Low mutation percentage of KRAS and BRAF genes in Brazilian anal tumors

  • Authors:
    • Lucas Tadeu Bidinotto
    • Carlos A R Véo
    • Edgar Aleman Loaiza
    • Alessandra Paulino Santos De França
    • Adriana Tarla Lorenzi
    • Luciana Albina Reis Rosa
    • Cristina Mendes De Oliveira
    • José Eduardo Levi
    • Cristovam Scapulatempo‑Neto
    • Adhemar Longatto‑Filho
    • Rui Manuel Reis
  • View Affiliations

  • Published online on: August 26, 2016     https://doi.org/10.3892/mmr.2016.5684
  • Pages: 3791-3797
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Anal cancer is a rare type of digestive tract disease, which has had a crescent incidence in a number of regions. Carcinomas are most frequently found, with squamous cell carcinoma (SCC) comprising ~95% of all anal tumors. The major risk factor for development of this type of tumor is human papillomavirus (HPV) infection. However, previous studies have identified patients with anal cancer that are HPV‑/p16‑and observed that they have a poorer outcome compared with HPV+/p16+ patients. This suggests that molecular profile may drive anal cancer progression. The aim of the present study was to evaluate the mutational status of two important oncogenes, KRAS and BRAF, in a series of anal cancer lesions. Resected tumors of the anal canal (n=43) were evaluated, nine of these were high‑grade squamous intra‑epithelial lesion cases (HSIL), 11 were adenocarcinomas, and 23 SCCs. Direct sequencing of KRAS proto‑oncogene, GTPase (KRAS; codons 12 and 13) and B‑Raf proto‑oncogene, serine/threonine kinase (BRAF; codon 600) was performed and associated with patient clinicopathological and molecular features. There was a trend of poorer prognosis of adenocarcinoma compared with HSIL and SCC. Analysis indicated one SCC patient (2.3%) exhibited a KRAS p.G13D mutation, and one adenocarcinoma patient (2.3%) exhibited a BRAF p.V600E mutation. It was observed that, these mutations are rare in anal tumors, and certain patients may be at a disadvantage using targeted therapies based on KRAS and BRAF mutational status. As there is a low mutation percentage in SCCs, adenocarcinomas and HSIL, there may exist other underlying molecular alterations that result in anal cancer development, which require further elucidation.

Introduction

Anal cancer is a rare type of digestive tract disease, which has had an increasing incidence in a number of regions (13). It is estimated a total of >7,200 new cases were diagnosed in the United States in 2015, with ~1,000 anal cancer-associated mortalities (4). Tumors in this site are classified, according to World Health Organization (WHO), as intraepithelial neoplasias, carcinomas and carcinoid tumors (5). Carcinomas are most frequently identified, with squamous cell carcinoma (SCC) comprising~95% of all the anal tumors (6), and ~5% of the lesions are adenocarcinomas (7). The age-standardized incidence is <1/100,000 people, and the mortality is 0.2/100,000 (1). In men who practice anal receptive intercourse, the incidence of anal cancer increases up to 35/100,000 (5,8). This is predominantly due to increased risk of human papillomavirus (HPV) infection (1), HPV16 is most frequently observed in anal SCC (9). HPV infection leads to intraepithelial neoplasia that progresses from low-grade to high-grade dysplasia and, finally, to invasive cancer. The regression of high-grade dysplastic lesions is rare (5). HPV infection results in high expression of cyclin-dependent kinase inhibitor 2A (p16), and disrupts the association between retinoblastoma protein and the E2F family of transcription factors, ultimately leading to cellular proliferation (10). Recently, it was demonstrated that a high frequency of women with cervical cancer also have infection of the anal canal by HPV16 (11). In addition to HPV infection, other known risk factors of anal cancer are immunodeficiency due to human immunodeficiency virus seropositivity, low cluster of differentiation 4 T cell count, immunosuppression following solid organ transplantation, and tobacco smoking (1,5).

Previous studies have indicated that patients presenting with no HPV infection and no p16 expression (HPV-/p16-) have a poorer outcome than patients that are HPV+/p16+, and suggest an optimization in the therapy to the former (12,13), and that improved molecular characterization should be performed. A previous study demonstrated that patients with SCC of the anal canal and HPV were irresponsive to standard chemoradiotherapy treatment and frequently presented with TP53 mutations (13). Furthermore, an additional study using immunohistochemistry (IHC), fluorescence in situ hybridization and next generation sequencing, observed alterations in molecular markers that may aid in the understanding of failure of certain therapeutic strategies, including overexpression of multidrug resistance-associated protein 1, DNA excision repair protein ERCC-1 and thymidylate synthetase, and suggest potential therapeutic targets, including the tyrosine kinase receptor, epidermal growth factor receptor (EGFR) (14). Molecular therapies targeting EGFR, such as cetuximab and panitumumab are currently used in colorectal cancer treatment, and tumor genetic make-up, including mutational status of KRAS and BRAF may predict patient response (15).

The aim of the present study is to evaluate the mutational status of two important oncogenes, KRAS and BRAF in a series of SCC, adenocarcinomas and high-grade squamous intra-epithelial lesions (HSILs), and whether they are associated with patient's clinicopathological features, and HPV status.

Materials and methods

In the current study, resected tumors of the anal canal from 43 patients were evaluated. The present study was approved by the ethics committee of Barretos Cancer Hospital (Barretos, Brazil) and informed consent was obtained from each patient. Histological review of the slides was performed by an expert pathologist (Dr Cristovam Scapulatempo-Neto), who confirmed the diagnosis and delimited the area of the slide containing the neoplastic lesion. Clinical data of the patients was obtained, and is summarized in Table I. HPV16 and HPV18 status, and immunohistochemical analysis of β-globin, p16, antigen Ki67 (Ki67), minichromosome maintenance protein complex (MCM) and DNA topoisomerase 2-α (TOP2A) were retrieved from a previous study (16).

Table I

Clinicopathological features of patients with anal lesions.

Table I

Clinicopathological features of patients with anal lesions.

ParameterFrequency (%)
HSILAdenocarcinomaSCC
Gender
 Female6 (20.7)7 (24.1)16 (55.2)
 Male3 (21.4)4 (28.6)7 (50.0)
Ethnicity
 Caucasian8 (21.6)8 (21.6)21 (56.8)
 Non-caucasian1 (16.7)3 (50)2 (33.3)
History of previous disease
 No2 (11.1)7 (38.9)9 (50.0)
 Yes7 (29.2)3 (12.5)14 (58.3)
 NA01 (100)0
History of tumor in the family
 No6 (21.4)6 (21.4)16 (57.1)
 Yes3 (21.4)4 (28.6)7 (50.0)
 NA01 (100)0
Tobacco consumption
 No1 (5.6)7 (38.9)10 (55.6)
 Yes7 (33.3)3 (14.3)11 (52.4)
 NA1 (25.0)1 (25.0)2 (50.0)
Surgery
 No6 (26.1)6 (26.1)11 (47.8)
 Yes3 (16.7)5 (27.8)10 (55.6)
 NA002 (100)
Radiotherapy
 No2 (40.0)1 (20.0)2 (40.0)
 Yes7 (19.4)10 (27.8)19 (52.8)
 NA002 (100)
Chemotherapy
 No2 (25.0)3 (37.5)3 (37.5)
 Yes7 (21.2)8 (24.2)18 (54.5)
 NA002 (100)
Response to the treatment
 No response4 (23.5)6 (35.3)7 (41.2)
 Complete response1 (8.3)2 (16.7)9 (75.0)
 Progression2 (22.2)2 (22.2)5 (55.6)
 NA2 (40.0)1 (20.0)2 (40.0)
Recurrence
 No7 (22.6)6 (19.4)18 (58.1)
 Yes2 (16.7)5 (41.7)5 (41.7)
Status
 Death by cancer3 (23.1)4 (30.8)6 (46.2)
 Death by other cause02 (66.7)1 (33.3)
 Alive, free of disease6 (27.3)3 (13.6)13 (59.1)
 Alive, with the disease02 (40.0)3 (60.0)
Age (years)
 <483 (33.3)1 (11.1)5 (55.6)
 48–665 (20.0)6 (24.0)14 (56.0)
 >661 (11.1)4 (44.4)4 (44.4)

[i] NA, not available.

The histological slides (10 µm) were processed, and DNA was isolated from macrodissected tumor area of one unstained section as previously described (17). The slides were placed at 80°C for deparaffinization for 10 min and hydrated with xylene and graded ethanol (100, 70 and 50%). DNA was isolated using QIAamp DNA Micro kit (Qiagen GmbH, Hilden, Germany), following the manufacturer's protocols, and quantified using NanoDrop 2000 (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The samples were diluted to a final concentration of 50 ng/µl and stored at −20°C for further analysis.

The hotspots of KRAS (codons 12 and 13) and BRAF (codon 600) were amplified using PCR and sequenced, as previously described (17). Amplification of KRAS was performed in a final reaction volume of 15 µl containing 1.5 µl buffer (Qiagen GmbH), 2 mM MgCl2 (Qiagen GmbH), 100 mM dNTPs (Invitrogen; Thermo Fisher Scientific, Inc.), 0.2 mM sense and 0.2 mM anti-sense primers (Sigma-Aldrich, St. Louis, MO, USA), 1 unit HotStarTaq DNA polymerase (Qiagen GmbH) and 1 µl DNA. The KRAS region was amplified using the following primers: Sense, 5′-GTGTGACATGTTCTAATATAGTCA-3′ and anti-sense, 5′-GAATGGTCCTGCACCAGTAA-3′. The BRAF amplification reaction was performed as described above, with 0.3 mM sense and anti-sense primers used. The region was amplified using the following primers: Sense, 5′-TCATAATGCTTG CTCTGATAGGA-3′ and anti-sense, 5′-GGCCAAAAATTTAATCAGTGGA-3′. The following cycling conditions were used: Initial denaturation at 96°C for 15 min, followed by 40 cycles of denaturation at 96°C for 45 sec, annealing at 55.5°C for 45 sec, then extension at 72°C for 10 min, all using a Veriti 96-Well thermal cycler (Applied Biosystems; Thermo Fisher Scientific, Inc.)

The PCR products were purified with EXO-SAP (GE Healthcare Life Sciences, Chalfont, UK), and sequenced using 1 µl BigDye (Applied Biosystems; Thermo Fisher Scientific, Inc.), 1.5 µl sequencing buffer (Applied Biosystems; Thermo Fisher Scientific, Inc.) and 1 µl primer (Thermo Fisher Scientific, Inc.). The sequencing reaction, which consisted of 30 cycles of denaturation at 96°C for 10 sec, annealing at 50°C for 5 sec and extension at 60°C for 4 min, was followed by post-sequencing purification with EDTA, alcohol and sodium citrate. The products of PCR were eluted in Hi-Di formamide (Thermo Fisher Scientific, Inc.) and incubated at 95°C for 5 min and at −4°C for at least 5 min. Direct sequencing was performed in a 3500 Genetic Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.). The mutations were confirmed with two independent reactions.

Survival analysis was performed considering the three different histology types using Kaplan-Meier plots and log rank statistical analysis using R.

Results

Histological review of the slides demonstrated that, from the 43 patients, 9 were diagnosed with HSIL, 11 patients were diagnosed with adenocarcinomas, and 23 with SCC. The mean age at diagnosis varied from 51 (HSIL) to 64 (adenocarcinoma) years. Overall survival (OS) and disease-free survival (DFS), based on the histologic type is presented in Fig. 1. There was a trend of poorer prognosis in adenocarcinoma compared with HSIL and SCC (P=0.176 and P=0.112 for OS and DFS, respectively).

Of the 43 patient samples examined, 1 (2.3%), exhibited a KRAS mutation, which was p.G13D (Fig. 2A and Table II). This case was a female SCC patient (age, 58) with previous gynecological or anal disease. The SCC was positive for β-globin and p16 using IHC, as well as 4+ Ki67 labeling. Notably, this patient also presented with HPV16 anal infection (Table II). Following surgery, radiotherapy and chemotherapy treatment, the patient exhibited complete response, and was free of disease at the last follow up of 140 months (Table II).

Table II

Molecular features of anal lesion patients.

Table II

Molecular features of anal lesion patients.

Histological typeGenderAge (years)HPV16aHPV18a Immunohistochemistrya
Mutation
TreatmentOncological responseDFS (months)Survival (months)Status
β-globinp16Ki67MCMTOP2AKRASBRAF
HSILM50+Ok+4+3+3+wtwtSurgery + RDT + QTProgression21.7425.10D
HSILF57+Weak+4+2+2+wtwtRDT + QTNo evidence47.1747.17A
HSILF57+Ok+4+3+2+wtwtRDT + QTNo evidence5.135.13D
HSILF47+Ok+4+1+2+wtwtSurgery + RDT + QTProgression12.2044.05D
HSILM38++Ok+4+3+2+wtwtRDT + QTNo evidence54.1154.11A
HSILF64+Ok4+2+3+wtwtNo treatmentNo evidence46.3846.38A
HSILF69+Ok+4+NAwtwtRDT + QTNA93.0393.03A
HSILF53+Ok+4+1+NAwtwtRDT + QTNA90.7990.79A
HSILM27+Ok4+2+NAwtwtSurgeryCR81.5181.51A
ADCF35+Ok4+3+wtwtSurgery + RDT + QTProgression1.7412.86D
ADCM69+Ok+4+3+wtwtRDT + QTNo evidence4.3810.82D
ADCM66OkNA1+2+wtwtRDTNo evidence1.884.97D
ADCF86+Ok3+2+wtwtRDTNo evidence31.1531.15D
ADCF60+Ok4+2+wtwtRDT + QTNo evidence5.765.76D
ADCM70+Ok+4+3+3+wtV600ESurgeryNo evidence0.132,11D
ADCF59+Ok+4+3+2+wtwtRDT + QTNA56.7856,78A
ADCM58+Ok+1++CB1+wtwtSurgery + RDT + QTCR24.7024.70A
ADCF63+Ok4+NAwtwtRDT + QTProgression3.4271.61A
ADCF52Ok4+1+2+wtwtSurgery + RDT + QNo evidence83.4283.42A
ADCF81+Ok2++CB2+wtwtSurgery + RDT + QTCR158.36158.36A
SCCM85+Ok+4+3+3+wtwtSurgery + RDTNo evidence19.0519.05D
SCCM64+Ok+3+3+2+wtwtSurgery + RDT + QTNo evidence59.6465.86D
SCCM50+OkNA4+2+2+wtwtRDT + QTProgression4.1423.49A
SCCF43+_Weak+4+2+wtwtRDT + QTProgression58.4258.42A
SCCM43+Ok1+1+wtwtSurgery + RDT + QTProgression31.9731.97A
SCCF44+Ok+4+3+3+wtwtNACR36.6136.61A
SCCF48+Ok+4+2+3+wtwtRDT + QTNo evidence18.3618.36D
SCCF62+Ok+4+1+2+wtwtSurgery +QTProgression15.8619.05D
SCCF56+Ok+4+3+2+wtwtRDT + QTCR64.3464.34A
SCCF64+Weak+4+3+3+wtwtSurgery + QTNo evidence60.3960.39D
SCCF68Ok+4+2+2+wtwtSurgery + RDT + QTNo evidence12.3712.37D
SCCF58+Ok+4+3+2+wtwtNANA69.3169.31A
SCCM56+Ok+4+2+NAwtwtRDT + QTNo evidence12.7099.61A
SCCM34Ok+4+3+3+wtwtRDT + QTCR54.7754.77A
SCCF44+OkNA2+1+1+wtwtRDT + QTNA43.1643.16A
SCCF48+Ok3+2+1+wtwtNo treatmentNo evidence20.8220.82D
SCCF56+Ok+4+2+2+wtwtSurgery + RDT + QTCR64.8064.80A
SCCF79+Ok+4+3+2+wtwtRDT + QTCR72.2772.27A
SCCF60+Ok+4++CB2+wtwtSurgery + RDTProgression10.46110.53A
SCCM57+Ok+4+2+3+wtwtSurgery + RDT + QTCR144.31144.31A
SCCF57+Weak+3+2+2+wtwtRDT + QTCR140.95140.95A
SCCF58+Ok+4+NAG13DwtSurgery + RDT + QTCR140.23140.23A
SCCF80+Ok+4+2+wtwtRDT + QTCR99.9399.93A

a As described in Scapulatempo-Neto et al (16). HSIL, high-grade squamous intra-epithelial lesion; ADC, adenocarcinoma; SCC, squamous cell carcinoma; F, female; M, male; HPV, human papillomavirus; p16, cyclin-dependent kinase inhibitor 2A; Ki67, antigen Ki67; MCM, minichromosome maintenance protein complex; TOP2A, DNA topoisomerase 2-α; KRAS, KRAS proto-oncogene, GTPase; BRAF, B-Raf proto-oncogene, serine/threonine kinase; wt, wild type; RDT, radiotherapy; QT, chemotherapy; CR, complete response; DFS, disease-free survival; D, deceased; A, alive.

A BRAF mutation, p.V600E, was also observed in only 1/43 patients (2.3%; Fig. 2B and Table II). This case was an adenocarcinoma of a male 70 year-old tobacco smoking patient. Similarly to the case described above, the patient also presented with HPV16 anal infection, and IHC indicated positivity for β-globin and p16 IHC labeling, as well as 4+ Ki67 labeling (Table II). In addition, the sample presented 3+ MCM and 3+ TOP2A IHC labeling. The patient relapsed and succumbed to the condition 2 months following surgery (Table II).

Discussion

The present study aimed to evaluate the mutational status of KRAS and BRAF in tumors arising in the anal canal, and to associate these findings with clinicopathological data. To the best of our knowledge, the majority of the mutational screening of KRAS and BRAF in anal tumors focused on SCC samples due to the predominance of this histological type in anal tumors.

Although tumors located in the anal region may be anatomically close to colorectal tumors, they exhibit different histological patterns, distinct features, and therefore distinct etiologies (18). Previous studies have observed a high incidence of KRAS mutation in colorectal cancer (CRC) worldwide (19), and in Brazilian populations (17,20). Overall, among 8,234 Brazilian CRC cases analyzed, the KRAS mutation frequency of 31.9%, with the majority of these samples exhibiting a p.G12D mutation (20). KRAS mutations generally arise in codons 12 or 13, and constitutively activate its pathway. The protein generated by the mutated gene is capable of transmitting the signal independently of tyrosine kinase receptor activation (21). Of the 43 patients analyzed, 1 patient with SCC was observed to have a KRAS mutation (2.3%). This low mutation rate is consistent with previous studies, which identified no KRAS mutation in SCC of the anal canal samples [n=36 samples (22), n=89 samples (23), n=53 samples (24) and n=66 samples (25)]. Additional studies observed a total of 1.6% (n=193 samples) (26), and 5% (n=84 samples) (27) of SCC to have a KRAS mutation. Furthermore, BRAF, another mitogen-activated protein kinase pathway gene, which was identified as mutated in ~50% of melanomas (28), presented a low mutation rate in the samples in the present study (2.3%). This gene was observed with a low percentage of mutation in anal tumors, varying from 0% (24,25,27) to 4.7% (26) consistent with the findings of the current study. In addition, this gene was also found mutated in a low percentage of precursor lesions of colorectal cancer (17) and colorectal cancer (29). It is important to highlight that no mutation was observed in patients with HSIL, which suggest that these mutations may occur preferentially in tumors with an invasive phenotype.

The standard treatment of anal SCC in the majority of patients is chemotherapy and radiotherapy, with a response rate of up to 80% (30). Metastatic and refractory cases are rare, although, it has been demonstrated that cetuximab-based treatment results in disease progression of KRAS-mutated tumors, while those with wild type KRAS exhibited partial or minor remission (31). This data is consistent with further studies in colorectal cancer that demonstrated KRAS and BRAF mutational status predicted tumor response to targeted therapies (15). Thus, an understanding of KRAS and BRAF mutational status is key in personalized medicine.

Using the mutational rate of KRAS and BRAF, the present study evaluated the anatomical association between anal and rectal tumors, tumors arising in the anus have different KRAS mutation percentage of the rectal counterparts, thus, the molecular differences require elucidation. To the best of our knowledge, the current study is the first to evaluate the percentage of KRAS and BRAF mutations in adenocarcinomas and HSIL of the anal canal, in addition to SCC. Furthermore, to date, the present study is the first to describe these mutations in tumors of anal canal of the Brazilian population. In addition to the well-known risk factor of HPV infection that drives anal cancer tumorigenesis, there are patients who develop these tumors in the absence of this infection. The present study evaluated the mutation percentage of two well-known drivers of colorectal cancer (KRAS) and melanoma (BRAF) to further elucidate other risk factors in anal cancer development. In conclusion, a low percentage of mutation was identified in SCCs, adenocarcinomas and HSIL, however, these tumors may exhibit other molecular alterations that result in anal cancer development, which require elucidation in future studies.

Acknowledgments

The present study was partially supported by the São Paulo Research Foundation (grant no. 2010/16795-4 to Dr Adhemar Longatto-Filho) and the Ministério da Ciência e Tecnologia/Financiadora de Estudos e Projetos (grant no. CT-INFRA-PROINFRA 01/2011). Dr Lucas Tadeu Bidinotto received a São Paulo Research Foundation fellowship (grant no. 2011/08523-7).

References

1 

Bosman FT; World Health Organization and International Agency for Research on Cancer: WHO Classification of Tumours of the Digestive System. IARC Press; Lyon: 2010

2 

Lampejo T, Kavanagh D, Clark J, Goldin R, Osborn M, Ziprin P and Cleator S: Prognostic biomarkers in squamous cell carcinoma of the anus: A systematic review. Br J Cancer. 103:1858–1869. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Rousseau DL Jr, Thomas CR Jr, Petrelli NJ and Kahlenberg MS: Squamous cell carcinoma of the anal canal. Surg Oncol. 14:121–132. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Leonard D, Beddy D and Dozois EJ: Neoplasms of anal canal and perianal skin. Clin Colon Rectal Surg. 24:54–63. 2011. View Article : Google Scholar :

6 

Deans GT, McAleer JJ and Spence RA: Malignant anal tumours. Br J Surg. 81:500–508. 1994. View Article : Google Scholar : PubMed/NCBI

7 

Franklin RA, Giri S, Valasareddy P, Lands LT and Martin MG: Comparative survival of patients with anal adenocarcinoma, squamous cell carcinoma of the anus, and rectal adenocarcinoma. Clin Colorectal Cancer. 15:47–53. 2016. View Article : Google Scholar

8 

Franceschi S and De Vuyst H: Human papillomavirus vaccines and anal carcinoma. Curr Opin HIV AIDS. 4:57–63. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Serup-Hansen E, Linnemann D, Skovrider-Ruminski W, Høgdall E, Geertsen PF and Havsteen H: Human papillo-mavirus genotyping and p16 expression as prognostic factors for patients with American joint committee on cancer stages I to III carcinoma of the anal canal. J Clin Oncol. 32:1812–1817. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Doorbar J: Molecular biology of human papillomavirus infection and cervical cancer. Clin Sci (Lond). 110:525–541. 2006. View Article : Google Scholar

11 

Veo CA, Saad SS, Fregnani JH, Scapulatempo-Neto C, Tsunoda AT, Resende JC, Lorenzi AT, Mafra A, Cinti C, Cotrim ID, et al: Clinical characteristics of women diagnosed with carcinoma who tested positive for cervical and anal high-risk human papillomavirus DNA and E6 RNA. Tumour Biol. 36:5399–5405. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Mai S, Welzel G, Ottstadt M, Lohr F, Severa S, Prigge ES, Wentzensen N, Trunk MJ, Wenz F, von Knebel-Doeberitz M and Reuschenbach M: Prognostic relevance of HPV infection and p16 overexpression in squamous cell anal cancer. Int J Radiat Oncol Biol Phys. 93:819–827. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Meulendijks D, Tomasoa NB, Dewit L, Smits PH, Bakker R, van Velthuysen ML, Rosenberg EH, Beijnen JH, Schellens JH and Cats A: HPV-negative squamous cell carcinoma of the anal canal is unresponsive to standard treatment and frequently carries disruptive mutations in TP53. Br J Cancer. 112:1358–1366. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Smaglo BG, Tesfaye A, Halfdanarson TR, Meyer JE, Wang J, Gatalica Z, Reddy S, Arguello D and Boland PM: Comprehensive multiplatform biomarker analysis of 199 anal squamous cell carcinomas. Oncotarget. 6:43594–43604. 2015.PubMed/NCBI

15 

De Roock W, Claes B, Bernasconi D, De Schutter J, Biesmans B, Fountzilas G, Kalogeras KT, Kotoula V, Papamichael D, Laurent-Puig P, et al: Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the effcacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: A retrospective consortium analysis. Lancet Oncol. 11:753–762. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Scapulatempo-Neto C, Veo CA, Fregnani JH, Lorenzi A, Mafra A, Melani A, Loaiza E, Rosa L, de Oliveira C, Levi J and Longatto-Filhø A: Characterization of topoisomerase II alpha (TOP2A) and minichromosome maintenance protein (MCM)2 expression in anal carcinoma. Oncol Lett. 2016.

17 

Yamane LS, Scapulatempo-Neto C, Alvarenga L, Oliveira CZ, Berardinelli GN, Almodova E, Cunha TR, Fava G, Colaiacovo W, Melani A, et al: KRAS and BRAF mutations and MSI status in precursor lesions of colorectal cancer detected by colonoscopy. Oncol Rep. 32:1419–1426. 2014.PubMed/NCBI

18 

Matalon SA, Mamon HJ, Fuchs CS, Doyle LA, Tirumani SH, Ramaiya NH and Rosenthal MH: Anorectal cancer: Critical anatomic and staging distinctions that affect use of radiation therapy. Radiographics. 35:2090–2107. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Ta n C and Du X: KRAS mutation testing in metastatic colorectal cancer. World J Gastroenterol. 18:5171–5180. 2012.

20 

Gil Ferreira C, Aran V, Zalcberg-Renault I, Victorino AP, Salem JH, Bonamino MH, Vieira FM and Zalis M: KRAS mutations: Variable incidences in a Brazilian cohort of 8,234 metastatic colorectal cancer patients. BMC Gastroenterol. 14:732014. View Article : Google Scholar : PubMed/NCBI

21 

Okumura S and Jänne PA: Molecular pathways: The basis for rational combination using MEK inhibitors in KRAS-mutant cancers. Clin Cancer Res. 20:4193–4199. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Gilbert DC, Williams A, Allan K, Stokoe J, Jackson T, Linsdall S, Bailey CM and Summers J: p16INK4A, p53, EGFR expression and KRAS mutation status in squamous cell cancers of the anus: Correlation with outcomes following chemo-radiotherapy. Radiother Oncol. 109:146–151. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Paliga A, Onerheim R, Gologan A, Chong G, Spatz A, Niazi T, Garant A, Macheto D, Alcindor T and Vuong T: EGFR and K-ras gene mutation status in squamous cell anal carcinoma: A role for concurrent radiation and EGFR inhibitors? Br J Cancer. 107:1864–1868. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Casadei Gardini A, Capelli L, Ulivi P, Giannini M, Freier E, Tamberi S, Scarpi E, Passardi A, Zoli W, Ragazzini A, et al: KRAS, BRAF and PIK3CA status in squamous cell anal carcinoma (SCAC). PLoS One. 9:e920712014. View Article : Google Scholar : PubMed/NCBI

25 

Prigge ES, Urban K, Stiegler S, Müller M, Kloor M, Mai S, Ottstadt M, Lohr F, Wenz F, Wagner S, et al: No evidence of oncogenic KRAS mutations in squamous cell carcinomas of the anogenital tract and head and neck region independent of human papillomavirus and p16 (INK4a) status. Hum Pathol. 45:2347–2354. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Serup-Hansen E, Linnemann D, Høgdall E, Geertsen PF and Havsteen H: KRAS and BRAF mutations in anal carcinoma. APMIS. 123:53–59. 2015. View Article : Google Scholar

27 

Martin V, Zanellato E, Franzetti-Pellanda A, Molinari F, Movilia A, Paganotti A, Deantonio L, De Dosso S, Assi A, Crippa S, et al: EGFR, KRAS, BRAF, and PIK3CA characterization in squamous cell anal cancer. Histol Histopathol. 29:513–521. 2014.

28 

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

29 

El-Deiry WS, Vijayvergia N, Xiu J, Scicchitano A, Lim B, Yee NS, Harvey HA, Gatalica Z and Reddy S: Molecular profiling of 6,892 colorectal cancer samples suggests different possible treatment options specific to metastatic sites. Cancer Biol Ther. 16:1726–1737. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Bartelink H, Roelofsen F, Eschwege F, Rougier P, Bosset JF, Gonzalez DG, Peiffert D, van Glabbeke M and Pierart M: Concomitant radiotherapy and chemotherapy is superior to radiotherapy alone in the treatment of locally advanced anal cancer: Results of a phase III randomized trial of the European Organization for Research and Treatment of Cancer Radiotherapy and Gastrointestinal Cooperative groups. J Clin Oncol. 15:2040–2049. 1997.PubMed/NCBI

31 

Lukan N, Ströbel P, Willer A, Kripp M, Dinter D, Mai S, Hochhaus A and Hofheinz RD: Cetuximab-based treatment of metastatic anal cancer: Correlation of response with KRAS mutational status. Oncology. 77:293–299. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 14 Issue 4

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
Bidinotto LT, Véo CA, Loaiza EA, De França AP, Lorenzi AT, Rosa LA, De Oliveira CM, Levi JE, Scapulatempo‑Neto C, Longatto‑Filho A, Longatto‑Filho A, et al: Low mutation percentage of KRAS and BRAF genes in Brazilian anal tumors. Mol Med Rep 14: 3791-3797, 2016
APA
Bidinotto, L.T., Véo, C.A., Loaiza, E.A., De França, A.P., Lorenzi, A.T., Rosa, L.A. ... Reis, R.M. (2016). Low mutation percentage of KRAS and BRAF genes in Brazilian anal tumors. Molecular Medicine Reports, 14, 3791-3797. https://doi.org/10.3892/mmr.2016.5684
MLA
Bidinotto, L. T., Véo, C. A., Loaiza, E. A., De França, A. P., Lorenzi, A. T., Rosa, L. A., De Oliveira, C. M., Levi, J. E., Scapulatempo‑Neto, C., Longatto‑Filho, A., Reis, R. M."Low mutation percentage of KRAS and BRAF genes in Brazilian anal tumors". Molecular Medicine Reports 14.4 (2016): 3791-3797.
Chicago
Bidinotto, L. T., Véo, C. A., Loaiza, E. A., De França, A. P., Lorenzi, A. T., Rosa, L. A., De Oliveira, C. M., Levi, J. E., Scapulatempo‑Neto, C., Longatto‑Filho, A., Reis, R. M."Low mutation percentage of KRAS and BRAF genes in Brazilian anal tumors". Molecular Medicine Reports 14, no. 4 (2016): 3791-3797. https://doi.org/10.3892/mmr.2016.5684