Analysis of somatic mutations in braf, CDKN2A/p16 and PI3KCA in patients with medullary thyroid carcinoma

  • Authors:
    • Fabrício P. Nascimento
    • Mirian G. Cardoso
    • Susan C. Lindsey
    • Ilda S. Kunii
    • Flávia O.F. Valente
    • Marina M.L. Kizys
    • Rosana Delcelo
    • Cléber P. Camacho
    • Rui M.B. Maciel
    • Magnus R. Dias‑Da‑Silva
  • View Affiliations

  • Published online on: December 29, 2015     https://doi.org/10.3892/mmr.2015.4731
  • Pages: 1653-1660
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Medullary thyroid carcinoma (MTC), a neuroendocrine tumor originating from thyroid parafollicular cells, has been demonstrated to be associated with mutations in RET, HRAS, KRAS and NRAS. However, the role of other genes involved in the oncogenesis of neural crest tumors remains to be fully investigated in MTC. The current study aimed to investigate the presence of somatic mutations in BRAF, CDKN2A and PI3KCA in MTC, and to investigate the correlation with disease progression. DNA was isolated from paraffin‑embedded tumors and blood samples from patients with MTC, and the hotspot somatic mutations were sequenced. A total of 2 novel HRAS mutations, p.Asp33Asn and p.His94Tyr, and polymorphisms within the 3' untranslated region (UTR) of CDKN2A (rs11515 and rs3088440) were identified, however, no mutations were observed in other genes. It was suggested that somatic point mutations in BRAF, CDKN2A and PI3KCA do not participate in the oncogenesis of MTC. Further studies are required in order to clarify the contribution of the polymorphisms identified in the 3'UTR of CDKN2A in MTC.

Introduction

Medullary thyroid carcinoma (MTC), a neuroendocrine tumor originating from thyroid parafollicular cells, accounts for ~4% of thyroid cancer cases (1). The majority are sporadic cases, however, 20-25% occur as a hereditary syndrome termed multiple endocrine neoplasia type 2 (MEN 2A and MEN 2B) and as familial MTC, both of which are associated with germline mutations in the RET oncogene (2).

Mutations in the RET oncogene have previously been identified in the tumor tissue of up to 64% of sporadic MTC cases (3). In addition, RAS gene mutations are observed in 10% of RET-negative cases and are associated with a subset of tumors with less aggressive behavior (4). While certain studies identified that ~90% of sporadic MTCs exhibited mutually exclusive mutations in RET, HRAS and KRAS (48), Moura et al (3) reported the presence of the RAS mutation in one case with RET-positive sporadic MTC and Rapa et al (9) identified no RAS mutations in 49 examined cases. Nevertheless, the clinical phenotype of sporadic and inherited MTCs is heterogeneous even in the presence of the same mutation; however the molecular mechanisms underlying the pathology remain to be fully elucidated.

In addition, it remains unclear whether there is a modulatory role in MTC tumor progression for additional genes such as BRAF, CDKN2A and PI3KCA. These genes participate in the tumorigenesis of several types of human malignancies such as tumors derived from neural crest cells, including melanoma, pheochromocytoma and paraganglioma (1012).

BRAF, like RET and RAS, is involved in the mitogen-activated protein kinase pathway and has a well-established role in the pathogenesis of malignancies such as melanoma and papillary thyroid cancer (13). Nevertheless, the contribution in the tumorigenesis of MTC remains controversial. A previous study reported a high prevalence of the p.Val600Glu BRAF mutation in sporadic MTC cases (14); however, subsequent studies did not confirm this observation (3,9,15,16).

An additional tumor suppressor gene, CDKN2A/p16INK4A, is involved in the G1/S transition in the cell cycle. Mutations and deletions have been identified in melanoma, and polymorphisms in its 3′ untranslated region (UTR) have been associated with earlier progression from primary to metastatic disease (17). By contrast, polymorphisms in another tumor suppressor gene, CDKN1B, which is in the same CDKN family, are associated with improved outcomes (18).

Additionally, PI3KCA is a gene that serves an important role in signaling pathways and cell growth, and contributes to tumorigenesis in several types of human malignancy (19,20). However, the role of this gene in the tumorigenesis of MTC remains to be fully understood.

Therefore, the current study aimed to verify the prevalence of somatic mutations in BRAF, CDKN2A and PI3KCA, which have already been described in other neural crest-derived tumors, and to determine the possible supporting role of these genes in the tumorigenesis of MTC.

Patients and methods

Patients and tissue samples

From 128 patients with MTC assessed at the Multiple Endocrine Neoplasia outpatient clinic at the Universidade Federal de Sao Paulo (Sao Paulo, Brazil) between February 2007 and June 2013, formalin-fixed paraffin-embedded (FFPE) tumor tissues were selected from 31 patients on the basis of the availability of tumor tissues, with no other selection criteria. DNA extraction was subsequently performed, using an in-house method as previously described (21). Subsequent to DNA extraction, 20 samples (from 13 males and 7 females; mean age, 40.55±16.74 years) provided the appropriate quantity and quality of DNA. The study was approved by the Ethics and Research Committee of the Universidade Federal de Sao Paulo (protocol number 1945/10), and all patients provided informed consent. Additionally, 1,092 genotypes of variant frequencies (single nucleotide polymorphisms; SNPs) were obtained from the 1000 Genomes database (http://www.1000genomes.org/) as a population genetics control.

DNA extraction and genotyping

DNA from peripheral blood and somatic DNA from 10-µm sections of FFPE MTC tissues was extracted using an in-house method as previously described (21). Polymerase chain reaction (PCR) was performed to amplify DNA corresponding to hotspot exons 2, 3 and 4 of HRAS; 2, 3 and 4 of KRAS; 2 and 3 of NRAS; 15 of BRAF; 9 and 20 of PI3KCA; and exons 2, 3 and the 3′UTR of the CDKN2A gene. The sequences of the primers are listed in Table I. The reactions were performed using 10 pM of each specific primer, 2.5 µl PCR buffer, 200 µM dNTP, 1.5 µM MgCl2 and 0.2 units Taq DNA polymerase (Invitrogen; Thermo Fisher Scientific, Waltham, MA, USA) in a 25-µl total reaction volume. The cycling conditions were as follows: 5 min at 95°C, 38 cycles of 45 sec at 95°C, 45 sec for annealing and 1 min at 72°C, and a final elongation for 10 min at 72°C. The PCR products were purified using the Illustra GFX PCR DNA and Gel Purification kit (GE Healthcare Life Sciences, Chalfont, UK) and were subject to sequencing using the Sanger method, with the Big Dye™ Terminator Cycle Sequencing Ready Reaction kit and the ABI PRISM 3130xl Genetic Analyzer (Applied Biosystems; Thermo Fisher Scientific). Gel electrophoresis of the PCR products was performed to analyze product quality and yield using a 1.8% agarose gel and a DNA ladder.

Table I

Primers used in the present study.

Table I

Primers used in the present study.

GeneForward primerReverse primer
BRAF exon 15 5′-AACTCAGCAGCATCTCAGGG-3′ 5′-CTTCATAATGCTTGCTCTGATAG-3′
CDKN2A exon 1 5′-ACCCTGGCTCTGACCATTC-3′ 5′-CAGGTCACGGGCAGAC-3′
CDKN2A exon 2 5′-GACCTCAGGTTTCTAACGCC-3′ 5′-CATATATCTACGTTAAAAGGCAGGAC-3′
PI3KCA exon 9 5′-TGGCAGTCAAACCTTCTCTC-3′ 5′-GAGAAAGTATCTACCTAAATCCACAGA-3′
PI3KCA exon 20 5′-AAATGTTTTGGTGTTCTTAATTTATTC-3′ 5′-GCAGCCAGAACTCTTTATTTTG-3′
C-kit exon 9 5′-GCCAGGGCTTTTGTTTTCTT-3′ 5′-AGCCTAAACATCCCCTTAAATTG-3′
C-kit exon 11 5′-AACCATTTATTTGTTCTCTCTCCA-3′ 5′-CCACTGGAGTTCCTTAAAGTCA-3′
C-kit exon 17 5′-TGGTTTTCTTTTCTCCTCCAAC-3′ 5′-GGACTGTCAAGCAGAGAATGG-3′
HRAS exon 2 5′-GGCAGGAGACCCTGTAGGAG-3′ 5′-AGCTGCTGGCACCTGGAC-3′
HRAS exon 3 5′-GTCCCTGAGCCCTGTCCTC-3′ 5′-CAGCCTCACGGGGTTCAC-3′
HRAS exon 4 5′-CTCTCGCTTTCCACCTCTCA-3′ 5′-GGGTGGAGAGCTGCCTCA-3′
KRAS exon 2 5′-TTAACCTTATGTGTGACATGTTCTAA-3′ 5′-GGTCCTGCACCAGTAATATGC-3′
KRAS exon 3 5′-AGACTGTGTTTCTCCCTTCTCA-3′ 5′-TGGCATTAGCAAAGACTCAAA-3′
KRAS exon 4 5′-GATATTTGTGTTACTAATGACTGTGCT-3′ 5′-TTATGATTTTGCAGAAAACAGATC-3′
NRAS exon 2 5′-TCGCCAATTAACCCTGATTAC-3′ 5′-TCCGACAAGTGAGAGACAGG-3′
NRAS exon 3 5′-TGGGCTTGAATAGTTAGATGC-3′ 5′-AGTGTGGTAACCTCATTTCCC-3′
In silico analysis of HRAS mutations and CDKN2A polymorphisms

Mutational analysis of HRAS was performed by the use of Project HOPE to obtain structural information from the analysis of PDB-file 1CTQ (22). The in silico analysis for the CDKN2A polymorphisms was performed using the Functional Single Nucleotide Polymorphism database (http://compbio.cs.queensu.ca/F-SNP/) as previously described (23). This database provides information regarding potential deleterious effects of SNPs with respect to splicing, transcription, translation and post-translation based on SNP functional significance (FS). The FS score for neutral SNPs is 0.1764, whereas the FS score for disease-associated SNPs is in the range of 0.5-1.

Statistical analysis

The allele and genotype frequencies were compared between patients with MTC and the 1000 Genomes database controls using a χ2 test. The clinicopathological features of patients carrying each of the polymorphisms rs11515 and rs3088440 were compared with those of patients without such polymorphisms using the χ2 test or the Student's unpaired t-test as appropriate. P<0.05 was considered to indicate a statistically significant difference, and the Hardy-Weinberg equilibrium was evaluated. Statistical analyses were performed using SPSS, version 22.0 (IBM SPSS, Armonk, NY, USA) and GraphPad Prism, version 3.0 (GraphPad Software, Inc., La Jolla, CA, USA).

Results

Screening of the RET, HRAS, KRAS and NRAS genes

Mutational screening of the RET gene was performed on all 20 patients. A total of 10 cases were identified to be familial tumors as confirmed by the presence of a germline mutation. In total, 30% of the sporadic cases (3/10) presented with a RET somatic mutation. The clinicopathological features and molecular analysis, including tumor staging based on the American Joint Committee in Cancer staging system (24), are summarized in Table II.

Table II

Summary of patient clinicopathological features and molecular analysis.

Table II

Summary of patient clinicopathological features and molecular analysis.

PatientGenderAge at diagnosis (y)pTNMaGermline RET alleleSomatic RET alleleSomatic H-, K-, NRAS alleleSomatic CDKN2A
1M28T2N1bMxWTWT HRAS_p.Asp33Asnrs11515
2F25T3N1bMxWTp.Met918Thr-WT
3M38T1N1aMxWTWTNA rs11515/rs3088440
4M56T3N1bMxWTWTWTWT
5F49T2NxMxWTp.Gln681Stop-WT
6M69T2N0MxWTWT HRAS_p.Gln61Argrs3088440
7M27T4N1MxWTWTWTWT
8M51T3N1bMxWTp.Met918Thr-rs11515
9F56T1N1bMxWTWT HRAS_p.Asp33AsnWT
10M41T4N1bMxWTWT HRAS_p.His94TyrWT
11M27T1N1aMxp.Cys634Arg-- rs11515/rs3088440
12F21T1N1aMxp.Gly533Cys--rs11515
13M61T1N1aMxp.Gly533Cys--WT
14F22T2N0Mxp.Cys634Arg-- rs11515/rs3088440
15M43T2N0Mxp.Cys634Arg--rs11515
16M72T1N0Mxp.Cys634Arg--WT
17M45T1N1aMxp.Cys634Arg--rs3088440
18F31T1NxMxp.Cys634Arg--rs3088440
19F15T1N1aMxp.Cys634Arg--rs3088440
20M40T1N0Mxp.Gly533Cys--WT

a TNM (Tumor, Node, Metastasis)/American Joint Committee on Cancer staging system. M, male; F, female; y, years; NA, not available; WT, wild-type.

To investigate exclusive causative mutations in cases of sporadic MTC other than RET mutations, HRAS, KRAS and NRAS were screened for somatic mutations in the hotspots. The majority of these patients had been previously analyzed for RET germline mutations as part of our routine evaluation, and for RET somatic mutations in a previous study (25) Two novel HRAS mutations, p.Asp33Asn and p.His94Tyr, were detected in RET-negative MTC tumors. Mutational analysis using Project HOPE suggests that the p.His94Tyr mutation is deleterious, and that the p.Asp33Asn mutation is likely to be damaging (Fig. 1). No differences in the clinical presentation or histological observations were noted between patients with MTC that had a mutation in the RAS gene (Table II).

No somatic mutations were identified in exon 15 of BRAF or in exons 9 and 20 of PI3KCA. Patient 9 was not analyzed for somatic mutations in PI3KCA due to an insufficient number of tumor samples.

Despite not having identified somatic mutations in CDKN2A hotspots, two polymorphisms in the 3′UTR regulatory region, 500 C→G (rs11515) and 540 C→T (rs3088440), were identified in the patients observed. The heterozygotic pattern of the two SNPs was observed in the same proportion, 7/20 MTC (35%). The genotype distribution was identified to be in the Hardy-Weinberg equilibrium and was not identified to exhibit linkage disequilibrium. To investigate whether the observed polymorphisms were limited to a somatic event, they were further analyzed in the peripheral blood, which confirmed germline inheritance. The in silico analysis demonstrated that the CDKN2A polymorphisms rs11515 and rs3088440 are located in the transcriptional regulatory region and that the nucleotide alterations may affect the binding of transcription factors.

In seven cases, it was possible to detect the presence of these polymorphisms in the secondary tumors in the lymph nodes (tumor metastases), however no differences between the genotypes of the primary and secondary tumors were observed, indicating that there was no additional somatic event in CDKN2A involved in the metastatic process. This analysis was additionally performed for BRAF and PI3KCA in metastatic tissues.

No associations between the polymorphisms and the clinicopathological features observed were identified (Table III). In addition, the frequency of the SNPs was compared with a population genetics control, and there was no significant difference between the two populations (Table IV).

Table III

Correlation between CDKN2A SNPs and clinicopathological features in the patient cohort.

Table III

Correlation between CDKN2A SNPs and clinicopathological features in the patient cohort.

Clinicopathological featurers11515 (n=20)
rs3088440 (n=20)
CC (n=13)CG (n=7)P-valueCC (n=13)CT (n=7)P-value
Gender0.5260.474
 Male (n=13)8/13 (61.5%)5/7 (71.4%)9/13 (69.2%)4/7 (57.1%)
 Female (n=7)5/13 (38.5%)2/7 (28.6%)4/13 (30.7%)3/7 (42.9%)
Age at diagnosis0.088a0.272a
Mean ± SD (y)45.41±17.4931.53±11.3644.062±17.4931.53±11.36
Tumor type0.5000.175
 Sporadic (n=10)7/13 (53.8%)3/7 (42.9%)8/13 (61.5%)2/7 (28.5%)
 Familial (n=10)6/13 (46.1%)4/7 (57.1%)5/13 (38.5%)5/7 (62.5%)
T category0.4640.291
 T17/13 (53.8%)2/7(28.5%)5/13 (38.5%)4/7 (57.1%)
 T22/13 (15.3%)3/7 (42.9%)3/13 (23.1%)2/7 (28.5%)
 T32/13 (15.3%)2/7 (28.5%)3/13 (23.1%)1/7 (14.4%)
 T42/13 (15.3%)0/7 (0%)2/13 (15.3%)0/7 (0%)
Tumor size0.421a0.689a
Mean ± SD (cm)1.954±1.112.34±1.032.315±1.221.671±0.59
 <28/13 (61.5%)2/7(28.6%)0.3787/13 (53.8%)4/7 (57.1%)0.339
 ≥25/13 (38.5%)5/7 (71.4%)6/13 (46.1%)3/7 (42.9%)
Lymph node metastases0.7420.742
 N04/13 (30.8%)5/7 (71.4%)3/13 (23.07%)3/7 (42.9%)
 N19/13 (69.2%)2/7 (28.5%)10/13 (76.9%)4/7 (57.1%)
AJCC stage0.7420.742
 I and II4/13 (30.7%)2/7 (28.5%)4/13 (30.7%)2/7 (28.5%)
 III and IV9/13 (69.2%)5/7 (71.4%)9/13 (69.2%)5/7 (71.4%)

{ label (or @symbol) needed for fn[@id='tfn2-mmr-13-02-1653'] } P-values were obtained using the χ2 test;

a continuous variables analyzed with Student's t-test. SNPs, single nucleotide polymorphisms; SD, standard deviation; y, years; AJCC, American Joint Committee on Cancer.

Table IV

Comparative analysis of the frequency of the non-coding CDKN2A germ line single nucleotide polymorphisms in patients with MTC and the control.

Table IV

Comparative analysis of the frequency of the non-coding CDKN2A germ line single nucleotide polymorphisms in patients with MTC and the control.

A, rs11515
Genotype frequency
Allele frequency
PopulationCCCGGGC (32)G (8)P-value
MTC0.600.40-0.800.200.25
1,000 genomesa0.790.190.020.880.12
B, rs3088440
Genotype frequency
Allele frequency
PopulationCCCTTTC (31)T (9)P-value

MTC0.550.45-0.780.220.65
1,000 genomesa0.730.240.030.850.15

a Sequences obtained from the 1000 Genomes database used as a population control. MTC, medullary thyroid carcinoma. The numbers in parentheses represent the frequency of each allele type in this locus in the studied cohort.

Discussion

The adjuvant role of additional genes in the tumorigenesis of MTC was investigated in the current study through analysis of tumor tissues from 20 patients. Screening in hotspot regions of BRAF, CDKN2A and PI3KCA did not identify any somatic mutations in the coding region. In addition, the results of the current study were not in agreement with the BRAF mutation frequency of 68.2% observed by Goutas et al (14). This suggests that BRAF does not serve an important role in the tumorigenesis of MTC. The observations of the current study concerning MTC are consistent with a previous study that demonstrated that somatic mutations in genes other than RET and RAS are very rare or even absent (5). Notably, the present study identified two novel HRAS mutations.

Additionally, two common polymorphisms in the 3′-UTR non-coding region of the gene CDKN2A were identified, rs11515 and rs3088440 (26). It is known that protein synthesis can be modulated by regulatory elements located in the 5′-UTR and 3′-UTR regions. The 3′-UTR, the site of the polymorphisms identified in the current study, serves an important role in translation and mRNA stability. Alterations in this region may be associated with the onset or progression of disease (27).

These polymorphisms have been investigated in various tumor types including urinary bladder neoplasm (28), esophageal adenocarcinoma (29) and cervical cancer (30) as presented in Table V. The two identified polymorphisms have been previously associated with an earlier progression from primary to metastatic disease in the case of melanoma (17), and rs3088440 was associated with the mechanism of tumor invasion in bladder cancer (28). Controversially, this polymorphism has been previously associated with a sub-group with reduced vertical growth of melanoma and a favorable outcome (31). However, additional studies have not identified a clinical correlation with tumor behavior (30,32,33).

Table V

Summary of the studies on CDKN2A polymorphisms in different tumor types.

Table V

Summary of the studies on CDKN2A polymorphisms in different tumor types.

Source, year (ref)rs11515 (%)rs3088440 (%)Tumor typenSampleMethod used
Sauroja et al, 2000 (17)16.6716.67Melanoma48Frozen/FFPE tissue PCR-SSCP/sequencing
Kumar et al, 2001 (26)2527.27Melanoma229FFPE tissuePCR-SSCP
Sakano et al, 2003 (28)18.112.9Bladder309BloodPCR-SSCP
Geddert et al, 2005 (29)13.3-ADC315FFPE tissuePCR-RFLP
Chansaenroj, et al 2013 (30)7.117.9Cervical56Cervical swabSequencing
Straume et al, 2002 (31)2523Melanoma185FFPE tissue PCR-SSCP/sequencing
Boonstra et al, 2011 (32)22.07-EAC214FFPE tissueSequencing
Pinheiro et al, 2014 (33)21.05-ESCC97FFPE tissueSequencing
15.63-HNSCC96FFPE tissuePCR-RFLP
Jin et al, 2012 (34)-16.7SGC156BloodPCR-RFLP
Polakova et al, 2008 (35)25.9813.07Colorectal612BloodPCR-RFLP
Royds et al, 2011 (36)31.78-GBM107BloodSequencing
Thakur et al, 2012 (37)13.64-Cervical150Fresh tissuePCR-RFLP
Zhang et al, 2011 (38)-17.0SCCHN1,287BloodPCR-RFLP
Zhang et al, 2013 (39)-20.5DTC303BloodPCR-RFLP
-20.9PTC273BloodPCR-RFLP
De Giorgi et al, 2014 (4 0)16.67-Melanoma12BloodSequencing
Song et al, 2014 (41)-33.88SCCOP552BloodPCR-RFLP
Nascimento et al, 2015a3535MTC20FFPE tissue + bloodSequencing

a Indicates the current study. ADC, gastric and esophageal adenocarcinomas; EAC, esophageal adenocarcinoma; ESCC, esophageal squamous cell carcinoma; GBM, glioblastoma multiforme; SCCHN, squamous cell carcinoma of the head and neck; SGC, salivary gland carcinoma; DTC, differentiated thyroid carcinoma; PTC, papillary thyroid cancer; HNSCC, head and neck squamous cell carcinoma; SCCOP, squamous cell carcinoma of the oropharynx; FFPE, formalin-fixed paraffin-embedded; PCR; polymerase chain reaction; SSCP, single-strand conformation polymorphism; RFLP, restriction fragment length polymorphism.

Using in silico analysis, the current study identified that the polymorphisms rs11515 and rs3088440 are located within a transcriptional regulatory region, and the alteration of nucleotides can affect the binding of potential transcriptional factors. For example, the presence of the C allele in rs3088440 favors the binding of the transcription factor c-Myb, which potentially results in the transcriptional repression of the CDKN2A gene, compromising its normal function in cell cycle control (42). However, no association was identified between this polymorphism and the clinicopathological parameters investigated in the cohort studied (Table III).

In conclusion, it is suggested that BRAF, CDKN2A and PI3KCA, listed as potential adjuvants in the tumorigenesis of MTC, do not participate through somatic mutations as modulators of oncogenesis. To the best of our knowledge, the current study is the first to investigate these two CDKN2A polymorphisms in the pathophysiology of MTC. Therefore, CDKN2A and its regulatory regions and the additional genes involved in tumorigenesis warrant further investigation in MTC.

Acknowledgments

The authors would like to thank the team of the Laboratory of Molecular and Translational Endocrinology, particularly Ms. Teresa Kasamatsu, Mr. Gilberto Furuzawa, Dr João Roberto Martins, Dr Ji Hoon Yang, Dr Fausto Germano Neto and Mr. Fernando Soares. The authors would additionally like to acknowledge Mr. Gilmar Miranda from Siratec Ltd. for graphic art design. The current study was supported grants from the Sao Paulo State Research Foundation (grant nos. 2012/11036-3, 2012/02465-8, 2012/01628-0, 2009/50575-4, 2012/00079-3 and 2011/20747-8).

References

1 

American Thyroid Association Guidelines Task Force; Kloos RT, Eng C, Evans DB, Francis GL, Gagel RF, Gharib H, Moley JF, Pacini F and Ringel MD: Medullary thyroid cancer: Management guidelines of the American thyroid association. Thyroid. 19:565–612. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Nosé V: Familial thyroid cancer: A review. Mod Pathol. 24(Suppl 2): S19–S33. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Moura MM, Cavaco BM, Pinto AE and Leite V: High prevalence of RAS mutations in RET-negative sporadic medullary thyroid carcinomas. J Clin Endocrinol Metab. 96:E863–E868. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Ciampi R, Mian C, Fugazzola L, Cosci B, Romei C, Barollo S, Cirello V, Bottici V, Marconcini G, Rosa PM, et al: Evidence of a low prevalence of RAS mutations in a large medullary thyroid cancer series. Thyroid. 23:50–57. 2013. View Article : Google Scholar

5 

Agrawal N, Jiao Y, Sausen M, Leary R, Bettegowda C, Roberts NJ, Bhan S, Ho AS, Khan Z, Bishop J, et al: Exomic sequencing of medullary thyroid cancer reveals dominant and mutually exclusive oncogenic mutations in RET and RAS. J Clin Endocrinol Metab. 98:E364–E369. 2013. View Article : Google Scholar :

6 

Tamburrino A, Molinolo AA, Salerno P, Chernock RD, Raffeld M, Xi L, Gutkind JS, Moley JF, Wells SA Jr and Santoro M: Activation of the mTOR pathway in primary medullary thyroid carcinoma and lymph node metastases. Clin Cancer Res. 18:3532–3540. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Simbolo M, Mian C, Barollo S, Fassan M, Mafficini A, Neves D, Scardoni M, Pennelli G, Rugge M, Pelizzo MR, et al: High-throughput mutation profiling improves diagnostic stratification of sporadic medullary thyroid carcinomas. Virchows Arch. 465:73–78. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Puppin C, Durante C, Sponziello M, Verrienti A, Pecce V, Lavarone E, Baldan F, Campese AF, Boichard A, Lacroix L, et al: Overexpression of genes involved in miRNA biogenesis in medullary thyroid carcinomas with RET mutation. Endocrine. 47:528–536. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Rapa I, Saggiorato E, Giachino D, Palestini N, Orlandi F, Papotti M and Volante M: Mammalian target of rapamycin pathway activation is associated to RET mutation status in medullary thyroid carcinoma. J Clin Endocrinol Metab. 96:2146–2153. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Berrocal A, Cabañas L, Espinosa E, Fernández-de-Misa R, Martín-Algarra S, Martínez-Cedres JC, Ríos-Buceta L and Rodríguez-Peralto JL: Melanoma: Diagnosis, staging and treatment. Consensus group recommendations. Adv Ther. 31:945–960. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Muscarella P, Bloomston M, Brewer AR, Mahajan A, Frankel WL, Ellison EC, Farrar WB, Weghorst CM and Li J: Expression of the p16INK4A/Cdkn2a gene is prevalently downregulated in human pheochromocytoma tumor specimens. Gene Expr. 14:207–216. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Güran S and Tali ET: p53 and p16INK4A mutations during the progression of glomus tumor. Pathol Oncol Res. 5:41–45. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW, Knauf JA, Basolo F, Zhu Z, Giannini R, Salvatore G, Fusco A, et al: BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J Clin Endocrinol Metab. 88:5399–5404. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Goutas N, Vlachodimitropoulos D, Bouka M, Lazaris AC, Nasioulas G and Gazouli M: BRAF and K-RAS mutation in a Greek papillary and medullary thyroid carcinoma cohort. Anticancer Res. 28:305–308. 2008.PubMed/NCBI

15 

Schulten HJ, Al-Maghrabi J, Al-Ghamdi K, Salama S, Al-Muhayawi S, Chaudhary A, Hamour O, Abuzenadah A, Gari M and Al-Qahtani M: Mutational screening of RET, HRAS, KRAS, NRAS, BRAF, AKT1 and CTNNB1 in medullary thyroid carcinoma. Anticancer Res. 31:4179–4183. 2011.PubMed/NCBI

16 

Boichard A, Croux L, Al Ghuzlan A, Broutin S, Dupuy C, Leboulleux S, Schlumberger M, Bidart JM and Lacroix L: Somatic RAS mutations occur in a large proportion of sporadic RET-negative medullary thyroid carcinomas and extend to a previously unidentified exon. J Clin Endocrinol Metab. 97:E2031–E2035. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Sauroja I, Smeds J, Vlaykova T, Kumar R, Talve L, Hahka-Kemppinen M, Punnonen K, Jansèn CT, Hemminki K and Pyrhönen S: Analysis of G(1)/S checkpoint regulators in metastatic melanoma. Genes Chromosomes Cancer. 28:404–414. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Pasquali D, Circelli L, Faggiano A, Pancione M, Renzullo A, Elisei R, Romei C, Accardo G, Coppola VR, De Palma M, et al: CDKN1B V109G polymorphism a new prognostic factor in sporadic medullary thyroid carcinoma. Eur J Endocrinol. 164:397–404. 2011. View Article : Google Scholar

19 

Tian Q, Frierson HF Jr, Krystal GW and Moskaluk CA: Activating c-kit gene mutations in human germ cell tumors. Am J Pathol. 154:1643–1647. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Samuels Y and Ericson K: Oncogenic PI3K and its role in cancer. Curr Opin Oncol. 18:77–82. 2006. View Article : Google Scholar

21 

Kizys MM, Cardoso MG, Lindsey SC, Harada MY, Soares FA, Melo MC, Montoya MZ, Kasamatsu TS, Kunii IS, Giannocco G, et al: Optimizing nucleic acid extraction from thyroid fine-needle aspiration cells in stained slides, formalin-fixed/paraffin-embedded tissues and long-term stored blood samples. Arq Bras Endocrinol Metabol. 56:618–626. 2012. View Article : Google Scholar

22 

Venselaar H, Te Beek TA, Kuipers RK, Hekkelman ML and Vriend G: Protein structure analysis of mutations causing inheritable diseases. An e-Science approach with life scientist friendly interfaces. BMC Bioinformatics. 11:5482010. View Article : Google Scholar : PubMed/NCBI

23 

Lee PH and Shatkay H: F-SNP: Computationally predicted functional SNPs for disease association studies. Nucleic Acids Res. 36(Database issue): D820–D824. 2008. View Article : Google Scholar :

24 

Edge SE, Byrd DR, Carducci MA, Compton CC, Fritz AG, Greene F and Trotti A: AJCC Cancer Staging Manual. Springer-Verlag; New York: 2009

25 

Lindsey SC, Kunii IS, Germano-Neto F, Sittoni MY, Camacho CP, Valente FO, Yang JH, Signorini PS, Delcelo R, Cerutti JM, et al: Extended RET gene analysis in patients with apparently sporadic medullary thyroid cancer: Clinical benefits and cost. Horm Cancer. 3:181–186. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Kumar R, Smeds J, Berggren P, Straume O, Rozell BL, Akslen LA and Hemminki K: A single nucleotide polymorphism in the 3′untranslated region of the CDKN2A gene is common in sporadic primary melanomas but mutations in the CDKN2B, CDKN2C, CDK4 and p53 genes are rare. Int J Cancer. 95:388–393. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Chatterjee S and Pal JK: Role of 5′- and 3′-untranslated regions of mRNAs in human diseases. Biol Cell. 101:251–262. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Sakano S, Berggren P, Kumar R, Steineck G, Adolfsson J, Onelöv E, Hemminki K and Larsson P: Clinical course of bladder neoplasms and single nucleotide polymorphisms in the CDKN2A gene. Int J Cancer. 104:98–103. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Geddert H, Kiel S, Zotz RB, Zhang J, Willers R, Gabbert HE and Sarbia M: Polymorphism of p16 INK4A and cyclin D1 in adenocarcinomas of the upper gastrointestinal tract. J Cancer Res Clin Oncol. 131:803–808. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Chansaenroj J, Theamboonlers A, Junyangdikul P, Swangvaree S, Karalak A, Chinchai T and Poovorawan Y: Polymorphisms in TP53 (rs1042522), p16 (rs11515 and rs3088440) and NQO1 (rs1800566) genes in Thai cervical cancer patients with HPV 16 infection. Asian Pac J Cancer Prev. 14:341–346. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Straume O, Smeds J, Kumar R, Hemminki K and Akslen LA: Significant impact of promoter hypermethylation and the 540 C>T polymorphism of CDKN2A in cutaneous melanoma of the vertical growth phase. Am J Pathol. 161:229–237. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Boonstra JJ, van Marion R, Tilanus HW and Dinjens WN: Functional polymorphisms associated with disease-free survival in resected carcinoma of the esophagus. J Gastrointest Surg. 15:48–56. 2011. View Article : Google Scholar :

33 

Pinheiro UB, de Carvalho Fraga CA, Mendes DC, Marques-Silva L, Farias LC, de Souza MG, Soares MB, Jones KM, Santos SH, de Paula AM, et al: p16 (CDKN2A) SNP rs11515 was not associated with head and neck carcinoma. Tumour Biol. 35:6113–6118. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Jin L, Xu L, Song X, Wei Q, Sturgis EM and Li G: Genetic variation in MDM2 and p14ARF and susceptibility to salivary gland carcinoma. PloS One. 7:e493612012. View Article : Google Scholar : PubMed/NCBI

35 

Polakova V, Pardini B, Naccarati A, Landi S, Slyskova J, Novotny J, Vodickova L, Bermejo JL, Hanova M, Smerhovsky Z, et al: Genotype and haplotype analysis of cell cycle genes in sporadic colorectal cancer in the czech republic. Hum Mutat. 30:661–668. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Royds JA, Al Nadaf S, Wiles AK, Chen YJ, Ahn A, Shaw A, Bowie S, Lam F, Baguley BC, Braithwaite AW, et al: The CDKN2A G500 allele is more frequent in GBM patients with no defined telomere maintenance mechanism tumors and is associated with poorer survival. PloS One. 6:e267372011. View Article : Google Scholar : PubMed/NCBI

37 

Thakur N, Hussain S, Nasare V, Das BC, Basir SF and Bharadwaj M: Association analysis of p16 (CDKN2A) and RB1 polymorphisms with susceptibility to cervical cancer in Indian population. Mol Biol Rep. 39:407–414. 2012. View Article : Google Scholar

38 

Zhang Y, Sturgis EM, Zafereo ME, Wei Q and Li G: p14ARF genetic polymorphisms and susceptibility to second primary malignancy in patients with index squamous cell carcinoma of the head and neck. Cancer. 117:1227–1235. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Zhang F, Xu L, Wei Q, Song X, Sturgis EM and Li G: Significance of MDM2 and P14 ARF polymorphisms in susceptibility to differentiated thyroid carcinoma. Surgery. 153:711–717. 2013. View Article : Google Scholar :

40 

De Giorgi V, Savarese I, D'Errico A, Gori A, Papi F, Colombino M, Cristina Sini M, Stanganelli I, Palmieri G and Massi D: CDKN2A mutations could influence the dermoscopic pattern of presentation of multiple primary melanoma: A clinical dermoscopic genetic study. J Eur Acad Dermatol Venereol. 29:574–580. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Song X, Sturgis EM, Huang Z, Li X, Li C, Wei Q and Li G: Potentially functional variants of p14ARF are associated with HPV-positive oropharyngeal cancer patients and survival after definitive chemoradiotherapy. Carcinogenesis. 35:62–68. 2014. View Article : Google Scholar

42 

Stenman G, Andersson MK and Andrén Y: New tricks from an old oncogene: Gene fusion and copy number alterations of MYB in human cancer. Cell Cycle. 9:2986–2995. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 13 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nascimento FP, Cardoso MG, Lindsey SC, Kunii IS, Valente FO, Kizys MM, Delcelo R, Camacho CP, Maciel RM, Dias‑Da‑Silva MR, Dias‑Da‑Silva MR, et al: Analysis of somatic mutations in braf, CDKN2A/p16 and PI3KCA in patients with medullary thyroid carcinoma. Mol Med Rep 13: 1653-1660, 2016
APA
Nascimento, F.P., Cardoso, M.G., Lindsey, S.C., Kunii, I.S., Valente, F.O., Kizys, M.M. ... Dias‑Da‑Silva, M.R. (2016). Analysis of somatic mutations in braf, CDKN2A/p16 and PI3KCA in patients with medullary thyroid carcinoma. Molecular Medicine Reports, 13, 1653-1660. https://doi.org/10.3892/mmr.2015.4731
MLA
Nascimento, F. P., Cardoso, M. G., Lindsey, S. C., Kunii, I. S., Valente, F. O., Kizys, M. M., Delcelo, R., Camacho, C. P., Maciel, R. M., Dias‑Da‑Silva, M. R."Analysis of somatic mutations in braf, CDKN2A/p16 and PI3KCA in patients with medullary thyroid carcinoma". Molecular Medicine Reports 13.2 (2016): 1653-1660.
Chicago
Nascimento, F. P., Cardoso, M. G., Lindsey, S. C., Kunii, I. S., Valente, F. O., Kizys, M. M., Delcelo, R., Camacho, C. P., Maciel, R. M., Dias‑Da‑Silva, M. R."Analysis of somatic mutations in braf, CDKN2A/p16 and PI3KCA in patients with medullary thyroid carcinoma". Molecular Medicine Reports 13, no. 2 (2016): 1653-1660. https://doi.org/10.3892/mmr.2015.4731