll or Hyde. Cancer Metastasis Reviews. 2007;26:373-400. DOI: 10.1007/s10555-007-9072-0
  110. 110. Azizi G, Keller JM, Lewis M, et al. Association of Hashimoto’s thyroiditis with thyroid cancer. Endocrine-Related Cancer. 2014;21:845-852. DOI: 10.1530/ERC-14-0258
  111. 111. Kim HJ, Park HK, Byun DW, Suh K, Yoo MH, Min YK, et al. Iodine intake as a risk factor for BRAF mutations in papillary thyroid cancer patients from an iodine-replete area. European Journal of Nutrition. 2018;57:809-815. DOI: 10.1007/s00394-016-1370-2
  112. 112. Russo D, Arturi F, Chiefari E, Filetti S. Thyrotropin receptor: A role for thyroid tumourigenesis? Forum (Genoa, Italy). 1999;9(2):166-175
  113. 113. García-Jiménez C, Santisteban P. TSH signalling and cancer. Arquivos Brasileiros de Endocrinologia e Metabologia. 2007;51(5):654-671
  114. 114. Matsuo K, Friedman E, Gejman PV, Fagin JA. The thyrotropin receptor (TSH-R) is not an oncogene for thyroid tumors: Structural studies of the TSH-R and the α-subunit of Gs in human thyroid neoplasms. The Journal of Clinical Endocrinology and Metabolism. 1993;76:1446-1451. DOI: 10.1210/jcem.76.6.8501149
  115. 115. Derwahl M, Broecker M, Kraiem Z. Thyrotropin may not be the dominant growth factor in benign and malignant thyroid tumors. The Journal of Clinical Endocrinology and Metabolism. 1998;84:829-834. DOI: 10.1210/jcem.84.3.5519
  116. 116. Shi Y, Zou M, Farid NR. Expression of thyrotropin receptor gene in thyroid carcinoma is associated with good prognosis. Clinical Endocrinology. 1993;39:269-274
  117. 117. Satta MA, DeRosa G, Testa A, Maussier ML, Valenza V, Rabitti C, et al. Thyroid cancer in suppressed contralateral lobe of patients with hot thyroid nodule. European Journal of Cancer. 1993;29A:1190-1192
  118. 118. Hoelting T, Tezelman S, Siperstein AE, Duh QY, Clark OH. Thyrotropin stimulates invasion and growth of follicular thyroid cancer cells via PKC- rather than PKA-activation. Biochemical and Biophysical Research Communications. 1993;195:1230-1236. DOI: 10.1006/bbrc.1993.2176
  119. 119. Ahn D, Sohn JH, Kim JH, Shin CM, Jeon JH, Park JY. Preoperative subclinical hypothyroidism in patients with papillary thyroid carcinoma. American Journal of Otolaryngology. 2013;34:312-319. DOI: 10.1016/j.amjoto.2012.12.013. Epub 2013 Jan 26
  120. 120. Gudmundsson J, Sulem P, Gudbjartsson DF, Jonasoon JG, Sigurdsson A, Bergthorsson JT, et al. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nature Genetics. 2009;41:460-464. DOI: 10.1038/ng.339
  121. 121. Mincer DL, Jialal I. Hashimoto thyroiditis. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2018
  122. 122. Pearce SH, Brabant G, Duntas LH, Monzani F, Peeters RP, Razvi S, et al. 2013 ETA guideline: Management of subclinical hypothyroidism. European Thyroid Journal. 2013;2:215-228

Written By

Maria V. Deligiorgi and Dimitrios T. Trafalis

Submitted: 02 December 2018 Reviewed: 12 February 2019 Published: 10 April 2019