Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

The impact of thyroid hormones on patients with hepatocellular carcinoma

  • Matthias Pinter,

    Roles Data curation, Formal analysis, Writing – original draft, Writing – review & editing

    Affiliations Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria, Liver Cancer (HCC) Study Group Vienna, Vienna, Austria

  • Lukas Haupt,

    Roles Data curation, Formal analysis, Writing – original draft, Writing – review & editing

    Affiliation Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria

  • Florian Hucke,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria, Liver Cancer (HCC) Study Group Vienna, Vienna, Austria, Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria

  • Simona Bota,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria, Liver Cancer (HCC) Study Group Vienna, Vienna, Austria, Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria

  • Theresa Bucsics,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria, Liver Cancer (HCC) Study Group Vienna, Vienna, Austria

  • Michael Trauner,

    Roles Supervision, Writing – review & editing

    Affiliation Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria

  • Markus Peck-Radosavljevic,

    Roles Supervision, Writing – review & editing

    Affiliations Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria, Liver Cancer (HCC) Study Group Vienna, Vienna, Austria, Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria

  • Wolfgang Sieghart

    Roles Conceptualization, Methodology, Supervision, Writing – review & editing

    wolfgang.sieghart@meduniwien.ac.at

    Affiliations Division of Gastroenterology & Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria, Liver Cancer (HCC) Study Group Vienna, Vienna, Austria

Abstract

Background & aims

Hypothyroidism has recently been proposed as predisposing factor for HCC development. However, the role of thyroid hormones (TH) in established HCC is largely unclear. We investigated the impact of TH on clinical characteristics and prognosis of HCC patients.

Methods

Of 838 patients diagnosed with nonsurgical HCC at the Division of Gastroenterology and Hepatology/Medical University of Vienna between 1992 and 2012, 667 patients fulfilled the inclusion criteria. The associations of thyroid function tests with patient, liver, and tumor characteristics as well as their impact on overall survival (OS) were investigated.

Results

Thyroid hormone substitution was more often observed in patients with low thyroid-stimulating hormone (TSH) concentration and in patients with elevated free tetraiodthyronine (fT4). Patients with high TSH (>3.77uU/ml) concentrations had larger tumors, while the opposite was true for patients with low TSH (<0.44uU/ml) concentrations. Subjects with elevated fT4 (>1.66ng/dl) were more likely to have elevated CRP. While TSH was only associated with OS in univariate analysis (≤1.7 vs. >1.7uU/ml, median OS (95%CI), 12.3 (8.9–15.7 months) vs. 7.3 months (5.4–9.2 months); p = 0.003), fT4 (≤1.66 vs. >1.66ng/dl, median OS (95%CI), 10.6 (7.5–13.6 months) vs. 3.3 months (2.2–4.3 months); p = 0.007) remained an independent prognostic factor for OS (HR (95%CI) for fT4>1.66ng/dl, 2.1 (1.3–3.3); p = 0.002) in multivariate analysis.

Conclusions

TSH and fT4 were associated with prognostic factors of HCC (i.e., tumor size, CRP level). Elevated fT4 concentrations were independently associated with poor prognosis in HCC. Further studies are needed to characterize the role of TH in HCC in detail.

Introduction

Components of thyroid hormone signaling are implicated in the development and progression, as well as in the prevention of various cancers including hepatocellular carcinoma (HCC) [13]. HCC usually develops in patients with liver cirrhosis [4, 5] and represents the second most common cause of cancer-related mortality in men globally [6].

The thyroid hormones (TH) tetraiodthyronine (T4) and, to a lesser extent, triiodthyronine (T3) are produced in the thyroid gland in a complex multi- step process, which is tightly controlled by the hypothalamus-pituitary-thyroid axis. Hypothalamic neurons produce the tripeptide thyrotropin-releasing-hormone (TRH), which uses the pituitary portal venous system to reach the anterior pituitary gland, where it binds to the TRH receptor in thyrotropic cells, and thereby stimulates both the production and pulsatile release of the glycoprotein thyrotropin (thyroid-stimulating hormone, TSH). By binding to the TSH-receptor situated at the basolateral membrane of thyrocytes, TSH activates thyroid hormone production and secretion.

The vast majority of both thyroid hormones is bound to the plasma proteins and therefore unable to bind to receptors in target tissues. This constellation allows preparation of a large pool of hormones, which can quickly be released when needed. Conversely, the smaller, unbound fraction of thyroid hormones is responsible for the actual physiological effects. The thyroid gland predominantly secretes T4, most of which is subjected to peripheral metabolism by deiodinase enzymes [7].

Since TH display extensive influence on regulatory mechanisms affecting the control of cellular growth and metabolism [710], there are myriad possible modes of interaction with both the development and progression of HCC. Although hypothyroidism has been shown to be associated with an elevated risk of HCC development [2, 11, 12], the role of TH in established HCC remains to be elucidated. Several preclinical studies have linked TH signaling to tumor-promoting actions, especially at advanced stages of hepatocarcinogenesis [1315]. However, little is known about both the prevalence and clinical role of TH in patients with manifest HCC. Hence, we aimed to investigate the association of TH with clinical patient and tumor characteristics as well as their impact on the prognosis of HCC patients.

Materials and methods

Patient selection

Data from all patients who were diagnosed with HCC by biopsy or radiological imaging according to the European Association for the Study of the Liver (EASL) [16] diagnostic criteria between August 1992 and February 2013 at the Medical University of Vienna were retrospectively collected and incorporated into a database. Patients who had received surgical treatment for HCC at any time after diagnosis were excluded from analysis. Only patients aged ≥18 years with TSH levels available at the time of diagnosis were eligible. Collection and retrospective analysis was approved by the Ethics Committee of the Medical University of Vienna.

Data acquisition

The date of HCC diagnosis (date of biopsy if available or diagnostic imaging) was considered the baseline of this study. Patient information was collected in a preexisting MS Access 2010 database (HCC database) in a pseudo-anonymous manner. Patient characteristics, laboratory parameters including thyroidal function (TSH, T3, T4, fT3, fT4), tumor characteristics, and parameters representing liver function were recorded from the patients’ charts and from the electronic patients’ information system. Determination of thyroid status was based on TSH and fT4 levels. We formed 5 subgroups: euthyreosis, hyperthyreosis (primary, secondary, and euthyroid hyperthyroxinemia), subclinical hyperthyreosis, subclinical hypothyreosis, hypothyreosis (primary and secondary), and thyroid hormone substitution. Liver function was assessed by MELD score and Child-Pugh score. The latter has been incorporated in the Barcelona Clinic Liver Cancer classification, the most widely used staging system for HCC, which has been endorsed by the American and European HCC guidelines [16, 17].

Statistics

Baseline characteristics were summarized using descriptive statistics. Chi square test or Fisher’s exact test were used to compare nominal data. Overall survival (OS) was defined as the time from date of diagnosis (date of biopsy if available or diagnostic imaging) until date of death or last contact. Survival curves were calculated using the Kaplan-Meier method and were compared by means of the log rank test (univariate analysis). Variables that reached a p-value of <0.05 in univariate analysis were entered into a multivariate analysis. The multivariate analysis was performed using a Cox proportional hazard regression model. Statistical tests were two-sided and a p-value <0.05 was considered significant. All statistical analyses were performed using SPSS version 17.0 (SPSS Inc., Chicago, IL).

Results

Patient characteristics

Of 667 patients included (Fig 1), 82% of the patients were male, with a male to female ratio of 4.6:1. The median age at diagnosis was 64 years (range, 32–87 years). Six percent of all patients were on thyroid hormone substitution. Detailed patient characteristics are shown in Table 1. Additional information on thyroid function is given in S1 Table. Mean follow-up was 65.5 months. Five hundred and forty-four (82%) patients died during the observation period.

thumbnail
Fig 1. Flow chart of patient selection.

Abbreviations: HCC, hepatocellular carcinoma; TSH, thyroid-stimulating hormone.

https://doi.org/10.1371/journal.pone.0181878.g001

Association of thyroid-stimulating hormone (TSH) and free tetraiodthyronine (fT4) with patient, liver, and tumor characteristics

TSH abnormalities significantly differed between age groups. While patients with low TSH concentrations were generally older (age >65 vs. ≤65years, 68 vs. 32%) those with high TSH (>3.77uU/ml) levels were predominantly younger (age >65 vs. ≤65years, 39 vs. 61%; p = 0.007). Thyroid hormone substitution was more often observed in patients with low TSH (<0.44uU/ml) concentration (24%) compared to those with normal (4%) or elevated TSH (13%) levels (p<0.001). Finally, patients with high TSH concentrations had larger tumors (tumor >5cm vs. ≤5cm, 61 vs. 39%) while the opposite was true for patients with low TSH concentrations (tumor >5cm vs. ≤5cm, 32 vs. 68%; p = 0.005). No association was observed between other variables including e.g. Child-Pugh stage, metastasis, or macrovascular invasion (Table 2). Notably, in patients with larger HCC (>5cm), TSH levels were only significantly associated with sex and thyroid hormone substitution, but not with other variables that might correlate with larger HCC (i.e., etiology, Child-Pugh stage) (S2 Table).

thumbnail
Table 2. Association between thyroid-stimulating hormone (TSH) and patient, liver, and tumor characteristics (n = 667).

https://doi.org/10.1371/journal.pone.0181878.t002

Free T4 (fT4) levels were available in 313 patients. Since sample size of hypothyroid patients (fT4 levels below 0.76ng/dl) was too small (n = 4) to allow for robust analysis, fT4 levels were divided into two groups (≤1.66 and >1.66ng/dl, hereafter referred to as ‘normal’ and ‘elevated’, respectively). The proportion of female patients was higher in patients with elevated fT4 levels (normal vs. elevated fT4, 16 vs. 36%; p = 0.025). Thyroid hormone substitution was more frequently observed in patients with elevated fT4 (normal vs. elevated fT4, 6 vs. 32%; p<0.001). Finally, subjects with elevated fT4 were more likely to have elevated CRP levels (normal vs. elevated fT4, 51 vs. 88%; p = 0.001). No association was observed between other variables representing liver function and tumor burden, respectively (S3 Table).

Since information on T3, T4, and fT3 was missing in 82–97% of patients due to the retrospective character of this analysis (S1 Table), we could not assess their association with patient, liver, and tumor characteristics.

Uni- and multivariate analyses of prognostic factors

Median survival of the study population (n = 667) was 9.3 months (95%CI, 7.6–11.0 months).

In univariate analysis (Table 3), both, TSH (TSH≤1.7 vs. >1.7uU/ml, median OS (95%CI), 12.3 (8.9–15.7 months) vs. 7.3 months (5.4–9.2 months); p = 0.003; Fig 2A) and fT4 (fT4≤1.66 vs. >1.66ng/dl, median OS (95%CI), 10.6 (7.5–13.6 months) vs. 3.3 months (2.2–4.3 months); p = 0.007; Fig 2B) were associated with OS. Patients with manifest hyperthyroid status (n = 17) had worse survival compared to those with normal fT4 levels (i.e., euthyreosis, subclinical hyperthyreosis). The cause of death in patients with hyperthyreosis was tumor progression in 6 patients, liver failure/decompensation in 4 subjects, and unknown in 7 patients. However, thyroid status was not significantly associated with OS, most likely due to small patient numbers in some subgroups.

thumbnail
Table 3. Univariate analysis of prognostic factors (N = 667).

https://doi.org/10.1371/journal.pone.0181878.t003

thumbnail
Fig 2. Kaplan-Meier survival curves.

Overall survival (OS) according to (A) thyroid-stimulating hormone (TSH) levels (TSH≤1.7 vs. >1.7uU/ml, median OS (95%CI), 12.3 (8.9–15.7 months) vs. 7.3 months (5.4–9.2 months); p = 0.003), and (B) free tetraiodthyronine (fT4) levels (fT4≤1.66 vs. >1.66ng/dl, median OS (95%CI), 10.6 (7.5–13.6 months) vs. 3.3 months (2.2–4.3 months); p = 0.007).

https://doi.org/10.1371/journal.pone.0181878.g002

Other variables that were significantly associated with OS included Child-Pugh class, tumor size, Eastern Cooperative Oncology Group (ECOG) performance status (PS), macrovascular invasion (MVI), extrahepatic metastases, first-line therapy, α-fetoprotein (AFP) level, and C-reactive protein (CRP) level. Importantly, thyroid hormone substitution had no impact on OS.

Given that fT4 levels were only available in 313 patients, we did not include both TSH and fT4 in the same multivariate Cox regression model, but analyzed them separately with all other variables that were significantly associated with OS in univariate analysis. Finally, fT4 (HR (95%CI) for fT4>1.66ng/dl, 2.1 (1.3–3.3); p = 0.002) remained an independent prognostic factor for OS (Table 4), while TSH was not significantly associated with OS in multivariate analysis (S4 Table).

After separating patients into early-intermediate (BCLC A-B) and advanced-terminal (BCLC C-D) stage, fT4 remained associated with OS. Median survival in the early-intermediate stage group was 25.3 months (95%CI, 20.1–30.6 months) for fT4 ≤1.66 (n = 97) and 15.5 months (95%CI, 9.2–21.8 months) for fT4 >1.66 (n = 8) (p = 0.043). Similarly, in the advanced-terminal stage group, patients with fT4 ≤1.66 (n = 191) had a significantly (p = 0.038) longer survival of 4.5 months (95%CI, 3.1–5.8 months) compared to 2.8 months (95%CI, 1.5–4.1 months) in patients with fT4 >1.66 (n = 17).

Discussion

In our cohort, 10% of patients had elevated TSH levels similar to the prevalence of hypothyroidism in patients with HCC reported by Hassan et al. (HCC patients vs. controls, 11.7 vs. 8%) [12]. Hypothyroidism has profound effects on metabolism and has therefore been linked to various conditions which either directly constitute an HCC risk factor or have the ability to contribute to the development of known predisposing conditions for HCC, such as obesity [1821], diabetes [2225], non-alcoholic fatty liver disease (NAFLD) [20, 26, 27], and hepatitis C infection [12, 28, 29]. Interestingly, Tarantino and colleagues reported that BMI predicted the presence of spleno-renal shunts and spleno-renal shunts were associated with an increased HCC incidence [30].

However, in our study, high serum TSH concentration was neither associated with type II diabetes mellitus nor BMI. Although etiology of liver disease was not significantly associated with TSH levels, HCV was more frequently observed in patients with high TSH levels (41%) compared to those with normal (29%) or low (20%) TSH. In contrast, Reddy and colleagues [31] found that hypothyroidism was more prevalent in patients with unknown etiology than in those with HCV or alcoholic liver disease. Additionally, we observed NASH only in patients with normal or elevated TSH but not in those with low TSH levels.

Notably, elevated TSH was associated with larger tumors in our study. In contrast, in breast cancer, hypothyroid patients were more likely to be diagnosed with a smaller tumor and at an earlier stage compared to euthyroid patients [32]. Moreover, in prostate cancer, serum T3 was higher in more advanced clinical stage, even though none of the men had levels above the normal range [33].

In terms of survival, patients with higher TSH levels showed a significantly worse outcome in univariate analysis. However, this effect did not hold true upon multivariate analysis when adjusting for other prognostic factors including Child-Pugh class, tumor size, performance status, macrovascular invasion, extrahepatic spread, tumor treatment, AFP, and CRP levels.

Next, we investigated the prevalence of abnormal fT4 levels, available in 313 of 667 patients. Elevated fT4 was found in 25 patients (4% of all patients, n = 667; 8% of those whose fT4 levels where available, n = 313). In comparison, a large study conducted in the United States reported a prevalence of only 0.5% for clinical hyperthyroidism. Notably, they used total instead of free T4 concentrations but adjusted for the common T4-confounders pregnancy and estrogen therapy [34]. In an epidemiologic study by Hassan et al., the prevalence of hyperthyroidism was 1.9% among HCC patients and 1.3% among controls [12].

In our cohort, elevated fT4 concentrations were not associated with etiology of liver disease and variables representing liver function or tumor burden. However, patients with elevated fT4 more frequently had elevated CRP levels which indicate worse prognosis in HCC and could be a reflection of the “inflammatory field effect” [35]. This effect could directly fuel tumor progression as CRP levels are an accepted surrogate marker for the release of Interleukin (IL) -6, an important regulator of CRP secretion, which in turn is associated with both acceleration of HCC development and metastasis [3538]. Additionally, thyroid hormone replacement was more common in patients with increased fT4 levels (fT4≤1.66 vs. >1.66, 6 vs. 32%).

We next investigated the impact of fT4 on the prognosis of HCC patients and found that elevated fT4 levels at the time of HCC diagnosis were significantly associated with poor OS. This effect held true even after adjusting for other known prognostic factors for HCC in a multivariate Cox regression model.

These results are supported by several preclinical studies showing that thyroid hormone signaling promotes tumor invasiveness and metastasis [2, 13, 15, 39, 40], and could be of clinical relevance mainly for two reasons. First, fT4 is a valuable prognostic parameter in HCC that is widely available, non-invasively collectable, and objective. Second, considering both the association between thyroid hormone substitution and elevated fT4 levels as well as the strong prognostic relevance of elevated fT4, our data could suggest some caution when replacing thyroid hormones in patients with established HCC, as hormone replacement-induced fT4 elevation might render tumors more aggressive. This should especially be considered in patients with advanced HCC receiving sorafenib therapy where hypothyroidism often occurs [4143], and might prompt physicians to initiate hormone replacement therapy.

Notably, other studies reported that T3 treatment led to regression of preneoplastic lesions in rodent models of heptocarcinogenesis [44, 45].

The main limitation of this study was the lack of patients with low fT4 levels, which precluded the investigation of effects of overt hypothyroidism on HCC and only allowed analyzing the effect of hypothyroidism using TSH levels. This especially impeded the analysis of the impact of thyroid hormones on HCC with metabolic background. Furthermore, the small sample size of patients with abnormal fT4 levels did not allow subgroup analyses within the BCLC stages and Child-Pugh class. As thyroid hormones may exert opposing effects at different stages of HCC development and progression some effects could evade detection when analysis is performed without differentiation according to tumor stage [2]. However, after grouping patients into early-intermediate (BCLC A-B) and advanced-terminal stage (BCLC C-D), the negative impact of elevated fT4 on survival remained significant in both subgroups. Additionally, non-thyroidal illness syndrome (NTIS), characterized by low serum T3 with normal T4 levels, is associated with HCC and other malignancies [46, 47]. The fact that T3 and fT3 levels were missing in most of our patients represents another potential bias we could not address in our analysis. In light of the stated limitations, this study could not adequately investigate the hypothesis of a dual role of thyroid hormones in HCC [2].

In conclusion, high TSH level was associated with larger tumor size but not with survival when adjusted for known prognostic factors for HCC. Elevation of fT4 resulted in poor survival and remained an independent prognostic factor for OS. These results can be considered as hypothesis-generating paving the path for further work. Prospective studies including clinically hypothyroid patients and subgroup analyses of all tumor stages are needed to further elucidate the role of thyroid hormones in HCC.

Supporting information

S2 Table. Association between TSH and patient, liver, and tumor characteristics in patients with the largest tumor being >5cm.

https://doi.org/10.1371/journal.pone.0181878.s002

(DOCX)

S3 Table. Association between free tetraiodthyronine (fT4) and patient, liver, and tumor characteristics (n = 313).

https://doi.org/10.1371/journal.pone.0181878.s003

(DOCX)

S4 Table. Multivariate analysis of prognostic factors.

https://doi.org/10.1371/journal.pone.0181878.s004

(DOCX)

References

  1. 1. Brown AR, Simmen RC, Simmen FA. The role of thyroid hormone signaling in the prevention of digestive system cancers. Int J Mol Sci. 2013;14(8):16240–57. Epub 2013/08/09. pmid:23924944;
  2. 2. Wu SM, Cheng WL, Lin CD, Lin KH. Thyroid hormone actions in liver cancer. Cell Mol Life Sci. 2013;70(11):1915–36. Epub 2012/09/08. pmid:22955376.
  3. 3. Perra A, Plateroti M, Columbano A. T3/TRs axis in hepatocellular carcinoma: new concepts for an old pair. Endocr Relat Cancer. 2016;23(8):R353–69. Epub 2016/06/30. pmid:27353037.
  4. 4. Hucke F, Sieghart W, Schoniger-Hekele M, Peck-Radosavljevic M, Muller C. Clinical characteristics of patients with hepatocellular carcinoma in Austria—is there a need for a structured screening program? Wien Klin Wochenschr. 2011;123(17–18):542–51. Epub 2011/07/30. pmid:21800047.
  5. 5. Pinter M, Trauner M, Peck-Radosavljevic M, Sieghart W. Cancer and liver cirrhosis: implications on prognosis and management. ESMO Open. 2016;1:e000042. pmid:27843598
  6. 6. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108. Epub 2015/02/06. pmid:25651787.
  7. 7. Hulbert AJ. Thyroid hormones and their effects: a new perspective. Biol Rev Camb Philos Soc. 2000;75(4):519–631. Epub 2000/12/16. pmid:11117200.
  8. 8. Gonzalez-Sancho JM, Garcia V, Bonilla F, Munoz A. Thyroid hormone receptors/THR genes in human cancer. Cancer Lett. 2003;192(2):121–32. Epub 2003/04/02. pmid:12668276.
  9. 9. Pucci E, Chiovato L, Pinchera A. Thyroid and lipid metabolism. Int J Obes Relat Metab Disord. 2000;24 Suppl 2:S109–12. Epub 2000/09/21. pmid:10997623.
  10. 10. Sinha RA, Singh BK, Yen PM. Thyroid hormone regulation of hepatic lipid and carbohydrate metabolism. Trends Endocrinol Metab. 2014;25(10):538–45. Epub 2014/08/17. pmid:25127738.
  11. 11. Frau C, Loi R, Petrelli A, Perra A, Menegon S, Kowalik MA, et al. Local hypothyroidism favors the progression of preneoplastic lesions to hepatocellular carcinoma in rats. Hepatology. 2015;61(1):249–59. Epub 2014/08/27. pmid:25156012.
  12. 12. Hassan MM, Kaseb A, Li D, Patt YZ, Vauthey JN, Thomas MB, et al. Association between hypothyroidism and hepatocellular carcinoma: a case-control study in the United States. Hepatology. 2009;49(5):1563–70. Epub 2009/04/29. pmid:19399911;
  13. 13. Chi HC, Chen SL, Liao CJ, Liao CH, Tsai MM, Lin YH, et al. Thyroid hormone receptors promote metastasis of human hepatoma cells via regulation of TRAIL. Cell Death Differ. 2012;19(11):1802–14. Epub 2012/05/12. pmid:22576662;
  14. 14. Lin YH, Wu MH, Liao CJ, Huang YH, Chi HC, Wu SM, et al. Repression of microRNA-130b by thyroid hormone enhances cell motility. J Hepatol. 2015;62(6):1328–40. Epub 2015/01/27. pmid:25617495.
  15. 15. Wu SM, Huang YH, Yeh CT, Tsai MM, Liao CH, Cheng WL, et al. Cathepsin H regulated by the thyroid hormone receptors associate with tumor invasion in human hepatoma cells. Oncogene. 2011;30(17):2057–69. Epub 2011/01/11. pmid:21217776.
  16. 16. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56(4):908–43. Epub 2012/03/20. pmid:22424438.
  17. 17. Heimbach J, Kulik LM, Finn R, Sirlin CB, Abecassis M, Roberts LR, et al. Aasld guidelines for the treatment of hepatocellular carcinoma. Hepatology. 2017. pmid:28130846.
  18. 18. Asvold BO, Bjoro T, Vatten LJ. Association of serum TSH with high body mass differs between smokers and never-smokers. J Clin Endocrinol Metab. 2009;94(12):5023–7. Epub 2009/10/23. pmid:19846737.
  19. 19. Biondi B. Thyroid and obesity: an intriguing relationship. J Clin Endocrinol Metab. 2010;95(8):3614–7. Epub 2010/08/06. pmid:20685890.
  20. 20. Chung GE, Kim D, Kim W, Yim JY, Park MJ, Kim YJ, et al. Non-alcoholic fatty liver disease across the spectrum of hypothyroidism. J Hepatol. 2012;57(1):150–6. Epub 2012/03/20. pmid:22425701.
  21. 21. Larsson SC, Wolk A. Overweight, obesity and risk of liver cancer: a meta-analysis of cohort studies. Br J Cancer. 2007;97(7):1005–8. Epub 2007/08/19. pmid:17700568;
  22. 22. Brenta G. Why can insulin resistance be a natural consequence of thyroid dysfunction? J Thyroid Res. 2011;2011:152850. Epub 2011/09/24. pmid:21941681;
  23. 23. Vyakaranam S, Vanaparthy S, Nori S, Palarapu S, Bhongir AV. Study of Insulin Resistance in Subclinical Hypothyroidism. Int J Health Sci Res. 2014;4(9):147–53. Epub 2015/01/13. pmid:25580384;
  24. 24. Perros P, McCrimmon RJ, Shaw G, Frier BM. Frequency of thyroid dysfunction in diabetic patients: value of annual screening. Diabet Med. 1995;12(7):622–7. Epub 1995/07/01. pmid:7554786.
  25. 25. El-Serag HB, Tran T, Everhart JE. Diabetes increases the risk of chronic liver disease and hepatocellular carcinoma. Gastroenterology. 2004;126(2):460–8. Epub 2004/02/06. pmid:14762783.
  26. 26. Eshraghian A, Hamidian Jahromi A. Non-alcoholic fatty liver disease and thyroid dysfunction: a systematic review. World J Gastroenterol. 2014;20(25):8102–9. Epub 2014/07/11. pmid:25009382;
  27. 27. Pagadala MR, Zein CO, Dasarathy S, Yerian LM, Lopez R, McCullough AJ. Prevalence of hypothyroidism in nonalcoholic fatty liver disease. Dig Dis Sci. 2012;57(2):528–34. Epub 2011/12/21. pmid:22183820;
  28. 28. Antonelli A, Ferri C, Pampana A, Fallahi P, Nesti C, Pasquini M, et al. Thyroid disorders in chronic hepatitis C. Am J Med. 2004;117(1):10–3. Epub 2004/06/24. pmid:15210382.
  29. 29. Rodriguez-Torres M, Rios-Bedoya CF, Ortiz-Lasanta G, Marxuach-Cuetara AM, Jimenez-Rivera J. Thyroid dysfunction (TD) among chronic hepatitis C patients with mild and severe hepatic fibrosis. Ann Hepatol. 2008;7(1):72–7. Epub 2008/04/01. pmid:18376370.
  30. 30. Tarantino G, Citro V, Conca P, Riccio A, Tarantino M, Capone D, et al. What are the implications of the spontaneous spleno-renal shunts in liver cirrhosis? BMC Gastroenterol. 2009;9:89. pmid:19930687;
  31. 31. Reddy A, Dash C, Leerapun A, Mettler TA, Stadheim LM, Lazaridis KN, et al. Hypothyroidism: a possible risk factor for liver cancer in patients with no known underlying cause of liver disease. Clin Gastroenterol Hepatol. 2007;5(1):118–23. Epub 2006/09/30. pmid:17008133.
  32. 32. Cristofanilli M, Yamamura Y, Kau SW, Bevers T, Strom S, Patangan M, et al. Thyroid hormone and breast carcinoma. Primary hypothyroidism is associated with a reduced incidence of primary breast carcinoma. Cancer. 2005;103(6):1122–8. Epub 2005/02/16. pmid:15712375.
  33. 33. Lehrer S, Diamond EJ, Bajwa AM, Kornreich R, Stagger S, Stone NN, et al. Association between serum triiodothyronine (t3) level and risk of disease recurrence in men with localized prostate cancer. Prostate Cancer Prostatic Dis. 2001;4(4):232–4. Epub 2002/12/24. pmid:12497024.
  34. 34. Hollowell JG, Staehling NW, Flanders WD, Hannon WH, Gunter EW, Spencer CA, et al. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab. 2002;87(2):489–99. Epub 2002/02/12. pmid:11836274.
  35. 35. Sieghart W, Pinter M, Hucke F, Graziadei I, Schoniger-Hekele M, Muller C, et al. Single determination of C-reactive protein at the time of diagnosis predicts long-term outcome of patients with hepatocellular carcinoma. Hepatology. 2013;57(6):2224–34. Epub 2012/09/11. pmid:22961713.
  36. 36. Wan S, Kuo N, Kryczek I, Zou W, Welling TH. Myeloid cells in hepatocellular carcinoma. Hepatology. 2015;62(4):1304–12. Epub 2015/04/29. pmid:25914264;
  37. 37. Maeda S, Kamata H, Luo JL, Leffert H, Karin M. IKKbeta couples hepatocyte death to cytokine-driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell. 2005;121(7):977–90. Epub 2005/07/02. pmid:15989949.
  38. 38. Rossi JF, Lu ZY, Jourdan M, Klein B. Interleukin-6 as a therapeutic target. Clin Cancer Res. 2015;21(6):1248–57. Epub 2015/01/16. pmid:25589616.
  39. 39. Chen RN, Huang YH, Lin YC, Yeh CT, Liang Y, Chen SL, et al. Thyroid hormone promotes cell invasion through activation of furin expression in human hepatoma cell lines. Endocrinology. 2008;149(8):3817–31. Epub 2008/05/10. pmid:18467449;
  40. 40. Lin KH, Shieh HY, Hsu HC. Negative regulation of the antimetastatic gene Nm23-H1 by thyroid hormone receptors. Endocrinology. 2000;141(7):2540–7. Epub 2000/06/30. pmid:10875256.
  41. 41. Riesenbeck LM, Bierer S, Hoffmeister I, Kopke T, Papavassilis P, Hertle L, et al. Hypothyroidism correlates with a better prognosis in metastatic renal cancer patients treated with sorafenib or sunitinib. World J Urol. 2011;29(6):807–13. Epub 2010/12/15. pmid:21153827.
  42. 42. Ahmadieh H, Salti I. Tyrosine kinase inhibitors induced thyroid dysfunction: a review of its incidence, pathophysiology, clinical relevance, and treatment. Biomed Res Int. 2013;2013:725410. Epub 2013/11/28. pmid:24282820;
  43. 43. Schmidinger M, Vogl UM, Bojic M, Lamm W, Heinzl H, Haitel A, et al. Hypothyroidism in patients with renal cell carcinoma: blessing or curse? Cancer. 2011;117(3):534–44. Epub 2010/09/17. pmid:20845482.
  44. 44. Ledda-Columbano GM, Perra A, Loi R, Shinozuka H, Columbano A. Cell proliferation induced by triiodothyronine in rat liver is associated with nodule regression and reduction of hepatocellular carcinomas. Cancer Res. 2000;60(3):603–9. Epub 2000/02/17. pmid:10676643.
  45. 45. Perra A, Kowalik MA, Pibiri M, Ledda-Columbano GM, Columbano A. Thyroid hormone receptor ligands induce regression of rat preneoplastic liver lesions causing their reversion to a differentiated phenotype. Hepatology. 2009;49(4):1287–96. Epub 2008/12/31. pmid:19115221.
  46. 46. Srivastava J, Robertson CL, Gredler R, Siddiq A, Rajasekaran D, Akiel MA, et al. Astrocyte Elevated Gene-1 (AEG-1) Contributes to Non-thyroidal Illness Syndrome (NTIS) Associated with Hepatocellular Carcinoma (HCC). J Biol Chem. 2015;290(25):15549–58. Epub 2015/05/07. pmid:25944909;
  47. 47. Warner MH, Beckett GJ. Mechanisms behind the non-thyroidal illness syndrome: an update. J Endocrinol. 2010;205(1):1–13. Epub 2009/12/18. pmid:20016054.