Next Article in Journal
Ferric-Chelate Reductase FRO3 Is Involved in Iron Homeostasis in Table Grape and Enhanced Plant Tolerance to Iron-Deficient Conditions
Previous Article in Journal
Eyes Are the Windows to the Soul: Reviewing the Possible Use of the Retina to Indicate Traumatic Brain Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Thyroid Cancer: Epidemiology, Classification, Risk Factors, Diagnostic and Prognostic Markers, and Current Treatment Strategies

1
Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
2
Department of Correct, Clinical, and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
3
Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(11), 5173; https://doi.org/10.3390/ijms26115173
Submission received: 16 April 2025 / Revised: 18 May 2025 / Accepted: 23 May 2025 / Published: 28 May 2025

Abstract

Thyroid cancer (TC) invariably remains the most prevalent endocrine cancer in the world. Major histological forms of TC include papillary (PTC), follicular (FTC), medullary (MTC), and anaplastic thyroid carcinoma (ATC), each of which has a unique clinical and molecular profile. The incidence rate of TC is higher in females, and unfortunately, it has tended to increase over the last several years. Yet the treatment of advanced or aggressive TC forms has improved recently because of developments in immunotherapy and targeted medicines, including PD-1 inhibitors and tyrosine kinase inhibitors (e.g., lenvatinib, sorafenib). Imaging, fine-needle aspiration biopsies, and molecular testing are implemented in the diagnostic process, e.g., in search of mutations that might affect prognosis and provide the most successful treatment option. Chemotherapy, immunotherapy, radioactive iodine therapy (RAI), surgery (such as a total thyroidectomy), and molecularly targeted therapies are currently standard treatment modalities in TC. Optimizing patient outcomes requires better diagnostic precision and individualized treatment regimens based on the genetic profile and tumor subtype. To improve survival and quality of life, it is critical to comprehend the complex etiology of TC and the changing therapeutic landscape.

1. Introduction

Thyroid cancer (TC) remains the most common endocrinological cancer worldwide, with a disturbingly increasing incidence rate over recent years [1,2]. The reason for this is not only a greater awareness of this malignancy and in-depth diagnosis leading to the early detection of less typical clinical symptoms but also its increasing occurrence due to a continually changing world and environmental threats associated with this. Environmental factors that are currently distinguished as potential risk factors for TC include the increasing usage of environmental chemicals as well as exposure to heavy metals, radiation, and air pollution [3,4,5,6,7]. Being exposed to the abovementioned factors constitutes an increasing global problem that might result in an increasing incidence of various endocrinological diseases and malignancies, including TC. An increase in the TC occurrence rate is associated with countries with a high or very high Human Development Index (HDI), where TC accounts for 91% of new cases [8]. In the following narrative review, we aimed to summarize the most recent knowledge regarding TC, including information about its epidemiology and changes in its trends, causes, prognostic and diagnostic factors, classification, and—most importantly—the current updates on TC treatment strategies and novel therapeutic approaches.

2. Epidemiology of Thyroid Cancer

Currently, TC is ranked as the 7th most prevalent cancer worldwide, with a mortality rate ranked as 24th highest among all cancers [9]. TC is three times more common in females than in males; the less aggressive histologic subtypes are more prevalent in females, while the more aggressive ones are equally frequent in both genders [10]. TC is often diagnosed at a younger age compared to any other adult cancer; the mean age at which TC is diagnosed is 51 [11]. Clinicopathological manifestations of TC differ depending on the age of the patient; e.g., younger patients more commonly present with lymph node metastasis, neurovascular, or capsular invasion, indicating that younger individuals usually present a more aggressive course of this malignancy [12]. Further, TC is half as common in black than in white individuals; the ranking of the incidence rate of TC nowadays is as follows, starting with the highest prevalence in white populations, followed by Asians/Pacific Islanders, American Indian/Alaskan natives, and black individuals [13]. Generally, TC occurrence is higher in non-Hispanic males and females than in Hispanic males and females [13]. TC incidence increased by over 200% from 1992 to 2018, while the mortality rate continually remained unchanged [14]. The incidence rate of TC increased in the majority of countries; this increase was the most significant in younger individuals, but generally, rates increased in all individual groups, irrespective of age [15]. The epidemiological landscapes based on the GLOBOCAN database indicate that the mortality rate of TC is less than 1 per 100,000 in most of the investigated countries for both genders; South Korea was indicated to have the highest incidence-to-mortality-rate ratio in both sexes [16]. It was also noted that there are no significant differences between the mortality rates in high-HDI countries and low- and medium-HDI countries [16].

3. Classification of Thyroid Cancer

3.1. Papillary Thyroid Cancer

Papillary thyroid cancer (papillary thyroid carcinoma, PTC) is the most common type of TC, accounting for nearly 90% of all TC cases [17,18]. Even though PTC is rare in children, it remains the most common pediatric thyroid malignancy, accounting for about 90% of TC in children [19]. Although it is the most prevalent type of TC and might occur in any age group, the peak of its most probable incidence is observed to be between 30 and 50 years of age [20]. The prognosis and the clinical outcome of PTC correlate with tumor size; it was observed that carcinomas that are less than 1.5 cm have a much better prognosis. PTC is observed to be a relatively mild cancer that can be easily operated on; however, in some cases, this type of carcinoma might present significant aggressive behavior associated with such factors as the induction of epithelial–mesenchymal transition, epigenetic modifications, or tumor cell metabolic programming leading to a higher probability of distant metastases and postoperative recurrence [17,21]. There are several aggressive variants of PTC, including the tall cell variant (TCV), diffuse sclerosing variant (DSV), columnar cell variant (CCV), hobnail variant (HV), and solid variant (SV) [22]. PTC usually spreads to the lymph nodes of the neck (primarily to the pretracheal and paratracheal lymph nodes), and the frequency of this type of metastasis is observed in approximately 70% of patients [23]. However, it must be considered that even though cancerous spread via lymph nodes might be associated with a worse recovery and higher risk of recurrence, in the case of PTC, the presence of lymph node metastasis is not associated with a higher mortality rate [24,25]. Only 10% of patients with PTC are estimated to present metastasis at the initial presentation of this malignancy [26]. Distant metastases are not common in PTC, but when they occur, the most common sites include the lungs, bones, liver, and brain [27]. There are several factors associated with a poor prognosis of PTC, but the most prevalent ones include older age at diagnosis (patients > 55 years old), distant metastases and extrathyroidal growth, large tumor size, male sex, aggressive subtypes of PTC, presence of an aneuploid cell population, vascular invasion, and solid and less differentiated areas [28,29]. The overall survival rate of PTC is very high irrespective of the age group; it was estimated that the cure rate might even reach 100% [30]. Similarly, in the case of pediatric patients, the mortality rate associated with PTC is estimated as low to even absent [19].

3.2. Follicular Thyroid Cancer

Follicular thyroid cancer (FTC) is the second most common malignancy of the thyroid gland, accounting for about 20% of all cases of TC [31]. It is estimated that FTC accounts for approximately 10% of thyroid malignancies in iodine-sufficient areas while accounting for about 25–40% in areas characterized by iodine deficiency [32,33]. This type of cancer, similar to PTC, more often occurs in females, with a female-to-male ratio equal to 3:1; the mean age at diagnosis is estimated to be 60 years old, while the peak onset age ranges from 40 to 60 years old [34,35,36]. It is estimated that about 80% of FTC cases present mild symptoms with a good prognosis, while the remaining 20% are characterized by aggressive behavior [31]. It was observed that the prognosis of FTC is associated with the size of the tumor, with carcinomas less than 1.5 cm indicating a good prognosis [37]. FTC can be divided into three subtypes, including minimally invasive, encapsulated angioinvasive, and widely invasive variants [38]. Previously, there was another subtype of FTC distinguished, namely Hurthle cell carcinoma; however, the World Health Organization has classified it as a different type of thyroid malignancy [38]. Cases of minimally invasive FTC are more often diagnosed in younger patients; however, it must be taken into consideration that this variant has also been observed to be a probable precursor of more aggressive variants [39]. Invasive follicular carcinoma is primarily characterized by vascular invasion and extension beyond the thyroid gland [40,41]. FTC is characterized by slower progression compared to other thyroid malignancies [42,43,44,45]. It was estimated that about 20% of nonfunctioning follicular adenomas might possess oncogenic mutations that can ultimately lead to the onset of FTC [46]. However, it must be taken into consideration that FTC cannot be distinguished from follicular thyroid adenoma based only on cytologic features [47]. FTC is more aggressive than PTC and tends to metastasize significantly more often; even though distant metastases are not that common for FTC, they are more often observed in this type of carcinoma than in PTC [48,49,50]. Approximately 6–20% of patients with FTC may present distant metastases, which are most often located in the lungs, followed by the bones, liver, brain, and skin [51,52]. The probability of metastases in the case of FTC is greater for older patients [49]. While metastasis to the lymph nodes is not common, vascular invasion is characteristic of FTC [53]. It was estimated that fewer than 10% of patients present metastases into the lymph nodes [54,55].

3.3. Medullary Thyroid Cancer

Medullary thyroid cancer (MTC) is a relatively rare neuroendocrine cancer originating from the thyroid parafollicular C cells responsible for calcitonin production [56]. This malignancy accounts for about 1–5% of all TC cases and may appear spontaneously or as a part of several hereditary syndromes, including multiple endocrine neoplasia (MEN) types 2A and 2B and familial medullary thyroid cancer (FMTC) [57]. Among all MTCs, the majority are sporadic, while about 25% account for those associated with MEN2 syndrome [58]. Sporadic MTC has been observed to be more prevalent in females, while the hereditary type of MTC is equally prevalent independently of gender [59]. It is estimated that MTC is the least common among all TCs, accounting for <5% of all thyroid malignancies [60]. The age peak for spontaneous MTC is between 40 and 60 years, with a mean age of 50 years; other trends are observed in cases of MTC associated with genetic diseases [61]. MTC associated with either MEN2A or MEN2B may appear in early childhood, while this malignancy usually occurs in the second to fourth decade when it is a component of FMTC [62,63,64,65,66,67]. In cases when a patient has a family history of MTC and/or MEN2 and presents positive for the RET gene mutation, prophylactic surgery may be performed to prevent the onset of MTC [68]. The prognosis of MTC depends on several factors, including the patient’s age, surgical resection status, and the histologic grade of the carcinoma. Generally, older patients presenting high-grade lesions with incomplete surgical treatment present significantly worse prognoses [69]. MTC is characterized by a rather poor prognosis primarily because of its high probability of metastases compared to other thyroid malignancies, as well as delayed diagnosis [57]. Further, high-grade tumors are characterized by poorer overall survival rates compared to low-grade carcinomas because of a higher incidence rate of distant metastases and a higher risk of local recurrence. In addition to metastases in the lymph nodes, distant metastases of medullary thyroid carcinoma can occur in the lungs, liver, or bones, as well as the brain [70].

3.4. Anaplastic Thyroid Cancer

Anaplastic thyroid carcinoma (ATC) is a very rare but also very aggressive form of undifferentiated thyroid cancer. This thyroid malignancy accounts for approximately 2% of all TCs [71]. Even though it is very infrequent, ATC accounts for approximately 50% of deaths associated with thyroid malignancies [72]. ATC is more often diagnosed in females; this type of cancer also usually occurs among people of the age over 65 years [73,74]. The etiopathology of ATC is still not fully explained. It is hypothesized that since most of these cancers occur in the setting of a long-standing goiter, its onset might be associated with an undiagnosed differentiated TC [75,76]. It is estimated that about 20–50% of patients with ATC present metastases to distant parts of the body, and the most common sites include the lungs, bones, and brain [77,78,79]. The average survival rate of patients diagnosed with ATC is about five to six months after diagnosis; fewer than 20% of patients are alive one year after diagnosis [80,81]. Factors that might facilitate a better prognosis include a younger age of the patient, tumor size less than 6 cm, unilateral tumor, and lack of lymph node and distant metastases [82].

4. Risk Factors and Causes of Thyroid Cancer

TC, being one of the most common malignancies, can be caused by multiple factors. These can be divided into modifiable and unmodifiable [83]. Among modifiable factors, we can distinguish obesity, smoking and secondhand smoking (SHS), heavy alcohol consumption, lack of exercise, and exposure to high levels of radiation. Within the unmodifiable factors, we can include sex, genetic factors such as gene mutations or inherited genetic syndromes, and preexisting benign thyroid disease (Figure 1) [84].

4.1. Modifiable Factors

4.1.1. Obesity

Obesity is an unquestionable risk factor for many diseases, including cancers. It can be highlighted as one of the primary TC causes [85,86]. Regardless of sex, a body mass index (BMI) of 25.0–29.9 is associated with an increased risk of thyroid cancer compared to that for a lower BMI. Obesity may contribute to thyroid cancer through multiple mechanisms, including chronic inflammation, oxidative stress, immune dysfunction, elevated thyroid-stimulating hormone (TSH), insulin resistance, adipokines, and increased aromatase activity. Persistent low-grade inflammation can generate reactive oxygen species, accelerate cell division, and impair tumor suppression. Higher TSH levels may encourage thyroid cell proliferation, genetic mutations, and cancer formation. Insulin resistance leads to increased insulin-like growth factor 1 (IGF-1), which activates cancer-promoting pathways like AKT/mTOR/PI3K and ERK/RAS/MAPK, supporting tumor growth and survival. Additionally, adipokines such as adiponectin, leptin, and resistin may play a role in thyroid carcinogenesis [87]. A 2018 study on obese mouse models showed a link between the overactivation of the JAK/STAT3 signaling pathway and lymphatic metastasis of TC. Obesity affects adipokine levels by increasing leptin and reducing adiponectin. Leptin stimulates cancer cell growth through JAK2/STAT3 and MAPK signaling, whereas adiponectin counteracts tumor progression by blocking the PI3K/AKT/mTOR pathway via AMPK activation. Lower adiponectin levels in obesity may play a key role in TC development and progression [88]. Chronic inflammation activates the transcription of nuclear factor kappa-light-chain-enhancer of activated B (NF-kB), STAT3, and activator protein 1 (AP1), which, cooperatively with hypoxia, promote cancer cell growth and angiogenesis. Tumors contain many immune cells such as tumor-associated lymphocytes, tumor-associated macrophages (TAMs), immature dendritic cells, mast cells, and myeloid-derived suppressor cells. The presence of mast cells and macrophages in tumors is linked to poor TC prognosis. Proinflammatory cytokines produced by NF-kB transcription play a crucial role in cancer progression. The BRAF V600E mutation and RET/PTC gene rearrangement enhance NF-kB activity, increase inflammatory mediator expression, and contribute to lymph node metastases in PTC (Figure 2) [89].

4.1.2. Smoking and Secondhand Smoking

Cigarette smoking and SHS are established risk factors for many diseases and have been implicated in TC. Even minimal SHS exposure induces immediate inflammatory responses, lasting for hours [90]. SHS contains toxic compounds such as thiocyanate, which disrupts thyroid hormone synthesis, and proinflammatory cytokines, for instance IL-1β and IL-6, which may exacerbate thyroid autoimmunity. Additionally, cigarette smoke delivers carcinogens like PAHs and nitrosamines into the bloodstream, leading to chronic inflammation, DNA damage, and potential malignant transformation in thyroid cells [83]. However, emerging evidence challenges the traditional view, suggesting that smoking may exert a protective effect on TC [91]. This protective effect is partly explained by the smoking-induced suppression of TSH, which normally promotes thyroid cell proliferation. Studies indicate that smoking lowers TSH levels while increasing free triiodothyronine (FT3) and free thyroxine (FT4). Conversely, TSH levels tend to rise after smoking cessation Additionally, smoking influences estrogen metabolism and activates the aryl hydrocarbon receptor (AHR) pathway, potentially reducing estrogen’s stimulatory effects on the thyroid. Another proposed mechanism includes increased sympathetic nervous system activity, leading to higher TSH levels [92,93,94,95]. What is more, heavy smokers who quit have a higher likelihood of developing TC than consistent smokers [96]. These conflicting findings highlight the complexity of the smoking–TC relationship and indicate the need for further research.

4.1.3. Alcohol Consumption

Despite popular beliefs, recent studies show that alcohol consumption is negatively correlated with the occurrence of TC [97]. Light to heavy alcohol consumption, when combined with smoking and SHS, is noted to have a submultiplicative effect on TC [95]. Potential mechanisms underlying alcohol’s protective role include alterations in thyroid hormone metabolism, disruptions in thyroid gland function, and impairment of the regulation of the hypothalamic–pituitary–thyroid axis [98]. Regular alcohol intake has been linked to lower TSH levels. Surprisingly, abstinence may increase the probability of developing TC [99]. Meta-analyses estimate that regular alcohol consumption may be associated with up to a 10% lower risk of TC [92]. Nonetheless, the toxic effects of alcohol on the thyroid and their broader systematic health risks must be taken into cautious consideration when interpreting these findings.

4.1.4. Lack of Exercise

There is strong scientific evidence that both physical inactivity and obesity independently elevate the risk of various cancers, whereas regular activity successfully reduces the risk of developing cancer [96]. Recent studies have highlighted the potential role of long non-coding RNAs (lncRNAs) as crucial molecular mediators linking physical activity to cancer prevention. Regular exercise has been shown to modulate the expression of multiple lncRNAs, which are typically upregulated in cancer. These lncRNAs participate in key cellular functions, including epigenetic modifications, cell proliferation, and apoptosis regulation, forming a complex network that may contribute to cancer protection. Abnormal expression of lncRNAs has been strongly associated with various diseases, including cancer, where they contribute to tumor progression, metastasis, and changes in the tumor microenvironment (TME). Emerging research suggests that both exercise and cancer can influence lncRNA levels, highlighting their potential role in disease regulation [100]. Regular exercise regulates the inflammatory response, lowering inflammatory markers like C-reactive protein (CRP), tumor necrosis factor-alpha, and interleukins such as IL-6. Additionally, exercise promotes an anti-inflammatory state by boosting the production of cytokines like IL-1ra and IL-10, which contribute to overall immune balance and disease prevention [101]. Another mechanism highlighting the importance of regular physical activity is the lowering of IGF1 levels, which helps regulate cell growth and prevents cancer progression. Exercise also boosts antioxidant enzyme production, strengthening the body’s defense against oxidative damage with adequate nutrient intake [91]. Furthermore, regular physical activity modulates key processes involved in tumorigenesis, such as proliferation, angiogenesis, and metastasis [96,102]. In TC patients, postoperative exercise significantly reduces the risk of bone fractures, a common complication following thyroidectomy [103]. Overall, physical activity offers many benefits for health and enhances the quality of life in patients being treated for TC [104].

4.1.5. Exposure to High Levels of Radiation

Historically, exposure to high levels of radiation was limited to specific populations. Today, the widespread use of mobile phones has introduced a new, persistent source of radiofrequency (RF) radiation. Emerging studies associate RF radiation from cell phones with an increased risk of TC [105,106]. RF exposure can alter gene-encoded proteins such as PAK6, MDM2, HDAC4, and DACT2, which are involved in tumor growth, progression, suppression, transcription, and apoptosis [105]. However, despite a better understanding of the process behind radiation-induced TC development, there are still no specific markers recognized. Similarities in histological types, oncogenic drivers, and gene expression profiles between radiation-induced and sporadic thyroid tumors suggest that they do not differ from typical markers. With the thyroid gland being highly sensitive to radiation, research has shown that radiation exposure in the head, neck, and chest increases the lifelong risk of developing thyroid malignancies. Radiation therapy in these areas can disrupt thyroid function, often leading to hypothyroidism, which in turn may contribute to cancer development [107]. With the rise in radiological procedures, particularly among pediatric patients, current research is shifting toward assessing risks associated with low-dose radiation exposure [108]. Recent genomic studies on Chernobyl-related TCs have revealed a dose-dependent rise in small deletions and structural variants, likely resulting from DNA double-strand breaks [108,109]. These enable PTC development after radiation exposure [109].

4.2. Unmodifiable Factors

4.2.1. Sex

Thyroid cancer incidence is significantly higher in women than in men, a difference thought to be related to reproductive activity and estrogen exposure. Throughout the menstrual cycle, fluctuations in estrogen levels affect circulating TSH concentrations by modulating thyroxine-binding globulin (TBG) levels, altering free thyroxine availability, and stimulating TSH secretion. Estrogens influence TC pathogenesis by promoting cell proliferation, inducing DNA damage, and modulating stress responses. The presence of estrogen receptors (ERα and ERβ) in thyroid tissue, along with the local conversion of androgens into estrogens, suggests direct hormonal involvement in tumor biology. ERα tends to promote thyroid tumor growth, while ERβ inhibits it. Additionally, estrogen-induced DNA adducts and changes in autophagy regulation contribute to tumor progression. Men with TC tend to have worse outcomes than women. This is partly due to later diagnosis, older age at presentation, and more advanced disease stages. However, male sex remains an independent risk factor, especially in patients with BRAF mutations [110]. A 2020 study reported that men with differentiated thyroid cancer (DTC) have a higher risk of recurrence than women. The risk was over three times higher in men with early stage disease but showed no significant difference in advanced stages. Despite receiving more aggressive treatment, men still had higher recurrence rates [111].

4.2.2. Genetic Factors

A 2021 population-based study shows global trends in TC incidence. It reveals various patterns among different subtypes. PTC has seen the most significant rise, especially in high-income and transitioning countries, mainly due to increased detection through advanced imaging. Other subtypes, including FTCs and MTCs, have remained relatively stable without clear temporal trends. The rarest and most aggressive form, ATC, has fortunately shown declining incidence rates in most regions. Overdiagnosis, particularly of PTC, has been a major factor in these trends, leading to calls for more cautious screening and diagnosis approaches [112].

4.3. Genes

4.3.1. Papillary Thyroid Cancer

PTC is the most common thyroid malignancy worldwide, regardless of age. Its diagnostic criteria have evolved over the years. Radiation exposure is a well-established risk factor, alongside environmental factors like obesity [113]. PTC is primarily driven by mutations that lead to the constitutive activation of the MAP kinase (MAPK) signaling pathway. These mutations occur in key oncogenes such as BRAF, RAS, RET, and NTRK, which function as upstream regulators of the pathway. The activation of MAPK effectors plays a critical role in tumor initiation, progression, and maintenance [114]. Although PTC generally has a favorable prognosis with a >90% 10-year survival rate, recurrence (especially in neck lymph nodes) occurs in 20–30% of cases. Early diagnosis is crucial, as traditional FNA cytology has limitations. A valuable biomarker, BRAF V600E mutation, may be crucial for PTC identification [115]. PTC can be hereditary, and some of these cases are linked to known syndromes such as Cowden syndrome, Gardner syndrome, and the Carney complex, where specific genetic mutations have been identified [116]. BRAF V600E is the most frequent mutation, resulting from a T-to-A transversion at nucleotide 1799. This mutation leads to the continuous activation of BRAF kinase, which subsequently phosphorylates MEK and ERK, driving uncontrolled cell proliferation. It is associated with aggressive tumor features, extrathyroidal invasion, and poorer prognosis. Additionally, AKAP9–BRAF fusion, caused by chromosomal inversion, has been identified in radiation-induced PTC [114]. RAS mutations (in NRAS, HRAS, and KRAS) occur in about 10–20% of cases and are more frequently associated with the follicular variant of PTC [114,117]. Additionally, RET/PTC rearrangements, resulting from chromosomal translocations, are present in 10–20% of cases. RET/PTC1 and RET/PTC3 mutations are the most common variants, particularly in radiation-induced pediatric PTC cases [114,117,118]. These mutations play a crucial role in tumor initiation and progression, influencing prognosis and potential therapeutic targets (Table 1) [117].

4.3.2. Medullary Thyroid Cancer

MTC is a rare neuroendocrine tumor, accounting for 3–5% of thyroid malignancies. Despite its low incidence, MTC is responsible for a significant proportion of TC-related deaths. It can occur sporadically or as part of hereditary syndromes like multiple endocrine neoplasia type 2 (MEN2) and FMTC. Calcitonin and carcinoembryonic antigen (CEA) serve as crucial diagnostic and prognostic markers for MTC [119,120]. Mutations in the RET proto-oncogene play a crucial role in the development of MTC and are classified into germline and somatic mutations. The RET gene encodes a receptor tyrosine kinase involved in cell proliferation, differentiation, and survival. Mutations can affect different RET domains, leading to distinct clinical outcomes. Extracellular RET mutations, especially in codon 634, are commonly linked to MEN2A syndrome, often resulting in early onset MTC. Codon 918 mutations are strongly associated with MEN2B syndrome, which is characterized by early and aggressive MTC, often with metastases in infancy. Intracellular mutations are usually seen in FMTC and result in milder disease progression. Genetic testing for RET mutations is recommended for all MTC patients to guide risk assessment, early diagnosis, and prophylactic thyroidectomy in at-risk individuals [119]. About 70% of RET wild-type sporadic MTCs harbor RAS mutations, with HRAS being the most common, followed by KRAS and rare NRAS mutations. RET and RAS mutations are usually mutually exclusive, though rare cases with both mutations exist. Within the other molecular alterations, we can distinguish CDKN2C loss, found in 20% of tumors and associated with distant metastases and reduced survival, and mTOR pathway activation, linked to lymph node metastases and more prevalent in RAS-mutant MTC. Increased EZH2 and SMYD3 expression are observed in aggressive tumors. MiRNA overexpression is linked to metastasis and worse outcomes [121]. A 2025 case report shows a possibility of a connection between a germline pathogenic variant in the CDKN1B gene [multiple endocrine neoplasia type 4 (MEN4)] and MTC. This case highlights the coexistence of MTC and MEN4 linked to a novel CDKN1B germline frameshift mutation, expanding the understanding of MEN4-associated endocrine tumors [122].

4.3.3. Follicular Thyroid Cancer

FTC is the second most prevalent type of differentiated TC. It originates from follicular cells, which are responsible for producing and secreting thyroid hormones. FTC is more common in women and typically presents during the fifth and sixth decades of life. Risk factors for FTC include iodine deficiency, age over 50 years, and female sex [34]. FTC is characterized by several genetic mutations that play pivotal roles in its pathogenesis. Mutations in the RAS family of oncogenes—specifically NRAS, HRAS, and KRAS—are among the most prevalent, detected in approximately half of FTC cases. They have been associated with poor prognosis and distant metastases. These mutations often occur at codon 61 and lead to the constitutive activation of signaling pathways that drive tumor development. Additionally, TERT promoter mutations are found in about 25% of FTCs and are linked to aggressive clinical outcomes, including advanced TNM stage, recurrence, and higher mortality rates. The PAX8-PPARγ fusion gene is present in approximately one-third of FTC cases and contributes to tumorigenesis through altered transcriptional regulation [123,124]. Furthermore, mutations in the EIF1AX gene have been identified in a subset of FTCs, often co-occurring with RAS mutations, suggesting a collaborative role in cancer progression [125]. Other mutations, such as EZH1, have been identified in follicular-patterned lesions, typically in lower-grade tumors [124].

4.3.4. Anaplastic Thyroid Cancer

ATC, representing 5–10% of TCs, is an extremely aggressive tumor with low survival rates. Distant metastases are present at the time of diagnosis in 30–40% of cases. Under microscopic examination, it exhibits a diverse cellular structure and a complete loss of follicular differentiation. ATC is frequently linked to coexisting DTC or a prior history of DTC, which suggests that certain DTC clones may undergo morphological changes leading to dedifferentiation. ATCs exhibit significantly higher frequencies of TP53, TERT promoter, PIK3CA, and PTEN mutations. Additionally, ATCs carry mutations in ATM, NF1, NF2, CDKN2A, CDKN2B, and RB1 [123,126,127]. ATCs frequently harbor TP53 mutations, disrupting tumor suppression and enabling uncontrolled growth. TERT promoter mutations, often co-occurring with BRAF or RAS alterations, enhance telomerase activity for indefinite proliferation. Disruptions in cell cycle regulators such as CDKN2A, CDKN2B, and RB1 further drive unregulated division. The PI3K/AKT/mTOR pathway is commonly activated via PIK3CA and PTEN mutations, promoting survival and metabolic reprogramming. DNA repair deficiencies, including ATM and NF1/2 mutations, increase genomic instability. Epigenetic regulators like ARID1A, SMARCA4, and histone-modifying enzymes are frequently mutated, contributing to aggressive phenotypes. Structural alterations, such as CNVs, impact tumor suppression. EIF1AX mutations, often with RAS alterations, may dysregulate protein synthesis. TAMs create an immunosuppressive microenvironment, further supporting tumor progression [127].

4.3.5. Preexisting Benign Thyroid Disease

Individuals with a history of benign thyroid diseases, including hyperthyroidism and goiter, are at an increased risk of developing TC. This association is likely mediated by chronic alterations in TSH levels, which may promote abnormal thyroid cell proliferation Additionally, studies have shown that a family history of benign thyroid conditions further raises the likelihood of TC, indicating a potential genetic link [107]. Patients with preexisting thyroid-specific autoimmune diseases, such as Hashimoto thyroiditis (HT) and Graves’ disease, have a significantly increased risk of developing TC [128,129]. A 2021 study showed a connection between HT and an increased risk of developing thyroid malignancies, especially PTC and MTC. Several mechanisms have been proposed to explain this association. One mentions the inflammatory process in HT resulting in DNA damage and further mutations. Another highlights the possible thyroid tissue proliferation caused by the constant increase in TSH levels. However, specific mechanisms are still unknown [130]. Interestingly, thyroid cancers arising in patients with preexisting autoimmune thyroid disease tend to present as smaller tumors with lower rates of lymph node metastasis. This observation suggests that immune-mediated factors may not only increase cancer risk but also modulate tumor behavior. Chronic inflammation, oxidative stress, and dysregulated TSH signaling are regarded as key contributors to thyroid carcinogenesis in this context (Table 2) [129].

5. Symptoms and Diagnosis of Thyroid Cancer

Thyroid cancer (TC) can present a wide range of symptoms. Research on the distress, anxiety, and depression experienced by individuals with advanced TC shows that over half of the participants experience various symptoms, with sleep disturbances, fatigue, and emotional distress being the most prevalent and severe. Moreover, symptoms like hoarseness, numbness, and a noticeable lump in the thyroid region are often observed, typically due to tumor interaction with the recurrent laryngeal nerve and parathyroid gland. Over 30% of patients report symptoms of anxiety and depression [131]. However, in many cases, an enlarged thyroid, which may be structurally abnormal, is diagnosed incidentally—often following an X-ray of the chest or neck conducted for reasons unrelated to thyroid malignancies. Approximately 50% of thyroid cancer cases are discovered incidentally, particularly in asymptomatic patients. These often involve small papillary thyroid cancers (PTCs). Additionally, TC is frequently identified histologically when thyroid glands are removed for benign conditions, revealing small tumors incidentally [132]. The diagnosis of TC typically involves a combination of physical examination, imaging studies (e.g., ultrasound), and fine-needle aspiration biopsy (FNAB). Further tests, including molecular markers and radioactive iodine scans, are used to determine the cancer’s type and extent [133]. The abnormal and continuous growth of cancerous cells within the thyroid leads to gland enlargement, which can be detected during regular check-ups. The enlarged thyroid may be unrecognized by the patient if the growth is minor and asymptomatic, or it may be easily identified if it causes discomfort or difficulty in swallowing [134]. The increase in the number of malignant cells can manifest as an enlarged gland or the presence of detectable nodules, which can be felt during a physical examination. Alarm symptoms identified during palpation often lead to further diagnostic testing, including serum TSH levels and neck ultrasound. Alarming ultrasound features that may prompt a biopsy include microcalcifications, neovascularization, disorganized internal vascularity, irregular margins, and a taller-than-wide shape [135]. Thyroid nodules, though often causing concern, are malignant in only a small percentage of cases, especially in adults. Ultrasonography (US) and fine-needle aspiration cytology (FNAC) remain the primary methods for assessing malignancy risk. Advances in US technology, such as elastography, have improved diagnostic accuracy, particularly for indeterminate nodules [136]. More recently, the introduction of molecular testing for genetic mutations has significantly enhanced the diagnosis of various thyroid malignancies and facilitated targeted therapy [137]. Molecular tests provide additional diagnostic accuracy beyond cytology alone. The Bethesda System for Reporting Thyroid Cytopathology (TBSRTC) classifies thyroid nodules into six categories based on cytologic evaluation. Molecular testing has been especially useful for Bethesda III and IV nodules, where cytology’s indeterminate nature presents a challenge in clinical decision-making. These tests can identify gene mutations and alterations correlated with varying malignancy risks. Aggressive TC is associated with BRAF V600E, RET/PTC, and TERT promoter mutations, whereas NRAS, HRAS, KRAS, BRAF K601E, and PAX8/PPARG alterations are more commonly linked to benign nodules, non-invasive follicular thyroid neoplasms with papillary-like nuclear features (NIFTPs), or low-risk differentiated thyroid cancers (DTCs). Preoperative molecular testing can optimize surgical decisions, particularly for Bethesda V and VI nodules, when compared to cases without molecular testing [123,138,139,140]. Both RNA and DNA–RNA molecular tests can be used to diagnose malignancy in indeterminate thyroid nodules, with no significant difference between the two. Both demonstrate high sensitivity and reasonable specificity [141]. Ultrasonography (US) remains the primary method for evaluating thyroid lesions. If malignancy is suspected or the ultrasound results are inconclusive, single-photon emission computed tomography (SPECT) or ultrasound-guided fine-needle aspiration (FNA) may be used for follow-up. Computer tomography (CT) is typically employed to assess metastasis, while magnetic resonance imaging (MRI), with its excellent soft-tissue contrast and high resolution, is helpful in distinguishing between benign and malignant nodules. Radioiodine is the preferred diagnostic and therapeutic approach due to the preservation of sodium–iodide symporter (NIS) in well-differentiated thyroid tumors. For poorly differentiated or dedifferentiated TC, PET/CT and PET/MRI are particularly useful, with emerging evidence suggesting that PET/MRI may offer superior staging for TC, both at initial diagnosis and during follow-up evaluations [142,143]. Papillary thyroid cancer (PTC) often lacks clinical manifestations, with neither hyperthyroidism nor hypothyroidism typically present, as PTC rarely affects hormone production. The diagnosis is often made when an enlarged gland is detected, either by the patient or during a clinical examination, typically due to swallowing or breathing difficulties [26,144]. Extrathyroidal growth of PTC occurs in about 10% of patients [145], and distant metastases are an uncommon manifestation of the disease [146,147]. Follicular thyroid cancer (FTC) usually presents as a solitary thyroid nodule, often accompanied by thyroiditis or nodular hyperplasia. Patients may remain asymptomatic for a long time, with symptoms appearing only in cases of significant gland enlargement, potentially leading to dyspnea, tracheal compression, or dysphagia. Currently, it is not possible to differentiate follicular thyroid adenomas from carcinomas based solely on cytomorphologic criteria. However, the use of reverse-transcriptase polymerase chain reaction (PCR) for TSH receptor and thyroglobulin messenger RNA can significantly improve the differentiation between these malignancies. FTC is confirmed through pathological examination showing follicular cells that lack nuclear atypia, with diagnosis primarily based on evidence of capsular and vascular invasion [34]. Medullary thyroid cancer (MTC) manifests similarly to PTC or FTC, presenting as a thyroid nodule with comparable symptoms. However, MTC is characterized by the excessive secretion of calcitonin, which can lead to symptoms such as diarrhea and flushing. Other substances, such as prostaglandins, serotonin, and vasoactive intestinal peptide (VIP), may also contribute to diarrhea in MTC. Serum levels of carcinoembryonic antigen (CEA) should be measured as part of the diagnostic workup for MTC [57,148,149]. Anaplastic thyroid cancer (ATC) typically presents with a rapidly growing mass that compresses surrounding structures, leading to dysphagia and dyspnea. A firm, painful neck lump, often accompanied by swollen lymph nodes, is common in ATC [150]. About half of ATC cases present with metastases at diagnosis and are classified as stage IV tumors. Therefore, it is crucial to conduct ultrasound, CT, MRI, and FNAB to assess tumor extent, local invasion, and possible distant metastases (Table 3) [71]. In recent years, high-throughput sequencing technologies have made major contributions to the molecular characterization of thyroid cancer [151]. Next-generation sequencing (NGS), whole-exome sequencing (WES), and RNA sequencing (RNA-Seq) provide a complete detection of point mutations, gene fusions, and expression profiles across numerous thyroid cancer subtypes [152]. NGS panels that target common changes like BRAF V600E, RAS mutations, RET/PTC rearrangements, and PAX8-PPARy fusions can improve diagnosis accuracy and inform treatment recommendations for intermediate cytology cases (Bethesda III/IV) [153]. RNA-Seq also aids in the discovery of uncommon fusion transcripts and gene expression profiles [154]. These technologies are progressively being integrated into clinical processes, led by ATA and NCCN recommendations, especially when conventional procedures produce unclear results [155].

6. Diagnostic Markers of Thyroid Cancer

Diagnostic indicators are critical in the diagnosis of TC because they allow for early detection of the disease, which is necessary for effective treatment and a better prognosis. The identification of relevant indicators enables exact diagnosis, which influences treatment selection and progress tracking.

6.1. Thyroglobulin

More than 90% of DTCs are PTCs, and thyroglobulin (TG), a key substrate for thyroid hormone production, is an important tumor marker for DTC [157]. Normally, differentiated thyroid cells produce TG [158], which is a 330 kD, 2750-amino-acid protein produced by the follicular epithelial cells of the thyroid [159]. These cells are found primarily in thyroid follicular cells and follicular lumens [159]. Autoantibodies to the TSH receptor can elevate TG levels, which can result in higher circulating levels in thyroid cancers that begin in follicular epithelial cells [160]. TG emphasizes the need for close monitoring and is used to identify recurrent disease and track residual disease [158]. Preoperative serum TG levels have the potential to differentiate between benign and malignant thyroid nodules in patients with equivocal cytology [161]. Validated immunoassays calibrated against approved reference materials should be used to measure serum TG levels. Laboratories performing TG tests must adhere to recognized national or international quality assurance programs [162]. Tg secretion is known to occur in both benign and well-differentiated malignant thyroid tissue [163]. In fact, the presence of anti-TG antibodies (TgAb) may affect TG test results, and the presence of normal thyroid remnants reduces the utility of TG [164]. According to a study, there is a nonlinear correlation between preoperative TG levels above 1.39 ng/mL and a significant increase in the risk of distant metastases in PCT [165]. In patients with diabetes and TC, serum TG levels may be associated with HbA1c levels; this association is stronger in individuals with fluctuating TG levels, which requires further study [166]. This association highlights the need for close monitoring and elucidating the complex links between TC and other metabolic disorders.

6.2. Calcitonin

MTC is an aggressive neuroendocrine tumor that releases the neurotransmitter calcitonin gene-related peptide (CGRP) and develops from thyroid parafollicular cells, usually with a poor prognosis [167]. CGRP expression in MTC is linked to aberrant dendritic cell (DC) formation [167]. Calcitonin (Ctn) secretion is recognized as one of the primary features of MTC [57]. Slight elevations in Ctn may be observed in individuals with C-cell hyperplasia, autoimmune thyroiditis, chronic renal illness, hyperparathyroidism, and some lung and neuroendocrine tumors, potentially leading to false-positive or false-negative results [168]. Ctn and CEA have been proposed as biochemical indicators for MTC according to the National Comprehensive Cancer Network and American Thyroid Association (ATA) Guidelines for Management of MTC [169]. Human plasma includes one or more Ctn-degrading enzymes, and serum proteases digest the hormone more quickly in plasma [170]. A drop in Ctn levels of less than 50% 30 min following thyroidectomy and central neck lymph node dissection (LND) suggests that tumor tissue persists in MTC patients [171]. With a 10-year survival rate of 97.7%, long-term postoperative Ctn normalization as a biochemical cure is a favorable prognostic factor associated with a better outcome [172]. Serial Ctn tests may be more sensitive than radiological follow-up in advanced MTC [173]. Serum Ctn levels are a reliable and accurate biochemical predictor of tumor development for postoperative monitoring [174]. In addition, higher Ctn levels may play an important role in the early diagnosis of MTC, especially in patients with a family history of medullary thyroid cancer [175,176]. Therefore, Ctn serves as both a diagnostic and prognostic biomarker, improving the precision of treatment and follow-up strategies in patients with MTC.

6.3. Carcinoembryonic Antigen

CEA is a glycoprotein initially identified as being involved in intercellular adhesion. It is expressed by neuroendocrine tissues of the gastrointestinal tract during fetal development [177]. CEA is commonly recognized as a marker of various malignant diseases, but it does not show specificity for MTC when used alone. However, its combined use with Ctn significantly increases diagnostic sensitivity for MTC [178]. Of note, serum CEA levels are commonly used to monitor disease progression in patients with MTC [179]. Normal CEA levels in healthy individuals range from 2 to 4 ng/mL, with levels above 10 ng/mL usually associated with malignant conditions [180]. More than 50% of patients with MTC have benign increases in CEA [179,181]. Immunohistochemical studies show that patients with stronger CEA staining have a more aggressive, diffuse subtype of MTC [168]. Interestingly, CEA can be detected in C cells at all stages of MTC progression [181]. The rare nature of MTC should be taken into account when examining CEA elevations [182]. Disease progression can vary considerably over time but can be relatively well estimated by examining the doubling time of Ctn and CEA levels [181]. Furthermore, the risk of central lymph node metastasis is significantly increased when CEA levels exceed 30 ng/mL [183]. Despite its role in MTC, CEA is also involved in various endothelial cell functions, including cell adhesion, proliferation, and migration, both in vivo and in vitro [184]. It is mainly metabolized in the liver, and hepatic or biliary dysfunction can lead to elevated CEA levels, resulting in false-positive results [185]. CEA levels above 271 ng/mL are significant for advanced tumor size and stage, central compartment metastasis, and a decreased likelihood of biochemical cure, whereas levels above 500 ng/mL are associated with significantly higher patient mortality [186]. This highlights the importance of CEA levels as markers for assessing prognosis in MTC. Interestingly, a study found that patients with sporadic MTC had higher postoperative CEA levels and lower trough Ctn levels compared with patients with hereditary forms of MTC [187]. The revised ATA guidelines indicate that CEA should not be considered a specific biomarker for MTC, although it is still useful in disease management. Surgical treatment of patients with elevated CEA levels, particularly those above 30 µg/L, typically includes total thyroidectomy, central cervical lymph node dissection, and unilateral lateral cervical LND [188]. Furthermore, monitoring the doubling time of Ctn and CEA levels after surgery provides sensitive markers for assessing the progression and aggressiveness of metastatic MTC [189].

6.4. Procalcitonin

Procalcitonin (Pct) is a 116-amino-acid precursor produced by thyroid C cells and is considered a more reliable and stable biomarker than Ctn for the diagnosis and monitoring of MTC, which is released by parafollicular cells in the thyroid [190]. In addition to its role in MTC, PCT is widely used to monitor inflammatory activity and is a key marker in differential diagnosis, especially in the identification of bacterial infections [191]. It has been recognized as a valuable biomarker for the early detection of systemic bacterial infections, and treatment is usually initiated when Pct levels exceed a threshold of 2 ng/mL [192]. Pct levels do not increase during viral infections, making it a more specific marker for bacterial infections [192]. It is also the most reliable marker for sepsis [193]. In healthy individuals, Pct levels are very low because prohormones are not secreted into the circulation [194]. Elevated Pct levels can be observed in various settings, such as trauma, surgery, heat stroke, immunomodulatory therapy, and malignant diseases, particularly neuroendocrine tumors and metastatic tumors [195]. Pct production is associated with the presence of tumor necrosis factor (TNF) and several inflammatory cytokines, including interleukin-1, interleukin-2, and interleukin-6 [196]. In addition to thyroid C cells, Pct is produced by other tissues in response to severe systemic inflammation, local bacterial infections, autoimmune diseases, trauma, surgery, and fungal or parasitic infections [197]. A major advantage of procalcitonin over other biomarkers is its stability. Pct has a longer half-life, better thermal stability, and standardized cutoff points, making it a valuable tumor marker in the diagnosis and monitoring of MTC, and it also correlates with tumor size and progression in patients with MTC [198]. During the COVID-19 pandemic, persistently high Pct levels have helped uncover previously undiagnosed cases of MTC, highlighting its potential for early detection and treatment [199]. Furthermore, when Pct levels remain elevated after infection, especially in combination with significant CEA levels, it may aid in the early detection of MTC [190]. Pct also plays a key role in calcium homeostasis [200]. However, one limitation is that Pct production is not exclusive to C cells, as it is also produced by neuroendocrine cells in the lung and gut, as well as in other tissues in response to various inflammatory and infectious conditions [191,192,193,201].

6.5. Thyroid-Stimulating Hormone

TSH is mainly produced by basophils in the distal part of the adenohypophysis and plays a vital role in thyroid function. It acts on thyroid tissue by binding to TSH receptors (TSHRs), which stimulate the production and release of thyroid hormones, including thyroxine (T4) and triiodothyronine (T3) [202]. Through its receptor, TSH promotes the growth and activity of thyroid follicular cells, influencing the synthesis and secretion of these hormones [202]. TSH also stimulates growth in thyroid follicular cells, resulting in thyroid enlargement [203]. TSH receptors are found on the cell membrane of DTC cells, and their activation encourages cell growth by increasing the expression of thyroid-related proteins such as TG [204]. It is widely recognized that high TSH levels stimulate thyroid cell proliferation, which can lead to lymph node metastasis and invasion of nearby tissues [205]. This characteristic of TSH has led to the implementation of TSH suppression strategies in postoperative patients to reduce tumor growth and minimize recurrence risks [206]. However, excessive TSH suppression, particularly in postmenopausal women, has been linked to negative effects such as osteoporosis and a higher risk of fractures [207]. Research also shows that maintaining serum TSH levels below 2 mU/L does not significantly affect the odds ratio for recurrence [204]. However, when serum TSH reaches 2 mU/L or higher, adverse outcomes, including the recurrence of DTC and cancer-related death, have been observed [208]. A retrospective study from 1977 indicated that TSH suppression treatment could significantly lower recurrence rates [209]. Interestingly, some studies suggest that lower TSH levels might actually increase the risk of thyroid cancer and goiter, a surprising finding since TSH is usually associated with promoting the growth of thyroid cancers [210]. Factors such as gender, BMI, T stage, and preoperative FT4 levels have been identified as independent risk factors that affect the success of TSH suppression [211]. These findings emphasize the complex role of TSH in thyroid cancer biology and the need for careful management of TSH levels in patients.

6.6. microRNA

Exosomal microRNAs (miRNAs) have emerged as promising biomarkers, offering significant potential to enhance prognostic assessments in various cancers, including PTC [212]. MiRNAs are small, non-coding RNA molecules, typically ranging from 19 to 25 nucleotides, that function as negative regulators of gene expression by binding to the 3′ untranslated regions of target messenger RNAs (mRNAs) in the cytoplasm [213]. This interaction results in translation repression and mRNA degradation, which profoundly influence gene expression within cells [214]. MiRNAs are known for their stability, making them accessible from a wide range of biological samples, such as blood, tissue biopsies, and even formalin-fixed paraffin-embedded tissues [215]. Furthermore, circulating miRNAs—those released into the bloodstream—serve as reliable indicators of cellular changes, with their levels potentially reflecting clinical features, treatment responses, and patient outcomes [216]. In thyroid cancer, miRNAs have been implicated both as oncogenes and tumor suppressors [217]. Specific miRNAs, like miR-31, have shown conflicting evidence in their relationship with the BRAFV600E mutation in PTC [218]. Other miRNAs, such as miR-129-5p, have demonstrated the ability to suppress the migration, proliferation, and lymph node metastasis (LNM) of PTC cells, although further research is needed to better understand their roles [219]. Additionally, the expression levels of miRNAs such as miR-221, miR-222, and miR-146 have been shown to distinguish between malignant and benign thyroid nodules, marking them as potential diagnostic biomarkers for early cancer detection [220,221]. Moreover, miRNAs like miR-136 are abnormally expressed in metastatic tumors, playing a significant role in tumor development and progression [222,223]. The interplay between miRNAs and genetic mutations in thyroid cancer is also linked to tumor aggressiveness and poorer prognosis [224], highlighting their importance in personalized medicine. Overall, miRNAs are excellent candidates for diagnostic, prognostic, and predictive patient stratification, offering hope for more accurate and less invasive cancer diagnostics [225].

6.7. BRAF

It has been demonstrated that the tumor cell proliferation index, measured by Ki67 expression, serves as an effective prognostic marker for MTC [226]. BRAFV600E is the most frequent genetic mutation in DTC, occurring in 60% of patients, and contributes to the development of malignant tumor cell phenotypes, including proliferation, metastasis, and immune escape [218]. This V600E mutation, for instance, reduces the expression of genes responsible for iodine metabolism, thereby diminishing tumors’ responsiveness to iodine-131 (131I) [227]. Papillary carcinoma is more commonly observed in patients with the BRAF mutation than in those without it [228]. In cases where a RAS-like mutation is detected, there is uncertainty regarding the malignancy risk of the nodule [218]. Anaplastic thyroid cancer, the most aggressive form of thyroid cancer, is found to harbor BRAF mutations in over 40% of cases [229]. In normal thyroid tissue, the wild-type BRAF gene is transcribed into the BRAF protein, which is activated by the RAS family [229]. BRAF is a cytoplasmic serine–threonine protein kinase that plays a crucial role in the MAPK signaling pathway. Among the RAF family members, BRAF is the only one activated by mutations in human cancers. However, BRAFV600E should not be used as the sole prognostic marker in low-risk thyroid cancer due to its low positive predictive value [230,231]. In conclusion, BRAF mutations, particularly BRAFV600E, are a significant risk factor for LNM in PTC. Research indicates that in conventional PTC, the presence of the BRAF mutation increases the risk of death related to LNM, a risk that is absent in BRAF wild-type cases [232]. Thyroid nodules with BRAF mutations that are not V600E exhibit a higher malignancy rate (73%), though still lower than that for nodules with BRAFV600E mutations. According to ATA guidelines, most of these nodules are classified as low risk, with only 8% being considered intermediate or high risk [233].

6.8. RAS

RAS mutations, particularly in the KRAS proto-oncogene, are crucial in the progression of ATC, a highly aggressive form of thyroid cancer. KRAS is part of a family of small GTP-binding proteins, and mutations in this gene can lead to the constitutive activation of the RAS/MAPK signaling pathway, promoting tumor growth and metastasis [234,235]. These mutations are observed in approximately 13% of PTC cases, where they often maintain some degree of follicular structural differentiation [236]. On the other hand, FTCs are more commonly associated with RAS mutations, placing them in the RAS-like category of thyroid tumors [237,238]. In both FTCs and PTCs, especially those with poorly differentiated areas, RAS mutations have been identified as important diagnostic markers. These mutations can be particularly useful when malignancy is not clearly evident through standard morphological evaluation [239,240]. Moreover, the presence of RAS mutations correlates with a significantly higher risk of distant metastasis and increased mortality, suggesting their potential as a prognostic marker in thyroid cancer [241]. It is also noteworthy that most tumors with RAS mutations are classified as the follicular variant of PTC, further emphasizing the role of these mutations in thyroid tumorigenesis [242]. Collectively, RAS mutations play a pivotal role in the development and progression of thyroid cancer, with important implications for diagnosis and prognosis.

6.9. RET

RET mutations play a significant role in the development of thyroid cancer, particularly in PTC [243]. One of the key genetic aberrations implicated in the development of PTC is the RET chromosomal rearrangement, commonly referred to as RET/PTC [244]. This rearrangement is most frequently associated with follicular cell-derived thyroid cancers [245]. RET/PTC fusions are known to result in the formation of a fusion gene, and they are present in more than 70% of radiation-induced thyroid cancers [246]. This highlights the crucial role of RET rearrangements in thyroid carcinogenesis, especially in the context of radiation exposure. The RET proto-oncogene encodes a cell membrane receptor tyrosine kinase, and when altered, it can lead to the activation of oncogenic signaling pathways that drive tumor formation [238,247]. There are various types of RET/PTC rearrangements, defined by different fusion partner genes, with RET/PTC1 and RET/PTC3 being the most common forms [248]. These rearrangements are more frequently found in malignant thyroid tissues than in benign nodules, and they are strongly associated with more aggressive tumor behavior, including a higher likelihood of lymph node metastasis [249]. RET/PTC is particularly prevalent in pediatric thyroid cancers, where it is the most common mutation found in children and adolescents with thyroid cancer [250]. In these patients, RET/PTC mutations are often linked to more aggressive tumor behavior and an increased frequency of metastatic dissemination [251]. Interestingly, benign thyroid lesions that harbor RET/PTC translocations are also characterized by a rapid growth rate, which often necessitates early surgical intervention [252]. This rapid progression underscores the potential malignancy of thyroid lesions harboring RET/PTC rearrangements, even if initially benign [253]. Moreover, while somatic RET mutations are generally considered negative prognostic indicators, some studies argue that these mutations do not necessarily correlate with compromised disease-specific survival (DSS) or overall survival (OS) in patients with MTC [254]. However, it is important to note that children who possess RET codon mutations exhibit the most severe manifestations of MTC, highlighting the potential for worse outcomes in this population [255]. In summary, RET/PTC rearrangements are a crucial molecular event in thyroid cancer, especially in PTC, and are strongly associated with aggressive disease, frequent metastasis, and rapid tumor growth. While the prognostic implications of RET mutations can vary, their role in the pathogenesis of thyroid cancer remains a significant focus of ongoing research [256].

6.10. Summary

Among the discussed biomarkers, thyroglobulin (Tg) and calcitonin (Ctn) hold the greatest clinical importance as the primary markers for differentiated thyroid cancer (DTC) and medullary thyroid cancer (MTC), respectively. While both are well-established tools for postoperative monitoring and assessing treatment response, their diagnostic accuracy may be limited by interfering factors such as Tg autoantibodies or comorbid conditions. CEA and procalcitonin (Pct) enhance the prognostic value of Ctn in MTC but lack sufficient specificity to serve as standalone markers. Emerging molecular markers—including microRNAs, BRAF and RAS mutations, and RET/PTC rearrangements—support subtype-specific diagnosis and risk stratification and may guide targeted therapy in the future, though further clinical validation is needed. Importantly, the diagnostic and prognostic relevance of each marker varies significantly across thyroid cancer subtypes, making histological context and individual patient characteristics essential for accurate interpretation (Table 4).

7. Current Treatment Strategies

7.1. Surgical Treatment of Thyroid Cancer

7.1.1. Papillary Thyroid Cancer

At present, surgery is the most common and effective method for treating PCT [26]. The choice of surgical procedure for PCT depends on the patient’s specific clinical condition and the characteristics of the carcinoma itself. In some situations, the entire thyroid gland must be removed, while in other cases, a thyroid lobectomy is performed to remove only the affected lobe. For patients with unilateral goiter, low baseline Ctn levels, and small tumors (≤2.5 cm), a lobectomy combined with central (±lateral) LND can be an effective alternative to more extensive procedures such as total thyroidectomy [257]. For unilateral tumors, lobectomy along with central (±lateral) LND may be sufficient, but for bilateral or larger (metastatic) tumors, more extensive surgery is required. The decision on the extent of surgery should be based on imaging results (such as ultrasound) and Ct levels [258]. In children with DTC, total thyroidectomy, along with bilateral LND of the central compartment and dissection of affected lateral compartments, is recommended [259]. While PCT generally has a favorable prognosis when diagnosed early, it is often associated with a high rate of LNM. There is no agreement on whether prophylactic LND should be a standard procedure for patients with cN0 PTC, though some research indicates it may improve outcomes and help avoid unnecessary subsequent surgeries, especially when risk factors are considered [260]. However, aggressive LND can lead to a range of postoperative complications, including hypoparathyroidism, chylous leakage, injury to the cervical plexus, recurrent laryngeal nerve damage, and neck numbness, which can all negatively affect the patient’s quality of life [261]. In terms of LNM in PTC, our study supports previous findings that younger age, multifocality, larger tumor size, and extrathyroidal invasion are independent risk factors for LNM in PTC patients [262]. Thyroid nodules are common in the general population, with prevalence rates ranging from 50% to 67% in autopsy studies [263]. Ultrasound imaging of thyroid nodules, followed by fine-needle aspiration biopsy for cytological analysis, remains the gold standard for pre-surgical diagnosis [264]. Additionally, a study showed that total thyroidectomy (TT) did not offer better therapeutic outcomes than thyroid lobectomy in low-risk DTC patients. Consequently, thyroid lobectomy was included as a treatment option in the 2015 ATA guidelines, which has led to an increase in both the number of low-risk DTC patients and the number of those undergoing thyroid lobectomy [204].

7.1.2. Follicular Thyroid Cancer

FTC often develops without noticeable symptoms in its early stages, making it challenging to detect early. The standard treatment for FTC is thyroid lobectomy, but this approach is not suitable for larger tumors (>4 cm) or those with significant extrathyroidal extension [265]. Additionally, surgical options are not recommended for patients with aggressive mutations associated with FTC [266]. In some cases, surgery might be considered for nodules that are initially classified as benign based on cytology and/or low risk on ultrasound but later become symptomatic [267]. However, for nodules with indeterminate cytology (Bethesda class III and IV), where active surveillance is not appropriate—such as in cases of large size, high suspicion of malignancy on ultrasound, or the presence of symptoms—surgical intervention may be necessary [268]. For minimally invasive FTC, the typical treatment involves thyroid lobectomy and isthmectomy, while for invasive FTC, the preferred treatment includes total thyroidectomy, radioiodine ablation, and the use of TSH-suppressing medications [34]. Studies indicate that for tumors ranging between 1 and 4 cm, the survival rates following TT and lobectomy are similar when various risk factors are taken into account [265]. In pediatric patients, lobectomy is often the preferred approach due to the lower likelihood of aggressive disease and the desire to preserve thyroid function [269]. The surgical strategy for FTC should be tailored to each individual, considering tumor characteristics, the patient’s age, and overall health to achieve the best outcomes and reduce the risk of complications [265]. Moreover, in the treatment of PTC with a dominant follicular variant (FVPTC), factors such as tumor size, extension outside the thyroid (extrathyroidal extension), and the patient’s gender are important. Research shows that for tumors smaller than 2 cm, lobectomy is typically sufficient, and more aggressive surgery is not necessary. However, for tumors larger than 2 cm, particularly those with extrathyroidal extension, TT is recommended, as it enhances survival rates [270].

7.1.3. Medullary Thyroid Cancer

The treatment for MTC primarily involves surgery, with TT being the preferred option in most cases, even for smaller unilateral tumors. When MTC is confined to the thyroid, TT should be performed along with bilateral central LND at level VI. This approach is recommended because research has demonstrated that CLND increases the likelihood of a cure [271]. For patients with MTC that is confined to the neck but includes lateral neck lymph node metastases, total thyroidectomy, bilateral CLND, and selective lateral LND (covering at least levels II to V) are recommended. During this procedure, it is crucial to protect key anatomical structures such as the sternocleidomastoid muscle, internal jugular vein, and accessory nerve [272]. In individuals with RET mutations, particularly children and younger adults, prophylactic TT may be considered, depending on the type of mutation and the patient’s age. In adult patients with RET mutations, TT along with removal of the appropriate lymph nodes based on Ctn levels is generally advised [56]. Recently, for sporadic MTC that is clinically confined to a single thyroid lobe, a less invasive option such as hemithyroidectomy with or without diagnostic ipsilateral central LND has been suggested as a risk-reducing approach [273]. For hereditary MTC, the most effective strategy involves a DNA and biochemical-based approach, which limits prophylactic surgery to TT to minimize surgical risks before the tumor has spread beyond the thyroid capsule [273]. In some cases of sporadic MTC, thyroid lobectomy may be suitable, particularly if preoperative neck ultrasound shows no evidence of metastasis to the opposite thyroid lobe. Postoperative monitoring of Ctn levels is necessary, and if levels drop to undetectable levels after lobectomy, further surgery may not be required [274].

7.1.4. Anaplastic Thyroid Cancer

The treatment of choice for ATC is thyroidectomy, yet the majority of cases are diagnosed at an advanced stage, rendering them unresectable at presentation [275]. Surgery plays a crucial role in localized ATC, as surgical resection is the preferred treatment for this stage [71]. However, for patients with more advanced disease, especially stage IVC, TT is the preferred method because it has been shown to improve OS and DSS [276]. In some cases, debulking surgery—removal of part of the tumor—is considered. However, in stage IVC, this approach is generally contraindicated because it does not offer significant survival benefits and may result in severe complications, such as delaying the initiation of systemic treatments, including chemotherapy and radiotherapy [276]. The first step in treating ATC is often surgery for local control, particularly in patients with stage IVB. In such cases, surgery may prevent immediate death from asphyxiation; however, these patients often succumb to the disease due to lung metastases or other complications as their condition deteriorates [277]. Radiation therapy may also be implemented postoperatively, in addition to levothyroxine treatment, which is necessary for patients following thyroidectomy. This postoperative care is crucial to manage hormonal imbalance and other complications [275]. While radical surgery, involving the resection of organs responsible for speech and swallowing, is sometimes performed in certain cases, its benefits are questionable, even when clear margins are achieved. Most patients still experience poor outcomes despite undergoing surgery [278]. ATC is commonly diagnosed at an advanced stage, often with invasion into intrathoracic vessels or prevertebral tissues, which is frequently used as a criterion for unresectability. However, opinions on the precise definition of resectability vary, making treatment decisions more complex (Figure 3) [279].

7.2. Radioiodine Therapy

After undergoing thyroidectomy, patients diagnosed with PCT generally receive both radioactive iodine (RAI) therapy and lifelong thyroid hormone replacement. The primary role of RAI therapy is to eliminate any remaining thyroid tissue or cancer cells that may be left after surgery. It uses RAI isotopes to target and destroy these cells, enhancing the treatment’s post-surgery effectiveness [280]. Subsequently, thyroid hormone replacement is essential to compensate for the lost thyroid gland and maintain normal bodily functions. On the other hand, MTC, which arises from the thyroid’s parafollicular C cells, does not concentrate iodine, rendering RAI therapy ineffective. In these cases, alternative treatments must be considered, as RAI therapy and ablation fail to achieve the desired results. While RAI therapy provides significant benefits for many patients with DTC, it is not without its limitations. Resistance to RAI therapy develops in 33–50% of thyroid cancer patients over time, leading to poorer prognosis and reduced survival [281]. This resistance is often linked to factors such as the primary tumor exceeding 40 mm in size, extrathyroidal extension, age over 55 years, and early rises in TG levels, all of which strongly predict a suboptimal response to treatment [281]. For high-risk DTC patients, particularly those with lymph node involvement or distant metastases, RAI therapy has been associated with improved long-term survival, with patients suffering from PTC and metastases showing a clear benefit [282]. However, RAI therapy is ineffective in treating ATC due to the lack of iodine uptake and radioresistance in these tumors [283]. Certain conditions contraindicate the use of RAI therapy, including pregnancy, breastfeeding, the absence of iodine uptake in TC, and thyroid issues like nystagmus or eye diseases in Graves’ disease, as well as thyrotoxicosis [284]. For patients without metastases prior to the 123-I scan, an empirical radioiodine dose is typically administered [285]. The decision to proceed with RAI therapy is guided by the recurrence risk classification from the ATA, though it can be challenging to apply in clinical practice due to the multitude of variables involved. Additional diagnostic tests, such as measuring TG or TgAb and performing neck ultrasound, may help better identify suitable candidates for therapy [286]. To ensure the success of RAI therapy, a careful analysis of diagnostic data is essential in determining the appropriate dosage to minimize complications while maximizing treatment effectiveness. In some instances, patients with low or very low risk may avoid systemic RAI therapy and instead receive low-dose protocols, as evidenced by clinical trials like ESTIMABLE2 [286]. RAI therapy is most beneficial for high-risk patients, such as those with metastatic disease or aggressive tumors. However, its benefits remain less clear and are a subject of ongoing debate for intermediate-risk patients [287]. Efforts to restore sensitivity to RAI therapy by redifferentiating thyroid cancer cells have shown limited success. Although some treatments may increase iodine uptake, they do not always lead to a clinically meaningful response [288]. For those with RAI-refractory disease, which includes patients with structural disease despite adequate preparation for RAI therapy, redifferentiation strategies involving kinase inhibitors (e.g., BRAF, MEK, RET) can potentially improve iodine uptake and treatment outcomes [289]. Additionally, RAI therapy can lead to significant alterations in hematological parameters, such as a reduction in white blood cells, neutrophils, lymphocytes, platelets, and red blood cells. This change increases the risk of anemia, infection, and bleeding [290]. Furthermore, I131 (radioactive iodine) primarily induces thyroid cell death through apoptosis, although necrosis may also occur in some cases. This therapy also impacts the immune response, shifting it from autoantibody production to a chemokine-driven immune response that attracts immune cells to inflamed thyroid tissues [291]. In conclusion, RAI therapy remains a cornerstone in treating DTC, especially for high-risk patients. However, its effectiveness can be hindered by factors like resistance and the presence of complications. Thus, precise patient selection and individualized treatment plans are crucial for optimizing outcomes.

7.3. Chemotherapy

Chemotherapy is infrequently used in the treatment of TC, primarily in advanced cases such as ATC, which is more aggressive and does not respond well to conventional treatments [292]. In these situations, when surgery or RAI ablation does not yield satisfactory results, chemotherapy may become the preferred treatment option [292]. Although chemotherapy plays a limited role in TC treatment, it is mainly employed in ATC, where chemotherapy alone has shown limited success [293]. Common chemotherapy drugs used in ATC treatment include doxorubicin, cisplatin, and taxanes such as docetaxel and paclitaxel [293]. Despite their relatively widespread use, these medications do not substantially improve patient survival [294]. Moreover, chemotherapy combinations like doxorubicin and cisplatin often come with significant side effects, including hematological and gastrointestinal issues [295]. However, the combination of doxorubicin and cisplatin tends to produce a higher response rate than doxorubicin alone. In one study, 20% of patients showed a partial response, and 30% had stable disease. The median progression-free survival was 6 months, and the overall survival was 9 months, with tolerable side effects [296]. Doxorubicin chemotherapy remains one of the few palliative options for patients with advanced or metastatic thyroid cancer resistant to RAI treatment. Administering doxorubicin at a dose of 60 mg/m2 every three weeks can result in disease stabilization, which, given the aggressive nature of the tumor, can be considered a therapeutic success [297]. However, chemotherapy for TC is generally of limited effectiveness, especially in radioiodine-resistant cases [298].

7.4. Immunotherapy

Recent advancements in TC treatment focus on the use of immunotherapies and targeted therapies. Immunotherapies, such as PD-1 inhibitors (e.g., pembrolizumab and spartalizumab), enhance the patient’s immune system to target the tumor, especially in cases where the cancer is resistant to other treatments. These therapies are particularly effective for cancers with high microsatellite instability or elevated PDL1 expression, as well as in ATC [299]. Immunotherapy’s effectiveness varies by tumor type, with ATC showing a better response due to its higher mutational load, in contrast to PCT or FTC, which have a lower mutational load and PD-L1 expression [300]. Targeted therapies are essential for treating rare and aggressive TC forms. Tyrosine kinase inhibitors (TKIs) such as sorafenib, lenvatinib, vandetanib, and cabozantinib have been approved for treating progressive TC. Clinical trials have demonstrated their effectiveness in improving progression-free survival and treatment response rates (ORR) [301]. However, while TKIs are effective, they can cause specific side effects such as liver issues, gastrointestinal problems, hypertension, proteinuria, and fatigue, though they avoid typical chemotherapy side effects like hair loss and nausea [302]. In addition to TKIs, treatments for specific mutations, such as TRK kinase inhibitors (larotrectinib and entrectinib), have shown success in treating cancers with NTRK gene mutations [133]. BRAFV600E inhibitors, such as vemurafenib, and their combination with MEK inhibitors (dabrafenib + trametinib), are particularly effective for patients with the BRAFV600E mutation in DTC [303]. However, the combination of dabrafenib and trametinib, while highly effective, can lead to more side effects, particularly in older patients, causing some to stop treatment [303]. Genetic factors can limit the effectiveness of these therapies. For example, mutations such as V804M-RET in MTC can lead to resistance to certain drugs [304]. Although tyrosine kinase inhibitors can stabilize the disease, they often do not result in complete remission [304]. In cases of unresectable MTC, systemic therapies like RET-specific inhibitors (pralsetinib and selpercatinib) or TKIs such as cabozantinib and vandetanib should be used. For tumors with microsatellite instability-high status or mismatch repair deficiencies, pembrolizumab therapy is also an option [304]. Targeted therapies continue to evolve, with new drugs like pralsetinib and selpercatinib focusing on RET gene mutations and entrectinib, larotrectinib, and repotrectinib targeting NTRK gene mutations. The use of BRAF inhibitors like vemurafenib and dabrafenib, MEK inhibitors such as trametinib, and anti-angiogenic multi-targeted kinase inhibitors (e.g., lenvatinib and sorafenib) is also becoming more widespread [305]. Ongoing monitoring is crucial in these treatments. Follow-up methods for DTC include serum TG measurements, neck ultrasonography, and occasionally I-131 whole-body scintigraphy [306]. Additionally, US-guided FNA-Tg has been shown to be a valuable tool for monitoring DTC patients, regardless of TSH levels or the presence of TgAb [307]. In summary, a modern approach to TC treatment involving immunotherapy and targeted therapies offers significant promise, especially for advanced or resistant cancer cases. However, these treatments’ success depends on genetic mutations, and close monitoring is essential to manage side effects and assess their ongoing effectiveness (Figure 4).

7.5. Comparison of First-Line and Second-Line Therapies

First-line therapies for TC typically include surgical resection, radioiodine ablation (RAI) (mainly in differentiated thyroid cancer (DTC), and TSH suppression [107,121]. These methods, especially when used in early and well-differentiated tumors such as PTC and FTC, are often curative therapies [107,110,115,121]. Total thyroidectomy with bilateral lymph node dissection remains the recommended approach in children with DTC, while thyroid lobectomy is commonly used in minimally invasive FTC [110,121]. In the case of MTC, surgical treatment—particularly TT with central neck dissection—is the mainstay of therapy, especially when the disease is limited to the thyroid gland [127]. Similarly, ATC, which is highly aggressive, is initially treated with thyroidectomy if possible, often followed by postoperative radiotherapy and levothyroxine therapy [107]. However, there are cases where the disease becomes refractory to first-line interventions—such as RAI-refractory DTC, unresectable tumors, or advanced/metastatic disease—in which case second-line therapies are required. RAI resistance develops in approximately 33–50% of patients with DTC over time, and this reduces survival and treatment efficacy [139]. In such cases, systemic therapies become necessary. These include tyrosine kinase inhibitors (TKIs) such as sorafenib and lenvatinib for progressive, RAI-refractory DTC and vandetanib or cabozantinib for advanced MTC [171]. In addition, newer RET-specific inhibitors such as pralsetinib and selpercatinib have shown efficacy in unresectable or metastatic MTC with RET mutations [171]. In ATC, BRAF and MEK inhibitors (e.g., dabrafenib and trametinib) are considered second-line options, particularly in BRAF-mutated tumors, with objective response rates above 60% [171]. Second-line treatments are often associated with a higher toxicity burden and a lower likelihood of achieving complete remission compared with first-line treatment. For example, lenvatinib in RAI-refractory DTC results in a median progression-free survival of approximately 18 months, which contrasts with the potential for complete remission with initial RAI therapy in early stage disease [139]. Furthermore, efforts to redifferentiate thyroid cancer cells to restore iodine sensitivity have had limited clinical impact, despite theoretical promise [146,147]. Chemotherapy, especially with doxorubicin, remains a palliative option in advanced, refractory cases [146]. As systemic treatment options evolve, early integration of targeted or immune-based therapies is increasingly being considered for aggressive variants such as tall cell thyroid cancer or poorly differentiated thyroid cancer (Table 5) [171].

7.6. Molecularly Targeted Therapies in Thyroid Cancer: Recent Advances

Recent advances in targeted therapies have resulted in the approval of new molecularly targeted drugs such as selpercatinib and pralsetinib, which have shown high efficiency in patients with MTC with RET mutations and DTC with RET fusions [308]. Both medications are selective RET kinase inhibitors that have been approved following the findings of trials such as LIBERTTO-001 (NTC03157128) and ARROW (NTC03037385) [309]. Concurrently, RET inhibitors such as pralsetinib have demonstrated success in patients with RET-altered thyroid tumors, including those who have received prior treatment [310]. Furthermore, PD-1 inhibitors like spartalizumab, when taken with lenvatinib, have demonstrated encouraging outcomes in the treatment of anaplastic thyroid carcinoma, a particularly aggressive kind of thyroid cancer (Table 6) [311].

7.7. Considerations in Resource-Limited Settings

In resource-limited settings, the management of thyroid cancer poses significant challenges due to various limitations in diagnostic tools, treatment modalities, and surgical expertise [313]. A lack of access to advanced diagnostic technologies such as molecular testing (e.g., BRAF or RET mutation panels), CT, and MRI is another intractable problem [314]. As a result, physicians often rely on more basic imaging modalities such as ultrasound, which are more affordable and available in many centers. However, they may not provide the same level of detail as more advanced methods. Additionally, essential therapies such as I-131 therapy, which is key in the treatment of DTC [315], may be unavailable or prohibitively expensive. Both targeted therapies and genetic testing are essential for personalized treatment plans but are often out of reach, leading to diagnostic limitations [316]. Surgical care may also be compromised, as a shortage of skilled endocrine surgeons increases the risk of suboptimal surgery, potentially affecting prognosis [317]. Furthermore, inadequate access to postoperative monitoring tools such as serum TG testing or high-quality ultrasound impedes the detection of recurrences and long-term management [161]. As a result, treatment protocols may be simplified, and lobectomy may be preferred over total thyroidectomy to reduce the dependence on lifelong hormonal therapy, which may not be consistently available [318]. In such circumstances, telemedicine consultations, regional centers of excellence, and international outreach programs play a key role in improving outcomes by facilitating access to both medical expertise and essential medications.

8. Future Perspectives

Future perspectives in TC therapy focus on the integration of genomic profiling and liquid biopsy to guide personalized treatment strategies and overcome resistance to some existing therapies. Continued research into novel actionable mutations and immune targets is essential to expand therapeutic options and improve outcomes for patients with advanced disease [319]. In the near future, artificial intelligence (AI) technology may be helpful in accessing the morphological and molecular features of TC. Recent advancements in AI have significantly enhanced the diagnostic accuracy of thyroid nodules by leveraging quantitative morphological and cytological features, aiding in the differentiation of challenging entities such as follicular adenoma, follicular carcinoma, and variants of papillary thyroid carcinoma. Furthermore, the integration of AI with molecular testing, including gene panels and protein-based classifiers, holds promise for improving risk stratification, personalized treatment planning, and the early identification of aggressive disease, thus setting the stage for more precise and individualized TC management [320]. Moreover, further research on TC risk factors is vital to elucidate the impact of endocrine-disrupting chemicals (EDCs) on the incidence and progression of TC and thyroid-related disorders. Scientists should focus on the role of various classes of EDCs, including heavy metals, cosmetic ingredients, industrial chemicals, pesticides, herbicides, pharmaceuticals, and both synthetic and naturally occurring hormones. Comprehensive studies are needed to better understand their mechanisms of action, exposure pathways, and potential effects on thyroid function and carcinogenesis [321]. Future perspectives of TC management also highlight the expanding role of PET/CT, particularly in identifying recurrent or metastatic disease in patients with non-iodine-avid tumors or elevated thyroglobulin levels. Advancements in novel radiotracers and theragnostic applications may further enhance its clinical utility, supporting more personalized and effective treatment strategies, though efforts to improve accessibility, cost-efficiency, and standardization remain essential [322].

9. Conclusions

Thyroid cancer has become a growing global health issue, with its incidence increasing notably in recent years. This rise is largely attributed to increased exposure to environmental factors such as chemicals, radiation, and pollution, especially in areas with a higher HDI. Despite the growing number of diagnoses, death rates have remained relatively stable, suggesting that while thyroid cancer is generally treatable, challenges persist in its early detection and management. The disease’s complex epidemiology shows a higher prevalence among women and younger populations, underscoring the need for targeted treatment approaches for different cancer subtypes, such as papillary, follicular, medullary, and anaplastic thyroid cancers. Recognizing key risk factors such as physical inactivity, obesity, radiation exposure, and genetic mutations is essential for understanding thyroid cancer’s development. Specifically, mutations in genes like BRAF, RAS, RET, and NTRK are crucial in assessing disease severity and prognosis. Genetic testing, especially for BRAF V600E and RET mutations, plays a critical role in tailoring treatment plans, offering better predictions of disease behavior and aiding therapeutic decisions. Various biomarkers, including TG, Ctn, CEA, and Pct, are vital for diagnosing and monitoring thyroid cancer. These markers are useful for early detection, tracking disease progression, and evaluating treatment effectiveness, particularly in differentiated and medullary thyroid cancers. The emerging role of microRNAs as exosomal biomarkers shows promising potential for enhancing diagnostic accuracy and patient classification. Although traditional treatments such as surgery and RAI therapy remain foundational, their effectiveness depends on cancer subtype, tumor size, and genetic characteristics. RAI therapy works well for many differentiated thyroid cancers but faces resistance, particularly in medullary and anaplastic types, making alternative treatments like chemotherapy and targeted therapies increasingly important. Immunotherapies and TKIs have shown potential in cases resistant to standard treatments, though their success relies heavily on genetic factors and precise patient selection. In conclusion, continued progress in genetic profiling, biomarker discovery, and personalized treatment strategies is crucial to improving thyroid cancer outcomes. While early detection and conventional therapies benefit many patients, those with advanced or resistant forms require more innovative treatments targeting specific genetic mutations for better management. The future of thyroid cancer care will likely combine genetic insights, novel therapies, and individualized approaches to optimize patient outcomes and mitigate the impact of this growing disease.

Author Contributions

Conceptualization, A.F. and J.B.; formal analysis, A.F., J.F. and J.B.; investigation, A.F., K.K., W.P., B.T. and J.J.; resources, A.F., K.K., W.P., B.T. and J.J.; data curation, A.F., K.K., W.P., B.T. and J.J.; writing—original draft preparation, A.F., K.K., W.P., B.T. and J.J.; writing—review and editing, A.F., J.F. and J.B.; visualization, A.F.; supervision, J.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
131Iiodine-131
AHRaryl hydrocarbon receptor
AIartificial intelligence
AP1activator protein 1
ATAAmerican Thyroid Association
ATCanaplastic thyroid cancer
BMIbody mass index
CEAcarcinoembryonic antigen
CGRPcalcitonin gene-related peptide
CRPC-reactive protein
CTcomputed tomography
Ctncalcitonin
DCdendritic cell
DSSdisease-specific survival
DSVdiffuse sclerosing variant
DTCdifferentiated thyroid cancer
EDCsendocrine-disrupting chemicals
ERαestrogen receptor alpha
ERβestrogen receptor beta
FMTCfamilial medullary thyroid cancer
FNAfine-needle aspiration
FNABfine-needle aspiration biopsy
FNACfine-needle aspiration cytology
FT3free triiodothyronine
FT4free thyroxine
FTCfollicular thyroid carcinoma
FTCfollicular thyroid cancer
FVPTCfollicular variant of papillary thyroid carcinoma
FVPTCPTC with a dominant follicular variant
HDIHuman Development Index
HTHashimoto thyroiditis
HVhobnail
IGF-1insulin-like growth factor 1
LNDlymph node dissection
LNMlymph node metastasis
MAPKMAP kinase
MEN2multiple endocrine neoplasia type 2
MEN4multiple endocrine neoplasia type 4
MRImagnetic resonance
MTCmedullary thyroid cancer
NF-kBnuclear factor kappa-light-chain-enhancer of activated B
NIFTPnon-invasive follicular thyroid neoplasm with papillary-like nuclear features
NISsodium–iodide symporter
ORRtreatment response rate
OSoverall survival
PCRreverse-transcriptase polymerase reaction
Pctprocalcitonin
PTCpapillary thyroid cancer
RAIradioactive iodine
RFradiofrequency
SHSsecondhand smoking
SPECTsingle-proton emission computed tomography
SVsolid variant
T3triiodothyronine
T4thyroxine
TAMstumor-associated macrophages
TBGthyroxine-binding globulin
TBSRTCThe Bethesda System for Reporting Thyroid Cytopathology
TCthyroid cancer
TCVtall cell variant
TGthyroglobulin
TKIstyrosine kinase inhibitors
TMEtumor microenvironment
TNFtumor necrosis factor
TSHthyroid-stimulating hormone
TSHRsTSH receptors
TTtotal thyroidectomy
TgAbanti-TG antibody
USultrasonography
VIPvasoactive intestinal peptide
lncRNAslong non-coding RNAs
mRNAmessenger RNA
miRNAmicroRNA

References

  1. Wells, S.A., Jr. Progress in Endocrine Neoplasia. Clin. Cancer Res. 2016, 22, 4981–4988. [Google Scholar] [CrossRef] [PubMed]
  2. Morris, L.G.; Sikora, A.G.; Tosteson, T.D.; Davies, L. The increasing incidence of thyroid cancer: The influence of access to care. Thyroid 2013, 23, 885–891. [Google Scholar] [CrossRef] [PubMed]
  3. Tang, Z.; Zhang, J.; Zhou, Q.; Xu, S.; Cai, Z.; Jiang, G. Thyroid Cancer "Epidemic": A Socio-Environmental Health Problem Needs Collaborative Efforts. Environ. Sci. Technol. 2020, 54, 3725–3727. [Google Scholar] [CrossRef] [PubMed]
  4. Kruger, E.; Toraih, E.A.; Hussein, M.H.; Shehata, S.A.; Waheed, A.; Fawzy, M.S.; Kandil, E. Thyroid Carcinoma: A Review for 25 Years of Environmental Risk Factors Studies. Cancers 2022, 14, 6172. [Google Scholar] [CrossRef]
  5. Ferrari, S.M.; Fallahi, P.; Antonelli, A.; Benvenga, S. Environmental Issues in Thyroid Diseases. Front. Endocrinol. 2017, 8, 50. [Google Scholar] [CrossRef]
  6. Bogović Crnčić, T.; Ilić Tomaš, M.; Girotto, N.; Grbac Ivanković, S. Risk Factors for Thyroid Cancer: What Do We Know So Far? Acta Clin. Croat. 2020, 59, 66–72. [Google Scholar] [CrossRef]
  7. Fiore, M.; Conti, G.O.; Caltabiano, R.; Buffone, A.; Zuccarello, P.; Cormaci, L.; Cannizzaro, M.A.; Ferrante, M. Role of Emerging Environmental Risk Factors in Thyroid Cancer: A Brief Review. Int. J. Environ. Res. Public Health 2019, 16, 1185. [Google Scholar] [CrossRef]
  8. Shank, J.B.; Are, C.; Wenos, C.D. Thyroid Cancer: Global Burden and Trends. Indian J. Surg. Oncol. 2022, 13, 40–45. [Google Scholar] [CrossRef]
  9. Ferlay, J.; Ervik, M.; Lam, F.; Laversanne, M.; Colombet, M.; Mery, L.; Pineros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today; International Agency for Research on Cancer: Lyon, France, 2024; Available online: https://gco.iarc.who.int/today (accessed on 13 January 2025).
  10. Rahbari, R.; Zhang, L.; Kebebew, E. Thyroid cancer gender disparity. Future Oncol. 2010, 6, 1771–1779. [Google Scholar] [CrossRef]
  11. Available online: https://www.cancer.org/cancer/types/thyroid-cancer/about/key-statistics.html (accessed on 5 April 2025).
  12. Girardi, F.M. Thyroid Carcinoma Pattern Presentation According to Age. Int. Arch. Otorhinolaryngol. 2017, 21, 38–41. [Google Scholar] [CrossRef]
  13. Magreni, A.; Bann, D.V.; Schubart, J.R.; Goldenberg, D. The Effects of Race and Ethnicity on Thyroid Cancer Incidence. JAMA Otolaryngol. Head Neck Surg. 2015, 141, 319–323. [Google Scholar] [CrossRef] [PubMed]
  14. Alagoz, O.; Zhang, Y.; Arroyo, N.; Fernandes-Taylor, S.; Yang, D.-Y.; Krebsbach, C.; Venkatesh, M.; Hsiao, V.; Davies, L.; Francis, D.O. Modeling Thyroid Cancer Epidemiology in the United States Using Papillary Thyroid Carcinoma Microsimulation Model. Value Health 2024, 27, 367–375. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, J.; Ngai, C.H.; Deng, Y.; Pun, C.N.; Lok, V.; Zhang, L.; Xu, Q.; Lucero-Prisno, D.E.; Xu, W.; Zheng, Z.J.; et al. Incidence and mortality of thyroid cancer in 50 countries: A joinpoint regression analysis of global trends. Endocrine 2023, 80, 355–365. [Google Scholar] [CrossRef]
  16. Pizzato, M.; Li, M.; Vignat, J.; Laversanne, M.; Singh, D.; La Vecchia, C.; Vaccarella, S. The epidemiological landscape of thyroid cancer worldwide: GLOBOCAN estimates for incidence and mortality rates in 2020. Lancet Diabetes Endocrinol. 2022, 10, 264–272. [Google Scholar] [CrossRef]
  17. Zhang, J.; Xu, S. High aggressiveness of papillary thyroid cancer: From clinical evidence to regulatory cellular networks. Cell Death Discov. 2024, 10, 378. [Google Scholar] [CrossRef]
  18. Available online: https://www.thyroidcancer.com/thyroid-cancer/papillary (accessed on 5 April 2025).
  19. Verburg, F.A.; Van Santen, H.M.; Luster, M. Pediatric papillary thyroid cancer: Current management challenges. OncoTargets Ther. 2016, 10, 165–175. [Google Scholar] [CrossRef]
  20. Hughes, D.T.; Haymart, M.R.; Miller, B.S.; Gauger, P.G.; Doherty, G.M. The most commonly occurring papillary thyroid cancer in the United States is now a microcarcinoma in a patient older than 45 years. Thyroid 2011, 21, 231–236. [Google Scholar] [CrossRef]
  21. Coca-Pelaz, A.; Shah, J.P.; Hernandez-Prera, J.C.; Ghossein, R.A.; Rodrigo, J.P.; Hartl, D.M.; Olsen, K.D.; Shaha, A.R.; Zafereo, M.; Suarez, C.; et al. Papillary Thyroid Cancer-Aggressive Variants and Impact on Management: A Narrative Review. Adv. Ther. 2020, 37, 3112–3128. [Google Scholar] [CrossRef] [PubMed]
  22. Lee, J.S.; Lee, J.S.; Yun, H.J.; Kim, S.M.; Chang, H.; Lee, Y.S.; Chang, H.-S.; Park, C.S. Aggressive Subtypes of Papillary Thyroid Carcinoma Smaller Than 1 cm. J. Clin. Endocrinol. Metab. 2023, 108, 1370–1375. [Google Scholar] [CrossRef]
  23. Maksimovic, S.; Jakovljevic, B.; Gojkovic, Z. Lymph Node Metastases Papillary Thyroid Carcinoma and their Importance in Recurrence of Disease. Med. Arch. 2018, 72, 108–111. [Google Scholar] [CrossRef]
  24. Smith, B.D.; Oyekunle, T.O.; Thomas, S.M.; Puscas, L.; Rocke, D.J. Association of Lymph Node Ratio with Overall Survival in Patients with Metastatic Papillary Thyroid Cancer. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 962–964. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, Y.; Wang, Y.; Zhao, K.; Li, D.; Chen, Z.; Jiang, R.; Wang, X.; He, X. Lymph node metastasis in young and middle-aged papillary thyroid carcinoma patients: A SEER-based cohort study. BMC Cancer 2020, 20, 181. [Google Scholar] [CrossRef] [PubMed]
  26. Limaiem, F.; Rehman, A.; Mazzoni, T. Papillary Thyroid Carcinoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK536943/ (accessed on 6 April 2025).
  27. Toraih, E.A.; Hussein, M.H.; Zerfaoui, M.; Attia, A.S.; Marzouk Ellythy, A.; Mostafa, A.; Ruiz, E.M.L.; Shama, M.A.; Russell, J.O.; Randolph, G.W.; et al. Site-Specific Metastasis and Survival in Papillary Thyroid Cancer: The Importance of Brain and Multi-Organ Disease. Cancers 2021, 13, 1625. [Google Scholar] [CrossRef] [PubMed]
  28. LiVolsi, V.A. Papillary thyroid carcinoma: An update. Mod. Pathol. 2011, 24, S1–S9. [Google Scholar] [CrossRef]
  29. Ito, Y.; Miyauchi, A. Prognostic factors of papillary and follicular carcinomas in Japan based on data of kuma hospital. J. Thyroid Res. 2012, 2012, 973497. [Google Scholar] [CrossRef]
  30. Ito, Y.; Miyauchi, A.; Kihara, M.; Fukushima, M.; Higashiyama, T.; Miya, A. Overall Survival of Papillary Thyroid Carcinoma Patients: A Single-Institution Long-Term Follow-Up of 5897 Patients. World J. Surg. 2018, 42, 615–622. [Google Scholar] [CrossRef]
  31. Luvhengo, T.E.; Bombil, I.; Mokhtari, A.; Moeng, M.S.; Demetriou, D.; Sanders, C.; Dlamini, Z. Multi-Omics and Management of Follicular Carcinoma of the Thyroid. Biomedicines 2023, 11, 1217. [Google Scholar] [CrossRef]
  32. Rosai, J.; Carcangiu, M.L.; DeLellis, R.A. Tumors of the thyroid gland. In Atlas of Tumor Pathology, 3rd Series, Fas 5; Armed Forces Institute of Pathology: Washington, DC, USA, 1992; pp. 21–48. [Google Scholar]
  33. Correa, P.; Chen, V.W. Endocrine gland cancer. Cancer 1995, 75, 338–352. [Google Scholar] [CrossRef]
  34. Ashorobi, D.; Anastasopoulou, C.; Lopez, P.P. Follicular Thyroid Cancer. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK539775/ (accessed on 6 April 2025).
  35. Seib, C.D.; Sosa, J.A. Evolving Understanding of the Epidemiology of Thyroid Cancer. Endocrinol. Metab. Clin. N. Am. 2019, 48, 23–35. [Google Scholar] [CrossRef] [PubMed]
  36. Available online: https://www.uptodate.com/contents/follicular-thyroid-cancer-including-oncocytic-carcinoma-of-the-thyroid (accessed on 6 April 2025).
  37. Available online: https://www.thyroidcancer.com/thyroid-cancer/follicular/diagnosis (accessed on 6 April 2025).
  38. Basolo, F.; Macerola, E.; Poma, A.M.; Torregrossa, L. The 5th edition of WHO classification of tumors of endocrine organs: Changes in the diagnosis of follicular-derived thyroid carcinoma. Endocrine 2023, 80, 470–476. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. Goffredo, P.; Cheung, K.; Roman, S.A.; Sosa, J.A. Can minimally invasive follicular thyroid cancer be approached as a benign lesion? A population-level analysis of survival among 1,200 patients. Ann. Surg. Oncol. 2013, 20, 767–772. [Google Scholar] [CrossRef] [PubMed]
  40. Yamazaki, H.; Sugino, K.; Katoh, R.; Matsuzu, K.; Kitagawa, W.; Nagahama, M.; Rino, Y.; Ito, K. New Insights on the Importance of the Extent of Vascular Invasion in Widely Invasive Follicular Thyroid Carcinoma. World J. Surg. 2023, 47, 2767–2775. [Google Scholar] [CrossRef] [PubMed]
  41. Leong, D.; Gill, A.J.; Turchini, J.; Waller, M.; Clifton-Bligh, R.; Glover, A.; Sywak, M.; Sidhu, S. The Prognostic Impact of Extent of Vascular Invasion in Follicular Thyroid Carcinoma. World J. Surg. 2023, 47, 412–420. [Google Scholar] [CrossRef] [PubMed]
  42. Lin, J.-D.; Chao, T.-C. Follicular thyroid carcinoma: From diagnosis to treatment. Endocr. J. 2006, 53, 441–448. [Google Scholar] [CrossRef] [PubMed]
  43. Turner, N.; Hamidi, S.; Ouni, R.; Rico, R.; Henderson, Y.C.; Puche, M.; Alekseev, S.; Colunga-Minutti, J.G.; Zafereo, M.E.; Lai, S.Y.; et al. Emerging therapeutic options for follicular-derived thyroid cancer in the era of immunotherapy. Front. Immunol. 2024, 15, 1369780. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Rajan, N.; Khanal, T.; Ringel, M.D. Progression and dormancy in metastatic thyroid cancer: Concepts and clinical implications. Endocrine 2020, 70, 24–35. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  45. Parameswaran, R.; Brooks, S.; Sadler, G.P. Molecular pathogenesis of follicular cell derived thyroid cancers. Int. J. Surg. 2010, 8, 186–193. [Google Scholar] [CrossRef] [PubMed]
  46. Kondo, T.; Ezzat, S.; Asa, S.L. Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nat. Rev. Cancer 2006, 6, 292–306. [Google Scholar] [CrossRef]
  47. McHenry, C.R.; Phitayakorn, R. Follicular adenoma and carcinoma of the thyroid gland. Oncologist 2011, 16, 585–593. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  48. Panda, S.K.; Patro, B.; Samantaroy, M.R.; Mishra, J.; Mohapatra, K.C.; Meher, R.K. Unusual presentation of follicular carcinoma thyroid with special emphasis on their management. Int. J. Surg. Case Rep. 2014, 5, 408–411. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Omar, B.; Yassir, H.; Youssef, O.; Sami, R.; Larbi, A.R.; Mohamed, R.; Mohamed, M. A rare case of follicular thyroid carcinoma metastasis to the sacral region: A case report with literature review. Int. J. Surg. Case Rep. 2022, 94, 107001. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Aboelnaga, E.M.; Ahmed, R.A. Difference between papillary and follicular thyroid carcinoma outcomes: An experience from Egyptian institution. Cancer Biol. Med. 2015, 12, 53–59. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  51. Parameswaran, R.; Hu, J.S.; En, N.M.; Tan, W.; Yuan, N. Patterns of metastasis in follicular thyroid carcinoma and the difference between early and delayed presentation. Ann. R. Coll. Surg. Engl. 2017, 99, 151–154. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Shen, J.; Yan, M.; Chen, L.; Ou, D.; Yao, J.; Feng, N.; Zhou, X.; Lei, Z.; Xu, D. Prognosis and influencing factors of follicular thyroid cancer. Cancer Med. 2024, 13, e6727. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Hirokawa, M.; Ito, Y.; Kuma, S.; Takamura, Y.; Miya, A.; Kobayashi, K.; Miyauchi, A. Nodal metastasis in well-differentiated follicular carcinoma of the thyroid: Its incidence and clinical significance. Oncol. Lett. 2010, 1, 873–876. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Phitayakorn, R.; McHenry, C.R. Follicular and Hurthle cell carcinoma of the thyroid gland. Surg. Oncol. Clin. 2006, 15, 603–623. [Google Scholar] [CrossRef]
  55. D’Avanzo, A.; Treseler, P.; Ituarte, P.H.G.; Wong, M.; Streja, L.; Greenspan, F.S.; Siperstein, A.E.; Duh, Q.; Clark, O.H. Follicular thyroid carcinoma: Histology and prognosis. Cancer 2004, 100, 1123–1129. [Google Scholar] [CrossRef]
  56. Kaliszewski, K.; Ludwig, M.; Ludwig, B.; Mikuła, A.; Greniuk, M.; Rudnicki, J. Update on the Diagnosis and Management of Medullary Thyroid Cancer: What Has Changed in Recent Years? Cancers 2022, 14, 3643. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Santillan, V.R.; Master, S.R.; Menon, G.; Burns, B. Medullary Thyroid Cancer. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK459354/ (accessed on 10 April 2025).
  58. Matrone, A.; Gambale, C.; Prete, A.; Elisei, R. Sporadic Medullary Thyroid Carcinoma: Towards a Precision Medicine. Front. Endocrinol. 2022, 13, 864253. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  59. Available online: https://www.thyroidcancer.com/thyroid-cancer/medullary#:~:text=Sporadic%20(not%20inherited)%20medullary%20thyroid,effect%20women%20equally%20to%20men (accessed on 10 April 2025).
  60. Gild, M.L.; Clifton-Bligh, R.J.; Wirth, L.J.; Robinson, B.G. Medullary Thyroid Cancer: Updates and Challenges. Endocr. Rev. 2023, 44, 934–946. [Google Scholar] [CrossRef]
  61. Nosé, V. Familial thyroid cancer: A review. Mod. Pathol. 2011, 24, S19–S33. [Google Scholar] [CrossRef] [PubMed]
  62. Jacob, M.; Rowland, D.; Lekarev, O.; Ergun-Longmire, B. Multiple Endocrine Neoplasia in Childhood: An Update on Diagnosis, Screening, Management and Treatment. Endocrines 2022, 3, 76–91. [Google Scholar] [CrossRef]
  63. Yasir, M.; Mulji, N.J.; Kasi, A. Multiple Endocrine Neoplasias Type 2. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK519054/ (accessed on 10 April 2025).
  64. Hyer, S.L.; Newbold, K.; Harmer, C. Familial medullary thyroid cancer: Clinical aspects and prognosis. Eur. J. Surg. Oncol. 2005, 31, 415–419. [Google Scholar] [CrossRef] [PubMed]
  65. Halling, K.C.; Bufill, J.A.; Cotter, M.; Artz, S.A.; Carpenter, A.B.; Schaid, D.; Hartman-Adams, H.; Chang, H.H.; Boustany, M.M.; Fithian, L.; et al. Age-Related Disease Penetrance in a Large Medullary Thyroid Cancer Family with a Codon 609 RET Gene Mutation. Mol. Diagn. 1997, 2, 277–286. [Google Scholar] [CrossRef] [PubMed]
  66. Skinner, M.A.; DeBenedetti, M.K.; Moley, J.F.; Norton, J.A.; Wells, S.A., Jr. Medullary thyroid carcinoma in children with multiple endocrine neoplasia types 2A and 2B. J. Pediatr. Surg. 1996, 31, 177–181. [Google Scholar] [CrossRef] [PubMed]
  67. Rich, T.A.; Feng, L.; Busaidy, N.; Cote, G.J.; Gagel, R.F.; Hu, M.; Jimenez, C.; Lee, J.E.; Perrier, N.; Sherman, S.I.; et al. Prevalence by age and predictors of medullary thyroid cancer in patients with lower risk germline RET proto-oncogene mutations. Thyroid 2014, 24, 1096–1106. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Skinner, M.A.; Moley, J.A.; Dilley, W.G.; Owzar, K.; Debenedetti, M.K.; Wells, S.A., Jr. Prophylactic thyroidectomy in multiple endocrine neoplasia type 2A. N. Engl. J. Med. 2005, 353, 1105–1113. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, K.; Wang, X.; Wei, T.; Li, Z.; Zhu, J.; Chen, Y.W. Well-defined survival outcome disparity across age cutoffs at 45 and 60 for medullary thyroid carcinoma: A long-term retrospective cohort study of 3601 patients. Front. Endocrinol. 2024, 15, 1393904. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Kazakou, P.; Simeakis, G.; Alevizaki, M.; Saltiki, K. Medullary thyroid carcinoma (MTC): Unusual metastatic sites. Endocrinol. Diabetes Metab. Case Rep. 2021, 21, 63. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  71. Limaiem, F.; Kashyap, S.; Naing, P.T.; Mathias, P.M.; Giwa, A.O. Anaplastic Thyroid Cancer. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar] [PubMed]
  72. Lowe, N.M.; Loughran, S.; Slevin, N.J.; Yap, B.K. Anaplastic thyroid cancer: The addition of systemic chemotherapy to radiotherapy led to an observed improvement in survival—A single centre experience and review of the literature. Sci. World J. 2014, 2014, 674583. [Google Scholar] [CrossRef]
  73. O’Neill, J.P.; Shaha, A.R. Anaplastic thyroid cancer. Oral Oncol. 2013, 49, 702–706. [Google Scholar] [CrossRef]
  74. Smallridge, R.C.; Copland, J.A. Anaplastic thyroid carcinoma: Pathogenesis and emerging therapies. Clin. Oncol. 2010, 22, 486–497. [Google Scholar] [CrossRef]
  75. Rao, S.N.; Smallridge, R.C. Anaplastic thyroid cancer: An update. Best Pract. Res. Clin. Endocrinol. Metab. 2023, 37, 101678. [Google Scholar] [CrossRef]
  76. Filetti, S.; Durante, C.; Hartl, D.; Leboulleux, S.; Locati, L.D.; Newbold, K.; Papotti, M.G.; Berruti, A.; ESMO Guidelines Committee. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 1856–1883. [Google Scholar] [CrossRef]
  77. Hassan, M.; Janjua, T.K.; Afridi, H.K.; Zahid, N.A. Anaplastic carcinoma of thyroid gland with widespread soft tissue metastasis: An unusual presentation. BMJ Case Rep. 2017, 2017, bcr2017220793. [Google Scholar] [CrossRef]
  78. Murayama, D.; Yamamoto, Y.; Matsui, A.; Yasukawa, M.; Okamoto, S.; Toda, S.; Iwasaki, H. Lung cavitation in patients with anaplastic thyroid cancer treated with lenvatinib. Gland Surg. 2022, 11, 963–969. [Google Scholar] [CrossRef]
  79. Lee, J.S.; Lee, J.S.; Yun, H.J.; Chang, H.; Kim, S.M.; Lee, Y.S.; Chang, H.S.; Park, C.S. Prognosis of Anaplastic Thyroid Cancer with Distant Metastasis. Cancers 2022, 14, 5784. [Google Scholar] [CrossRef]
  80. Available online: https://my.clevelandclinic.org/health/diseases/23539-anaplastic-thyroid-cancer-atc#:~:text=The%20average%20survival%20rate%20of,alive%20one%20year%20after%20diagnosis (accessed on 10 April 2025).
  81. Moreno, F.; Reyes, C.; Pineda, C.A.; Castellanos, G.; Cálix, F.; Calderón, J.; Vasquez-Bonilla, W.O. Anaplastic thyroid carcinoma with unusual long-term survival: A case report. J. Med. Case Rep. 2022, 16, 39. [Google Scholar] [CrossRef]
  82. Glaser, S.M.; Mandish, S.F.; Gill, B.S.; Balasubramani, G.K.; Clump, D.A.; Beriwal, S. Anaplastic thyroid cancer: Prognostic factors, patterns of care, and overall survival. Head Neck 2016, 38, E2083–E2090. [Google Scholar] [CrossRef] [PubMed]
  83. Maleki, Z.; Hassanzadeh, J.; Ghaem, H. Relationship of modifiable risk factors with the incidence of thyroid cancer: A worldwide study. BMC Res. Notes 2025, 18, 22. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Z.; He, H.; Shan, G.; Lin, Y. Research progress in epidemiology and risk factors of thyroid cancer. China Oncol. 2025, 35, 21–29. [Google Scholar]
  85. Hisan, U.K.; Myung, S.K.; Nguyen, G.V. Associations Between Obesity and Risk of Thyroid Cancer: A Meta-Analysis of Cohort Studies. Nutr. Cancer 2025, 77, 288–298. [Google Scholar] [CrossRef]
  86. Matrone, A.; Ferrari, F.; Santini, F.; Elisei, R. Obesity as a risk factor for thyroid cancer. Curr. Opin. Endocrinol. Diabetes Obes. 2020, 27, 358–363. [Google Scholar] [CrossRef]
  87. Kwon, H.; Han, K.D.; Park, C.Y. Weight change is significantly associated with risk of thyroid cancer: A nationwide population-based cohort study. Sci. Rep. 2019, 9, 1546. [Google Scholar] [CrossRef]
  88. Kim, W.G.; Cheng, S.Y. Mechanisms Linking Obesity and Thyroid Cancer Development and Progression in Mouse Models. Horm. Cancer 2018, 9, 108–116. [Google Scholar] [CrossRef]
  89. Le Moli, R.; Vella, V.; Tumino, D.; Piticchio, T.; Naselli, A.; Belfiore, A.; Frasca, F. Inflammasome activation as a link between obesity and thyroid disorders: Implications for an integrated clinical management. Front. Endocrinol. 2022, 13, 959276. [Google Scholar] [CrossRef]
  90. Flor, L.S.; Anderson, J.A.; Ahmad, N.; Aravkin, A.; Carr, S.; Dai, X.; Gil, G.F.; Hay, S.I.; Malloy, M.J.; McLaughlin, S.A.; et al. Health effects associated with exposure to secondhand smoke: A Burden of Proof study. Nat. Med. 2024, 30, 149–167. [Google Scholar] [CrossRef]
  91. Wang, M.; Gong, W.W.; Lu, F.; He, Q.F.; Hu, R.Y.; Zhong, J.M.; Yu, M. Associations of intensity, duration, cumulative dose, and age at start of smoking, with thyroid cancer in Chinese males: A hospital-based case-control study in Zhejiang Province. Tob. Induc. Dis. 2020, 18, 97. [Google Scholar] [CrossRef]
  92. Shen, Y.; Wang, X.; Wang, L.; Xiong, D.; Wu, C.; Cen, L.; Xie, L.; Li, X. Modifiable risk factors for thyroid cancer: Lifestyle and residence environment. Endokrynol. Pol. 2024, 75, 119–129. [Google Scholar] [CrossRef] [PubMed]
  93. Leko, M.B.; Gunjača, I.; Pleić, N.; Zemunik, T. Environmental Factors Affecting Thyroid-Stimulating Hormone and Thyroid Hormone Levels. Int. J. Mol. Sci. 2021, 22, 6521. [Google Scholar] [CrossRef]
  94. An, S.-Y.; Kim, S.Y.; Oh, D.J.; Min, C.; Sim, S.; Choi, H.G. Obesity is positively related and tobacco smoking and alcohol consumption are negatively related to an increased risk of thyroid cancer. Sci. Rep. 2020, 10, 19279. [Google Scholar] [CrossRef]
  95. Yeo, Y.; Shin, D.W.; Han, K.; Kim, D.; Kim, T.H.; Chun, S.; Jeong, S.M.; Song, Y.M. Smoking, Alcohol Consumption, and the Risk of Thyroid Cancer: A Population-Based Korean Cohort Study of 10 Million People. Thyroid 2022, 32, 440–448. [Google Scholar] [CrossRef]
  96. Friedenreich, C.M.; Ryder-Burbidge, C.; McNeil, J. Physical activity, obesity and sedentary behavior in cancer etiology: Epidemiologic evidence and biologic mechanisms. Mol. Oncol. 2021, 15, 790–800. [Google Scholar] [CrossRef]
  97. Huang, L.; Feng, X.; Yang, W.; Li, X.; Zhang, K.; Feng, S.; Wang, F.; Yang, X. Appraising the Effect of Potential Risk Factors on Thyroid Cancer: A Mendelian Randomization Study. J. Clin. Endocrinol. Metab. 2022, 107, e2783–e2791. [Google Scholar] [CrossRef]
  98. Feng, X.; Wang, F.; Yang, W.; Zheng, Y.; Liu, C.; Huang, L.; Li, L.; Cheng, H.; Cai, H.; Li, X.; et al. Association Between Genetic Risk, Adherence to Healthy Lifestyle Behavior, and Thyroid Cancer Risk. JAMA Netw. Open 2022, 5, e2246311. [Google Scholar] [CrossRef]
  99. Yeo, Y.; Han, K.; Shin, D.W.; Kim, D.; Jeong, S.M.; Chun, S.; Choi, I.Y.; Jeon, K.H.; Kim, T.H. Changes in Smoking, Alcohol Consumption, and the Risk of Thyroid Cancer: A Population-Based Korean Cohort Study. Cancers 2021, 13, 2343. [Google Scholar] [CrossRef]
  100. Liu, Q. Role of exercise on the reduction of cancer development: A mechanistic review from the lncRNA point of view. Clin. Exp. Med. 2025, 25, 77. [Google Scholar] [CrossRef]
  101. Fiore, M.; Cristaldi, A.; Okatyeva, V.; Bianco, S.L.; Conti, G.O.; Zuccarello, P.; Copat, C.; Caltabiano, R.; Cannizzaro, M.; Ferrante, M. Physical Activity and Thyroid Cancer Risk: A Case-Control Study in Catania (South Italy). Int. J. Environ. Res. Public Health 2019, 16, 1428. [Google Scholar] [CrossRef]
  102. Hong, B.S.; Lee, K.P. A systematic review of the biological mechanisms linking physical activity and breast cancer. Phys. Act. Nutr. 2020, 24, 25–31. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, J.; Han, K.; Jung, J.-H.; Ha, J.; Jeong, C.; Heu, J.-Y.; Lee, S.-W.; Lee, J.; Lim, Y.; Kim, M.K.; et al. Physical activity and reduced risk of fracture in thyroid cancer patients after thyroidectomy—Anationwide cohort study. Front. Endocrinol. 2023, 14, 1173781. [Google Scholar] [CrossRef] [PubMed]
  104. Ferrante, M.; Distefano, G.; Distefano, C.; Copat, C.; Grasso, A.; Conti, G.O.; Cristaldi, A.; Fiore, M. Benefits of Physical Activity during and after Thyroid Cancer Treatment on Fatigue and Quality of Life: A Systematic Review. Cancers 2022, 14, 3657. [Google Scholar] [CrossRef] [PubMed]
  105. Luo, J.; Li, H.; Deziel, N.C.; Huang, H.; Zhao, N.; Ma, S.; Ni, X.; Udelsman, R.; Zhang, Y. Genetic susceptibility may modify the association between cell phone use and thyroid cancer: A population-based case-control study in Connecticut. Environ. Res. 2020, 182, 109013. [Google Scholar] [CrossRef]
  106. Carlberg, M.; Koppel, T.; Hedendahl, L.K.; Hardell, L. Is the Increasing Incidence of Thyroid Cancer in the Nordic Countries Caused by Use of Mobile Phones? Int. J. Environ. Res. Public Health 2020, 17, 9129. [Google Scholar] [CrossRef]
  107. Parad, M.T.; Fararouei, M.; Mirahmadizadeh, A.R.; Afrashteh, S. Thyroid cancer and its associated factors: A population-based case-control study. Int. J. Cancer 2021, 149, 514–521. [Google Scholar] [CrossRef] [PubMed]
  108. Saenko, V.; Mitsutake, N. Radiation-Related Thyroid Cancer. Endocr. Rev. 2024, 45, 1–29. [Google Scholar] [CrossRef]
  109. Morton, L.M.; Karyadi, D.M.; Stewart, C.; Bogdanova, T.I.; Dawson, E.T.; Steinberg, M.K.; Dai, J.; Hartley, S.W.; Schonfeld, S.J.; Sampson, J.N.; et al. Radiation-related genomic profile of papillary thyroid carcinoma after the Chernobyl accident. Science 2021, 372, eabg2538. [Google Scholar] [CrossRef]
  110. Suteau, V.; Munier, M.; Briet, C.; Rodien, P. Sex Bias in Differentiated Thyroid Cancer. Int. J. Mol. Sci. 2021, 22, 12992. [Google Scholar] [CrossRef] [PubMed]
  111. Zahedi, A.; Bondaz, L.; Rajaraman, M.; Leslie, W.D.; Jefford, C.; Young, J.E.; Pathak, K.A.; Bureau, Y.; Rachinsky, I.; Badreddine, M.; et al. Risk for Thyroid Cancer Recurrence Is Higher in Men Than in Women Independent of Disease Stage at Presentation. Thyroid 2020, 30, 871–877. [Google Scholar] [CrossRef]
  112. Miranda-Filho, A.; Lortet-Tieulent, J.; Bray, F.; Cao, B.; Franceschi, S.; Vaccarella, S.; Dal Maso, L. Thyroid cancer incidence trends by histology in 25 countries: A population-based study. Lancet Diabetes Endocrinol. 2021, 9, 225–234. [Google Scholar] [CrossRef]
  113. Lam, A.K. Papillary Thyroid Carcinoma: Current Position in Epidemiology, Genomics, and Classification. In Papillary Thyroid Carcinoma; Lam, A.K., Ed.; Humana: New York, NY, USA, 2022. [Google Scholar] [CrossRef]
  114. Fagin, J.A. Genetics of papillary thyroid cancer initiation: Implications for therapy. Trans. Am. Clin. Climatol. Assoc. 2005, 116, 259–269. [Google Scholar]
  115. Sheng, D.; Yu, X.; Li, H.; Zhang, M.; Chen, J. BRAF V600E mutation and the Bethesda System for Reporting Thyroid Cytopathology of fine-needle aspiration biopsy for distinguishing benign from malignant thyroid nodules. Medicine 2021, 100, e27167. [Google Scholar] [CrossRef] [PubMed]
  116. Capezzone, M.; Robenshtok, E.; Cantara, S.; Castagna, M.G. Familial non-medullary thyroid cancer: A critical review. J. Endocrinol. Investig. 2021, 44, 943–950. [Google Scholar] [CrossRef] [PubMed]
  117. Abdullah, M.I.; Junit, S.M.; Ng, K.L.; Jayapalan, J.J.; Karikalan, B.; Hashim, O.H. Papillary Thyroid Cancer: Genetic Alterations and Molecular Biomarker Investigations. Int. J. Med. Sci. 2019, 16, 450–460. [Google Scholar] [CrossRef] [PubMed]
  118. Uno, D.; Endo, K.; Yoshikawa, T.; Hirai, N.; Kobayashi, E.; Nakanishi, Y.; Kondo, S.; Yoshizaki, T. Correlation between gene mutations and clinical characteristics in papillary thyroid cancer: A retrospective analysis of BRAF mutations and RET rearrangements. Thyroid Res. 2024, 17, 21. [Google Scholar] [CrossRef]
  119. Pavlidis, E.; Sapalidis, K.; Chatzinikolaou, F.; Kesisoglou, I. Medullary thyroid cancer: Molecular factors, management and treatment. Rom. J. Morphol. Embryol. 2020, 61, 681–686. [Google Scholar] [CrossRef]
  120. Wu, Z.; Jiang, Y.; Gong, H.; Jiang, T.; Su, A.; Yi, L. Diagnostic value of preoperative systemic inflammatory markers and carcinoembryonic antigen in medullary thyroid carcinoma and the risk factors affecting its prognosis. Gland Surg. 2025, 14, 13–27. [Google Scholar] [CrossRef]
  121. Barletta, J.A.; Nosé, V.; Sadow, P.M. Genomics and Epigenomics of Medullary Thyroid Carcinoma: From Sporadic Disease to Familial Manifestations. Endocr. Pathol. 2021, 32, 35–43. [Google Scholar] [CrossRef]
  122. Mercè, F.; Asla, Q.; Illana, F.J.; Victòria, F.; Javier, H.-L.; Marta, S.; Iglesias, C.; Webb, S.M.; Aulinas, A. A novel likely pathogenic germline variant in CDKN1B in a patient with MEN4 and medullary thyroid cancer. Fam. Cancer 2025, 24, 24. [Google Scholar] [CrossRef]
  123. Macerola, E.; Poma, A.M.; Vignali, P.; Basolo, A.; Ugolini, C.; Torregrossa, L.; Santini, F.; Basolo, F. Molecular Genetics of Follicular-Derived Thyroid Cancer. Cancers 2021, 13, 1139. [Google Scholar] [CrossRef]
  124. Soares, P.; Póvoa, A.A.; Melo, M.; Vinagre, J.; Máximo, V.; Eloy, C.; Cameselle-Teijeiro, J.M.; Sobrinho-Simões, M. Molecular Pathology of Non-familial Follicular Epithelial–Derived Thyroid Cancer in Adults: From RAS/BRAF-like Tumor Designations to Molecular Risk Stratification. Endocr. Pathol. 2021, 32, 44–62. [Google Scholar] [CrossRef]
  125. Ahmadi, S.; Landa, I. The prognostic power of gene mutations in thyroid cancer. Endocr. Connect. 2025, 13, e230297. [Google Scholar] [CrossRef] [PubMed]
  126. Xu, B.; Ghossein, R.A. Advances in Thyroid Pathology: High Grade Follicular Cell-derived Thyroid Carcinoma and Anaplastic Thyroid Carcinoma. Adv. Anat. Pathol. 2023, 30, 3–10. [Google Scholar] [CrossRef] [PubMed]
  127. 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]
  128. Park, S.K.; Ryoo, J.H.; Kim, M.H.; Jung, J.Y.; Jung, Y.S.; Kim, K.N.; Shin, S.; Oh, C.M. Association Between Eight Autoimmune Diseases and Thyroid Cancer: A Nationwide Cohort Study. Thyroid 2024, 34, 206–214. [Google Scholar] [CrossRef]
  129. McLeod, D.S.; Bedno, S.A.; Cooper, D.S.; Hutfless, S.M.; Ippolito, S.; Jordan, S.J.; Matos, P.G.; Neale, R.E.; Sabini, E.; Whiteman, D.C.; et al. Pre-existing Thyroid Autoimmunity and Risk of Papillary Thyroid Cancer: A Nested Case-Control Study of US Active-Duty Personnel. J. Clin. Oncol. 2022, 40, 2578–2587. [Google Scholar] [CrossRef]
  130. Abbasgholizadeh, P.; Naseri, A.; Nasiri, E.; Sadra, V. Is Hashimoto thyroiditis associated with increasing risk of thyroid malignancies? A systematic review and meta-analysis. Thyroid Res. 2021, 14, 26. [Google Scholar] [CrossRef] [PubMed]
  131. Cai, M.; Gou, J. Symptoms and negative emotions in patients with advanced thyroid cancer: A prospective cross-sectional study. BMC Cancer 2024, 24, 1418. [Google Scholar] [CrossRef]
  132. Lincango-Naranjo, E.; Solis-Pazmino, P.; El Kawkgi, O.; Salazar-Vega, J.; Garcia, C.; Ledesma, T.; Rojas, T.; Alvarado-Mafla, B.; Young, G.; Dy, B.; et al. Triggers of thyroid cancer diagnosis: A systematic review and meta-analysis. Endocrine 2021, 72, 644–659. [Google Scholar] [CrossRef]
  133. Nabhan, F.; Dedhia, P.H.; Ringel, M.D. Thyroid cancer, recent advances in diagnosis and therapy. Int. J. Cancer 2021, 149, 984–992. [Google Scholar] [CrossRef]
  134. Soares, L.; Gomes, K.; dos Santos Silva, I. Thyroid Cancer and Quality of Life: A Literature Review. Clin. J. Obstet. Gynecol. 2024, 7, 007–013. [Google Scholar]
  135. Nelim, R. Thyroid Cancer: Current Understanding and Emerging Trends in Diagnosis and Treatment. Rep. Thyroid Res. 2023, 7, 36. [Google Scholar]
  136. Ulisse, S.; Baldini, E.; Lauro, A.; Pironi, D.; Tripodi, D.; Lori, E.; Ferent, I.C.; Amabile, M.I.; Catania, A.; Di Matteo, F.M.; et al. Papillary Thyroid Cancer Prognosis: An Evolving Field. Cancers 2021, 13, 5567. [Google Scholar] [CrossRef] [PubMed]
  137. Ferraz, C. Molecular testing for thyroid nodules: Where are we now? Rev. Endocr. Metab. Disord. 2024, 25, 149–159. [Google Scholar] [CrossRef] [PubMed]
  138. Rajab, M.; Payne, R.J.; Forest, V.-I.; Pusztaszeri, M. Molecular Testing for Thyroid Nodules: The Experience at McGill University Teaching Hospitals in Canada. Cancers 2022, 14, 4140. [Google Scholar] [CrossRef]
  139. Lukyanov, S.A.; Titov, S.E.; Kozorezova, E.S.; Demenkov, P.S.; Veryaskina, Y.A.; Korotovskii, D.V.; Ilyina, T.E.; Vorobyev, S.L.; Zhivotov, V.A.; Bondarev, N.S.; et al. Prediction of the Aggressive Clinical Course of Papillary Thyroid Carcinoma Based on Fine Needle Aspiration Biopsy Molecular Testing. Int. J. Mol. Sci. 2024, 25, 7090. [Google Scholar] [CrossRef]
  140. Belaiche, A.; Morand, G.B.; Turkdogan, S.; Kang, E.S.; Forest, V.-I.; Pusztaszeri, M.P.; Hier, M.P.; Mlynarek, A.M.; Richardson, K.; Sadeghi, N.; et al. Molecular Markers in Follicular and Oncocytic Thyroid Carcinomas: Clinical Application of Molecular Genetic Testing. Curr. Oncol. 2024, 31, 5919–5928. [Google Scholar] [CrossRef]
  141. Livhits, M.J.; Zhu, C.Y.; Kuo, E.J.; Nguyen, D.T.; Kim, J.; Tseng, C.-H.; Leung, A.M.; Rao, J.; Levin, M.; Douek, M.L.; et al. Effectiveness of Molecular Testing Techniques for Diagnosis of Indeterminate Thyroid Nodules: A Randomized Clinical Trial. JAMA Oncol. 2021, 7, 70–77. [Google Scholar] [CrossRef]
  142. Chakrabarty, N.; Mahajan, A.; Basu, S.; D’Cruz, A.K. Comprehensive Review of the Imaging Recommendations for Diagnosis, Staging, and Management of Thyroid Carcinoma. J. Clin. Med. 2024, 13, 2904. [Google Scholar] [CrossRef]
  143. David, E.; Grazhdani, H.; Tattaresu, G.; Pittari, A.; Foti, P.V.; Palmucci, S.; Spatola, C.; Lo Greco, M.C.; Inì, C.; Tiralongo, F.; et al. Thyroid Nodule Characterization: Overview and State of the Art of Diagnosis with Recent Developments, from Imaging to Molecular Diagnosis and Artificial Intelligence. Biomedicines 2024, 12, 1676. [Google Scholar] [CrossRef]
  144. Ali, M.; Al Harbawi, L.; Abed, Z.; Jawhar, N. Neck Masses as The First Presentation of Occult Papillary Thyroid Carcinoma: Case Series. Egypt. J. Hosp. Med. 2023, 90, 465–470. [Google Scholar] [CrossRef]
  145. Spyroglou, A.; Kostopoulos, G.; Tseleni, S.; Toulis, K.; Bramis, K.; Mastorakos, G.; Konstadoulakis, M.; Vamvakidis, K.; Alexandraki, K.I. Hobnail Papillary Thyroid Carcinoma, A Systematic Review and Meta-Analysis. Cancers 2022, 14, 2785. [Google Scholar] [CrossRef] [PubMed]
  146. Dhanani, R.; Faisal, M.; Akram, M.; Shakeel, O.; Zahid, M.T.; Hassan, A.; Hussain, R. Differentiated Thyroid Carcinoma: Distant Metastasis as an Unusual Sole Initial Manifestation. Turk. Arch. Otorhinolaryngol. 2021, 59, 188–192. [Google Scholar] [CrossRef] [PubMed]
  147. Li, D.; Li, J.; Zhou, J.; Xiao, Q.; Gao, H. Metastatic papillary thyroid carcinoma with no primary tumor in the thyroid gland: A case report and review of literature. Transl. Cancer Res. 2022, 11, 299–305. [Google Scholar] [CrossRef]
  148. Shakir, M.K.M.; Spiro, A.J.; Mai, V.Q.; Hoang, T.D. Diarrhea as an Initial Presentation in Patients with Medullary Thyroid Cancer: Delaying the Diagnosis. Case Rep. Gastroenterol. 2020, 14, 391–401. [Google Scholar] [CrossRef] [PubMed]
  149. Pelizzo, M.R.; Mazza, E.I.; Mian, C.; Boschin, I.M. Medullary thyroid carcinoma. Expert Rev. Anticancer. Ther. 2023, 23, 943–957. [Google Scholar] [CrossRef]
  150. Pawęska, W.; Dominik, H.; Dominik, B. Anaplastic thyroid cancer with life-threatening symptoms in an older female—A case report. J. Educ. Health Sport. 2023, 41, 117–122. [Google Scholar] [CrossRef]
  151. Cha, Y.J.; Koo, J.S. Next-generation sequencing in thyroid cancer. J Transl Med. 2016, 14, 322. [Google Scholar] [CrossRef]
  152. Newfield, R.S.; Jiang, W.; Sugganth, D.X.; Hantash, F.M.; Lee, E. Newbury RO. Mutational analysis using next generation se-quencing in pediatric thyroid cancer reveals BRAF and fusion oncogenes are common. Int J PediatrOtorhinolaryngol. 2022, 157, 111121. [Google Scholar] [CrossRef]
  153. Bellevicine, C.; Migliatico, I.; Sgariglia, R.; Nacchio, M.; Vigliar, E.; Pisapia, P.; Iaccarino, A.; Bruzzese, D.; Fonderico, F.; Salvatore, D.; et al. Evaluation of BRAF, RAS, RET/PTC, and PAX8/PPARg alterations in different Bethesda diagnostic cate-gories: A multicentric prospective study on the validity of the 7-gene panel test in 1172 thyroid FNAs deriving from different hospitals in South Italy. Cancer Cytopathol. 2020, 128, 107–118. [Google Scholar] [CrossRef]
  154. Qiu, J.; Zhang, W.; Xia, Q.; Liu, F.; Li, L.; Zhao, S.; Gao, X.; Zang, C.; Ge, R.; Sun, Y. RNA sequencing identifies crucial genes in papil-lary thyroid carcinoma (PTC) progression. Exp. Mol. Pathol. 2016, 100, 151–159. [Google Scholar] [CrossRef]
  155. Patel, S.G.; Carty, S.E.; Lee, A.J. Molecular Testing for Thyroid Nodules Including Its Interpretation and Use in Clinical Practice. Ann Surg Oncol. 2021, 28, 8884–8891. [Google Scholar] [CrossRef] [PubMed]
  156. Moore, A.; Bar, Y.; Maurice-Dror, C.; Finkel, I.; Goldvaser, H.; Dudnik, E.; Goldstein, D.A.; Gordon, N.; Billan, S.; Gutfeld, O.; et al. Next-generation sequencing in thyroid cancers: Do targetable alterations lead to a therapeutic advantage?: A multicenter experience. Medicine 2021, 100, e26388. [Google Scholar] [CrossRef] [PubMed]
  157. Pacini, F.; Castagna, M.G. Approach to and treatment of differentiated thyroid carcinoma. Med. Clin. N. Am. 2012, 96, 369–383. [Google Scholar] [CrossRef] [PubMed]
  158. Giovanella, L.; D’Aurizio, F.; Petranović Ovčariček, P.; Görges, R. Diagnostic, Theranostic and Prognostic Value of Thyroglobulin in Thyroid Cancer. J. Clin. Med. 2024, 13, 2463. [Google Scholar] [CrossRef]
  159. Lin, J.-D. Thyroglobulin and human thyroid cancer. Clin. Chim. Acta 2008, 388, 15–21. [Google Scholar] [CrossRef]
  160. Berlińska, A.; Świątkowska-Stodulska, R. Clinical use of thyroglobulin: Not only thyroid cancer. Endocrine 2024, 84, 786–799. [Google Scholar] [CrossRef]
  161. Lu, Y.; Zhao, H.; Liu, C.; Kuang, Z.; Li, X. The role of preoperative serum thyroglobulin in the diagnosis and treatment of differentiated thyroid cancer: A systematic review and meta-analysis. Front. Oncol. 2024, 14, 1426785. [Google Scholar] [CrossRef]
  162. Prpić, M.; Franceschi, M.; Romić, M.; Jukić, T.; Kusić, Z. Thyroglobulin as a tumor marker in differentiated thyroid cancer—Clinical considerations. Acta Clin. Croat. 2018, 57, 518–527. [Google Scholar] [CrossRef]
  163. Li, S.; Ren, C.; Gong, Y.; Ye, F.; Tang, Y.; Xu, J.; Guo, C.; Huang, J. The Role of Thyroglobulin in Preoperative and Postoperative Evaluation of Patients with Differentiated Thyroid Cancer. Front. Endocrinol. 2022, 13, 872527. [Google Scholar] [CrossRef]
  164. Verrienti, A.; Pecce, V.; Grani, G.; Del Gatto, V.; Barp, S.; Maranghi, M.; Giacomelli, L.; Di Gioia, C.; Biffoni, M.; Filetti, S.; et al. Serum microRNA-146a-5p and microRNA-221-3p as potential clinical biomarkers for papillary thyroid carcinoma. J. Endocrinol. Investig. 2025, 48, 619–631. [Google Scholar] [CrossRef]
  165. Hou, F.; Zhu, Y.; Zhao, H.; Cai, H.; Wang, Y.; Peng, X.; Lu, L.; He, R.; Hou, Y.; Li, Z.; et al. Development and validation of an interpretable machine learning model for predicting the risk of distant metastasis in papillary thyroid cancer: A multicenter study. eClinicalMedicine 2024, 77, 102913. [Google Scholar] [CrossRef] [PubMed]
  166. Hataya, Y.; Fujishima, Y.; Fujimoto, K.; Iwakura, T.; Matsuoka, N. Association between serum thyroglobulin levels and glycemic control in patients with thyroid cancer after radioiodine therapy. Endocr. J. 2025, 72, 637–643, epub ahead of printing. [Google Scholar] [CrossRef] [PubMed]
  167. Hou, Y.; Lin, B.; Xu, T.; Jiang, J.; Luo, S.; Chen, W.; Chen, X.; Wang, Y.; Liao, G.; Wang, J.; et al. The neurotransmitter calcitonin gene-related peptide shapes an immunosuppressive microenvironment in medullary thyroid cancer. Nat. Commun. 2024, 15, 5555. [Google Scholar] [CrossRef] [PubMed]
  168. Wells, S.A.; Asa, S.L.; Dralle, H.; Elisei, R.; Evans, D.B.; Gagel, R.F.; Lee, N.; Machens, A.; Moley, J.F.; Pacini, F.; et al. American Thyroid Association Guidelines Task Force on Medullary Thyroid Carcinoma. Revised American Thyroid Association guidelines for the management of medullary thyroid carcinoma. Thyroid 2015, 25, 567–610. [Google Scholar] [CrossRef]
  169. Jeong, I.Y.; Yun, H.J.; Kim, S.M.; Park, Y. Diagnostic Performance of Preoperative Calcitonin and Procalcitonin Tests for Differential Diagnosis of Medullary Thyroid Cancer. Diagnostics 2024, 14, 1809. [Google Scholar] [CrossRef]
  170. Baylin, S.B.; Bailey, A.L.; Hsu, T.H.; Foster, G.V. Degradation of human calcitonin in human plasmas. Metabolism 1977, 26, 1345–1354. [Google Scholar] [CrossRef]
  171. Faggiano, A.; Milone, F.; Ramundo, V.; Chiofalo, M.G.; Ventre, I.; Giannattasio, R.; Severino, R.; Lombardi, G.; Colao, A.; Pezzullo, L. A decrease of calcitonin serum concentrations less than 50 percent 30 minutes after thyroid surgery suggests incomplete C-cell tumor tissue removal. J. Clin. Endocrinol. Metab. 2010, 95, E32–E36. [Google Scholar] [CrossRef]
  172. Modigliani, E.; Cohen, R.; Campos, J.M.; Conte-Devolx, B.; Maes, B.; Boneu, A.; Schlumberger, M.; Bigorgne, J.C.; Dumontier, P.; Leclerc, L.; et al. Prognostic factors for survival and for biochemical cure in medullary thyroid carcinoma: Results in 899 patients. The GETC Study Group. Groupe d’étude des tumeurs à calcitonine. Clin. Endocrinol. 1998, 48, 265–273. [Google Scholar] [CrossRef]
  173. Réti, Z.; Tabák, Á.G.; Garami, M.; Kalina, I.; Kiss, G.; Sápi, Z.; Tóth, M.; Tőke, J. Spontaneous and Treatment-Related Changes of Serum Calcitonin in Medullary Thyroid Cancer: Long-Term Experience in a Patient with Multiple Endocrine Neoplasia Type 2B. JCO Precis. Oncol. 2024, 8, e2300675. [Google Scholar] [CrossRef]
  174. Jiao, Z.; Wu, T.; Jiang, M.; Jiang, S.; Jiang, K.; Peng, J.; Luo, G.; Yu, Y.; Chen, W.; Yang, A. Early postoperative calcitonin-to-preoperative calcitonin ratio as a predictive marker for structural recurrence in sporadic medullary thyroid cancer: A retrospective study. Front. Endocrinol. 2022, 13, 1094242. [Google Scholar] [CrossRef]
  175. Verbeek, H.H.; de Groot, J.W.B.; Sluiter, W.J.; Muller Kobold, A.C.; van den Heuvel, E.R.; Plukker, J.T.; Links, T.P. Calcitonin testing for detection of medullary thyroid cancer in people with thyroid nodules. Cochrane Database Syst. Rev. 2020, 3, CD010159. [Google Scholar] [CrossRef] [PubMed]
  176. Shepet, K.; Alhefdhi, A.; Lai, N.; Mazeh, H.; Sippel, R.; Chen, H. Hereditary medullary thyroid cancer: Age-appropriate thyroidectomy improves disease-free survival. Ann. Surg. Oncol. 2013, 20, 1451–1455. [Google Scholar] [CrossRef] [PubMed]
  177. Desai, S.; Guddati, A.K. Carcinoembryonic Antigen, Carbohydrate Antigen 19-9, Cancer Antigen 125, Prostate-Specific Antigen and Other Cancer Markers: A Primer on Commonly Used Cancer Markers. World J. Oncol. 2023, 14, 4–14. [Google Scholar] [CrossRef]
  178. Wang, B.; Huang, J.; Chen, L. Management of medullary thyroid cancer based on variation of carcinoembryonic antigen and calcitonin. Front. Endocrinol. 2024, 15, 1418657. [Google Scholar] [CrossRef]
  179. Chen, L.; Zhao, K.; Li, F.; He, X. Medullary Thyroid Carcinoma with Elevated Serum CEA and Normal Serum Calcitonin After Surgery: A Case Report and Literature Review. Front. Oncol. 2020, 10, 526716. [Google Scholar] [CrossRef]
  180. Passos, I.; Stefanidou, E.; Meditskou-Eythymiadou, S.; Mironidou-Tzouveleki, M.; Manaki, V.; Magra, V.; Laskou, S.; Mantalovas, S.; Pantea, S.; Kesisoglou, I.; et al. A Review of the Significance in Measuring Preoperative and Postoperative Carcinoembryonic Antigen (CEA) Values in Patients with Medullary Thyroid Carcinoma (MTC). Medicina 2021, 57, 609. [Google Scholar] [CrossRef]
  181. Zhang, D.; Liang, N.; Sun, H.; Frattini, F.; Sui, C.; Yang, M.; Wang, H.; Dionigi, G. Critically evaluated key points on hereditary medullary thyroid carcinoma. Front. Endocrinol. 2024, 15, 1412942. [Google Scholar] [CrossRef] [PubMed]
  182. Mendelsohn, G.; Wells, S.A., Jr.; Baylin, S.B. Relationship of tissue carcinoembryonic antigen and calcitonin to tumor virulence in medullary thyroid carcinoma. An immunohistochemical study in early, localized, and virulent disseminated stages of disease. Cancer 1984, 54, 657–662. [Google Scholar] [CrossRef]
  183. Shaghaghi, A.; Salari, A.; Jalaeefar, A.; Shirkhoda, M. Management of lymph nodes in medullary thyroid carcinoma: A review. Ann. Med. Surg. 2022, 81, 104538. [Google Scholar] [CrossRef]
  184. Zi-Yang, Y.; Kaixun, Z.; Dongling, L.; Zhou, Y.; Chengbin, Z.; Jimei, C.; Caojin, Z. Carcinoembryonic antigen levels are increased with pulmonary output in pulmonary hypertension due to congenital heart disease. J. Int. Med. Res. 2020, 48, 300060520964378. [Google Scholar] [CrossRef]
  185. Dilek, O.N.; Arslan Kahraman, D.İ.; Kahraman, G. Carcinoembryonic antigen in the diagnosis, treatment, and follow-up of focal liver lesions. World J. Gastrointest. Surg. 2024, 16, 999–1007. [Google Scholar] [CrossRef] [PubMed]
  186. Turkdogan, S.; Forest, V.I.; Hier, M.P.; Tamilia, M.; Florea, A.; Payne, R.J. Carcinoembryonic antigen levels correlated with advanced disease in medullary thyroid cancer. J. Otolaryngol. Head Neck Surg. 2018, 47, 55. [Google Scholar] [CrossRef] [PubMed]
  187. Kotwal, A.; Erickson, D.; Geske, J.R.; Hay, I.D.; Castro, M.R. Predicting Outcomes in Sporadic and Hereditary Medullary Thyroid Carcinoma over Two Decades. Thyroid 2021, 31, 616–626. [Google Scholar] [CrossRef] [PubMed]
  188. Kankanala, V.L.; Zubair, M.; Mukkamalla, S.K.R. Carcinoembryonic Antigen. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  189. Pishdad, R.; Vahidi Rad, M.; Cespedes, L. A Benign Medullary Thyroid Cancer. Cureus 2022, 14, e21038. [Google Scholar] [CrossRef]
  190. Kaya, F.; Alsafdi, T. Elevated Procalcitonin Levels can Occur in Bacterial Infections and also in Medullary Thyroid Carcinoma. Eur. J. Case Rep. Intern. Med. 2024, 11, 004679. [Google Scholar] [CrossRef]
  191. Huang, J.; Zu, Y.; Zhang, L.; Cui, W. Progress in Procalcitonin Detection Based on Immunoassay. Research 2024, 7, 0345. [Google Scholar] [CrossRef]
  192. Lisieska-Żołnierczyk, S.; Gajęcka, M.; Zielonka, Ł.; Dąbrowski, M.; Gajęcki, M.T. Blood levels of zearalenone, thyroid-stimulating hormone, and thyroid hormones in patients with colorectal cancer. Toxicon 2024, 251, 108125. [Google Scholar] [CrossRef]
  193. Adıgüzel Akman, Ö.; Esnafoglu, E. Evaluation of procalcitonin and C-reactive protein levels in children with autism spectrum disorder and attention deficit hyperactivity disorder. Int. J. Dev. Disabil. 2023, 71, 159–167. [Google Scholar] [CrossRef]
  194. Kiriakopoulos, A.; Giannakis, P.; Menenakos, E. Calcitonin: Current concepts and differential diagnosis. Ther. Adv. Endocrinol. Metab. 2022, 13, 20420188221099344. [Google Scholar] [CrossRef]
  195. Jandric, M.; Zlojutro, B.; Momcicevic, D.; Dragic, S.; Topolovac, S.; Kovacevic, T.; Kovacevic, P. Incidental diagnosis of medullary thyroid microcarcinoma in COVID-19 patient with elevated procalcitonin levels. Arch. Clin. Cases 2024, 11, 98–101. [Google Scholar] [CrossRef]
  196. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef] [PubMed]
  197. Censi, S.; Manso, J.; Benvenuti, T.; Piva, I.; Iacobone, M.; Mondin, A.; Torresan, F.; Basso, D.; Crivellari, G.; Zovato, S.; et al. The role of procalcitonin in the follow-up of medullary thyroid cancer. Eur. Thyroid J. 2023, 12, e220161. [Google Scholar] [CrossRef] [PubMed]
  198. Bucci, I.; Di Dalmazi, G.; Giuliani, C.; Russo, P.; Ciappini, B.; Amatetti, C.; Guarino, P.; Napolitano, G. Advanced medullary thyroid carcinoma uncovered by persistently elevated procalcitonin in a patient with COVID-19. Endocrinol. Diabetes Metab. Case Rep. 2024, 2024, 24–0052. [Google Scholar] [CrossRef]
  199. Saha, A.; Mukhopadhyay, M.; Paul, S.; Bera, A.; Bandyopadhyay, T. Incidental diagnosis of medullary thyroid carcinoma due to persistently elevated procalcitonin in a patient with COVID-19 pneumonia: A case report. World J. Clin. Cases 2022, 10, 7171–7177. [Google Scholar] [CrossRef]
  200. Shahzad, H.; Khokhar, A.J.; Ibrahim, S.; Farooq, M.U.; Rashid, R.; Raheem, H.U.; Singh, G. High Procalcitonin Does Not Always Indicate a Bacterial Infection. Cureus 2024, 16, e72274. [Google Scholar] [CrossRef]
  201. Maruna, P.; Nedelníková, K.; Gürlich, R. Physiology and genetics of procalcitonin. Physiol. Res. 2000, 49, S57–S61. [Google Scholar]
  202. Wang, X.; Wu, Z.; Liu, Y.; Wu, C.; Jiang, J.; Hashimoto, K.; Zhou, X. The role of thyroid-stimulating hormone in regulating lipid metabolism: Implications for body-brain communication. Neurobiol. Dis. 2024, 201, 106658. [Google Scholar] [CrossRef]
  203. Pirahanchi, Y.; Toro, F.; Jialal, I. Physiology, Thyroid Stimulating Hormone. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  204. Won, H.R.; Jeon, E.; Chang, J.W.; Kang, Y.E.; Song, K.; Kim, S.W.; Lim, D.M.; Ha, T.K.; Chung, K.W.; Kim, H.J.; et al. Is Maintaining Thyroid-Stimulating Hormone Effective in Patients Undergoing Thyroid Lobectomy for Low-Risk Differentiated Thyroid Cancer? A Systematic Review and Meta-Analysis. Cancers 2022, 14, 1470. [Google Scholar] [CrossRef]
  205. Xiang, Y.; Xu, Y.; Bhandari, A.; Sindan, N.; Hirachan, S.; Yang, Q.; Guo, G.; Shen, Y. Serum TSH levels are associated with postoperative recurrence and lymph node metastasis of papillary thyroid carcinoma. Am. J. Transl. Res. 2021, 13, 6108–6116. [Google Scholar] [PubMed]
  206. Yan, C.; Sun, J.; He, X.; Jia, L. An age-and sex-matched postoperative therapy should be required in thyroid papillary carcinoma. Front. Endocrinol. 2024, 15, 1339191. [Google Scholar] [CrossRef]
  207. Sugitani, I.; Fujimoto, Y. Effect of postoperative thyrotropin suppressive therapy on bone mineral density in patients with papillary thyroid carcinoma: A prospective controlled study. Surgery 2011, 150, 1250–1257. [Google Scholar] [CrossRef] [PubMed]
  208. Haymart, M.R.; Esfandiari, N.H.; Stang, M.T.; Sosa, J.A. Controversies in the Management of Low-Risk Differentiated Thyroid Cancer. Endocr. Rev. 2017, 38, 351–378. [Google Scholar] [CrossRef] [PubMed]
  209. Mazzaferri, E.L.; Young, R.L.; Oertel, J.E.; Kemmerer, W.T.; Page, C.P. Papillary thyroid carcinoma: The impact of therapy in 576 patients. Medicine 1977, 56, 171–196. [Google Scholar] [CrossRef]
  210. Zhou, W.; Brumpton, B.; Kabil, O.; Gudmundsson, J.; Thorleifsson, G.; Weinstock, J.; Zawistowski, M.; Nielsen, J.B.; Chaker, L.; Medici, M.; et al. G.W.A.S of thyroid stimulating hormone highlights pleiotropic effects and inverse association with thyroid cancer. Nat. Commun. 2020, 11, 3981. [Google Scholar] [CrossRef]
  211. Zhang, S.; Niu, S.; Zhou, L. Gender, FT4 levels, T stage, and BMI as predictors of TSH levels in thyroid cancer patients. Front. Endocrinol. 2025, 16, 1422464. [Google Scholar] [CrossRef]
  212. Zhu, M.; Gao, Y.; Zhu, K.; Yuan, Y.; Bai, H.; Meng, L. Exosomal miRNA as biomarker in cancer diagnosis and prognosis: A review. Medicine 2024, 103, e40082. [Google Scholar] [CrossRef] [PubMed]
  213. Kazlauskiene, M.; Klimaite, R.; Kondrotiene, A.; Dauksa, A.; Dauksiene, D.; Verkauskiene, R.; Zilaitiene, B. Plasma miRNA-146b-3p, -222-3p, -221-5p, -21a-3p Expression Levels and TSHR Methylation: Diagnostic Potential and Association with Clinical and Pathological Features in Papillary Thyroid Cancer. Int. J. Mol. Sci. 2024, 25, 8412. [Google Scholar] [CrossRef]
  214. Cabané, P.; Correa, C.; Bode, I.; Aguilar, R.; Elorza, A.A. Biomarkers in Thyroid Cancer: Emerging Opportunities from Non-Coding RNAs and Mitochondrial Space. Int. J. Mol. Sci. 2024, 25, 6719. [Google Scholar] [CrossRef]
  215. Armos, R.; Bojtor, B.; Papp, M.; Illyes, I.; Lengyel, B.; Kiss, A.; Szili, B.; Tobias, B.; Balla, B.; Piko, H.; et al. MicroRNA Profiling in Papillary Thyroid Cancer. Int. J. Mol. Sci. 2024, 25, 9362. [Google Scholar] [CrossRef]
  216. Gayosso-Gómez, L.V.; Ortiz-Quintero, B. Circulating MicroRNAs in Blood and Other Body Fluids as Biomarkers for Diagnosis, Prognosis, and Therapy Response in Lung Cancer. Diagnostics 2021, 11, 421. [Google Scholar] [CrossRef]
  217. Calin, G.A.; Croce, C.M. MicroRNA signatures in human cancers. Nat. Rev. Cancer 2006, 6, 857–866. [Google Scholar] [CrossRef] [PubMed]
  218. Zhang, P.; Guan, L.; Sun, W.; Zhang, Y.; Du, Y.; Yuan, S.; Cao, X.; Yu, Z.; Jia, Q.; Zheng, X.; et al. Targeting miR-31 represses tumourigenesis and dedifferentiation of BRAFV600E-associated thyroid carcinoma. Clin. Transl. Med. 2024, 14, e1694. [Google Scholar] [CrossRef] [PubMed]
  219. Shen, Y.; Xie, R.; Chen, Y.; Han, X.; Li, X.E. Diagnostic value of microRNA-129-5p and TSH combination for papillary thyroid cancer with cervical lymph node metastasis. Int. J. Biol. Markers 2025, 40, 46–54. [Google Scholar] [CrossRef]
  220. de Matos, M.L.G.; Pinto, M.; Gonçalves, A.; Canberk, S.; Bugalho, M.J.M.; Soares, P. Insights in biomarkers complexity and routine clinical practice for the diagnosis of thyroid nodules and cancer. PeerJ 2025, 13, e18801. [Google Scholar] [CrossRef]
  221. Silaghi, C.A.; Lozovanu, V.; Silaghi, H.; Georgescu, R.D.; Pop, C.; Dobrean, A.; Georgescu, C.E. The Prognostic Value of MicroRNAs in Thyroid Cancers-A Systematic Review and Meta-Analysis. Cancers 2020, 12, 2608. [Google Scholar] [CrossRef]
  222. Yan, M.; Li, X.; Tong, D.; Han, C.; Zhao, R.; He, Y.; Jin, X. miR-136 suppresses tumor invasion and metastasis by targeting RASAL2 in triple-negative breast cancer. Oncol. Rep. 2016, 36, 65–71. [Google Scholar] [CrossRef]
  223. Chen, X.; Huang, Z.; Chen, R. Microrna-136 promotes proliferation and invasion ingastric cancer cells through Pten/Akt/P-Akt signaling pathway. Oncol. Lett. 2018, 15, 4683–4689. [Google Scholar] [CrossRef]
  224. Yip, L.; Kelly, L.; Shuai, Y.; Armstrong, M.J.; Nikiforov, Y.E.; Carty, S.E.; Nikiforova, M.N. MicroRNA signature distinguishes the degree of aggressiveness of papillary thyroid carcinoma. Ann. Surg. Oncol. 2011, 18, 2035–2041. [Google Scholar] [CrossRef] [PubMed]
  225. Galuppini, F.; Censi, S.; Moro, M.; Carraro, S.; Sbaraglia, M.; Iacobone, M.; Fassan, M.; Mian, C.; Pennelli, G. MicroRNAs in Medullary Thyroid Carcinoma: A State of the Art Review of the Regulatory Mechanisms and Future Perspectives. Cells 2021, 10, 955. [Google Scholar] [CrossRef]
  226. Tisell, L.E.; Oden, A.; Muth, A.; Altiparmak, G.; Mõlne, J.; Ahlman, H.; Nilsson, O. The Ki67 index a prognostic marker in medullary thyroid carcinoma. Br. J. Cancer 2003, 89, 2093–2097. [Google Scholar] [CrossRef]
  227. Durante, C.; Puxeddu, E.; Ferretti, E.; Morisi, R.; Moretti, S.; Bruno, R.; Barbi, F.; Avenia, N.; Scipioni, A.; Verrienti, A.; et al. BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism. J. Clin. Endocrinol. Metab. 2007, 92, 2840–2843. [Google Scholar] [CrossRef] [PubMed]
  228. Li, X.; Abdel-Mageed, A.B.; Kandil, E. BRAF mutation in papillary thyroid carcinoma. Int. J. Clin. Exp. Med. 2012, 5, 310–315. [Google Scholar] [PubMed]
  229. Li, Z.; Song, L.; Yang, Y.; Zhao, Y.; Ma, S. Mannose enhances anti-tumor effect of PLX4032 in anaplastic thyroid cancer. Endocr.-Relat. Cancer 2025, 32, e240209. [Google Scholar] [CrossRef]
  230. Nagano, H.; Matsumoto, H.; Ando, Y.; Nakajo, M.; Yamashita, M. Mutations in Anaplastic Thyroid Carcinoma: An Analysis of the Japanese National Genomic Database. Laryngoscope Investig. Otolaryngol. 2025, 10, e70110. [Google Scholar] [CrossRef]
  231. Nguyen, L.T.T.; Thompson, E.K.; Bhimani, N.; Duong, M.C.; Nguyen, H.G.; Bullock, M.; Gild, M.L.; Glover, A. Prognostic Significance of Key Molecular Markers in Thyroid Cancer: A Systematic Literature Review and Meta-Analysis. Cancers 2025, 17, 939. [Google Scholar] [CrossRef]
  232. Tang, H.; Guo, D.; Yang, B.; Huang, S.H. Location based BRAF V600E mutation status and dimension patterns of sporadic thyroid nodules: A population-based study. BMC Cancer 2025, 25, 406. [Google Scholar] [CrossRef]
  233. Instrum, R.; Swartzwelder, C.E.; Ghossein, R.A.; Xu, B.; Givi, B.; Wong, R.J.; Untch, B.R.; Morris, L.G.T. Clinical and Pathologic Characteristics of Cytologically Indeterminate Thyroid Nodules with Non-V600E BRAF Alterations. Cancers 2025, 17, 741. [Google Scholar] [CrossRef] [PubMed]
  234. Mannino, D.; Basilotta, R.; De Luca, F.; Casili, G.; Esposito, E.; Paterniti, I. KRAS-SOS-1 Inhibition as New Pharmacological Target to Counteract Anaplastic Thyroid Carcinoma (ATC). Int. J. Mol. Sci. 2025, 26, 2579. [Google Scholar] [CrossRef]
  235. Bikas, A.; Ahmadi, S.; Pappa, T.; Marqusee, E.; Wong, K.; Nehs, M.A.; Cho, N.L.; Haase, J.; Doherty, G.M.; Sehgal, K.; et al. Additional Oncogenic Alterations in RAS-Driven Differentiated Thyroid Cancers Associate with Worse Clinicopathologic Outcomes. Clin. Cancer. Res. 2023, 29, 2678–2685. [Google Scholar] [CrossRef]
  236. Xiong, S.; Liu, S.; Zhang, W.; Zeng, C.; Liao, D.; Tang, T.; Wang, S.; Guo, Y. Annotation-free genetic mutation estimation of thyroid cancer using cytological slides from multi-centers. Diagn. Pathol. 2025, 20, 22. [Google Scholar] [CrossRef]
  237. McGoron, A.J.; Garcia, J.M.; Uluvar, B.; Gulec, S.A. Thyroid differentiation profile for differentiated thyroid cancer. Endocr. Oncol. 2025, 5, e240072. [Google Scholar] [CrossRef] [PubMed]
  238. Sabi, E.M. The role of genetic and epigenetic modifications as potential biomarkers in the diagnosis and prognosis of thyroid cancer. Front. Oncol. 2024, 14, 1474267. [Google Scholar] [CrossRef] [PubMed]
  239. Nikiforov, Y.E. Genetic alterations involved in the transition from well-differentiated to poorly differentiated and anaplastic thyroid carcinomas. Endocr. Pathol. 2004, 15, 319–327. [Google Scholar] [CrossRef] [PubMed]
  240. Burkhardt, A.; Smith, C.L.; Singh, R.R. Validation of a Compact and Self-Contained Pyrosequencing Platform for Clinical Screening of RAS Mutations in Thyroid Cancers. Diagnostics 2025, 15, 390. [Google Scholar] [CrossRef]
  241. Riccio, I.; Laforteza, A.; Landau, M.B.; Hussein, M.H.; Linhuber, J.; Staav, J.; Issa, P.P.; Toraih, E.A.; Kandil, E. Decoding RAS mutations in thyroid cancer: A meta-analysis unveils specific links to distant metastasis and increased mortality. Am. J. Otolaryngol. 2025, 46, 104570. [Google Scholar] [CrossRef]
  242. Tsou, P.; Wu, C.J. Classifying driver mutations of papillary thyroid carcinoma on whole slide image: An automated workflow applying deep convolutional neural network. Front. Endocrinol. 2024, 15, 1395979. [Google Scholar] [CrossRef]
  243. Santoro, M.; Carlomagno, F. Central role of RET in thyroid cancer. Cold Spring Harb. Perspect. Biol. 2013, 5, a009233. [Google Scholar] [CrossRef]
  244. Nikiforov, Y.E. RET/PTC rearrangement in thyroid tumors. Endocr. Pathol. 2002, 13, 3–16. [Google Scholar] [CrossRef]
  245. Khonrak, T.; Watcharadetwittaya, S.; Chamgramol, Y.; Intarawichian, P.; Deenonpoe, R. RET rearrangements are relevant to histopathologic subtypes and clinicopathological features in Thai papillary thyroid carcinoma patients. Pathol. Oncol. Res. 2023, 29, 1611138. [Google Scholar] [CrossRef]
  246. Liu, Y.; Zhu, J.; Zhou, S.; Hou, Y.; Yan, Z.; Ao, X.; Wang, P.; Zhou, L.; Chen, H.; Liang, X.; et al. Low-dose ionizing radiation-induced RET/PTC1 rearrangement via the non-homologous end joining pathway to drive thyroid cancer. MedComm 2024, 5, e690. [Google Scholar] [CrossRef]
  247. Zhang, J.; Chen, Y.H.; Lu, Q. Pro-oncogenic and anti-oncogenic pathways: Opportunities and challenges of cancer therapy. Future Oncol. 2010, 6, 587–603. [Google Scholar] [CrossRef]
  248. Zhang, W.; Lin, S.; Wang, Z.; Zhang, W.; Xing, M. Coexisting RET/PTC and TERT Promoter Mutation Predict Poor Prognosis but Effective RET and MEK Targeting in Thyroid Cancer. J. Clin. Endocrinol. Metab. 2024, 109, 3166–3175. [Google Scholar] [CrossRef]
  249. Luciano, N.; Orlandella, F.M.; Braile, M.; Cavaliere, C.; Aiello, M.; Franzese, M.; Salvatore, G. Association of radiomic features with genomic signatures in thyroid cancer: A systematic review. J. Transl. Med. 2024, 22, 1088. [Google Scholar] [CrossRef] [PubMed]
  250. Nikita, M.E.; Jiang, W.; Cheng, S.M.; Hantash, F.M.; McPhaul, M.J.; Newbury, R.O.; Phillips, S.A.; Reitz, R.E.; Waldman, F.M.; Newfield, R.S. Mutational Analysis in Pediatric Thyroid Cancer and Correlations with Age, Ethnicity, and Clinical Presentation. Thyroid 2016, 26, 227–234. [Google Scholar] [CrossRef] [PubMed]
  251. Cendra, A.S.; Pekarek, L.; Pedrejón, B.I.; Bernier, L.; Cervantes, E.D.R.; Cendra, C.S.; Cassinello, J.; López-Gonzalez, L.; Mañez, F.; Carrero, C.B.; et al. New Horizons of Biomarkers in Metastatic Thyroid Cancer. J. Cancer 2025, 16, 241–264. [Google Scholar] [CrossRef]
  252. Sapio, M.R.; Guerra, A.; Marotta, V.; Campanile, E.; Formisano, R.; Deandrea, M.; Motta, M.; Limone, P.P.; Fenzi, G.; Rossi, G.; et al. High growth rate of benign thyroid nodules bearing RET/PTC rearrangements. J. Clin. Endocrinol. Metab. 2011, 96, E916–E919. [Google Scholar] [CrossRef]
  253. Ashwini, B.R.; Nirmala, C.; Natarajan, M.; Biligi, D.S. A study to evaluate association of nuclear grooving in benign thyroid lesions with RET/PTC1 and RET/PTC3 gene translocation. Thyroid Res. 2023, 16, 21. [Google Scholar] [CrossRef]
  254. Frank-Raue, K.; Rondot, S.; Raue, F. Molecular genetics and phenomics of RET mutations: Impact on prognosis of MTC. Mol Cell Endocrinol. 2010, 322, 2–7. [Google Scholar] [CrossRef] [PubMed]
  255. Sachdev, C.; Gattani, R.G.; Agrawal, J. Carney Complex and Its Association with Thyroid Cancer, Molecular Pathway, and Treatment. Cureus 2023, 15, e48503. [Google Scholar] [CrossRef]
  256. Fagin, J.A.; Mitsiades, N. Molecular pathology of thyroid cancer: Diagnostic and clinical implications. Best Pract. Res. Clin. Endocrinol. Metab. 2008, 22, 955–969. [Google Scholar] [CrossRef]
  257. Lugaresi, M.; Moneta, C.; Saruggia, G.; Dionigi, G.; Gazzano, G.; Fugazzola, L. Changing the paradigm: Lobectomy for sporadic medullary thyroid cancer. Eur. Thyroid J. 2025, 14, e250040. [Google Scholar] [CrossRef] [PubMed]
  258. Abuahmed, M.Y.; Rashid, R.; Aboelwafa, W.A.; Hamza, Y.M. The Oncologic Outcomes of Bilateral Central Lymph Node Dissection in Unilobar Papillary Thyroid Cancer and Its Risks: A Prospective Cohort Study. Cureus 2024, 16, e65443. [Google Scholar] [CrossRef] [PubMed]
  259. Hay, I.D.; Gonzalez-Losada, T.; Reinalda, M.S.; Honetschlager, J.A.; Richards, M.L.; Thompson, G.B. Long-term outcome in 215 children and adolescents with papillary thyroid cancer treated during 1940 through 2008. World J. Surg. 2010, 34, 1192–1202. [Google Scholar] [CrossRef] [PubMed]
  260. Ozel, T.M.; Soytas, Y.; Akbulut, S.; Celik, A.; Yildiz, G.; Karatay, H.; Sari, S. The necessity of prophylactic central lymph node dissection in clinically n0 papillary thyroid carcinoma: Perspective from the endemic region. Langenbecks Arch. Surg. 2025, 410, 109. [Google Scholar] [CrossRef]
  261. Moo, T.A.; McGill, J.; Allendorf, J.; Lee, J.; Fahey, T., 3rd; Zarnegar, R. Impact of prophylactic central neck lymph node dissection on early recurrence in papillary thyroid carcinoma. World J. Surg. 2010, 34, 1187–1191. [Google Scholar] [CrossRef]
  262. Yang, Y.; Wang, D.S.; Yang, L.; Huang, Y.H.; He, Y.; Chen, M.S.; Wang, Z.Y.; Fan, L.; Yang, H.W. A nomogram based on the 3-gene signature and clinical characteristics for predicting lymph node metastasis in papillary thyroid cancer. Cancer Biomark. 2025, 42, 18758592241311195. [Google Scholar] [CrossRef]
  263. Bomeli, S.R.; LeBeau, S.O.; Ferris, R.L. Evaluation of a thyroid nodule. Otolaryngol. Clin. N. Am. 2010, 43, 229–238. [Google Scholar] [CrossRef]
  264. Jasim, S.; Dean, D.S.; Gharib, H. Fine-Needle Aspiration of the Thyroid Gland. In Endotext; Feingold, K.R., Ahmed, S.F., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  265. Gopalan, V.; Deshpande, S.G.; Zade, A.A.; Tote, D.; Rajendran, R.; Durge, S.; Bhargava, A. Advances in the Diagnosis and Treatment of Follicular Thyroid Carcinoma: A Comprehensive Review. Cureus 2024, 16, e66186. [Google Scholar] [CrossRef]
  266. Mekel, M.; Nucera, C.; Hodin, R.A.; Parangi, S. Surgical implications of B-RafV600E mutation in fine-needle aspiration of thyroid nodules. Am. J. Surg. 2010, 200, 136–143. [Google Scholar] [CrossRef]
  267. Yamazaki, H.; Sugino, K.; Katoh, R.; Matsuzu, K.; Kitagawa, W.; Nagahama, M.; Saito, A.; Ito, K. Management of follicular thyroid carcinoma. Eur. Thyroid J. 2024, 13, e240146. [Google Scholar] [CrossRef]
  268. Durante, C.; Hegedüs, L.; Czarniecka, A.; Paschke, R.; Russ, G.; Schmitt, F.; Soares, P.; Solymosi, T.; Papini, E. 2023 European Thyroid Association Clinical Practice Guidelines for thyroid nodule management. Eur. Thyroid J. 2023, 12, e230067. [Google Scholar] [CrossRef] [PubMed]
  269. Kaplan, E.; Angelos, P.; Applewhite, M.; Mercier, F.; Grogan, R.H. Chapter 21 SURGERY OF THE THYROID. In Endotext; Feingold, K.R., Ahmed, S.F., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  270. Tang, J.; Kong, D.; Bu, L.; Wu, G. Surgical management for follicular variant of papillary thyroid carcinoma. Oncotarget 2017, 8, 79507–79516. [Google Scholar] [CrossRef] [PubMed]
  271. Konstantinidis, A.; Stang, M.; Roman, S.A.; Sosa, J.A. Surgical management of medullary thyroid carcinoma. Updates Surg. 2017, 69, 151–160. [Google Scholar] [CrossRef] [PubMed]
  272. Taniuchi, M.; Kawata, R.; Terada, T.; Higashino, M.; Aihara, T.; Jinnin, T. Central node dissection from the perspective of lateral neck node metastasis in papillary thyroid carcinoma. Auris Nasus Larynx 2024, 51, 266–270. [Google Scholar] [CrossRef]
  273. Machens, A.; Dralle, H. Surgical Treatment of Medullary Thyroid Cancer. Recent Results Cancer Res. 2025, 223, 247–266. [Google Scholar] [CrossRef]
  274. Mao, Y.V.; Hughes, E.G.; Steinmetz, D.; Troob, S.; Kim, J.; Tseng, C.H.; Fishbein, G.A.; Sajed, D.P.; Livhits, M.J.; Yeh, M.W.; et al. Extent of Surgery for Medullary Thyroid Cancer and Prevalence of Occult Contralateral Foci. JAMA Otolaryngol. Head Neck Surg. 2024, 150, 209–214. [Google Scholar] [CrossRef]
  275. Cleere, E.F.; Crotty, T.J.; Hintze, J.M.; Fitzgerald, C.W.R.; Kinsella, J.; Lennon, P.; Timon, C.V.I.; Woods, R.S.R.; Shine, N.P.; O’Neill, J.P. The role of surgery for anaplastic thyroid carcinoma in the era of targeted therapeutics: A scoping review. Laryngoscope Investig. Otolaryngol. 2023, 8, 1673–1684. [Google Scholar] [CrossRef]
  276. Oliinyk, D.; Augustin, T.; Rauch, J.; Koehler, V.F.; Belka, C.; Spitzweg, C.; Käsmann, L. Role of surgery to the primary tumor in metastatic anaplastic thyroid carcinoma: Pooled analysis and SEER-based study. J. Cancer Res. Clin. Oncol.. 2023, 149, 3527–3547. [Google Scholar] [CrossRef]
  277. Koda, K.; Katoh, M.; Yasuhara, K. Management of anaplastic thyroid cancer and proposed treatment guidelines-A 5-year case series study. Cancer Rep. 2022, 5, e1727. [Google Scholar] [CrossRef]
  278. McIver, B.; Hay, I.D.; Giuffrida, D.F.; Dvorak, C.E.; Grant, C.S.; Thompson, G.B.; van Heerden, J.A.; Goellner, J.R. Anaplastic thyroid carcinoma: A 50-year experience at a single institution. Surgery 2001, 130, 1028–1034. [Google Scholar] [CrossRef]
  279. Hu, S.; Helman, S.N.; Hanly, E.; Likhterov, I. The role of surgery in anaplastic thyroid cancer: A systematic review. Am. J. Otolaryngol. 2017, 38, 337–350. [Google Scholar] [CrossRef] [PubMed]
  280. Luster, M.; Clarke, S.E.; Dietlein, M.; Lassmann, M.; Lind, P.; Oyen, W.J.; Tennvall, J.; Bombardieri, E.; European Association of Nuclear Medicine (EANM). Guidelines for radioiodine therapy of differentiated thyroid cancer. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 1941–1959. [Google Scholar] [CrossRef] [PubMed]
  281. Erten, A.; Abidin Sayiner, Z.; Erkılıç, S.; Oğuzkan Balcı, S.; Akarsu, E. The effect of SLC5A5 gene expression in tumor tissues on refractoriness to radioactive iodine treatment of differentiated thyroid carcinomas. Qatar Med. J. 2025, 2025, 5. [Google Scholar] [CrossRef] [PubMed]
  282. Weis, H.; Weindler, J.; Schmidt, K.; Hellmich, M.; Drzezga, A.; Schmidt, M. Impact of Radioactive Iodine Treatment on Long-Term Relative Survival in Patients with Papillary and Follicular Thyroid Cancer: A SEER-Based Study Covering Histologic Subtypes and Recurrence Risk Categories. J. Nucl. Med. 2025, 66, 525–530. [Google Scholar] [CrossRef]
  283. Zhang, L.; Zhu, C.; Huang, S.; Xu, M.; Li, C.; Fu, H.; Yin, Y.; Liang, S.; Wang, H.; Cui, Z.; et al. Efficient delivery of anlotinib and radioiodine by long circulating nano-capsules for active enhanced suppression of anaplastic thyroid carcinoma. J. Nanobiotechnol. 2025, 23, 180. [Google Scholar] [CrossRef]
  284. Padda, I.S.; Nguyen, M. Radioactive Iodine Therapy. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  285. Van Nostrand, D.; Wartofsky, L. Radioiodine in the treatment of thyroid cancer. Endocrinol. Metab. Clin. N. Am. 2007, 36, 807–822. [Google Scholar] [CrossRef]
  286. Sparano, C.; Moog, S.; Hadoux, J.; Dupuy, C.; Al Ghuzlan, A.; Breuskin, I.; Guerlain, J.; Hartl, D.; Baudin, E.; Lamartina, L. Strategies for Radioiodine Treatment: What’s New. Cancers 2022, 14, 3800. [Google Scholar] [CrossRef]
  287. Orosco, R.K.; Hussain, T.; Noel, J.E.; Chang, D.C.; Dosiou, C.; Mittra, E.; Divi, V.; Orloff, L.A. Radioactive iodine in differentiated thyroid cancer: A national database perspective. Endocr. Relat. Cancer 2019, 26, 795–802. [Google Scholar] [CrossRef]
  288. Liu, Y.; Wang, J.; Hu, X.; Pan, Z.; Xu, T.; Xu, J.; Jiang, L.; Huang, P.; Zhang, Y.; Ge, M. Radioiodine therapy in advanced differentiated thyroid cancer: Resistance and overcoming strategy. Drug Resist. Updat. 2023, 68, 100939. [Google Scholar] [CrossRef]
  289. Mayson, S.E.; Chan, C.M.; Haugen, B.R. Tailoring the approach to radioactive iodine treatment in thyroid cancer. Endocr. Relat. Cancer. 2021, 28, T125–T140. [Google Scholar] [CrossRef]
  290. Berta, D.M.; Teketelew, B.B.; Cherie, N.; Tamir, M.; Abriham, Z.Y.; Ayele Angelo, A.; Tarekegne, A.M.; Chane, E.; Mulatie, Z.; Walle, M. Effect of radioactive iodine therapy on hematological parameters in patients with thyroid cancer: Systematic review and meta-analysis. Front. Endocrinol. 2025, 16, 1562851. [Google Scholar] [CrossRef] [PubMed]
  291. Riley, A.S.; McKenzie, G.A.G.; Green, V.; Schettino, G.; England, R.J.A.; Greenman, J. The effect of radioiodine treatment on the diseased thyroid gland. Int. J. Radiat. Biol. 2019, 95, 1718–1727. [Google Scholar] [CrossRef] [PubMed]
  292. Albero, A.; Lopéz, J.E.; Torres, A.; de la Cruz, L.; Martín, T. Effectiveness of chemotherapy in advanced differentiated thyroid cancer: A systematic review. Endocr. Relat. Cancer. 2016, 23, R71–R84. [Google Scholar] [CrossRef] [PubMed]
  293. Veiga, L.H.; Bhatti, P.; Ronckers, C.M.; Sigurdson, A.J.; Stovall, M.; Smith, S.A.; Weathers, R.; Leisenring, W.; Mertens, A.C.; Hammond, S.; et al. Chemotherapy and thyroid cancer risk: A report from the childhood cancer survivor study. Cancer Epidemiol. Biomarkers Prev. 2012, 21, 92–101. [Google Scholar] [CrossRef]
  294. Wang, B.C.; Lin, G.H.; Kuang, B.H.; Cao, R.B. Emerging chemotherapy-based treatments in anaplastic thyroid cancer: An updated analysis of prospective studies. Front. Endocrinol. 2024, 15, 1385747. [Google Scholar] [CrossRef]
  295. Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–738. [Google Scholar] [CrossRef]
  296. Hussein, O.; Karen, D.; Zidan, J. Cisplatin based chemotherapy in patients with advanced differentiated thyroid carcinoma refractory to I131 treatment. Indian J. Med. Paediatr. Oncol. 2013, 34, 234–237. [Google Scholar] [CrossRef]
  297. Matuszczyk, A.; Petersenn, S.; Bockisch, A.; Gorges, R.; Sheu, S.Y.; Veit, P.; Mann, K. Chemotherapy with doxorubicin in progressive medullary and thyroid carcinoma of the follicular epithelium. Horm. Metab. Res. 2008, 40, 210–213. [Google Scholar] [CrossRef]
  298. Fullmer, T.; Cabanillas, M.E.; Zafereo, M. Novel Therapeutics in Radioactive Iodine-Resistant Thyroid Cancer. Front. Endocrinol. 2021, 12, 720723. [Google Scholar] [CrossRef]
  299. Angelousi, A.; Tzoulis, P.; Tsoli, M.; Chatzellis, E.; Koumarianou, A.; Kaltsas, G. Immunotherapy for endocrine tumours: A clinician’s perspective. Endocr.-Relat. Cancer 2024, 31, e230296. [Google Scholar] [CrossRef]
  300. Han, P.Z.; Ye, W.D.; Yu, P.C.; Tan, L.C.; Shi, X.; Chen, X.F.; He, C.; Hu, J.Q.; Wei, W.J.; Lu, Z.W.; et al. A distinct tumor microenvironment makes anaplastic thyroid cancer more lethal but immunotherapy sensitive than papillary thyroid cancer. JCI Insight 2024, 9, e173712. [Google Scholar] [CrossRef] [PubMed]
  301. Valerio, L.; Pieruzzi, L.; Giani, C.; Agate, L.; Bottici, V.; Lorusso, L.; Cappagli, V.; Puleo, L.; Matrone, A.; Viola, D.; et al. Targeted Therapy in Thyroid Cancer: State of the Art. Clin. Oncol. 2017, 29, 316–324. [Google Scholar] [CrossRef] [PubMed]
  302. Puliafito, I.; Esposito, F.; Prestifilippo, A.; Marchisotta, S.; Sciacca, D.; Vitale, M.P.; Giuffrida, D. Target Therapy in Thyroid Cancer: Current Challenge in Clinical Use of Tyrosine Kinase Inhibitors and Management of Side Effects. Front. Endocrinol. 2022, 13, 860671. [Google Scholar] [CrossRef]
  303. Jeon, Y.; Park, S.; Lee, S.H.; Kim, T.H.; Kim, S.W.; Ahn, M.J.; Jung, H.A.; Chung, J.H. Combination of Dabrafenib and Trametinib in Patients with Metastatic BRAFV600E-Mutated Thyroid Cancer. Cancer Res. Treat. 2024, 56, 1270–1276. [Google Scholar] [CrossRef] [PubMed]
  304. Viola, D.; Valerio, L.; Molinaro, E.; Agate, L.; Bottici, V.; Biagini, A.; Lorusso, L.; Cappagli, V.; Pieruzzi, L.; Giani, C.; et al. Treatment of advanced thyroid cancer with targeted therapies: Ten years of experience. Endocr.-Relat. Cancer 2016, 23, R185–R205. [Google Scholar] [CrossRef]
  305. Zhang, L.; Feng, Q.; Wang, J.; Tan, Z.; Li, Q.; Ge, M. Molecular basis and targeted therapy in thyroid cancer: Progress and opportunities. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188928. [Google Scholar] [CrossRef]
  306. El Omri, M.; Gabsi, O.; Bellakhdher, M.; Ghammem, M.; Kermani, W.; Abdelkefi, M. Management of Thyroid Nodules in Children: A single-center experience. Iran J. Otorhinolaryngol. 2025, 37, 65–71. [Google Scholar]
  307. Duval, M.A.D.S.; Zanella, A.B.; Cristo, A.P.; Faccin, C.S.; Graudenz, M.S.; Maia, A.L. Impact of Serum TSH and Anti-Thyroglobulin Antibody Levels on Lymph Node Fine-Needle Aspiration Thyroglobulin Measurements in Differentiated Thyroid Cancer Patients. Eur. Thyroid J. 2017, 6, 292–297. [Google Scholar] [CrossRef]
  308. Subbiah, V.; Hu, M.I.; Mansfield, A.S.; Taylor, M.H.; Schuler, M.; Zhu, V.W.; Hadoux, J.; Curigliano, G.; Wirth, L.; Gainor, J.F.; et al. Pralsetinib in Patients with Advanced/Metastatic Rearranged During Transfection (RET)-Altered Thyroid Cancer: Updated Efficacy and Safety Data from the ARROW Study. Thyroid. 2024, 34, 26–40. [Google Scholar] [CrossRef]
  309. Ratajczak, M.; Gaweł, D.; Godlewska, M. Novel Inhibitor-Based Therapies for Thyroid Cancer—An Update. Int. J. Mol. Sci. 2021, 22, 11829. [Google Scholar] [CrossRef]
  310. Chen, M.F.; Repetto, M.; Wilhelm, C.; Drilon, A. RET Inhibitors in RET Fusion-Positive Lung Cancers: Past, Present, and Future. Drugs. 2024, 84, 1035–1053. [Google Scholar] [CrossRef] [PubMed]
  311. Capdevila, J.; Wirth, L.J.; Ernst, T.; Aix, S.P.; Lin, C.-C.; Ramlau, R.; Butler, M.O.; Delord, J.-P.; Gelderblom, H.; Ascierto, P.A.; et al. PD-1 Blockade in Anaplastic Thyroid Carcinoma. J. Clin. Oncol. 2020, 38, 2620–2627. [Google Scholar] [CrossRef] [PubMed]
  312. ClinicalTrials.gov [Internet]. U.S. National Library of Medicine: Bethesda, MD, USA. Available online: https://clinicaltrials.gov/ (accessed on 8 April 2025).
  313. Pulumati, A.; Pulumati, A.; Dwarakanath, B.S.; Verma, A.; Papineni, R.V.L. Technological advancements in cancer diagnostics: Improvements and limitations. Cancer Rep. 2023, 6, e1764. [Google Scholar] [CrossRef] [PubMed]
  314. Hoang, J.K.; Branstetter, B.F., 4th; Gafton, A.R.; Lee, W.K.; Glastonbury, C.M. Imaging of thyroid carcinoma with CT and MRI: Approaches to common scenarios. Cancer Imaging 2013, 13, 128–139. [Google Scholar] [CrossRef]
  315. Lv, R.B.; Wang, Q.G.; Liu, C.; Liu, F.; Zhao, Q.; Han, J.G.; Ren, D.L.; Liu, B.; Li, C.L. Low versus high radioiodine activity for ablation of the thyroid remnant after thyroidectomy in Han Chinese with low-risk differentiated thyroid cancer. OncoTargets Ther. 2017, 10, 4051–4057. [Google Scholar] [CrossRef]
  316. Su, J.; Yang, L.; Sun, Z.; Zhan, X. Personalized Drug Therapy: Innovative Concept Guided with Proteoformics. Mol. Cell. Proteom. 2024, 23, 100737. [Google Scholar] [CrossRef]
  317. Conroy, P.C.; Wilhelm, A.; Calthorpe, L.; Ullmann, T.M.; Davis, S.; Huang, C.Y.; Shen, W.T.; Gosnell, J.; Duh, Q.Y.; Roman, S.; et al. Endocrine surgeons are performing more thyroid lobectomies for low-risk differentiated thyroid cancer since the 2015 ATA guidelines. Surgery 2022, 172, 1392–1400. [Google Scholar] [CrossRef] [PubMed]
  318. Park, S.; Jeon, M.J.; Song, E.; Oh, H.S.; Kim, M.; Kwon, H.; Kim, T.Y.; Hong, S.J.; Shong, Y.K.; Kim, W.B.; et al. Clinical Features of Early and Late Postoperative Hypothyroidism After Lobectomy. J. Clin. Endocrinol. Metab. 2017, 102, 1317–1324. [Google Scholar] [CrossRef]
  319. Gil, M.S.R.; Pozas, J.; Molina-Cerrillo, J.; Gómez, J.; Pian, H.; Pozas, M.; Carrato, A.; Grande, E.; Alonso-Gordoa, T. Current and Future Role of Tyrosine Kinases Inhibition in Thyroid Cancer: From Biology to Therapy. Int. J. Mol. Sci. 2020, 21, 4951. [Google Scholar] [CrossRef]
  320. Li, L.-R.; Du, B.; Liu, H.-Q.; Chen, C. Artificial Intelligence for Personalized Medicine in Thyroid Cancer: Current Status and Future Perspectives. Front. Oncol. 2021, 10, 604051. [Google Scholar] [CrossRef]
  321. Modica, R.; Benevento, E.; Colao, A. Endocrine-disrupting chemicals (EDCs) and cancer: New perspectives on an old relationship. J. Endocrinol. Investig. 2022, 46, 667–677. [Google Scholar] [CrossRef] [PubMed]
  322. Panagiotidis, E.; Zhang-Yin, J.T. The Role of Positron Emission Tomography/Computed Tomography in the Management of Differentiated Thyroid Cancer: Current Applications and Future Perspectives. J. Clin. Med. 2024, 13, 6918. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Modifiable and unmodifiable factors in developing thyroid cancer.
Figure 1. Modifiable and unmodifiable factors in developing thyroid cancer.
Ijms 26 05173 g001
Figure 2. Molecular obesity mechanisms associated with thyroid cancer.
Figure 2. Molecular obesity mechanisms associated with thyroid cancer.
Ijms 26 05173 g002
Figure 3. Thyroid cancers, genetics, and surgical treatment.
Figure 3. Thyroid cancers, genetics, and surgical treatment.
Ijms 26 05173 g003
Figure 4. Immunotherapy in thyroid cancers.
Figure 4. Immunotherapy in thyroid cancers.
Ijms 26 05173 g004
Table 1. The most common mutations in chosen types of thyroid cancer.
Table 1. The most common mutations in chosen types of thyroid cancer.
Type of Cancer/MutationPapillary (PTC)Medullary (MTC)Follicular (FTC)Anaplastic (ATC)
RET/PTC+++
RAS+++
BRAF V600E++
PAX8-PPARγ++
NTRK++
MET+
CDKN1B+
TERT+
TP53+
PTEN+
ALK+
PIK3CA+
EIF1AX+
Table 2. Comparison of four major types of thyroid cancer.
Table 2. Comparison of four major types of thyroid cancer.
Cancer TypePTCFTCMTCATC
Incidence
(% of all TC cases)
90%20%1–5%2%
Age 30–50 years old40–50 years old40–60 years oldOver 65 years old
PrognosisRelatively mild cancer80% mild; 20% aggressivePoor prognosisVery poor prognosis
Metastatic potential70% of patients present metastases to the lymph nodes of the neckMetastases present more often than in PTC casesHigh probability of metastasis20–50% of ATC cases present metastases
Table 3. Diagnostic techniques for thyroid cancer. Overview of diagnostic techniques used in the evaluation of thyroid cancer, summarizing their diagnostic value, limitations, and current recommendations based on the latest guidelines from the American Thyroid Association (ATA) and the National Comprehensive Cancer Network (NCCN). The table aims to provide a systematic comparison of conventional and advanced methods, with particular emphasis on their role in assessing malignancy risk and guiding clinical decision-making in indeterminate and advanced cases.
Table 3. Diagnostic techniques for thyroid cancer. Overview of diagnostic techniques used in the evaluation of thyroid cancer, summarizing their diagnostic value, limitations, and current recommendations based on the latest guidelines from the American Thyroid Association (ATA) and the National Comprehensive Cancer Network (NCCN). The table aims to provide a systematic comparison of conventional and advanced methods, with particular emphasis on their role in assessing malignancy risk and guiding clinical decision-making in indeterminate and advanced cases.
Diagnostic TechniqueDiagnostic ValueLimitationsRecommendations (ATA, NCCN)
Ultrasound (US)Primary method for evaluating thyroid nodules, detects features suspicious for malignancy (e.g., microcalcifications) [131]May not be sufficient for definitive diagnosis, especially in indeterminate nodules [136]Recommended as the first step in diagnosis (ATA, NCCN) [133]
Fine-needle aspiration biopsy (FNAB)Cytological evaluation helps assess malignancy risk, particularly in indeterminate nodules [133]Results can be indeterminate for Bethesda III/IV nodules [137]Essential for indeterminate nodules (ATA, NCCN) [135]
Molecular genetic testingIncreases diagnostic accuracy, particularly for Bethesda III/IV nodules, aiding in treatment planning [137]High cost, availability may be limited, not always accessible in every center [137]Recommended for indeterminate nodules (ATA, NCCN) [137]
Single-photon emission computed tomography (SPECT)Helps assess malignant changes and potential metastasis [135]Requires specialized equipment, less useful for evaluating small nodules [136]Recommended in advanced disease, particularly for metastasis assessment (ATA) [71]
Computed tomography (CT)Useful for assessing metastasis, especially in advanced cases [71]Limited value for diagnosing early thyroid tumors [135]Recommended for metastasis assessment (NCCN) [71]
Magnetic resonance imaging (MRI)Helps differentiate between benign and malignant nodules, especially in advanced disease [134]Expensive, time-consuming, requires specialized equipment [134]Recommended for assessing local invasion and metastasis (NCCN) [134]
PET/CT, PET/MRIAdvanced techniques useful for assessing poorly differentiated or dedifferentiated TC [142,143]Expensive, limited availability in some centers [142,143]Recommended in advanced TC, particularly for staging and follow-up (ATA, NCCN) [142,143]
High-throughput sequencing (NGS, WES, RNA-Seq)Allows for the complete profiling of genetic changes (mutations, fusions, expression), which enhances diagnosis and tailored therapy selection in intermediate or aggressive situations [151,152,154]Expensive, requires bioinformatics support, limited access in some contexts [156]Suggested in cases of ambiguous cytology or probable advanced illness (ATA, NCCN) [153,155]
Table 4. Diagnostic and prognostic markers in thyroid cancer subtypes and their clinical application. A summary of significant diagnostic and prognostic biomarkers in thyroid cancer, organized by histological subtype and clinical applicability. The table summarizes the primary value of each marker—such as diagnosis, monitoring, or risk assessment—as well as any pertinent limitations, such as specificity, variability, or clinical application. The purpose of this overview is to clarify the role of each biomarker in subtype-specific thyroid cancer therapy.
Table 4. Diagnostic and prognostic markers in thyroid cancer subtypes and their clinical application. A summary of significant diagnostic and prognostic biomarkers in thyroid cancer, organized by histological subtype and clinical applicability. The table summarizes the primary value of each marker—such as diagnosis, monitoring, or risk assessment—as well as any pertinent limitations, such as specificity, variability, or clinical application. The purpose of this overview is to clarify the role of each biomarker in subtype-specific thyroid cancer therapy.
MarkerSubtype(s)Clinical ApplicationLimitations/Notes
Thyroglobulin (Tg)PTC, FTC (DTC) [157,158,159,160]Recurrence detection, residual disease monitoring, preoperative risk assessment [158,161,165]Affected by TgAb; also secreted by benign thyroid remnants [163,164]
Calcitonin (Ctn)MTC [167,168]Early diagnosis, postoperative monitoring, biochemical cure assessment [169,171,172,173,175]Elevated in non-malignant conditions; rapidly degraded in plasma [168,170]
Carcinoembryonic antigen (CEA)MTC [177,179,181]Disease progression monitoring, aggressiveness estimation [179,183,186,187,189]Non-specific; influenced by liver function; high levels correlate with poor prognosis [180,181,185,186]
Procalcitonin (Pct)MTC [190,198]Tumor size correlation, progression monitoring; adjunct to Ctn and CEA [190,198,199]Elevated in infections, trauma, surgery; not exclusive to C cells [191,192,193,195]
TSHPTC, FTC (DTC) [202,204]Postoperative management, recurrence risk stratification [205,206,208,209]Oversuppression risks (e.g., osteoporosis); complex relationship with recurrence [207,210,211]
miRNAsPTC, FTC, ATC [212,217]Diagnostic and prognostic value, patient stratification [216,220,221,224,225]Experimental; conflicting evidence; requires clinical validation [218,219,222,223]
BRAF V600EPTC, ATC [218,229,232]Risk of lymph node metastasis, iodine-refractory tumor indication [227,228,232]Not reliable as a sole prognostic marker in low-risk disease [230,231,233]
RAS mutationsFTC, PTC (follicular variant), ATC [234,236,237]Diagnostic aid, metastatic risk, mortality prediction [239,240,241]Common in follicular-pattern tumors; also present in benign lesions [238,242]
RET/PTC rearrangementsPTC (esp. radiation-induced), pediatric MTC [244,246,250]Diagnostic relevance, aggressive behavior predictor, pediatric cases [249,250,251]Present in both benign and malignant lesions; strongly linked to radiation exposure [252,253]
Table 5. Comparison of first-line and second-line therapies in thyroid cancer.
Table 5. Comparison of first-line and second-line therapies in thyroid cancer.
Type of Thyroid CancerFirst-Line TherapySecond-Line Therapy
PCT
-
Surgery: lobectomy or total thyroidectomy [107]
-
Postoperative radioactive iodine (RAI) therapy [138]
-
Thyroid hormone replacement [138]
-
Redifferentiation (BRAF, MEK, RET inhibitors) [147]
-
Tyrosine kinase inhibitors (TKIs), immunotherapy in resistant cases [163,165]
FTC
-
Surgery: lobectomy, isthmectomy, total thyroidectomy (TT) [117,121]
-
RAI therapy [121]
-
TSH suppression [121]
-
Targeted therapies in resistant cases (TKI) [165]
MTC
-
Surgery: total thyroidectomy (TT) + central lymph node dissection (CLND) [125,126,127]
-
RET inhibitors (pralsetinib, selpercatinib) [164]
-
Tyrosine kinase inhibitors (TKIs): cabozantinib, vandetanib [165,170]
ATC
-
Surgery (if operable) [131,132]
-
Radiotherapy [131]
-
Thyroid hormone replacement [131]
-
Chemotherapy: doxorubicin, cisplatin, paclitaxel [150,151,160]
-
Immunotherapy: PD-1 inhibitors (pembrolizumab, spartalizumab) [163]
RAI-refractory DTC
-
Radioactive iodine (RAI) therapy (if still effective) [138,140]
-
Kinase inhibitors: sorafenib, lenvatinib [165]
-
Redifferentiation: BRAF/MEK/RET inhibitors [146,147]
RET/NTRK mutated cancers
-
Surgery (if resectable)
-
RET inhibitors: pralsetinib, selpercatinib [170,171]
-
NTRK inhibitors: larotrectinib, entrectinib, repotrectinib [167,171]
Table 6. Selected current clinical trials [312].
Table 6. Selected current clinical trials [312].
Study ID (NCT)Drug CombinationMolecular TargetThyroid Cancer TypePhaseStatus
NCT04760288Pralsetinib vs. standard careRETRET-mutant MTC3Recruiting
NCT04006676PralsetinibRETMTC1/2Active, not recruiting
NCT03954791EntrectinibNTRKNTRK fusion-positive thyroid cancers2Completed
NCT04222972Spartalizumab + lenvatinibPD-1 + VEGFATC1/2Recruiting
NTC03157128SelpercatinibRET inhibitorMTC, DTC1/2Completed
NTC03037385PralsetinibRET inhibitorMTC, DTC1/2Completed
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Forma, A.; Kłodnicka, K.; Pająk, W.; Flieger, J.; Teresińska, B.; Januszewski, J.; Baj, J. Thyroid Cancer: Epidemiology, Classification, Risk Factors, Diagnostic and Prognostic Markers, and Current Treatment Strategies. Int. J. Mol. Sci. 2025, 26, 5173. https://doi.org/10.3390/ijms26115173

AMA Style

Forma A, Kłodnicka K, Pająk W, Flieger J, Teresińska B, Januszewski J, Baj J. Thyroid Cancer: Epidemiology, Classification, Risk Factors, Diagnostic and Prognostic Markers, and Current Treatment Strategies. International Journal of Molecular Sciences. 2025; 26(11):5173. https://doi.org/10.3390/ijms26115173

Chicago/Turabian Style

Forma, Alicja, Karolina Kłodnicka, Weronika Pająk, Jolanta Flieger, Barbara Teresińska, Jacek Januszewski, and Jacek Baj. 2025. "Thyroid Cancer: Epidemiology, Classification, Risk Factors, Diagnostic and Prognostic Markers, and Current Treatment Strategies" International Journal of Molecular Sciences 26, no. 11: 5173. https://doi.org/10.3390/ijms26115173

APA Style

Forma, A., Kłodnicka, K., Pająk, W., Flieger, J., Teresińska, B., Januszewski, J., & Baj, J. (2025). Thyroid Cancer: Epidemiology, Classification, Risk Factors, Diagnostic and Prognostic Markers, and Current Treatment Strategies. International Journal of Molecular Sciences, 26(11), 5173. https://doi.org/10.3390/ijms26115173

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