SARS-CoV-2 in Thyroid Disorders: Comparison
Please note this is a comparison between Version 3 by Jessie Wu and Version 2 by Jessie Wu.
A properly functioning thyroid gland is crucial for health, influencing growth, neuronal development, reproduction and as a key regulator of energy metabolism, although thyroid disorders/diseases are extremely common and affect 200 million people worldwide. Viral infection may represent one of the major environmental factors related to common thyroid disorders, including subacute thyroiditis (SAT), nonthyroidal illness syndrome (NTIS) and autoimmune thyroid diseases (AITDs) including Graves’ disease (GD) and Hashimoto’s thyroiditis (HT).
  • COVID-19
  • SARS-CoV-2
  • vaccine
  • virus
  • thyroid
  • thyroid disorder

1. Subacute Thyroiditis

From the first clinical case of subacute thyroiditis (SAT) (also known as De Quervain thyroiditis or granulomatous thyroiditis) associated with COVID-19 published in May 2020 [1], a large number of SAT cases have been progressively reported [2][3][4]. The incidence of non-coronavirus SAT has been estimated at 12.1 cases/100,000 per year, with a female/male ratio of 5:1 [3]. Based on a systematic review of 15 studies (case reports and case series) published between December 2019 and February 2021, on 17 subjects, Christensen and colleagues (2022) [5] estimated that patient ages generally ranged from 18 to 69 years in COVID-19-associated SAT. Comparable results were found in a narrative review by Popescu et al. (2022) [3] including 2 retrospective studies, 5 case series, and 29 case reports (a total of 81 patients), which reported that the youngest patient was 18 years old and the oldest 85 years old, albeit using a postmortem diagnosis [6]. The majority of cases with typical thyroiditis were females [3][7], however the precise incidence of SAT among subjects infected with COVID-19 virus is currently indefinite [8]. Consistent with the Classical-SAT that typically follows a viral infection, the time recorded from diagnosis of COVID-19 infection and symptoms of SAT was usually from six to eight weeks (and up to six months), though SAT also occurred simultaneously with COVID-19 disease, and even in the absence of respiratory symptoms [3]. Indeed, in a systematic review comprising 38 patients from 26 case reports, case series, and letters on SAT associated with COVID-19 and published by the end of 2021, Ando et al. (2022) showed that SAT may develop regardless of the severity of COVID-19, and in most patients with SAT, infection from SARS-CoV-2 was classified as mild or moderate in severity [8]. Similar findings were reported in the analysis by Popescu et al. (2022), with SAT associated with various COVID-19 presentations, from asymptomatic to complicated forms [3].
In the majority of cases, the clinical picture appeared similar to that reported for typical SAT cases, with fever, anterior neck pain, fatigue, tremors, anosmia, sweating, and palpitations characterizing thyrotoxicosis, which results from destruction of thyroid follicles and release of thyroid hormones [2][9]. In addition to clinical symptoms, specific laboratory and imaging features of SAT included thyroid high erythrocyte sedimentation rate (ESR), common elevation of C-reactive protein (CRP), increase in white blood count, negativity for thyroid autoantibodies, enlarged hypoechoic thyroid with decreased vascularity found on ultrasound, and markedly reduced or absent iodine uptake in the gland as detected by thyroid scintigraphy [2][5]. In the systematic review by Stasiak and Lewiński (2021) [2] including 73 studies, 5 of them reported cases of painless SAT, therefore neck pain, which is considered as the key diagnostic criterion, may also be absent in the COVID-19-associated disorder, probably as a consequence of the frequent use of analgesics and nonsteroidal anti-inflammatory drugs (NSAIDs) in COVID-19 patients [10][11]. In particular, in one of the first published case series, Muller et al. (2020) [11] documented that 75% of the eight patients with thyroiditis/thyrotoxicosis after COVID-19 infection requiring high intensity of care and followed up at a mean of 55 days, had ever experienced neck pain, and instead of lymphocytosis, displayed the lymphopenia occurring in COVID-19 patients. Furthermore, these patients had low or suppressed levels of TSH with normal levels of T3 and T4, indicating that SAT may have overlapped with nonthyroidal illness syndrome (NTIS), a condition described as thyroxine thyrotoxicosis [11]. Lymphopenia in COVID-19 may be triggered by various processes including a direct effect of the virus on the apoptosis of lymphocytes and bone marrow impairment, cytokine-induced lymphocyte apoptosis, metabolic and biochemical abnormalities resulting in decreased production, functionality and survival of lymphocytes, all postulated mechanisms that in turn can influence the HPT axis [12]. Thus, lymphopenia, a marker of extrapulmonary signs in COVID-19, not only can predict the onset of thyroid dysfunction but the lack of lymphocytic infiltration and the formation of giant cells (congregates of lymphocytes, histiocytes, and colloid) in the thyroid prevents tension in the thyroid capsule and appearance of pain in early post-COVID SAT [11][13]. In SAT that occurs later, after normal lymphocyte counts have recovered, the resulting lymphocyte infiltration can rapidly lead to pain in the anterior cervical region [13].
Consistently, in a recent combined retrospective–prospective study conducted over a period of 20 months, among 6.8% of patients with COVID-19 developing SAT, those with painless SAT (n = 5) presented earlier, had more severe thyrotoxic manifestations and exhibited higher CRP, interleukin-6 (IL-6) and neutrophil-lymphocyte ratio (NLR) and lower absolute lymphocyte count than those with painful SAT (n = 6) [14]. Besides, the authors reported significant correlations of IL-6 with free and total T4 and free and total T3, suggesting that destructive effects of cytokines may play a key role in painless SAT [14].
Similar results were observed in a retrospective single-center study, in which the close relationship between thyrotoxicosis and higher serum IL-6 in patients hospitalized for COVID-19 in non-intensive care units, is indicative of the cytokine storm action associated with COVID-19 in triggering and sustaining gland inflammation [10]. In this research, however, contrary to typical SAT parameters, 74.6% of patients with thyrotoxicosis (20.2% of the total) had normal TSH levels, while subjects with overt thyrotoxicosis exhibited higher serum free T4 levels than those with a subclinical disorder [10]. A cohort study instead reported that, in a small subset of COVID-19 survivors (n = 55) followed up with a median of 79 days, none had overt thyrotoxicosis (using a conservative definition of TSH < 0.30 mU/L and free T4 > 23.0 pmol/L), even patients admitted to the intensive care unit [15]. Overall, these data suggest that COVID-19 infection gives rise to a novel entity of SAT, which could often be underdiagnosed also due to its self-limiting nature and especially in subjects with severe COVID-19 disease, where multiple manifestations of infection may hide inflammation of the thyroid gland [3][16]. On the other hand, in most post-viral forms, SAT may present as a first or unique manifestation of SARS-CoV-2 infection or, alternatively, within a clinic picture dominated by SAT rather than COVID-19 disease [3]. Of note, thyrotoxicosis may increase the cardiovascular risk even after short exposure to thyroid hormone excess, favoring the development of arrhythmias and thromboembolic events described in patients with SARS-CoV-2 infection, aggravating the general status of severe infections [10][17][18][19]. After a diagnosis of COVID-19-induced SAT, patients should be treated with beta blockers and NSAIDs, while the efficacy of antithyroid drugs during the thyrotoxic stage of the disease has not been demonstrated [3][5]. Actually, most patients with SAT are treated with glucocorticoids, which are effective at relieving symptoms and reducing the incidence of relapse [20][21]. Glucocorticoids, now considered the gold standard in the treatment of COVID-19 patients requiring invasive mechanical ventilation or oxygen supplementation [22], may instead lead to a decrease of serum TSH through the direct release suppression of the TSH-releasing factor in the hypothalamus, providing both an additional mechanism of thyroid dysfunction and delaying the diagnosis of SAT [20][23][24].

2. Nonthyroidal Illness Syndrome

NTIS has been frequently observed in COVID-19 patients. Chen and colleagues [24] reported that 56% of patients with COVID-19 had lower TSH than the normal range and serum TSH levels in the severe and critical patient group were significantly lower compared to no-COVID-19 pneumonia patients with a similar degree of severity. In addition, the degree of decrease in TSH and total T3 levels was positively correlated with disease severity [24]. Consistently, TSH and fT3 were significantly lower in COVID-19 deceased patients than in those moderately to severely ill, or critically ill but recovered [25] as well as in living COVID-19 patients, compared to healthy controls [26]. A study by Gong et al. (2021) [27] reported that among 150 patients with COVID-19 and low fT3 levels, those in the low TSH group had higher mortality and critical illness rates compared to those in the normal TSH group, and low TSH levels were independently associated with 90-day mortality. The mortality rate was also significantly higher in the low fT4 group [25]. Although these findings might suggest peculiar effects of COVID-19 on TSH levels, both directly on the pituitary secreting cells and indirectly on the pituitary-thyroid axis as a consequence of systemic inflammation caused by virus infection, the observed decrease in TSH level could be induced by hypoxemia or by glucocorticoids with which most patients were treated [24], as previously described. In contrast, Muller et al. (2020) [11] found that patients admitted to high intensive care units(HICUs) because of COVID-19 had lower TSH levels than those admitted to HICUs in the absence of COVID-19 and those in the group with COVID-19 but admitted to low intensive care units, while no significant difference was found in fT3 levels. Importantly, low concentrations of TSH and T3 in the HICU group were associated with normal or elevated concentrations of T4, indicating a transient thyroxine increase related to the thyrotoxicosis, in the underlying context of NTIS [11]. NTIS was also experienced by patients with mild to moderate COVID-19 not requiring intensive care, with low fT3 having prognostic significance as associated with worsening clinical severity of COVID-19 [28]. The analysis of the clinical data of 100 patients with COVID-19 showed that severely or critically ill patients had lower concentrations of serum albumin, fT3, TSH and fT3/free FT4 than non-severely ill patients, and their fT3 reduction was independently associated with all-cause mortality [29]. Moreover, subjects with low fT3 had lower fT3/free T4 ratio, higher inflammatory markers (CRP and ESR) and higher indices of tissue injury (i.e., aspartate aminotransferase and lactate dehydrogenase—LDH), while the inverse correlation between ESR and fT3/free T4 ratio was indicative of the effect of systemic inflammation on the deiodinase activity [30]. Similar results were found in patients with COVID-19 and NTIS (27.5% of the total), who were characterized by higher levels of ESR, CRP and procalcitonin, and a lower lymphocyte count than in COVID-19 non-NTIS patients [31]. Conversely, in a study by Gao et al. (2020) [29], fT3 was negatively associated with CRP and IL-6 only in non-severely ill patients and survivors, and with tumor necrosis factor alpha (TNF-α) only in survivors, while not related to CRP, IL-6 and TNF-α in non-survivors, possibly due to a different role of the inflammatory response in NTIS at different stages of COVID-19. Recently, Lui and co-authors (2022) [12] reported that in COVID-19 patients with fT3 levels compatible with NTIS, TSH showed a significant correlation with lower lymphocyte count, suggesting a potential interaction between the HPT axis and the immune system, which is supported by the parallel recovery in TSH and fT3 with lymphocyte counts in those subjects reassessed after a median of nine days. In addition, although patients who had both NTIS and lymphopenia were more likely to have severe COVID-19 outcomes compared to those who only had either one of NTIS or lymphopenia, only NTIS was an independent predictor of severe outcomes in COVID-19 [12]. Sciacchitano et al. (2021) [32] showed that, among patients hospitalized for COVID-19, low fT3 levels (61.2% of the total) were significantly associated with increased NLR and absolute neutrophil count and with reduced levels of T lymphocytes, especially of the helper-inducer T cell subpopulations that were observed in the more severe group of patients. Low fT3 values were also correlated with increased levels of inflammation (high-sensitivity CRP), tissue damage (LDH, ferritin, high-sensitivity cardiac troponin I) and coagulation (prothrombin time, fibrinogen, D-dimer) serum markers, as well as with higher radiological scores of disease severity (Lung Immune Prognostic Index, Sequential Organ Failure Assessment Score and Tomographic severity score), clearly indicating that reduced fT3 levels can be considered as a prognostic biomarker of COVID-19 [32]. Of interest, the authors also noted a different expression pattern of a small subset of genes (i.e., CD38, CD79B, IFIT3 and NLRP3) involved in the immune reaction and expressed in immune cells in two patients that, in addition to COVID-19, also presented hematological malignancies [32]. In a subsequent study, Sciacchitano et al. (2021) [33] evaluated the thyroid hormone function and body composition by Bioelectrical Impedance Analysis in 74 critically ill COVID-19 patients and in 96 outpatients affected by thyroid diseases in different functional conditions. In patients with COVID-19, a significant inverse correlation was observed between fT3 serum levels and the hydration status (through the Total Body Water/Free Fat Mass—TBW/FFM ratio). Furthermore, reduced fT3 serum values in COVID-19 patients were associated with the increase in TBW, extracellular water and sodium/potassium exchangeable ratio (Nae:Ke), and with the reduction of the intracellular water. Conversely, these alterations were not seen in non-COVID-19 patients, except for the single patient affected by severe hyperthyroidism and myxedema. In particular, Na+-K+ pump could represent a possible target of T3 action, as patients with COVID-19 with NTIS had lower mRNA expression levels of the genes coding for the two major isoforms of this pump. Overall, the results indicate that low fT3 serum levels may lead to the altered distribution of salt and water in the body as a consequence of reduced peripheral thyroid hormone activity and a clinical picture resembling that observed in myxedema [33].
Importantly, more than 90% of children with COVID-19 disease in a picture of severe multisystem inflammatory syndrome exhibited NTIS; however, variables related to organ damage, inflammation and severity of multisystemic involvement and other hematological parameters, such as hemoglobin, platelets, leukocytes, were unrelated to thyroid function, possibly suggesting that low T3 syndrome may act as an adaptive mechanism to conserve energy during a long period of critical illness rather than during acute events [34].

3. Autoimmune Thyroiditis

A number of case reports, case series, and observational studies have investigated the impact of COVID-19 infection on autoimmune thyroid diseases (AITDs) that in turn are associated with thyrotoxicosis. A systematic review comprising a total of 13 studies published between December 2019 and October 2021, reported 14 cases of Graves’ disease (GD) and 5 cases of hypothyroidism due to HT occurring concomitantly or following COVID-19 [35]. In particular, eight cases were diagnosed with GD, the most frequent cause of hyperthyroidism in iodine-sufficient areas and especially in middle-aged women, characterized by low TSH and raised serum concentrations of thyroid hormones, presence of TSH receptor antibodies and thyroid stimulating immunoglobulins, and hypoechoic pattern and increased vascularity at ultrasound [35][36]. Four cases also had Graves’ ophthalmopathy (GO), the most common GD extrathyroidal manifestation [37][38][39][40], while more than half of the cases with GD-associated COVID-19 had a previous history of GD or hyperthyroidism [35]. All subjects were treated with antithyroid drugs, mainly methimazole, with most cases showing symptom recovery and thyroid function improvement after therapy [35]. However, despite administering adequate treatment for four patients manifesting thyroid storm (a severe thyrotoxicosis that may cause high mortality even among patients without COVID-19 infection), one patient died of acute respiratory distress syndrome [40][41]. GD is likely a multifactorial disease, and the complex interplay between genetic and non-genetic factors (e.g., iodine, infections, psychological stress, gender, smoking, vitamin D, selenium, immune modulating agents) underlying this condition may cause activation of an immune response involving innate and adaptive immune pathways [39][42]. Notably, the hyperinflammatory status associated with COVID-19 seems to be primarily mediated by T helper (Th) 1-type cytokines as well as IL-6, and a prevalent Th1 immune response (not related to the hyperthyroidism per se, but to the autoimmune process) has also been reported in the immune-pathogenesis of GD [38][43]. On the other hand, the confounder effect of certain drugs should be considered, as reported above. Indeed, while glucocorticoids, heparin and dopamine interfere with HPT function inhibiting the secretion of TSH, NSAIDs administration may result in a transient elevation of thyroid hormones by inducing their displacement from plasma-binding proteins [37][44]. Furthermore, drugs such as tocilizumab which acts as an antibody IL-6 receptor inhibitor, while preventing autoimmune inflammatory responses, might cause some adverse effects that could be confused with the possible onset of thyroid autoimmunity [45][46].
Of the five HT cases included in the systematic review by Tutal et al. (2022) [35], two had previous hypothyroidism. All subjects were treated with LT4 and euthyroidism was achieved within two weeks to four months [35]; however, one patient, diagnosed with myxedema coma, a life-threatening and emergency presentation of hypothyroidism, with concomitant COVID-19, died of cardiac arrest [47][48]. In light of the temporal relationship between COVID-19 infection and onset of HT (concomitant or up to a few weeks; [35]), a possible link between HT and SARS-CoV-2 is based on the hypothesis that the inflammatory status and the consequent oxidative stress from COVID-19 infection determines an impaired signaling in the SIRT1 (a NAD+-dependent Class III deacetylase enzyme) pathway, which in turn results in an altered expression of the transcription factor Forkhead Box P3 (FOXP3) [49][50]. Therefore, as Foxp3 is a critical regulator of the development and function of CD4+CD25+ regulatory T cells (Tregs) and affected Tregs lead to an autoimmune response involving autoreactive T cells, this environment may promote HT development [51][52][53]. In the reassessment of 122 non-critically ill patients after a median interval of 90 days from COVID-19, Lui et al. (2021) [54] observed that anti-TPO titers and anti-TG titers increased significantly and four patients became positive for anti-TPO, evidencing the utility of thyroid function surveillance post COVID-19 and longer follow-up to monitor potential incidents of thyroid dysfunction among COVID-19 survivors.

4. SARS-CoV-2 Vaccine-Associated Thyroid Disorders

In previous chapters, wresearchers attempted to summarize the current evidence on the association between COVID-19 infection and the onset of thyroid dysfunction. However, recent studies have reported emerging findings on SAT occurrence also following SARS-CoV-2 vaccination and some possible mechanisms have been proposed to support this relationship  [55]. The first of these is based on the effects produced by adjuvants, immunological or pharmacological substances contained in vaccines, which, if they enhance the response to vaccination, may lead to the insurgence of serious side effects, called “autoimmune/inflammatory syndrome by adjuvants” (ASIA) or Shoenfeld’s syndrome in genetically susceptible and predisposed individuals [56]. ASIA originates from dysregulation of both the innate and adaptive immune systems and is responsible for several autoimmune diseases including systemic lupus erythematosus, rheumatoid arthritis, or autoimmune endocrinopathies like HT and SAT, as described after administration of other vaccines, such as the influenza, human papillomavirus and hepatitis B [56][57]. A second more convincing hypothesis is related to molecular mimicry, as demonstrated by an in vitro study revealing a strong cross-reactivity between antibodies against protein S (the protein that binds to ACE2 receptors and allows the virus to enter the cell) and a number of antigens, including TPO, which could account for autoimmunity and inflammatory reactions observed in both SARS-CoV-2 infection and vaccination in genetically predisposed individuals [55][58][59]. In a systematic review of 30 studies, for a total of 51 patients with diagnosis of SAT having received a recent (within 4 weeks) injection of SARS-CoV-2 vaccine, 74.5% were women with a median age at onset of 40 years, and most subjects were vaccinated with an mRNA vaccine [55]. Another review of the literature (16 reports with 22 SAT cases after vaccination against COVID-19), in addition to confirming the greatest apparent vulnerability of females, reported that the median number of days for the development of symptoms after vaccination was 7 days, and approximately 2 weeks from the onset of symptoms to diagnosis [60]. Therefore, symptoms of thyroiditis appear in a few days, unlike the SAT triggered by COVID-19 infection, which commonly develops within 2–3 weeks from infection, and Classical-SAT that has been recognized to manifest 2–8 weeks after upper respiratory tract viral infections [61][62]. The early presentation of SAT associated with the SARS-CoV-2 vaccination could be a consequence of the maximal concentration of viral proteins, and subsequently of autoimmunity, reached within a few days after vaccination [63][64]. Clinical symptoms recorded were those of painful SAT, namely neck pain, palpitations, fatigue, fever, weight loss, anxiety, or insomnia, accompanied by TSH suppression (88.2% of cases), and increased CRP and ESR, the latter significantly associated with the severity of thyrotoxicosis. Different findings were shown in a retrospective cohort study which included 23 patients with SAT detected within 90 days of a COVID-19 vaccination (CoronaVac or Pfizer/BioNTech) and grouped as Vac-SAT, and 62 patients with SAT detected before the COVID-19 pandemic and grouped as Classical-SAT [65]. The authors found that, among the inflammatory markers, only ESR was significantly higher in the Classical-SAT group than the Vac-SAT group, while the others (e.g., CRP, NLR, platelet to lymphocyte ratio) were similar between the two groups [65]. Moreover, SAT-duration was 28 (10–150) days, and higher in Vac-SAT than in Classical-SAT (p = 0.023), while a previous history of LT4 use and increased TSH after resolution were more frequent in Vac-SAT than in Classical-SAT (p = 0.027 and p = 0.041, respectively). Consistently, Ippolito et al., (2022) [55] reported that hypothyroidism was detected in about 26% of the patients after remission of SAT associated with COVID-19 vaccine, and LT4 was indicated in approximately 60% of them. In addition, same as SAT triggered by COVID-19 infection, patients were treated with NSAIDs, beta blockers and corticosteroids for a median of four weeks, leading to a substantial decrease of patients with thyrotoxicosis at follow-up (31.6%) [55]. Of note, while thyroid function and inflammation did not vary with vaccine type, there was a significant difference in geographic origin, with mostly Asians among the patients receiving non-mRNA vaccines [55]. On the other hand, all patients with a history of autoimmune thyroid disease and who developed SAT, had received the mRNA vaccine [55], which, compared to the other traditional vaccines, is encapsulated by lipid nanoparticles that preserve and maintain mRNA stability and could also have adjuvant capacity [59][66]. Adjuvants have the ability to enhance the half-life and efficacy of therapeutic molecules in cells by increasing the immunogenicity of the active ingredient but, at the same time, some of them (e.g., polyethylene glycol) might induce immune responses in predisposed individuals, including anaphylactic reactions [67][68].
Notably, the SARS-CoV-2 vaccination may precipitate a thyrotoxicosis with different underlying etiologies, as shown by Pla Peris et al. (2022) [62] who reported eight cases with thyrotoxicosis after SARS-CoV-2 vaccination—four cases of GD, two of SAT, one case of concurrent GD and SAT and one of atypical SAT—with the onset of symptoms following vaccination ranging from 10 to 14 days in six out of eight patients and 7–8 weeks in two patients. The other two cases of GD were observed in two female health care workers who had received a SARS-CoV-2 vaccine and three days later manifested clinical signs of thyroid hyperactivity, with increased thyroid hormone levels, suppressed TSH, and elevated antithyroid antibodies [69]. More recently, Chaudhary et al. (2022) [70] published a case series of four patients developing GD following the administration of SARS-CoV-2 vector vaccine. Three cases were females and had a family/self-history of autoimmune disease, and, although all patients responded well to medical treatment and became euthyroid after two to four months, two of them showed worsening of thyrotoxicosis after the second dose of vaccine, demonstrating that further vaccination may result in an amplification of the autoimmune response [70]. Concurrent with hyperthyroidism, two of these patients had mild thyroid eye disease, a debilitating condition that occurs frequently in patients suffering from GD and appears to be more common in individuals of Asian ethnicity [70][71]. In summary, like SAT, GD may also present both after infection with SARS-CoV-2 and vaccine against SARS-CoV-2, suggesting that protein S has the potential to induce or unmask autoimmunity in genetically predisposed individuals, although it is still unclear if its onset may also depend on the bystander effect of adjuvants [70]. Furthermore, to date, cases of GD after SARS-CoV-2 vaccine administration have only been observed with mRNA and viral vector vaccine, but not following inactivated vaccine [71][72].
Although the number of cases of thyroid disorders, especially SAT, recorded following COVID-19 vaccination is negligible compared to the billions of vaccine doses administered at a global level, their incidence could be underestimated due to a general lack of awareness among clinicians, as well as the possible misdiagnosis of these conditions that could be confused with other side effects occurring after vaccination. Furthermore, while the association between SARS-CoV-2 vaccination and SAT is far from proven and a better understanding of etiopathogenesis will contribute to optimizing patient management, it should be considered that the reported cases have shown a temporal relationship with vaccination, which probably excludes the possibility of other triggering factors. On the other hand, if thyroid disorders following COVID-19 vaccination are a clinical event apparently with a time association with vaccine administration, this does not imply a cause–effect relationship.

References

  1. Brancatella, A.; Ricci, D.; Viola, N.; Sgrò, D.; Santini, F.; Latrofa, F. Subacute Thyroiditis after Sars-COV-2 Infection. J. Clin. Endocrinol. Metab. 2020, 105, dgaa276.
  2. Stasiak, M.; Lewiński, A. New aspects in the pathogenesis and management of subacute thyroiditis. Rev. Endocr. Metab. Disord. 2021, 22, 1027–1039.
  3. Popescu, M.; Ghemigian, A.; Vasile, C.M.; Costache, A.; Carsote, M.; Ghenea, A.E. The New Entity of Subacute Thyroiditis amid the COVID-19 Pandemic: From Infection to Vaccine. Diagnostics 2022, 12, 960.
  4. Yorulmaz, G.; Sahin Tekin, M. SARS-CoV-2 vaccine-associated subacute thyroiditis. J. Endocrinol. Investig. 2022, 45, 1341–1347.
  5. Christensen, J.; O’Callaghan, K.; Sinclair, H.; Hawke, K.; Love, A.; Hajkowicz, K.; Stewart, A.G. Risk factors, treatment and outcomes of subacute thyroiditis secondary to COVID-19: A systematic review. Intern. Med. J. 2022, 52, 522–529.
  6. Jakovac, H.; Ferenčić, A.; Stemberger, C.; Vitezi’c, B.M.; Cuculi’c, D. Detection of SARS-CoV-2 antigens in thyroid gland showing histopathological features of subacute thyroiditis. Eur. Thyroid J. 2022, 11, e220005.
  7. Desailloud, R.; Hober, D. Viruses and thyroiditis: An update. Virol. J. 2009, 6, 5.
  8. Ando, Y.; Ono, Y.; Sano, A.; Fujita, N.; Ono, S. Subacute Thyroiditis after COVID-19: A Literature Review. Am. J. Trop. Med. Hyg. 2022, 107, 1074.
  9. Chen, W.; Tian, Y.; Li, Z.; Zhu, J.; Wei, T.; Lei, J. Potential Interaction Between SARS-CoV-2 and Thyroid: A Review. Endocrinology 2021, 162, bqab004.
  10. Lania, A.; Sandri, M.T.; Cellini, M.; Mirani, M.; Lavezzi, E.; Mazziotti, G. Thyrotoxicosis in patients with COVID-19: The THYRCOV study. Eur. J. Endocrinol. 2020, 183, 381–387.
  11. Muller, I.; Cannavaro, D.; Dazzi, D.; Covelli, D.; Mantovani, G.; Muscatello, A.; Ferrante, E.; Orsi, E.; Resi, V.; Longari, V.; et al. SARS-CoV-2-related atypical thyroiditis. Lancet Diabetes Endocrinol. 2020, 8, 739–741.
  12. Lui, D.T.W.; Lee, C.H.; Chow, W.S.; Lee, A.C.H.; Tam, A.R.; Pang, P.; Ho, T.Y.; Cheung, C.Y.Y.; Fong, C.H.Y.; Law, C.Y.; et al. The Independent Association of TSH and Free Triiodothyronine Levels with Lymphocyte Counts among COVID-19 Patients. Front. Endocrinol. 2022, 12, 774346.
  13. Lui, D.T.W.; Lee, C.H.; Chow, W.S.; Lee, A.C.H.; Tam, A.R.; Cheung, C.Y.Y.; Fong, C.H.Y.; Kwok, S.T.M.; Law, C.Y.; To, K.K.W.; et al. Development of a prediction score (ThyroCOVID) for identifying abnormal thyroid function in COVID-19 patients. J. Endocrinol. Investig. 2022, 45, 2149–2156.
  14. Mondal, S.; DasGupta, R.; Lodh, M.; Ganguly, A. Subacute thyroiditis following recovery from COVID-19 infection: Novel clinical findings from an Eastern Indian cohort. Postgrad. Med. J. 2022.
  15. Khoo, B.; Tan, T.; Clarke, S.A.; Mills, E.G.; Patel, B.; Modi, M.; Phylactou, M.; Eng, P.C.; Thurston, L.; Alexander, E.C.; et al. Thyroid Function Before, During, and After COVID-19. J. Clin. Endocrinol. Metab. 2021, 106, e803–e811.
  16. Dworakowska, D.; Morley, S.; Mulholland, N.; Grossman, A.B. COVID-19-related thyroiditis: A novel disease entity? Clin. Endocrinol. 2021, 95, 369–377.
  17. Mattar, S.A.M.; Koh, S.J.Q.; Rama Chandran, S.; Cherng, B.P.Z. Subacute thyroiditis associated with COVID-19. BMJ Case Rep. 2020, 13, e237336.
  18. Dekkers, O.M.; Horváth-Puhó, E.; Cannegieter, S.C.; Vandenbroucke, J.P.; Sørensen, H.T.; Jørgensen, J.O. Acute cardiovascular events and all-cause mortality in patients with hyperthyroidism: A population-based cohort study. Eur. J. Endocrinol. 2017, 176, 1–9.
  19. Madjid, M.; Safavi-Naeini, P.; Solomon, S.D.; Vardeny, O. Potential Effects of Coronaviruses on the Cardiovascular System: A Review. JAMA Cardiol. 2020, 5, 831–840.
  20. Naguib, R. Potential relationships between COVID-19 and the thyroid gland: An update. J. Int. Med. Res. 2022, 50.
  21. Arao, T.; Okada, Y.; Torimoto, K.; Kurozumi, A.; Narisawa, M.; Yamamoto, S.; Tanaka, Y. Prednisolone Dosing Regimen for Treatment of Subacute Thyroiditis. J. UOEH 2015, 37, 103–110.
  22. RECOVERY Collaborative Group; Horby, P.; Lim, W.S.; Emberson, J.R.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Brightling, C.; Ustianowski, A.; et al. Dexamethasone in Hospitalized Patients with Covid-19. N. Engl. J. Med. 2021, 384, 693–704.
  23. Clarke, S.A.; Abbara, A.; Dhillo, W.S. Impact of COVID-19 on the Endocrine System: A Mini-review. Endocrinology 2022, 163, bqab203.
  24. Chen, M.; Zhou, W.; Xu, W. Thyroid Function Analysis in 50 Patients with COVID-19: A Retrospective Study. Thyroid 2021, 31, 8–11.
  25. Chen, T.; Wu, D.; Chen, H.; Yan, W.; Yang, D.; Chen, G.; Ma, K.; Xu, D.; Yu, H.; Wang, H.; et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: Retrospective study. BMJ 2020, 368, m1091.
  26. Li, M.Y.; Li, L.; Zhang, Y.; Wang, X.S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect. Dis. Poverty 2020, 9, 45.
  27. Gong, J.; Wang, D.K.; Dong, H.; Xia, Q.S.; Huang, Z.Y.; Zhao, Y.; Chen, X.; Yuan, F.; Li, J.B.; Lu, F.E. Prognostic significance of low TSH concentration in patients with COVID-19 presenting with non-thyroidal illness syndrome. BMC Endocr. Disord. 2021, 21, 111.
  28. Lui, D.T.W.; Lee, C.H.; Chow, W.S.; Lee, A.C.H.; Tam, A.R.; Fong, C.H.Y.; Law, C.Y.; Leung, E.K.H.; To, K.K.W.; Tan, K.C.B.; et al. Role of non-thyroidal illness syndrome in predicting adverse outcomes in COVID-19 patients predominantly of mild-to-moderate severity. Clin. Endocrinol. 2021, 95, 469–477.
  29. Gao, W.; Guo, W.; Guo, Y.; Shi, M.; Dong, G.; Wang, G.; Ge, Q.; Zhu, J.; Zhou, X. Thyroid hormone concentrations in severely or critically ill patients with COVID-19. J. Endocrinol. Investig. 2021, 44, 1031–1040.
  30. Lui, D.T.W.; Lee, C.H.; Chow, W.S.; Lee, A.C.H.; Tam, A.R.; Fong, C.H.Y.; Law, C.Y.; Leung, E.K.H.; To, K.K.W.; Tan, K.C.B.; et al. Thyroid Dysfunction in Relation to Immune Profile, Disease Status, and Outcome in 191 Patients with COVID-19. J. Clin. Endocrinol. Metab. 2021, 106, e926–e935.
  31. Zou, R.; Wu, C.; Zhang, S.; Wang, G.; Zhang, Q.; Yu, B.; Wu, Y.; Dong, H.; Wu, G.; Wu, S.; et al. Euthyroid Sick Syndrome in Patients with COVID-19. Front. Endocrinol. 2020, 11, 566439.
  32. Sciacchitano, S.; De Vitis, C.; D’Ascanio, M.; Giovagnoli, S.; De Dominicis, C.; Laghi, A.; Anibaldi, P.; Petrucca, A.; Salerno, G.; Santino, I.; et al. Gene signature and immune cell profiling by high-dimensional, single-cell analysis in COVID-19 patients, presenting Low T3 syndrome and coexistent hematological malignancies. J. Transl. Med. 2021, 19, 139.
  33. Sciacchitano, S.; Capalbo, C.; Napoli, C.; Negro, A.; De Biase, L.; Marcolongo, A.; Anibaldi, P.; Salvati, V.; Petrella, L.; Merlo, L.; et al. Nonthyroidal illness syndrome (NTIS) in severe COVID-19 patients: Role of T3 on the Na/K pump gene expression and on hydroelectrolytic equilibrium. J. Transl. Med. 2021, 19, 491.
  34. Calcaterra, V.; Biganzoli, G.; Dilillo, D.; Mannarino, S.; Fiori, L.; Pelizzo, G.; Zoia, E.; Fabiano, V.; Carlucci, P.; Camporesi, A.; et al. Non-thyroidal illness syndrome and SARS-CoV-2-associated multisystem inflammatory syndrome in children. J. Endocrinol. Investig. 2022, 45, 199–208.
  35. Tutal, E.; Ozaras, R.; Leblebicioglu, H. Systematic review of COVID-19 and autoimmune thyroiditis. Travel Med. Infect. Dis. 2022, 47, 102314.
  36. De Leo, S.; Lee, S.Y.; Braverman, L.E. Hyperthyroidism. Lancet 2016, 388, 906–918.
  37. Allam, M.M.; El-Zawawy, H.T.; Ahmed, S.M.; Aly Abdelhamid, M. Thyroid disease and covid-19 infection: Case series. Clin. Case Rep. 2021, 9, e04225.
  38. Jiménez-Blanco, S.; Pla-Peris, B.; Marazuela, M. COVID-19: A cause of recurrent Graves’ hyperthyroidism? J. Endocrinol. Investig. 2021, 44, 387–388.
  39. Lanzolla, G.; Marcocci, C.; Marinò, M. Graves’ disease and Graves’ orbitopathy following COVID-19. J. Endocrinol. Investig. 2021, 44, 2011–2012.
  40. Milani, N.; Najafpour, M.; Mohebbi, M. Case series: Rare cases of thyroid storm in COVID-19 patients. Clin. Case Rep. 2021, 9, e04772.
  41. Edwards, K.; Hussain, I. Two Cases of Severe Autoimmune Thyrotoxicosis Following SARS-CoV-2 Infection. J. Investig. Med. High Impact Case Rep. 2021, 9.
  42. Marinò, M.; Latrofa, F.; Menconi, F.; Chiovato, L.; Vitti, P. Role of genetic and non-genetic factors in the etiology of Graves’ disease. J. Endocrinol. Investig. 2015, 38, 283–294.
  43. Fallahi, P.; Ferrari, S.M.; Elia, G.; Ragusa, F.; Paparo, S.R.; Patrizio, A.; Camastra, S.; Miccoli, M.; Cavallini, G.; Benvenga, S.; et al. Cytokines as Targets of Novel Therapies for Graves’ Ophthalmopathy. Front. Endocrinol. 2021, 12, 654473.
  44. Burch, H.B. Drug Effects on the Thyroid. N. Engl. J. Med. 2019, 381, 749–761.
  45. Çabuk, S.A.; Cevher, A.Z.; Küçükardalı, Y. Thyroid Function during and after COVID-19 Infection: A Review. touchREV Endocrinol. 2022, 18, 58–62.
  46. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295.
  47. Ono, Y.; Ono, S.; Yasunaga, H.; Matsui, H.; Fushimi, K.; Tanaka, Y. Clinical characteristics and outcomes of myxedema coma: Analysis of a national inpatient database in Japan. J. Epidemiol. 2017, 27, 117–122.
  48. Dixit, N.M.; Truong, K.P.; Rabadia, S.V.; Li, D.; Srivastava, P.K.; Mosaferi, T.; Calfon Press, M.A.; Donangelo, I.; Kelesidis, T. Sudden Cardiac Arrest in a Patient with Myxedema Coma and COVID-19. J. Endocr. Soc. 2020, 4, bvaa130.
  49. Motta, M.C.; Divecha, N.; Lemieux, M.; Kamel, C.; Chen, D.; Gu, W.; Bultsma, Y.; McBurney, M.; Guarente, L. Mammalian SIRT1 represses forkhead transcription factors. Cell 2004, 116, 551–563.
  50. Salminen, A.; Kaarniranta, K.; Kauppinen, A. Crosstalk between Oxidative Stress and SIRT1: Impact on the Aging Process. Int. J. Mol. Sci. 2013, 14, 3834–3859.
  51. Xue, H.; Yu, X.; Ma, L.; Song, S.; Li, Y.; Zhang, L.; Yang, T.; Liu, H. The possible role of CD4⁺CD25(high)Foxp3⁺/CD4⁺IL-17A⁺ cell imbalance in the autoimmunity of patients with Hashimoto thyroiditis. Endocrine 2015, 50, 665–673.
  52. Yagi, H.; Nomura, T.; Nakamura, K.; Yamazaki, S.; Kitawaki, T.; Hori, S.; Maeda, M.; Onodera, M.; Uchiyama, T.; Fujii, S.; et al. Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int. Immunol. 2004, 16, 1643–1656.
  53. Knack, R.S.; Hanada, T.; Knack, R.S.; Mayr, K. Hashimoto’s thyroiditis following SARS-CoV-2 infection. BMJ Case Rep. 2021, 14, e244909.
  54. Lui, D.T.W.; Lee, C.H.; Chow, W.S.; Lee, A.C.H.; Tam, A.R.; Fong, C.H.Y.; Law, C.Y.; Leung, E.K.H.; To, K.K.W.; Tan, K.C.B.; et al. Insights from a Prospective Follow-up of Thyroid Function and Autoimmunity among COVID-19 Survivors. Endocrinol. Metab. 2021, 36, 582–589.
  55. Ippolito, S.; Gallo, D.; Rossini, A.; Patera, B.; Lanzo, N.; Fazzino, G.F.M.; Piantanida, E.; Tanda, M.L. SARS-CoV-2 vaccine-associated subacute thyroiditis: Insights from a systematic review. J. Endocrinol. Investig. 2022, 45, 1189–1200.
  56. Bragazzi, N.L.; Hejly, A.; Watad, A.; Adawi, M.; Amital, H.; Shoenfeld, Y. ASIA syndrome and endocrine autoimmune disorders. Best Pract. Res. Clin. Endocrinol. Metab. 2020, 34, 101412.
  57. Pipitone, G.; Rindi, L.V.; Petrosillo, N.; Foti, N.A.M.; Caci, G.; Iaria, C.; Donno, D.R.; Boumis, E.; Paviglianiti, G.; Taglietti, F. Vaccine-Induced Subacute Thyroiditis (De Quervain’s) after mRNA Vaccine against SARS-CoV-2: A Case Report and Systematic Review. Infect. Dis. Rep. 2022, 14, 142–154.
  58. Vojdani, A.; Kharrazian, D. Potential antigenic cross-reactivity between SARS-CoV-2 and human tissue with a possible link to an increase in autoimmune diseases. Clin. Immunol. 2020, 217, 108480.
  59. Pujol, A.; Gómez, L.A.; Gallegos, C.; Nicolau, J.; Sanchís, P.; González-Freire, M.; López-González, Á.A.; Dotres, K.; Masmiquel, L. Thyroid as a target of adjuvant autoimmunity/inflammatory syndrome due to mRNA-based SARS-CoV2 vaccination: From Graves’ disease to silent thyroiditis. J. Endocrinol. Investig. 2022, 45, 875–882.
  60. Jhon, M.; Lee, S.H.; Oh, T.H.; Kang, H.C. Subacute Thyroiditis After Receiving the mRNA COVID-19 Vaccine (Moderna): The First Case Report and Literature Review in Korea. J. Korean Med. Sci. 2022, 37, e39.
  61. Ross, D.S.; Burch, H.B.; Cooper, D.S.; Greenlee, M.C.; Laurberg, P.; Maia, A.L.; Rivkees, S.A.; Samuels, M.; Sosa, J.A.; Stan, M.N.; et al. 2016 American Thyroid Association Guidelines for Diagnosis and Management of Hyperthyroidism and Other Causes of Thyrotoxicosis. Thyroid 2016, 26, 1343–1421.
  62. Pla Peris, B.; Merchante Alfaro, A.Á.; Maravall Royo, F.J.; Abellán Galiana, P.; Pérez Naranjo, S.; González Boillos, M. Thyrotoxicosis following SARS-COV-2 vaccination: A case series and discussion. J. Endocrinol. Investig. 2022, 45, 1071–1077.
  63. Dotan, A.; Muller, S.; Kanduc, D.; David, P.; Halpert, G.; Shoenfeld, Y. The SARS-CoV-2 as an instrumental trigger of autoimmunity. Autoimmunity Rev. 2021, 20, 102792.
  64. İremli, B.G.; Şendur, S.N.; Ünlütürk, U. Three Cases of Subacute Thyroiditis Following SARS-CoV-2 Vaccine: Postvaccination ASIA Syndrome. J. Clin. Endocrinol. Metab. 2021, 106, 2600–2605.
  65. Topaloğlu, Ö.; Tekin, S.; Topaloğlu, S.N.; Bayraktaroglu, T. Differences in Clinical Aspects Between Subacute Thyroiditis Associated with COVID-19 Vaccines and Classical Subacute Thyroiditis. Horm. Metab. Res. 2022, 54, 380–388.
  66. Chung, Y.H.; Beiss, V.; Fiering, S.N.; Steinmetz, N.F. COVID-19 Vaccine Frontrunners and Their Nanotechnology Design. ACS Nano 2020, 14, 12522–12537.
  67. Chen, B.M.; Cheng, T.L.; Roffler, S.R. Polyethylene Glycol Immunogenicity: Theoretical, Clinical, and Practical Aspects of Anti-Polyethylene Glycol Antibodies. ACS Nano 2021, 15, 14022–14048.
  68. Sellaturay, P.; Nasser, S.; Islam, S.; Gurugama, P.; Ewan, P.W. Polyethylene glycol (PEG) is a cause of anaphylaxis to the Pfizer/BioNTech mRNA COVID-19 vaccine. Clin. Exp. Allergy 2021, 51, 861–863.
  69. Vera-Lastra, O.; Ordinola Navarro, A.; Cruz Domiguez, M.P.; Medina, G.; Sánchez Valadez, T.I.; Jara, L.J. Two Cases of Graves’ Disease Following SARS-CoV-2 Vaccination: An Autoimmune/Inflammatory Syndrome Induced by Adjuvants. Thyroid 2021, 31, 1436–1439.
  70. Chaudhary, S.; Dogra, V.; Walia, R. Four cases of Graves’ disease following viral vector severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) vaccine. Endocr. J. 2022, 69, 1431–1435.
  71. Chin, Y.H.; Ng, C.H.; Lee, M.H.; Koh, J.W.H.; Kiew, J.; Yang, S.P.; Sundar, G.; Khoo, C.M. Prevalence of thyroid eye disease in Graves’ disease: A meta-analysis and systematic review. Clin. Endocrinol. 2020, 93, 363–374.
  72. Sriphrapradang, C. Aggravation of hyperthyroidism after heterologous prime-boost immunization with inactivated and adenovirus-vectored SARS-CoV-2 vaccine in a patient with Graves’ disease. Endocrine 2021, 74, 226–227.
More
Video Production Service