Open Access

B‑Raf mutation and papillary thyroid carcinoma patients (Review)

  • Authors:
    • Lixin Jiang
    • Haidi Chu
    • Haitao Zheng
  • View Affiliations

  • Published online on: March 1, 2016     https://doi.org/10.3892/ol.2016.4298
  • Pages: 2699-2705
  • Copyright: © Jiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Thyroid carcinoma is the most prevalent endocrine neoplasm globally. In the majority of thyroid carcinoma cases, a positive prognosis is predicted following administration of the appropriate treatment. A wide range of genetic alterations present in thyroid carcinoma exert their oncogenic actions partially through the activation of the mitogen‑activated protein kinase pathway, with the B‑Raf mutation in particular being focused on by experts for decades. The B‑Raf gene has numerous mutations, however, V600E presents with the highest frequency. It is believed that the existence of the V600E mutation may demonstrate an association with the clinicopathological characteristics of patients, however, inconsistencies remain in the literature. A number of explanatory theories have been presented in order to resolve these discrepancies. Recently, it has been suggested that the V600E mutation may function as a target in a novel approach that may aid the diagnosis and prognosis of thyroid carcinoma, with a number of vying methods put forward to that effect. The current review aims to assist researchers in further understanding the possible association between B‑Raf mutations and thyroid carcinoma.

Introduction

Thyroid cancer accounts for 1% of all epithelial malignancies worldwide, and it is currently understood to be the most frequently occurring endocrine neoplasm (1). The incidence of the disease continues to rise, with papillary thyroid cancer (PTC) being the most prevalent histological subtype of thyroid malignancy (2). It is currently unknown as to whether the increase in PTC occurrence is a valid result or a false-positive result possibly occurring due to improved diagnostic techniques and other procedures, or due to increased screening methods for small nodules (3,4).

The majority of thyroid cancers have a positive prognosis following administration of the appropriate treatment. Such treatments may include surgery, and adjuvant radioactive iodine and thyroid-stimulating hormone (TSH) suppression therapy. However, in the literature, following initial treatment, the recurrence rate of differentiated thyroid cancer increased to 30% and the mortality rate was 8% at 30 years of follow-up (5,6).

The Ras/Raf/mitogen-activated protein kinase (MAPK) kinase/MAPK/extracellular signal-regulated kinase (Ras/Raf/MEK/MAPK/ERK) signaling pathway serves a fundamental role in cell cycling, proliferation and survival, and if there is constitutive activation of the MAPK/ERK pathway, it functions in the upregulation of cell division and proliferation, and the subsequent tumorigenesis (79). The MAPK/ERK pathway is an important mechanism in the initiation and progression of human cancer, with mutagenic variations of Ras, B-Raf and rearranged during transfection (RET)/PTC all reported to be associated with papillary thyroid tumorigenesis (9).

B-Raf and B-RafV600E mutation

B-Raf belongs to the Raf kinase family, with a specific mutation, V600E, typically resulting in the constitutive activation of the MAPK/ERK pathway and the subsequent development of PTC. V600E mutation, however, has not yet been identified in other histological subtypes of thyroid cancer, including medullar thyroid cancer and follicular thyroid cancer (1013).

Mutations of B-Raf, a serine-threonine kinase and downstream signaling molecule of Ras and RET, are potent activators of the MAPK/ERK pathway (9,14). B-Raf somatic mutations have previously been reported in a broad range of human cancers, with the highest prevalence observed in melanoma and thyroid cancer (14). One mutation in particular, located on chromosome 7, occurs in the kinase domain of B-Raf and is a T1799A transversion mutation, which results in a single amino acid substitution of valine to glutamic acid (V600E). This V600E mutation accounts for 25–85% of B-Raf activating mutations, dependent on the patient population and histological subtype (15). The B-RafV600E mutation potently increases the kinase activity of B-Raf by evoking the phosphorylation of ERK1/2 at 480-fold higher than the B-Raf wild-type (16). This mutant stimulates constitutive signaling, which may then bypass the requirement for extracellular growth factors. The phosphorylation of downstream MEK1/2 and ERK1/2 results in the expression of a number of genes that are involved in cell proliferation, differentiation, survival, tumorigenesis and the promotion of epithelial-mesenchymal transition (17,18).

The results of the study by Schweppe et al (19), amongst others, are consistent with the possibility that the B-RafV600E mutant, which lies downstream from Ras and RET/PTC, serves a more significant function than RET/PTC and Ras mutants in the activation of the MAPK signaling pathway (19).

Association of B-Raf mutation with clinicopathological PTC characteristics

A previous study reported that in comparison with Ras or RET/PTC mutations, the B-RafV600E mutation was frequently more potent in promoting aggressive pathogenesis and poorer clinicopathological outcomes in patients with PTC (20).

In thyroid cancer, the B-RafV600E mutation has been demonstrated to be associated with tumor recurrence, high-risk clinicopathological characteristics and reduced sensitivity to radioiodine therapy. Conventional factors that indicated high-risk clinicopathological characteristics included the male gender, an increased age, a larger tumor size, the presence of extrathyroidal invasion, local lymph node metastasis, distant metastasis and advanced disease stages (21). Studies have demonstrated a significant correlation between the B-RafV600E mutation and reliable prognostic predictors, including extrathyroidal invasion, lymph nodal metastasis and an advanced tumor-node-metastasis (TNM) stage (2224). Lupi et al (23) reported that PTC patients who presented with a B-RafV600E mutation achieved a 1.5 to 2.1-fold increase in extrathyroidal extension, lymph node metastasis and advanced TNM stages when compared with those who exhibited wild-type B-Raf. Frasca et al (25) observed that the aggressive features of PTC correlated independently with the B-RafV600E mutation when the general data of patients, including gender, age, tumor size, residence, multifocality and histological subtype, were selected for multivariate logistic regression analysis. There is partial agreement over the aforementioned conclusions (26), however, conflicting results have also been reported, stating that no significant association was observed between the B-RafV600E mutation and poorer clinicopathological factors (27,28). Despite this, in a Finish cohort of TNM stage I or II PTC patients following a 16-year follow-up, the presentation of a similar outcome suggested that there was no association between B-RafV600E mutation and PTC recurrence following primary treatment with total thyroidectomy and radioiodine remnant ablation (29). However, the question as to why there are so many differing arguments remains. It has been suggested that the number of cases, the enrollment criteria and the tumor classification involved in these studies may be the reason (30). In B-RafV600E mutation research, it has been reported that B-Raf overexpression or downregulation is a protective event. B-Raf expression serves an important role in benign and malignant thyroid disease, by delaying their development and progression in the absence of activating mutations by at least 10 years (31).

Regarding PTC with a diameter <10 mm (PTMC), typically exhibiting a satisfactory prognosis, a B-Raf mutation is suggested to be more likely to manifest with aggressive clinicopathological characteristics. This may be useful in the evaluation and estimation of the risk stratification and management of PTMC (32). It has also been observed that the B-RafV600E mutation strongly correlates with radioiodine resistance in patients with PTC due to the reduced expression of sodium iodide symporter (NIS) and TSH receptor (TSHR), which results in a reduced capacity for iodine uptake (33).

Subsequently, several alternative published risk factors for the occurrence of thyroid cancer have included radiation exposure, diets low in iodine and certain hereditary conditions, such as multiple endocrine neoplasia type 2A (MEN-2A), MEN-2B and familial medullary thyroid carcinoma (34).

BRAF mutation in association with other factors

Shimamura et al concluded that B-RafV600E itself may not be sufficient for PTC development. This, however, does not mean that B-RafV600E is not the driver mutation, but rather that additional genetic and/or epigenetic changes may be required for full transformation (35). Several other studies also agreed with this theory, stating that the increased expression of several tumor-promoting molecules, including vimentin (36), matrix metalloproteinase (25,3739), nuclear factor-κB (39), Ki-67 (40), prohibitin (41), vascular endothelial growth factor (42) and hepatocyte growth factor receptor (43), is associated with BRAF mutations in carcinoma.

In one previous study, hypermethylation was observed to occur in 33% (76/231) of PTCs, and was also linked with multifocality (40%) and extrathyroidal invasion (40%) (44). It is believed that hypermethylation occurs during the later stages of PTC and is specific to the classical variant; thus, this epigenetic event may be secondary to alternative genetic modifications, and be dependent on tumor type (45). Furthermore, a strong association has been observed between low solute carrier family 5 member 8 (SLC5A8) expression and the presence of the B-RafV600E mutation (45,46) or advanced clinicopathological features (44), suggesting that there is an association with the progression to aggressive PTC.

Hu et al (44) demonstrated that the methylation of several tumor suppressor genes, including SLC5A8, retinoic acid receptor b2, tissue inhibitor of metalloproteinase-3 and death-associated protein kinase, were closely associated with B-Raf mutation in PTC. The inhibition of these particular tumor suppressor genes represented an important molecular mechanism of the B-Raf mutation-induced invasiveness and progression of PTC (44), including lymph node metastasis, extrathyroidal invasion and an advanced tumor stage at diagnosis, and their epigenetic silencing may be an important mechanism by which B-Raf mutation promotes aggressive progression (47,48).

NIS is located in the basal membrane, functioning to absorb and accumulate radioiodine in the cells from the blood stream. The B-Raf mutation in PTC has been frequently associated with the aberrant silencing or decreased expression of thyroid iodide-handling genes, including the genes for NIS thyroid peroxidase, pendrin, thyroglobulin and TSHR (43,4951).

Detection of B-RafV600E as a powerful marker in PTC

Thyroid nodules are common, and with the increasing use of diagnostic imaging, the number of thyroid nodules that are detected is growing, as is the number that require further diagnostic evaluation via fine-needle aspiration biopsy (FNAB). Diagnostic imaging is an office-based, straightforward, safe and sensitive procedure that represents an accepted standard of practice (52).

Only 5% of all thyroid nodules are malignant; therefore, the presurgical determination as to whether nodules are benign or malignant is imperative (52). In the aspirate test of nodules following FNAB, 60–70% are considered benign and 5% are considered malignant, whilst 10–30% are considered as uncertain or suspicious. The indeterminate thyroid nodules on FNABs are grouped into three subcategories: Follicular lesions of undetermined significance, follicular and oncocytic neoplasms, and suspicious nodules for malignancy (53). However, this presents its own limitation. In practice, the use of FNAB cytopathological categorization of indeterminate nodules is required as a definitive diagnosis of malignancy; it necessitates a morphological finding of vascular and/or capsular invasion by the tumor that is solely identifiable in a resected thyroid sample (52).

Recently, FNAB has been utilized in conjunction with molecular testing to improve the accuracy of cytological diagnosis. The current challenge is to develop a highly accurate diagnostic test for early-stage thyroid cancer, and also to produce effective molecular-directed therapies for advanced thyroid cancer. The early detection of cancer, prior to the occurrence of metastasis, is crucial for patients and clinicians, as it improves prognosis and patient quality of life, and may provide additional treatment options. One of the most promising tools for the early detection of cancer are biomarkers (54). Tumor markers may include functional subgroups of proteins (glycoproteins, enzymes and receptors), hormones, molecular markers (genetic mutations, translocations or amplifications) (55), and epigenetic markers (tumor suppressor gene hypermethylation) (56). Beyond their screening and diagnostic value, biomarkers may also be utilized for the estimation of tumor volume, the evaluation of response to treatment or as prognostic indicators of disease progression. Biomarkers therefore represent a non-invasive approach that may provide important information during the diagnosis and follow up of PTC (55,5759). Among the nodules suspected of being cancerous, 20–25% eventually exhibit thyroid cancer following surgery, and therefore, 75–80% of patients in this subgroup undergo a redundant thyroidectomy (53,6062). Following extensive research, particularly in genetic alterations, a number of biomarkers have been used to improve diagnostic accuracy in the cases of undetermined or suspected cytological tests, and mutation detection for B-Raf, RAS, RET/PTC and PAX8/PPARγ mutations in clinical FNAB samples from thyroid nodules may contribute to the cancer diagnosis (63). B-Raf may also be utilized as a tool for the diagnosis and follow-up of PTC. Pupilli et al (64) observed that patients with a histopathological diagnosis of PTC demonstrated a higher percentage of circulating B-RafV600E, as cell-free DNA, compared to those with benign tumors.

Techniques that are utilized for the detection of B-RafV600E mutation include the colorimetric gene detection method, direct DNA sequencing, PCR-based single-strand conformation polymorphism, pyrosequencing, immunohistochemical detection, dual-priming oligonucleotide (DPO)-based multiplex PCR analysis and restriction fragment length analysis, which identifies B-Raf mutations in 2–20% of cells with a wild-type background (30). The liability and diversity of such techniques may contribute to the variability of the outcomes. It has been suggested that the pre-operative acquaintance of B-Raf mutations of the thyroid nodules may aid surgical decisions regarding the extent of surgery required, i.e., a subtotal or total thyroidectomy, and whether neck dissection is necessary. An appropriate surgical plan prior to surgery appears to be possible, particularly for patients presenting with an early stage of thyroid carcinoma, and may subsequently reduce any post-operative complications. The testing of B-Raf mutations has been identified as an independent predictor of treatment failure, and may also be used to estimate the risk of tumor recurrence (22,65,66).

Treatments for PTC patients with B-Raf mutation

Based on the evidence that B-Raf is involved in the development of papillary carcinoma, and also the progression to anaplastic cancer, B-Raf is an appealing target in thyroid cancer. B-RafV600E has been observed to induce thyroid cancer in vivo and thyroid cell transformation in vitro, verifying that this mutation functions as an oncogene for thyroid cancer (67,68).

Since the B-RafV600E mutation is highly prevalent in PTC, particularly in aggressive subtypes (9), recurrent PTC (69) and radioiodine-refractory PTC (22,33,51,70), B-Raf mutation-directed targeting of the MAPK pathway using MEK and Raf inhibitors will be likely to have wide applicability for thyroid cancer (71).

Numerous studies have demonstrated the role of the MAPK signaling pathway in thyroid tumorigenesis, and therefore, B-Raf has become an inviting target (72). Several molecule inhibitors of B-Raf have been developed, including sorafenib, PLX4032, PLX4720, XL281 and RAF265, each with varying selectivity (73). Encouraging results regarding the B-Raf inhibitors PLX4032 and sorafenib have been reported in clinical trials treating malignant melanoma, a disease that has a high frequency of B-Raf mutations (74,75). These drugs exhibit a marked inhibition of cell proliferation, motility, survival and invasion in vitro and in vivo (76,77).

Potent B-RafV600E specific inhibitors are currently available as a novel treatment against melanoma (78). Therapeutic drugs that have been thoroughly tested in various clinical trials and have been approved by the Food and Drug Administration are dabrafenib, vemurafenib and trametinib, all proving to be highly specific for the B-Raf kinase, particularly for the V600E mutant. During phase III clinical trials comparing vemurafenib/dabrafenib and dacarbazine in the treatment of V600E mutant melanomas, each drug demonstrated significant activity in terms of response rate, however, the survival benefits were limited by the rapid acquisition of resistance (79,80). It is likely that B-Raf mutation-targeted treatment using such inhibitors, similar to MEK inhibitors, will be particularly effective for thyroid cancer. However, it has been reported that B-Raf mutation-directed therapy lacks the ability to induce the significant apoptosis of thyroid cancer cells (8184). Consistent with this, Schweppe et al (19) observed that MEK inhibitors inhibited cell proliferation, but not apoptosis. Analogous to MEK inhibitors, these types of inhibitors are also inclined to have a similar limitation, namely a lack of pro-apoptotic effects. Cancer cells are arrested only at the G0/G1 stage of the cell cycle if they do not undergo apoptosis, and tumor growth will persist once the administration of the inhibitors has ceased (85).

Despite a lack of additional supporting studies, it was observed that 38% of B-Raf-positive PTCs demonstrated RET/PTC rearrangement, contrary to previous reports stating that the B-RafV600E mutation does not occur alongside RET/PTC or Ras mutations in cancer. Concurrent RET/PTC and B-Raf mutations have also been reported in PTC. This phenomenon suggests that mutations occurring in the joint pathway of B-Raf and RET/PTC1 may cooperate in PTC development (86,87).

Schweppe et al (19) demonstrated that MEK inhibitors only inhibited cell proliferation and not apoptosis. Such results are consistent with the possibility that the targeting of a limited number of pathways, including the MAPK pathway, may not be sufficient enough to induce apoptosis, based on evidence from several studies (8184). Therefore, given the frequent disturbance in multiple major signaling pathways in thyroid tumorigenesis, this may be an effective therapeutic strategy for thyroid cancer, as previously proposed (20).

Despite an increasing number of clinical trials with selective pathway inhibitors demonstrating promising results in patients with B-RafV600E mutations, acquired resistance to such agents is an emerging problem. It is clear that primary or acquired resistance to B-Raf inhibitors is a clinical problem. Based on previous study data, acquired resistance to B-Raf inhibitors in melanoma has been reported to occur through a number of different mechanisms, resulting in reactivation of the MAPK pathway and/or upregulation of the phosphoinositide 3-kinase pathway (88). The current mechanistic explanations are as follows: i) Intratumoral heterogeneity in the B-Raf genotype, with wild-type and mutant B-Raf within the same tumor; ii) a tumor microenvironment in which stromal cell secretion of hepatocyte growth factor (HGF) results in the activation of the HGF receptor, reactivation of the MAPK and phosphoinositide 3-kinase-AKT signalling pathways and resistance to B-Raf inhibition; iii) the upregulation of specific tyrosine kinase receptors, which has been demonstrated to be a mechanism of primary and acquired resistance to B-Raf inhibitors; and iv) rapid upregulation and activation of the epidermal growth factor receptor caused by B-Raf inhibition (8992).

The clinical effectiveness and safety of the inhibitors tested is typically limited, raising the question of the effectiveness of inhibiting only the MAPK pathway to target resistant and aggressive tumors presenting with B-Raf mutations (21). Therefore, novel concepts are being analyzed, including the combined treatment with dabrafenib and the MEK inhibitor trametinib, delaying the emergence of resistance and resulting in more positive outcomes (93).

There are a number of uncertainties that require further study. Firstly, the association between B-Raf mutations and PTC clinical clinicopathological characteristics does not produce a clear correlation. Secondly, the use of B-Raf inhibitors is a specific treatment for patients presenting with mutations, particularly those with incurable thyroid cancer; however, when and how to use two or more B-Raf inhibitors simultaneously, and the possibility of primary or acquired resistance, are difficult tasks to overcome. Thirdly, the mechanisms of how B-Raf interacts with other signal networks requires further investigation. Finally, how to apply testing for B-Raf mutations in clinical settings, and how to use this as a guideline for surgeons to draft personal treatment strategies are questions that require prolonged future investigation and discussion.

The identification of B-Raf mutations, particularly V600E, is a promising revolution for the research into human cancer. The identification of associations between mutations and cancer may be taken advantage of in order to aid the investigation of thyroid cancer tumorigenesis and possibly the development of appropriate therapies in the future.

References

1 

Pelizzo MR, Dobrinja C, Casal Ide E, Zane M, Lora O, Toniato A, Mian C, Barollo S, Izuzquiza M, Guerrini J, et al: The role of BRAF(V600E) mutation as poor prognostic factor for the outcome of patients with intrathyroid papillary thyroid carcinoma. Biomed Pharmacother. 68:413–417. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Davies L and Welch HG: Increasing incidence of thyroid cancer in the United States, 1973–2002. JAMA. 295:2164–2167. 2006. View Article : Google Scholar : PubMed/NCBI

3 

American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated Thyroid Cancer. Cooper DS, Doherty GM, Haugen BR, Kloos RT, Lee SL, Mandel SJ, Mazzaferri EL, McIver B, Pacini F, et al: Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid. 19:1167–1214. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Pacini F, Schlumberger M, Dralle H, Elisei R, Smit JW and Wiersinga W: European Thyroid Cancer Taskforce: European consensus for the management of patients with differentiated thyroid carcinoma of the follicular epithelium. Eur J Endocrinol. 154:787–803. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Mazzaferri EL and Jhiang SM: Long-term impact of initial surgical and medical therapy on papillary and follicular thyroid cancer. Am J Med. 97:418–428. 1994. View Article : Google Scholar : PubMed/NCBI

6 

Santoro M and Vecchio G: Thyroid cancer: A molecular perspective. Mol Cell Endocrinol. 321:1–2. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Robinson MJ and Cobb MH: Mitogen-activated protein kinase pathways. Curr Opin Cell Biol. 9:180–186. 1997. View Article : Google Scholar : PubMed/NCBI

8 

Kohno M and Pouyssegur J: Targeting the ERK signaling pathway in cancer therapy. Ann Med. 38:200–211. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Xing M: BRAF mutation in thyroid cancer. Endocr Relat Cancer. 12:245–262. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Pizzolanti G, Russo L, Richiusa P, Bronte V, Nuara RB, Rodolico V, Amato MC, Smeraldi L, Sisto PS, Nucera M, et al: Fine-needle aspiration molecular analysis for the diagnosis of papillary thyroid carcinoma through BRAF V600E mutation and RET/PTC rearrangement. Thyroid. 17:1109–1115. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Chiosea S, Nikiforova M, Zuo H, Ogilvie J, Gandhi M, Seethala RR, Ohori NP and Nikiforov Y: A novel complex BRAF mutation detected in a solid variant of papillary thyroid carcinoma. Endocr Pathol. 20:122–126. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Santarpia L, Sherman SI, Marabotti A, Clayman GL and El-Naggar AK: Detection and molecular characterization of a novel BRAF activated domain mutation in follicular variant of papillary thyroid carcinoma. Hum Pathol. 40:827–833. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Namba H, Nakashima M, Hayashi T, Hayashida N, Maeda S, Rogounovitch TI, Ohtsuru A, Saenko VA, Kanematsu T and Yamashita S: Clinical implication of hot spot BRAF mutation, V599E, in papillary thyroid cancers. J Clin Endocrinol Metab. 88:4393–4397. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, et al: Mutations of the BRAF gene in human cancer. Nature. 417:949–954. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Nikiforova MN and Nikiforov YE: Molecular genetics of thyroid cancer: Implications for diagnosis, treatment and prognosis. Expert Rev Mol Diagn. 8:83–95. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Project CG, Jones CM, Marshall CJ, Springer CJ, et al: Cancer Genome Project: Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell. 116:855–867. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Vasko V, Espinosa AV, Scouten W, He H, Auer H, Liyanarachchi S, Larin A, Savchenko V, Francis GL, de la Chapelle A, et al: Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci USA. 104:2803–2808. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Nucera C, Lawler J and Parangi S: BRAF(V600E) and microenvironment in thyroid cancer: A functional link to drive cancer progression. Cancer Res. 71:2417–2422. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Schweppe RE, Kerege AA, Sharma V, Poczobutt JM, Gutierrez-Hartmann A, Grzywa RL and Haugen BR: Distinct genetic alterations in the mitogen-activated protein kinase pathway dictate sensitivity of thyroid cancer cells to mitogen-activated protein kinase kinase 1/2 inhibition. Thyroid. 19:825–835. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Xing M: BRAF mutation in papillary thyroid cancer: Pathogenic role, molecular bases, and clinical implications. Endocr Rev. 28:742–762. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Tang KT and Lee CH: BRAF mutation in papillary thyroid carcinoma: Pathogenic role and clinical implications. J Chin Med Assoc. 73:113–128. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Xing M, Westra WH, Tufano RP, Cohen Y, Rosenbaum E, Rhoden KJ, Carson KA, Vasko V, Larin A, Tallini G, et al: BRAF mutation predicts a poorer clinical prognosis for papillary thyroid cancer. J Clin Endocrinol Metab. 90:6373–6379. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Lupi C, Giannini R, Ugolini C, Proietti A, Berti P, Minuto M, Materazzi G, Elisei R, Santoro M, Miccoli P and Basolo F: Association of BRAF V600E mutation with poor clinicopathological outcomes in 500 consecutive cases of papillary thyroid carcinoma. J Clin Endocrinol Metab. 92:4085–4090. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Kim SJ, Lee KE, Myong JP, Park JH, Jeon YK, Min HS, Park SY, Jung KC, Koo Do H and Youn YK: BRAF V600E mutation is associated with tumor aggressiveness in papillary thyroid cancer. World J Surg. 36:310–317. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Frasca F, Nucera C, Pellegriti G, Gangemi P, Attard M, Stella M, Loda M, Vella V, Giordano C, Trimarchi F, et al: BRAF(V600E) mutation and the biology of papillary thyroid cancer. Endocr Relat Cancer. 15:191–205. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Lee KC, Li C, Schneider EB, Wang Y, Somervell H, Krafft M, Umbricht CB and Zeiger MA: Is BRAF mutation associated with lymph node metastasis in patients with papillary thyroid cancer? Surgery. 152:977–983. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Kim TY, Kim WB, Song JY, Rhee YS, Gong G, Cho YM, Kim SY, Kim SC, Hong SJ and Shong YK: The BRAF mutation is not associated with poor prognostic factors in Korean patients with conventional papillary thyroid microcarcinoma. Clin Endocrinol (Oxf). 63:588–593. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Liu RT, Chen YJ, Chou FF, Li CL, Wu WL, Tsai PC, Huang CC and Cheng JT: No correlation between BRAFV600E mutation and clinicopathological features of papillary thyroid carcinomas in Taiwan. Clin Endocrinol (Oxf). 63:461–466. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Pelttari H, Schalin-Jäntti C, Arola J, Löyttyniemi E, Knuutila S and Välimäki MJ: BRAF V600E mutation does not predict recurrence after long-term follow-up in TNM stage I or II papillary thyroid carcinoma patients. APMIS. 120:380–386. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Li X, Abdel-Mageed AB and Kandil E: BRAF mutation in papillary thyroid carcinoma. Int J Clin Exp Med. 5:310–315. 2012.PubMed/NCBI

31 

Derdas SP, Soulitzis N, Balis V, Sakorafas GH and Spandidos DA: Expression analysis of B-Raf oncogene in V600E-negative benign and malignant tumors of the thyroid gland: Correlation with late disease onset. Med Oncol. 30:3362013. View Article : Google Scholar : PubMed/NCBI

32 

Li F, Chen G, Sheng C, Aaron GM, Huang Y, Lv Z, Xu H, Xing M and Qu S: BRAFV600E mutation in papillary thyroid microcarcinoma: A meta-analysis. Endocr Relat Cancer. 22:159–168. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Durante C, Puxeddu E, Ferretti E, Morisi R, Moretti S, Bruno R, Barbi F, Avenia N, Scipioni A, Verrienti A, et al: BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism. J Clin Endocrinol Metab. 92:2840–2843. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Stephen JK, Chitale D, Narra V, Chen KM, Sawhney R and Worsham MJ: DNA methylation in thyroid tumorigenesis. Cancers (Basel). 3:1732–1743. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Shimamura M, Nakahara M, Orim F, Kurashige T, Mitsutake N, Nakashima M, Kondo S, Yamada M, Taguchi R, Kimura S, Nagayama Y, et al: Postnatal Expression of BRAFV600E does not induce thyroid cancer in mouse models of thyroid papillary carcinoma. Endocrinology. 154:4423–4430. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Watanabe R, Hayashi Y, Sassa M, Kikumori T, Imai T, Kiuchi T and Murata Y: Possible involvement of BRAFV600E in altered gene expression in papillary thyroid cancer. Endocr J. 56:407–414. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Melillo RM, Castellone MD, Guarino V, De Falco V, Cirafici AM, Salvatore G, Caiazzo F, Basolo F, Giannini R, Kruhoffer M, et al: The RET/PTC-RAS-BRAF linear signaling cascade mediates the motile and mitogenic phenotype of thyroid cancer cells. J Clin Invest. 115:1068–1081. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Mesa C Jr, Mirza M, Mitsutake N, Sartor M, Medvedovic M, Tomlinson C, Knauf JA, Weber GF and Fagin JA: Conditional activation of RET/PTC3 and BRAFV600E in thyroid cells is associated with gene expression profiles that predict a preferential role of BRAF in extracellular matrix remodeling. Cancer Res. 66:6521–6529. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Palona I, Namba H, Mitsutake N, Starenki D, Podtcheko A, Sedliarou I, Ohtsuru A, Saenko V, Nagayama Y, Umezawa K and Yamashita S: BRAFV600E promotes invasiveness of thyroid cancer cells through nuclear factor kappaB activation. Endocrinology. 147:5699–5707. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Nakayama H, Yoshida A, Nakamura Y, Hayashi H, Miyagi Y, Wada N, Rino Y, Masuda M and Imada T: Clinical significance of BRAF (V600E) mutation and Ki-67 labeling index in papillary thyroid carcinomas. Anticancer Res. 27:3645–3649. 2007.PubMed/NCBI

41 

Franzoni A, Dima M, D'Agostino M, Puppin C, Fabbro D, Di Loreto C, Pandolfi M, Puxeddu E, Moretti S, Celano M, et al: Prohibitin is overexpressed in papillary thyroid carcinomas bearing the BRAF(V600E) mutation. Thyroid. 19:247–255. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Jo YS, Li S, Song JH, Kwon KH, Lee JC, Rha SY, Lee HJ, Sul JY, Kweon GR, Ro H, et al: Influence of the BRAF V600E mutation on expression of vascular endothelial growth factor in papillary thyroid cancer. J Clin Endocrinol Metab. 91:3667–3670. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Giordano TJ, Kuick R, Thomas DG, Misek DE, Vinco M, Sanders D, Zhu Z, Ciampi R, Roh M, Shedden K, et al: Molecular classification of papillary thyroid carcinoma: Distinct BRAF, RAS, and RET/PTC mutation-specific gene expression profiles discovered by DNA microarray analysis. Oncogene. 24:6646–6656. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Hu S, Liu D, Tufano RP, Carson KA, Rosenbaum E, Cohen Y, Holt EH, Kiseljak-Vassiliades K, Rhoden KJ, Tolaney S, et al: Association of aberrant methylation of tumor suppressor genes with tumor aggressiveness and BRAF mutation in papillary thyroid cancer. Int J Cancer. 119:2322–2329. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Porra V, Ferraro-Peyret C, Durand C, Selmi-Ruby S, Giroud H, Berger-Dutrieux N, Decaussin M, Peix J-L, Bournaud C, Orgiazzi J, et al: Silencing of the tumor suppressor gene SLC5A8 is associated with BRAF mutations in classical papillary thyroid carcinomas. J Clin Endocrinol Metab. 90:3028–3035. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Zane M, Agostini M, Enzo MV, Casal Ide E, Del Bianco P, Torresan F, Merante Boschin I, Pennelli G, Saccani A, Rubello D, et al: Circulating cell-free DNA, SLC5A8 and SLC26A4 hypermethylation, BRAF(V600E): A non-invasive tool panel for early detection of thyroid cancer. Biomed Pharmacother. 67:723–730. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Liu D, Hu S, Hou P, Jiang D, Condouris S and Xing M: Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant. Clin Cancer Res. 13:1341–1349. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Puxeddu E, Durante C, Avenia N, Filetti S and Russo D: Clinical implications of BRAF mutation in thyroid carcinoma. Trends Endocrinol Metab. 19:138–145. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Oler G and Cerutti JM: High prevalence of BRAF mutation in a Brazilian cohort of patients with sporadic papillary thyroid carcinomas: Correlation with more aggressive phenotype and decreased expression of iodide-metabolizing genes. Cancer. 115:972–980. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Mian C, Barollo S, Pennelli G, Pavan N, Rugge M, Pelizzo MR, Mazzarotto R, Casara D, Nacamulli D, Mantero F, et al: Molecular characteristics in papillary thyroid cancers (PTCs) with no 131I uptake. Clin Endocrinol (Oxf). 68:108–116. 2008. View Article : Google Scholar : PubMed/NCBI

51 

Romei C, Ciampi R, Faviana P, Agate L, Molinaro E, Bottici V, Basolo F, Miccoli P, Pacini F, Pinchera A and Elisei R: BRAFV600E mutation, but not RET/PTC rearrangements, is correlated with a lower expression of both thyroperoxidase and sodium iodide symporter genes in papillary thyroid cancer. Endocr Relat Cancer. 15:511–520. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Gómez Sáez JM: Diagnostic and prognostic markers in differentiated thyroid cancer. Curr Genomics. 12:597–608. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Baloch ZW, LiVolsi VA, Asa SL, Rosai J, Merino MJ, Randolph G, Vielh P, DeMay RM, Sidawy MK and Frable WJ: Diagnostic terminology and morphologic criteria for cytologic diagnosis of thyroid lesions: A synopsis of the National Cancer Institute Thyroid Fine-Needle Aspiration State of the Science Conference. Diagn Cytopathol. 36:425–437. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Hayes DF, Bast RC, Desch CE, Fritsche H Jr, Kemeny NE, Jessup JM, Locker GY, Macdonald JS, Mennel RG, Norton L, et al: Tumor marker utility grading system: A framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst. 88:1456–1466. 1996. View Article : Google Scholar : PubMed/NCBI

55 

Kulasingam V and Diamandis EP: Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat Clin Pract Oncol. 5:588–599. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Xing M: Gene methylation in thyroid tumorigenesis. Endocrinology. 148:948–953. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Dalle Carbonare L, Frigo A, Francia G, Davì MV, Donatelli L, Stranieri C, Brazzarola P, Zatelli MC, Menestrina F and Valenti MT: Runx2 mRNA expression in the tissue, serum, and circulating non-hematopoietic cells of patients with thyroid cancer. J Clin Endocrinol Metab. 97:E1249–E1256. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Yu S, Liu Y, Wang J, Guo Z, Zhang Q, Yu F, Zhang Y, Huang K, Li Y, Song E, et al: Circulating microRNA profiles as potential biomarkers for diagnosis of papillary thyroid carcinoma. J Clin Endocrinol Metab. 97:2084–2092. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Milas M, Shin J, Gupta M, Novosel T, Nasr C, Brainard J, Mitchell J, Berber E and Siperstein A: Circulating thyrotropin receptor mRNA as a novel marker of thyroid cancer: Clinical applications learned from 1758 samples. Ann Surg. 252:643–651. 2010.PubMed/NCBI

60 

Haugen BR, Woodmansee WW and McDermott MT: Towards improving the utility of fine-needle aspiration biopsy for the diagnosis of thyroid tumors. Clin Endocrinol (Oxf). 56:281–290. 2002. View Article : Google Scholar : PubMed/NCBI

61 

Sahin M, Gursoy A, Tutuncu NB and Guvener DN: Prevalence and prediction of malignancy in cytologically indeterminate thyroid nodules. Clin Endocrinol (Oxf). 65:514–518. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Letsas KP, Andrikoula M and Tsatsoulis A: Fine needle aspiration biopsy-RT-PCR molecular analysis of thyroid nodules: A useful preoperative diagnostic tool. Minerva Endocrinol. 31:179–182. 2006.PubMed/NCBI

63 

Santoro M, Dathan NA, Berlingieri MT, Bongarzone I, Paulin C, Grieco M, Pierotti MA, Vecchio G and Fusco A: Molecular characterization of RET/PTC3; a novel rearranged version of the RETproto-oncogene in a human thyroid papillary carcinoma. Oncogene. 9:509–516. 1994.PubMed/NCBI

64 

Pupilli C, Pinzani P, Salvianti F, Fibbi B, Rossi M, Petrone L, Perigli G, De Feo ML, Vezzosi V, Pazzagli M, et al: Circulating BRAFV600E in the diagnosis and follow-up of differentiated papillary thyroid carcinoma. J Clin Endocrinol Metab. 98:3359–3365. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Kebebew E, Weng J, Bauer J, Ranvier G, Clark OH, Duh QY, Shibru D, Bastian B and Griffin A: The prevalence and prognostic value of BRAF mutation in thyroid cancer. Ann Surg. 246:466–471. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Zagzag J, Pollack A, Dultz L, Dhar S, Ogilvie JB, Heller KS, Deng FM and Patel KN: Clinical utility of immunohistochemistry for the detection of the BRAF v600e mutation in papillary thyroid carcinoma. Surgery. 154:1199–1205. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Espinosa AV, Porchia L and Ringel MD: Targeting BRAF in thyroid cancer. Br J Cancer. 96:16–20. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Cohen Y, Xing M, Mambo E, Guo Z, Wu G, Trink B, Beller U, Westra WH, Ladenson PW and Sidransky D: BRAF mutation in papillary thyroid carcinoma. J Natl Cancer Inst. 95:625–627. 2003. View Article : Google Scholar : PubMed/NCBI

69 

Henderson YC, Shellenberger TD, Williams MD, El-Naggar AK, Fredrick MJ, Cieply KM and Clayman GL: High rate of BRAF and RET/PTC dual mutations associated with recurrent papillary thyroid carcinoma. Clin Cancer Res. 15:485–491. 2009. View Article : Google Scholar : PubMed/NCBI

70 

Ricarte-Filho JC, Ryder M, Chitale DA, Rivera M, Heguy A, Ladanyi M, Janakiraman M, Solit D, Knauf JA, Tuttle RM, et al: Mutational profile of advanced primary and metastatic radioactive iodine-refractory thyroid cancers reveals distinct pathogenetic roles for BRAF, PIK3CA, and AKT1. Cancer Res. 69:4885–4893. 2009. View Article : Google Scholar : PubMed/NCBI

71 

Hoeflich KP, Herter S, Tien J, Wong L, Berry L, Chan J, O'Brien C, Modrusan Z, Seshagiri S, Lackner M, et al: Antitumor efficacy of the novel RAF inhibitor GDC-0879 is predicted by BRAFV600E mutational status and sustained extracellular signal-regulated kinase/mitogen-activated protein kinase pathway suppression. Cancer Res. 69:3042–3051. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Saji M and Ringel MD: The PI3K-Akt-mTOR pathway in initiation and progression of thyroid tumors. Mol Cell Endocrinol. 321:20–28. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Montagut C and Settleman J: Targeting the RAF-MEK-ERK pathway in cancer therapy. Cancer Lett. 283:125–34. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Gupta-Abramson V, Troxel AB, Nellore A, Puttaswamy K, Redlinger M, Ransone K, Mandel SJ, Flaherty KT, Loevner LA, O'Dwyer PJ and Brose MS: Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol. 26:4714–4719. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Smalley KS: PLX-4032, a small-molecule B-Raf inhibitor for the potential treatment of malignant melanoma. Curr Opin Investig Drugs. 11:699–706. 2010.PubMed/NCBI

76 

Salerno P, De Falco V, Tamburrino A, Nappi TC, Vecchio G, Schweppe RE, Bollag G, Santoro M and Salvatore G: Cytostatic activity of adenosine triphosphate-competitive kinase inhibitors in BRAF mutant thyroid carcinoma cells. J Clin Endocrinol Metab. 95:450–455. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Xing J, Liu R, Xing M and Trink B: The BRAFT1799A mutation confers sensitivity of thyroid cancer cells to the BRAFV600E inhibitor PLX4032 (RG7204). Biochem Biophys Res Commun. 404:958–962. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, et al: Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc Natl Acad Sci USA. 105:3041–3046. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, et al: BRIM-3 Study Group: Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 364:2507–2516. 2011. View Article : Google Scholar : PubMed/NCBI

80 

Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, Millward M, Rutkowski P, Blank CU, Miller WH Jr, Kaempgen E, et al: Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet. 380:358–365. 2012. View Article : Google Scholar : PubMed/NCBI

81 

Ball DW, Jin N, Rosen DM, Dackiw A, Sidransky D, Xing M and Nelkin BD: Selective growth inhibition in BRAF mutant thyroid cancer by the mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244. J Clin Endocrinol Metab. 92:4712–4718. 2007. View Article : Google Scholar : PubMed/NCBI

82 

Liu D, Liu Z, Jiang D, Dackiw AP and Xing M: Inhibitory effects of the mitogen-activated protein kinase kinase inhibitor CI-1040 on the proliferation and tumor growth of thyroid cancer cells with BRAF or RAS mutations. J Clin Endocrinol Metab. 92:4686–4695. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Leboeuf R, Baumgartner JE, Benezra M, Malaguarnera R, Solit D, Pratilas CA, Rosen N, Knauf JA and Fagin JA: BRAFV600E mutation is associated with preferential sensitivity to mitogen-activated protein kinase kinase inhibition in thyroid cancer cell lines. J Clin Endocrinol Metab. 93:2194–2201. 2008. View Article : Google Scholar : PubMed/NCBI

84 

Liu D and Xing M: Potent inhibition of thyroid cancer cells by the MEK inhibitor PD0325901 and its potentiation by suppression of the PI3K and NF-kappaB pathways. Thyroid. 18:853–864. 2008. View Article : Google Scholar : PubMed/NCBI

85 

Xing M: Genetic-targeted therapy of thyroid cancer: A real promise. Thyroid. 19:805–809. 2009. View Article : Google Scholar : PubMed/NCBI

86 

Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE and Fagin JA: 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. 63:1454–1457. 2003.PubMed/NCBI

87 

Musholt TJ, Schönefeld S, Schwarz CH, Watzka FM, Musholt PB, Fottner C, Weber MM, Springer E and Schad A: Impact of pathognomonic genetic alterations on the prognosis of papillary thyroid carcinoma. ESES vienna presentation. Langenbecks Arch Surg. 395:877–883. 2010. View Article : Google Scholar : PubMed/NCBI

88 

Sullivan RJ and Flaherty KT: Resistance to BRAF-targeted therapy in melanoma. Eur J Cancer. 49:1297–1304. 2013. View Article : Google Scholar : PubMed/NCBI

89 

Guerra A, Sapio MR, Marotta V, Campanile E, Rossi S, Forno I, Fugazzola L, Budillon A, Moccia T, Fenzi G and Vitale M: The primary occurrence of BRAF(V600E) is a rare clonal event in papillary thyroid carcinoma. J Clin Endocrinol Metab. 97:517–524. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J, Frederick DT, et al: Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 487:500–504. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Corcoran RB, Ebi H, Turke AB, Coffee EM, Nishino M, Cogdill AP, Brown RD, Della Pelle P, Dias-Santagata D, Hung KE, et al: EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. Cancer Discov. 2:227–235. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, Beijersbergen RL, Bardelli A and Bernards R: Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 483:100–103. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Flaherty KT, Infante JR, Daud A, Gonzalez R, Kefford RF, Sosman J, Hamid O, Schuchter L, Cebon J, Ibrahim N, et al: Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med. 367:1694–703. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2016
Volume 11 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang L, Chu H and Zheng H: B‑Raf mutation and papillary thyroid carcinoma patients (Review). Oncol Lett 11: 2699-2705, 2016
APA
Jiang, L., Chu, H., & Zheng, H. (2016). B‑Raf mutation and papillary thyroid carcinoma patients (Review). Oncology Letters, 11, 2699-2705. https://doi.org/10.3892/ol.2016.4298
MLA
Jiang, L., Chu, H., Zheng, H."B‑Raf mutation and papillary thyroid carcinoma patients (Review)". Oncology Letters 11.4 (2016): 2699-2705.
Chicago
Jiang, L., Chu, H., Zheng, H."B‑Raf mutation and papillary thyroid carcinoma patients (Review)". Oncology Letters 11, no. 4 (2016): 2699-2705. https://doi.org/10.3892/ol.2016.4298