Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3540 2024-01-17 19:21:23 |
2 update references and layout -1127 word(s) 2413 2024-01-18 03:04:42 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Cortas, C.; Charalambous, H. Radioactive Iodine Refractory Differentiated Thyroid Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/53992 (accessed on 06 July 2024).
Cortas C, Charalambous H. Radioactive Iodine Refractory Differentiated Thyroid Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/53992. Accessed July 06, 2024.
Cortas, Christos, Haris Charalambous. "Radioactive Iodine Refractory Differentiated Thyroid Cancer" Encyclopedia, https://encyclopedia.pub/entry/53992 (accessed July 06, 2024).
Cortas, C., & Charalambous, H. (2024, January 17). Radioactive Iodine Refractory Differentiated Thyroid Cancer. In Encyclopedia. https://encyclopedia.pub/entry/53992
Cortas, Christos and Haris Charalambous. "Radioactive Iodine Refractory Differentiated Thyroid Cancer." Encyclopedia. Web. 17 January, 2024.
Radioactive Iodine Refractory Differentiated Thyroid Cancer
Edit

Patients with differentiated thyroid cancer usually present with early-stage disease and undergo surgery followed by adjuvant radioactive iodine ablation, resulting in excellent clinical outcomes and prognosis.

thyroid cancer differentiated thyroid cancer radioactive iodine refractory

1. Introduction

Thyroid cancer (TC) is the most common malignancy of the endocrine system, and it is estimated that approximately 585,000 new cases were diagnosed globally in 2020, which is about 3% of all cancer cases [1]. Around 75% of patients that are diagnosed with TC are women, with an incidence rate of 10.1/100,000 females per year whereas the incidence rate for males is 3.1/100,000, annually. Over the last three decades there has been a worldwide increase in the incidence of thyroid cancer which may be partly the result of overdiagnosis due to increased use of radiological investigations, including neck ultrasound and cross-sectional imaging; however there is also evidence of an increase in larger clinically significant tumors [2].
TC is divided according to the cell of origin, if it originates from follicular epithelial cells or parafollicular C cells. Parafollicular C cells give rise to medullary thyroid cancer, whereas follicular cells give rise to four histological types: papillary thyroid cancer (PTC), which accounts for 80–85% of TC, follicular thyroid cancer (FTC) accounts for 10–15%, poorly-differentiated thyroid cancer (PDTC) accounts for 1–2%, and anaplastic thyroid cancer (ATC), which accounts for less than <2%. Well-differentiated thyroid cancers (DTC) include PTC and FTC, as well as subtypes like follicular–oncocytic thyroid carcinomas (FTC-OV) and Hurthle cell carcinoma. As DTC accounts for most TCs, researchers are going to discuss systemic treatment options for DTC [3].
DTC is mainly diagnosed at an early stage and is treated with surgery with or without adjuvant radioactive iodine (RAI) treatment and TSH suppression. DTC is one of the most curable cancers, with excellent long-term prognosis, but up to 20% of patients develop local or regional recurrences, and up to 10% develop distant metastases [4]. With longer follow ups, up to 35% of patients with DTC can recur after 40 years, with 20% failing to concentrate iodine, resulting in overall close to 10% of patients dying of their disease [5][6][7]. A study looking at the SEER database showed that independent predictive factors for distant metastasis are tumor size, age at diagnosis, thyroidectomy, node positive disease, T3-T4 stage, and histopathology for female DTC patients [8].
It is worth pointing out that even patients with metastatic disease have a much better prognosis compared with other solid tumors, with the 10-year survival being about 45% due to the use of RAI for metastatic disease. A little more than half of patients with metastatic disease, around 60%, will eventually develop disease refractory to RAI, because cancer cells lose their ability to concentrate iodine. Resistance to RAI is a poor prognostic factor and once patients develop RAIR TC, the five-year overall survival is quoted to decrease down to 10% [9].

2. Molecular Biology of Differentiated Thyroid Cancer

In the pathogenesis of DTC, somatic mutations are involved affecting the RAS (Rat Sarcoma Virus)/BRAF (B-Raf proto-oncogene, Serine/Threonine Kinase), MAPK (Mitogen-Activated Protein Kinase) pathway, and PI3K (phosphatidylinositol 3-kinase)/Akt (Protein Kinase B) pathways. In PTC, beyond mutations in BRAF and RAS, fusions of the RET (Rearranged during Transfection) and NTRK (Neurotrophic Tyrosine Receptor Kinase) transmembrane proteins are also seen. In FTC, the most common events are mutations to RAS and PAX8/PPARy (Paired Box 8/Peroxisome Proliferator-Activated Receptors) rearrangements. During cancer development, proliferation and dedifferentiation, additional mutations that affect the PI3K-AKT pathway, are also generated [10].
Data from the Cancer Genome Atlas (TCGA) project, published in 2014, identified a large number of tumor driver mutations [11]. In fact, in only 3.5% of PTC cases there were no putative pathologic mutations identified. Known driver mutations identified include RAS, RET/PTC, TP53, TRK, PTEN, β-catenin, PAX8/PPARy, BRAF, PIK3CA, BRAF/AKAP9, AKT1, ETV6/NTRK3, and STRN/ALK [12]. According to the TCGA, driver mutations are mostly point mutations accounting for 75% of all cases, while approximately 15% are gene fusions and the rest are gene amplifications.
The TCGA provides evidence that BRAF alterations are the most common causative molecular event in PTCs, and it is found in approximately 58.5% of PTC cases [11]. BRAF V600E is the most common mutation found in the BRAF gene. BRAF mutations are also associated with worse prognosis in terms of overall survival, need of re-operation, and higher risk of local recurrence in patients older than 65 years old [13][14][15]. Also, BRAF mutations are thought to lead to loss of the ability of follicular cells to concentrate iodine uptake, hence leading to dedifferentiated RAIR disease [15][16][17][18]. Finally, it should be borne in mind that BRAF mutations on nodules with inconclusive cytology results from FNA are rarely found in benign neoplasms, since 99% of nodules with BRAF mutations are positive for malignancy on final histopathology [19].
The second most common genetic alteration in DTC are point mutations in RAS family genes (KRAS, NRAS, and HRAS). NRAS mutations are the most common in TC and they are predominantly alterations in exons 12 and 61 [20]. Mutations in RAS genes are identified as driver mutations in approximately 12.7% of cases according to TCGA. They are found especially with follicular histology, and sometimes in patients with PTC too [11]. Most of the cases of RAS-mutated FTCs are follicular variant PTCs. RAS mutation is also found in follicular adenomas of the thyroid, which have a high risk of progressing to malignancy. This suggests that RAS mutations are an early event in the carcinogenesis pathways, and further mutational events are needed for cancer development [20].
RET/PTC rearrangements are accounted for as the causative genetic alteration in 6.3% of PTC, according to TCGA [10]. RET is a transmembrane protein with tyrosine kinase activity to its intracellular domain which regulates both RAS and PI3K-AKT pathways. During RET/PTC rearrangement the protein product remains in a constant activated form, leading to enhanced cell proliferation. They are the result of chromosomal rearrangements and the most common are paracentric chromosomal inversions leading to RET/PTC1 and RET/PTC3 [21]. The prevalence of these mutations is higher in younger patients [22]. RET/PTC is directly correlated with exposure to ionizing radiation, based on data gathered after the Chernobyl nuclear factory accident [23]. RET/PTC rearrangements are more common in patients younger than 45 years old [24].
Another genomic alteration in PTC is the amplification of PIK3CA, present in 53.1% of patients [25]. PIK3CA amplification leads to enhanced signaling of PI3K, which has a fundamental role in thyroid cancer carcinogenesis. PIK3CA is normally activated by tyrosine kinase proteins or by RAS signaling [26]. There are also mutations in PIK3CA in 2% of patients diagnosed with PTC. They are more frequent in FTC, poorly differentiated, anaplastic, and undifferentiated TCs, reaching 10–15% [25][27].
Telomerase reverse transcriptase (TERT) mutations and TP53 mutations were also identified in patients with DTC. TERT mutations lead to amplified telomerase activity and enhanced proliferative potential of cancer cells. TERT mutations are found more frequently in poorly differentiated TC and only account for 1% of DTC, however they are associated with poor prognosis and higher risk of relapse [12]. A synergistic effect as a poor prognostic factor was observed between TERT, RAS, and BRAF mutations, leading to tumors with more aggressive behaviors [28].
TRK fusions are found in 2% of adult patients diagnosed with PTC but are higher in pediatric and young adult PTCs, where they account for about 6–15% of all PTCs [29][30][31]. TRK is normally expressed in embryogenesis and in adult life it takes part in neurological functions such as pain, proprioception, appetite, and memory. TRK fusions lead to overexpression of the chimeric protein, resulting in constitutively active, ligand-independent downstream signaling which promotes carcinogenesis.
ALK fusions are found in about 0.8% of PTCs. Case reports of RAIR thyroid cancer treated with Crizotinib are described in the literature. The most typical histologic appearance of ALK fusion-positive thyroid cancer is PTC, however, it is found with increased relative incidence in up to 4–9% of patients with PDTC [32].
Finally, PAX8/PPARy rearrangements are the result of a fusion of PAX8, which is a transcription factor, with the PPARy gene. This mutation is mainly found in FTC and in a follicular variant of PTC. PAX8/PPARy rearrangements have an incident rate of 30–35% in patients with FTC [33].

3. Radioactive Iodine Treatment (RAI) and Radioactive Iodine Refractory (RAIR) Disease

The hallmark of treatment of DTC in patients with localized and locally advanced disease remains surgery followed by RAI treatment and TSH suppression. RAI is part of the adjuvant treatment for patients with high-risk features. In the adjuvant setting, RAI ablates both residual cancer cells to reduce the risk of cancer recurrence and also ablates remnant thyroid tissue to facilitate follow up with thyroglobulin monitoring [34]. Post-RAI treatment whole body iodine imaging should be assessed in all cases of RAI treatment to evaluate efficacy and evidence of residual thyroid cancer.
RAI is also the standard first-line treatment for recurrent or metastatic disease. The treatment outcome and response to RAI treatment should be assessed with an iodine whole body scan. However, in up to 15% of patients, RAI treatment is no longer effective as a result of loss of the expression of the sodium iodide symporter (NIS), which occurs following loss of thyroid differentiation [35]. This results in no RAI uptake in the post-RAI scan. In patients with iodine-negative post-treatment scans, but with strong clinical or radiological suspicion of recurrence of metastatic disease, there is the option of undertaking an FDG PET/CT scan, because FDG PET/CT scans can identify lesions from tissues that are non-iodine avid [36]. In these patients, the disease is classified as RAI Refractory (RAIR) disease.
There is no global agreement regarding the definition of RAIR. It has been proposed that RAIR can occur in any of the following four conditions: (a) the absence of uptake of RAI in all lesions on scintigraphy, (b) the absence of RAI uptake in some but not all lesions, (c) progression despite uptake of RAI, or (d) reaching the maximum recommended activity of RAI [37].
On the cellular level, iodine is used by follicular cells to synthesize thyroid hormones and enters the cell by passing through the basal membrane, making use of the sodium iodine symporter mechanism (NIS). Once iodine is inside the thyroid follicle, it gets oxidized by TPO (thyroid peroxidase) and then is further processed by TG (thyroglobulin) before becoming T3 and T4 hormones. The whole process is regulated by the thyroid stimulating hormone (TSH) which binds on the TSH receptor (TSH-R). TSH-R activates adenylyl cyclase, resulting in the accumulation of cyclic AMP (cAMP) within thyroid cells. cAMP induces NIS transcription by stimulating thyroid-specific transcriptional factors (TTFs) including paired box 8 (PAX8). Activation of both MAPK/ERK and PI3K/AKT pathways results in inhibition of TTFs and loss of NIS expression [35]. The BRAF V600E mutation has been correlated to the loss of NIS expression and RAIR [38]. BRAF activation represses PAX8 binding to the NIS promoter, and results in dysregulation of involved proteins in this process e.g., NIS, TPO, TG, TSH-R, and thus results in the emergence of RAIR TC.

4. Localized Treatment and Timing of Initiation of Systemic Therapy for RAIR DTC

Not all patients with a rising thyroglobulin and a negative diagnostic iodine scan have RAIR metastatic disease. These patients require further investigations, including conventional neck, brain, thorax, abdomen, and bone imaging, while, increasingly, PET scans are being used to detect recurrent or metastatic disease. Causes of a false negative iodine scan also need to be excluded; the TSH level at the time of the diagnostic scan must be elevated to or above 30 mU/L, and iodine contamination (e.g., history of recent iodine contrast radiography) or a high iodine diet also need to be excluded [39]. Finally, some centers, to be absolutely sure that this is iodine refractory disease, advocate the use of high-dose iodine therapy in patients with raised thyroglobulin and a negative diagnostic iodine scan [39].
Once RAIR has been confirmed, all patients with recurrent RAIR disease, and in the absence of specific contraindications, should have TSH suppression aimed at a serum level of <0.1 μIU/mL [40]. In a small minority of patients with RAIR and isolated recurrence, confirmed on PET to be in the neck or with single or very limited oligometastatic disease, an attempt to offer surgery to render the patient disease-free may be indicated and is normally reserved for patients with excellent performance status and lack of significant comorbidity [40][41].
For patients with more extensive disease or patients with oligometastatic disease who cannot have surgery, other local modality therapies, e.g., radiotherapy or ablative techniques, may be considered with the aim to obtain disease control or palliate disease-related symptoms. Radiofrequency ablation (RFA) can be considered for solitary lesions, or for lesions causing symptoms and for lesions <2–3 cm in patients not eligible for surgery or requiring extensive resection [41]. Vertebroplasty can also be considered for patients with vertebral metastases. Bisphosphonates or denosumab should be considered in patients with TC and multiple bone metastases [41]. These decisions should be taken jointly at the Multi-Disciplinary Team (MDT) meeting, after discussion with surgical, medical, and radiation oncologists, alongside nuclear medicine physicians and endocrinologists.
For patients with multiple foci of metastatic disease, e.g., multiple lung metastases or multiple bone metastases, the question then becomes as to the timing of the initiation of the systemic therapy. An increase in serum thyroglobulin (Tg) levels in the absence of radiologically evident disease progression should not be used to select patients requiring systemic therapy [41]. Instead, the rate of Tg rise or Tg doubling time should be used to guide frequency of imaging during follow up, in conjunction with tempo of disease as judged by previous imaging. For patients with Tg antibodies, these commonly result in false negative Tg results, but they can also produce false positive Tg results [39], thus, physicians should depend on radiological imaging for monitoring disease activity/tempo of the disease and not make decisions regarding follow up of patients based on the Tg result.
Care should be exercised regarding the timing of the initiation of systemic therapy for patients with RAIR, as current treatments are non-curative, they are aimed at palliation of symptoms and prolongation of survival; these treatments are essentially life-long and are associated with significant toxicity [40][41][42]. Given that many patients may be asymptomatic and with a slow disease tempo, many patients may not need to start systemic therapy immediately, and instead a period of observation would be indicated. This period of observation in some patients may extend for years, as the natural history of DTC is quite variable, with rates of disease progression ranging from a few months to many years. However, for patients with heavy disease burden, more rapid disease tempo, and who are symptomatic from their disease, initiation of systemic therapy with tyrosine kinase inhibitors is appropriate [40][41].

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. Pellegriti, G.; Frasca, F.; Regalbuto, C.; Squatrito, S.; Vigneri, R. Worldwide increasing incidence of thyroid cancer: Update on epidemiology and risk factors. J. Cancer Epidemiol. 2013, 2013, 965212.
  3. Noone, A.M.; Cronin, K.A.; Altekruse, S.F.; Howlader, N.; Lewis, D.R.; Petkov, V.I.; Penberthy, L. Cancer Incidence and Survival Trends by Subtype Using Data from the Surveillance Epidemiology and End Results Program, 1992–2013. Cancer Epidemiol Biomark. Prev. 2017, 264, 632–641.
  4. Schlumberger, M.J. Diagnostic follow-up of well-differentiated thyroid carcinoma: Historical perspective and current status. J. Endocrinol. Investig. 1999, 22, 3–7.
  5. Mazzaferri, E.L.; Kloos, R.T. Clinical review 128: Current approaches to primary therapy for papillary and follicular thyroid cancer. J. Clin. Endocrinol. Metab. 2001, 86, 1447–1463.
  6. Mazzaferri, E.L.; Jhiang, S.M. Long-term impact of initial surgical and medical therapy on papillary and follicular thyroid cancer. Am. J. Med. 1994, 97, 418–428.
  7. Schlumberger, M.J. Papillary and follicular thyroid carcinoma. N. Engl. J Med. 1998, 338, 297–306.
  8. Wang, W.; Shen, C.; Yang, Z. Nomogram individually predicts the risk for distant metastasis and prognosis value in female differentiated thyroid cancer patients: A SEER-based study. Front. Oncol. 2022, 12, 800639.
  9. Haugen, B.R.; Alexander, E.K.; Bible, K.C.; Doherty, G.M.; Mandel, S.J.; Nikiforov, Y.E.; Pacini, F.; Randolph, G.W.; Sawka, A.M.; Schlumberger, M.; et al. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid 2016, 26, 1–133.
  10. Nikiforov, Y.E.; Nikiforova, M.N. Molecular genetics and diagnosis of thyroid cancer. Nat. Rev. Endocrinol. 2011, 7, 569–580.
  11. Agrawal, N.; Akbani, R.; Aksoy, B.A.; Ally, A.; Arachchi, H.; Asa, S.L.; Auman, J.T.; Balasundaram, M.; Balu, S.; Baylin, S.B.; et al. Integrated genomic characterization of papillary thyroid carcinoma. Cell 2014, 159, 676–690.
  12. Marotta, V.; Sciammarella, C.; Colao, A.; Faggiano, A. Application of molecular biology of differentiated thyroid cancer for clinical prognostication. Endocr. Relat. Cancer 2016, 23, 499–515.
  13. Xing, M.; Westra, W.H.; Tufano, R.P.; Cohen, Y.; Rosenbaum, E.; Rhoden, K.J.; Carson, K.A.; Vasko, V.; Larin, A.; Tallini, G.; et al. BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J. Clin. Endocrinol. Metab. 2005, 90, 6373–6379.
  14. Yip, L.; Nikiforova, M.N.; Carty, S.E.; Yim, J.H.; Stang, M.T.; Tublin, M.J.; Lebeau, S.O.; Hodak, S.P.; Ogilvie, J.B.; Nikiforov, Y.E. Optimizing surgical treatment of papillary thyroid carcinoma associated with BRAF mutation. Surgery 2009, 146, 1215–1223.
  15. Howell, G.M.; Carty, S.E.; Armstrong, M.J.; Lebeau, S.O.; Hodak, S.P.; Coyne, C.; Stang, M.T.; McCoy, K.L.; Nikiforova, M.N.; Nikiforov, Y.E.; et al. Both BRAF V600E mutation and older age (≥ 65 years) are associated with recurrent papillary thyroid cancer. Ann. Surg. Oncol. 2011, 18, 3566–3571.
  16. Kimura, E.T.; Nikiforova, M.N.; Zhu, Z.; Knauf, J.A.; Nikiforov, Y.E.; Fagin, J.A. High prevalence of BRAF mutations in thyroid cancer: Genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 2003, 63, 1454–1457.
  17. Knauf, J.A.; Fagin, J.A. Role of MAPK pathway oncoproteins in thyroid cancer pathogenesis and as drug targets. Curr. Opin. Cell Biol. 2009, 21, 296–303.
  18. Soares, P.; Trovisco, V.; Rocha, A.S.; Lima, J.; Castro, P.; Preto, A.; Máximo, V.; Botelho, T.; Seruca, R.; Sobrinho-Simões, M. BRAF mutations and RET/PTC rearrangements are alternative events in the etiopathogenesis of PTC. Oncogene 2003, 22, 4578–4580.
  19. Nikiforova, M.N.; Nikiforov, Y.E. Molecular diagnostics and predictors in thyroid cancer. Thyroid 2009, 19, 1351–1361.
  20. Xing, M. Molecular pathogenesis and mechanisms of thyroid cancer. Nat. Rev. Cancer 2013, 13, 184–199.
  21. Zhu, Z.; Ciampi, R.; Nikiforova, M.N.; Gandhi, M.; Nikiforov, Y.E. Prevalence of RET/PTC rearrangements in thyroid papillary carcinomas: Effects of the detection methods and genetic heterogeneity. J. Clin. Endocrinol. Metab. 2006, 91, 3603–3610.
  22. Witt, R.L.; Ferris, R.L.; Pribitkin, E.A.; Sherman, S.I.; Steward, D.L.; Nikiforov, Y.E. Diagnosis and management of differentiated thyroid cancer using molecular biology. Laryngoscope 2013, 123, 1059–1064.
  23. Nikiforov, Y.E.; Rowland, J.M.; Bove, K.E.; Monforte-Munoz, H.; Fagin, J.A. Distinct pattern of ret oncogene rearrangements in morphological variants of radiation-induced and sporadic thyroid papillary carcinomas in children. Cancer Res. 1997, 57, 1690–1694.
  24. 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.
  25. Abubaker, J.; Jehan, Z.; Bavi, P.; Sultana, M.; Al-Harbi, S.; Ibrahim, M.; Al-Nuaim, A.; Ahmed, M.; Amin, T.; Al-Fehaily, M.; et al. Clinicopathological analysis of papillary thyroid cancer with PIK3CA alterations in a Middle Eastern population. J. Clin. Endocrinol. Metab. 2008, 93, 611–618.
  26. Xing, M. Recent advances in molecular biology of thyroid cancer and their clinical implications. Otolaryngol. Clin. N. Am. 2008, 41, 1135–1146.
  27. Xing, M. Genetic alterations in the phosphatidylinositol-3 kinase/Akt pathway in thyroid cancer. Thyroid 2010, 20, 697–706.
  28. Song, Y.S.; Lim, J.A.; Choi, H.; Won, J.K.; Moon, J.H.; Cho, S.W.; Lee, K.E.; Park, Y.J.; Yi, K.H.; Park, D.J.; et al. Prognostic effects of TERT promoter mutations are enhanced by coexistence with BRAF or RAS mutations and strengthen the risk prediction by the ATA or TNM staging system in differentiated thyroid cancer patients. Cancer 2016, 122, 1370–1379.
  29. Solomon, J.P.; Benayed, R.; Hechtman, J.F.; Ladanyi, M. Identifying patients with NTRK fusion cancer. Ann. Oncol. 2019, 30, viii16–viii22.
  30. Pekova, B.; Sykorova, V.; Dvorakova, S.; Vaclavikova, E.; Moravcova, J.; Katra, R.; Astl, J.; Vlcek, P.; Kodetova, D.; Vcelak, J.; et al. RET, NTRK, ALK, BRAF, and MET Fusions in a Large Cohort of Pediatric Papillary Thyroid Carcinomas. Thyroid 2020, 30, 1771–1780.
  31. Cocco, E.; Scaltriti, M.; Drilon, A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat. Rev. Clin. Oncol. 2018, 15, 731–747.
  32. Panebianco, F.; Nikitski, A.V.; Nikiforova, M.N.; Kaya, C.; Yip, L.; Condello, V.; Wald, A.I.; Nikiforov, Y.E.; Chiosea, S.I. Characterization of thyroid cancer driven by known and novel ALK fusions. Endocr. Relat. Cancer 2016, 26, 803–814.
  33. Santarpia, L.; Myers, J.N.; Sherman, S.I.; Trimarchi, F.; Clayman, G.L.; El-Naggar, A.K. Genetic alterations in the RAS/RAF/mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt signaling pathways in the follicular variant of papillary thyroid carcinoma. Cancer 2010, 116, 2974–2983.
  34. Tuttle, R.M.; Ahuja, S.; Avram, A.M.; Bernet, V.J.; Bourguet, P.; Daniels, G.H.; Dillehay, G.; Draganescu, C.; Flux, G.; Führer, D.; et al. Controversies, Consensus, and Collaboration in the Use of 131I Therapy in Differentiated Thyroid Cancer: A Joint Statement from the American Thyroid Association, the European Association of Nuclear Medicine, the Society of Nuclear Medicine and Molecular Imaging, and the European Thyroid Association. Thyroid 2019, 29, 461–470.
  35. Paladino, S.; Melillo, R.M. Editorial: Novel Mechanism of Radioactive Iodine Refractivity in Thyroid Cancer. J. Natl. Cancer Inst. 2017, 109, djx106.
  36. Feine, U.; Lietzenmayer, R.; Hanke, J.P.; Held, J.; Wöhrle, H.; Müller-Schauenburg, W. Fluorine-18-FDG and iodine-131-iodide uptake in thyroid cancer. J. Nucl. Med. 1996, 37, 1468–1472.
  37. Fugazzola, L.; Elisei, R.; Fuhrer, D.; Jarzab, B.; Leboulleux, S.; Newbold, K.; Smit, J. 2019 European Thyroid Association Guidelines for the Treatment and Follow-Up of Advanced Radioiodine-Refractory Thyroid Cancer. Eur. Thyroid J. 2019, 8, 227–245.
  38. Liu, J.; Liu, Y.; Lin, Y.; Liang, J. Radioactive Iodine-Refractory Differentiated Thyroid Cancer and Redifferentiation Therapy. Endocrinol. Metab. 2019, 34, 215–225.
  39. Mallick, U.K.; Charalambous, H. Current issues in the management of differentiated thyroid cancer. Nucl. Med. Commun. 2004, 25, 873–881.
  40. Filetti, S.; Durante, C.; Hartl, D.; Leboulleux, S.; Locati, L.D.; Newbold, K.; Papotti, M.G.; Berruti, A.; ESMO Guidelines Committee. Electronic address: . Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up †. Ann. Oncol. 2019, 30, 1856–1883.
  41. Filetti, S.; Durante, C.; Hartl, D.M.; Leboulleux, S.; Locati, L.D.; Newbold, K.; Papotti, M.G.; Berruti, A.; ESMO Guidelines Committee. Electronic address: . ESMO Clinical Practice Guideline update on the use of systemic therapy in advanced thyroid cancer. Ann. Oncol. 2022, 33, 674–684.
  42. Haddad, R.I.; Bischoff, L.; Ball, D.; Bernet, V.; Blomain, E.; Busaidy, N.L.; Campbell, M.; Dickson, P.; Duh, Q.Y.; Ehya, H.; et al. Thyroid Carcinoma, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 925–951.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 118
Revisions: 2 times (View History)
Update Date: 18 Jan 2024
1000/1000
Video Production Service