SciELO - Scientific Electronic Library Online

 
vol.72 número6Precision Oncology in Pancreatic Cancer. From Surgery- to Genetic-Based ChemotherapyUpdating Zika Diagnostic Methods: The Point-of-Care Approach índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay artículos similaresSimilares en SciELO

Compartir


Revista de investigación clínica

versión On-line ISSN 2564-8896versión impresa ISSN 0034-8376

Rev. invest. clín. vol.72 no.6 Ciudad de México nov./dic. 2020  Epub 09-Abr-2021

https://doi.org/10.24875/ric.20000111 

Brief communications

Complete Screening of Exons 2, 3, and 4 of KRAS and NRAS Genes Reveals a Higher Number of Clinically Relevant Mutations than Food and Drug Administration Quantitative Polymerase Chain Reaction-Based Commercial Kits

Héctor E. Sánchez-Ibarra1 

Luisa M. Reyes-Cortes1 

Esteban López-Tavera1 

Claudia M. Luna-Aguirre1 

Hugo A. Barrera-Saldaña1  * 

1Genetics Laboratory, Vitagénesis SA de CV., Monterrey, NL, Mexico


Abstract

Background:

The presence of clinically relevant mutations in KRAS and NRAS genes determines the response of anti-epidermal growth factor receptor antibody therapy for metastatic colorectal cancer (mCRC). The only quantitative polymerase chain reaction (qPCR)-based diagnostic tests approved by the Food and Drug Administration (FDA) screen merely for mutations in codons 12 and 13 of KRAS.

Objective:

The objective of the study was to study the frequency of clinically relevant mutations in KRAS and NRAS genes that are not included in FDA-approved qPCR tests.

Methods:

Formalin-fixed paraffin-embedded tumor specimens from 1113 mCRC Mexican patients from different health institutions across the country were analyzed by Sanger sequencing for KRAS mutations in exons 2, 3, and 4. Furthermore, 83 were analyzed in exons 2, 3, and 4 of NRAS.

Results:

From the specimens tested for KRAS, 33.69% harbored a mutation. From these, 71.77% were in codon 12 and 27.69% in codon 13 (both located in exon 2). Codons 59 (exon 3) and 146 (exon 4) accounted for the remaining 0.54%. From the 83 specimens, in which NRAS was analyzed, three mutations were found in codon 12 (3.61%). Approximately 6% of RAS mutated specimens would have been falsely reported as RAS wild type if an FDA-approved qPCR diagnostic test had been used.

Conclusions:

While these kits based on qPCR can be very practical and highly sensitive, their mutation coverage ignores mutations from poorly genetically characterized populations.

Key words: Colorectal cancer; Molecular diagnostics; RAS genes; Real-time polymerase chain reaction; Sanger sequencing

INTRODUCTION

Colorectal cancer (CRC) is the third most common neoplasia in men and the second in women (10.9% and 9.5% of all cancers, respectively) worldwide. In North America, CRC is the fourth most common cancer with an estimated incidence in 2018 of 49.9 for every 100,000 individuals1. Precursor lesions of the colonic mucosa known as adenomatous polyps (adenomas) may develop into colorectal tumors; however, < 10% of colorectal adenomas develop into invasive cancers. The progression from adenoma to adenocarcinoma has been associated with many different molecular mechanisms, including epigenetic events, DNA mismatch repair defects, chromosomal rearrangements, and mutations in key oncogenes and tumor suppressor genes2,3.

Colon cancer is one of the main diseases treated by targeted therapy as there is extensive information about the genes affected by cancer-causing mutations, their normal functions, and their carcinogenic effects when mutated2,4. Epidermal growth factor receptor (EGFR) triggers a signaling pathway related to the proliferation of tumor cells. The EGFR signaling cascade promotes cell proliferation by activation of Ras-GTPase and Erk/Map kinase. Because of this, medical treatment consists of blocking EGFR with monoclonal antibodies (mAbs), such as cetuximab and panitumumab5,6. However, mutational screening is necessary to identify resistant patients and is mandatory as a companion diagnostic test7. Even though EFGR is commonly overexpressed in all human cancers, its abnormality fails to predict the success of anti-EGFR therapy. On the other hand, the RAS gene family has become widely accepted as a biomarker for the response to treatment of several types of cancer, such as metastatic CRC (mCRC). Correspondingly, a patient with mCRC not harboring a KRAS or NRAS mutation could benefit from the treatment with anti-EGFR mAbs8,9 and vice versa.

RAS is one of the most studied and best characterized mCRC-associated gene families. Oncogenic mutations of three of its members, H, K, and NRAS, were among the first genetic changes identified in human tumors. These three members encode membrane-associated guanine nucleotide-binding proteins (p21ras) and are closely related, having 85% amino acid sequence identity and very similar functions10. At the molecular level, the above-mentioned KRAS and NRAS mutations result in reduced intrinsic GTPase activity, which, in turn, leads to permanent activation of KRAS or NRAS itself and of its downstream signaling pathways, thereby mediating malignant transformation. KRAS gene is in the short arm of chromosome 12, in p12.1, and is composed of 46,148 nucleotides divided into six exons. In contrast, the NRAS gene is in 1p13.2 of chromosome 1 and consists of seven exons spread over 12,431 nucleotides. Of the three human RAS isoforms, KRAS (Kirsten ras or Ki-ras) is the most frequently mutated oncogene in mCRC11,12, from which codons 12 and 13 of the second exon and 61 of the third exon have been found to harbor mutations in roughly 40-50% of tumors, with approximately 85-90% of mutations occurring in codons 12 and 134. Mutations involving codon 61 in the third exon and codon 146 in the fourth exon of the KRAS gene have also been described, with frequencies ranging from 1 to 6.6%13. An approximately 10% of the remaining KRAS negative tumors have been found mutated, mostly in NRAS codon 6114.

Around the world, several research groups are studying and publishing data on the relation of RAS genes with mCRC15, revealing that mutations in these genes might be associated with an increase of tumor aggressiveness and poor survival (prognostic value), as well as with resistance to gene-targeted therapies (predictive value)16.

A number of methods to detect KRAS mutations are available, including real-time or quantitative polymerase chain reaction (qPCR) with or without melting curve analysis, pyrosequencing, and Sanger sequencing (SS), the latter being the “gold standard” in clinical mutation testing17. Sanger sequencing can detect essentially all base substitutions, small insertions, and deletions; however, its modest limit of detection (≥ 10% mutated DNA in a wild-type background) could be highly variable depending on the DNA extraction method and the laboratory performing the test18.

The only qPCR-based companion diagnostic tests approved by the U.S. Food and Drug Administration (FDA) are the Cobas® KRAS Mutation Test from Roche (Pleasanton, CA, USA) and the therascreen KRAS RGQ PCR Kit from QIAGEN (Manchester, UK), which screen only for mutations in codon 12 and 13 of KRAS. This study aims to state the frequency of mutations that are not included in FDA-approved qPCR-based commercial companion diagnostic devices.

METHODS

Tumor samples

Tumor samples consisted of formalin-fixed paraffin-embedded (FFPE) tissue of primary tumor lesions from 1030 consecutive mCRC patients received in our laboratory for routine KRAS gene mutation screening from April 2010 to December 2013. This study also included 83 consecutive mCRC patient tumor specimens received from 2013 to 2016 that were routinely analyzed for both, KRAS and NRAS genes. The patients, 646 men and 447 women, were residents from Mexico and had Stage IV carcinoma. Patients signed informed consent before the performance of tests where it is agreed that their results may be used for research purposes. The study was approved by the Ethics and Research Committee from Hospital La Misión private hospital (17CI19039096). The specimens came from High Specialty Regional Hospitals of Bajío (HRAEB), Oaxaca (HRAEO), Yucatan’s peninsula (HRAEPY), Chiapas (HRAEC), Ciudad Victoria (HRAECV), and Ixtapaluca (HRAEI), as well as from Doctor’s Hospital, Opción Oncología, ONCARE, OCA Hospital, Hospital San José, Hospital Zambrano Hellion, and Hospital La Misión, Monterrey, NL, Mexico. Information on previous or current treatments received was not available.

Biological samples

In most cases, the FFPE blocks were accompanied with their respective slide stained with hematoxylin and eosin; the tumor area was bounded by the pathologist of the institution where the sample came from. For FFPE blocks that had < 20% of tumoral tissue, the tumoral area was microdissected until reaching an area of approximately 20 mm² of tumoral tissue since the limit of detection of SS mutation screening is equal or greater than 10% of mutated cells in a background of wild-type cells. When only blocks were received, one section was obtained and stained with hematoxylin and eosin; a certified pathologist was on hand to identify the tumor region of interest. Additional histological information about the tumors was not available.

DNA extraction and mutation analysis

DNA extraction from scrapped tumor areas performed with the QIAamp DNA FFPE Tissue Kit (QIAGEN, Mexico City, Mexico) was used as a template to amplify the exons of interest of RAS genes by PCR followed by SS using an automated capillary electrophoresis sequencer from Applied Biosystems (Mexico City, Mexico) as described in our previous study9. Primer pairs for amplification of exons 2, 3, and 4 of KRAS and NRAS genes are described in Table S1.

Commercial kits

Conceptual analysis of the sequencing revealed mutations was performed based on the information in the fact sheets from Cobas KRAS Mutation Test and the therascreen KRAS RGQ PCR Kit describing the mutations that can be detected by them, as well as the reported limits of detection.

Public and patient involvement statement

Patients signed informed consent before the performance of tests where it is agreed that their results may be used for research purposes. This study was carried out as a result of the finding of uncommon mutations in routine tests. This is a retrospective study; patients were not specifically recruited to perform this analysis. The information obtained will not be notified to patients unless it represents a risk to their health.

RESULTS

In our laboratory, of the 1113 samples tested for KRAS, 375 (~33.69%) harbored a mutation. From these, 267 were in codon 12 (~72%) and 103 in codon 13 (~28%) (exon 2). Codons 59 and 146 (exon 3 and 4, respectively) accounted for the remaining ~0.1%, with incidences of one case for each codon. In KRAS, we found c.36T > A and c.39C > T mutations in codons 12 and 13, respectively, which are synonymous mutations not available for screening by the FDA-approved qPCR kits.

These devices can detect all non-synonymous mutations in codon 12 (Table 1). However, they cannot detect the mutations c.37G > A (p.G13S) and c.38G > C (p.G13A), which account for 0.45% of false negatives according to our results. In addition, we found unique cases of the mutations c.176C > T (p.A59V) and c.183A > C (p.Q61H) in exon 3 and c.436G > A (p.A146T) in exon 4.

Table 1 Comparison of Sanger sequencing versus commercial kits for detecting RAS mutations 

Mutationa COSMIC ID Type of mutationb Sanger sequencing Commercial kits mutation coveragee

Incidencec %d % p/codon 1 2
KRAS Codon 12 (n = 1113)

c.35G > A p.(G12D) 55497369 NS 129 11.59 48.31
c.35G > T p.(G12V) 55497419 NS 66 5.93 24.72
c.34G > T p.(G12C) 55497469 NS 28 2.52 10.49
c.35G > C p.(G12A) 55497479 NS 25 2.25 9.36
c.34G > A p.(G12S) 55497461 NS 12 1.08 4.49
c.34G > C p.(G12R) 55497582 NS 5 0.45 1.87
c.36T > A p.(G12=) 55510792 S 2 0.18 0.75
267 23.99 100.00

KRAS Codon 13 (n = 1113)

c.38G > A p.(G13D) 55497388 NS 80 7.19 77.67
c.39C > T p.(G13=) 55756103 S 8 0.72 7.77
c.37G > T p.(G13C) 55497378 NS 9 0.81 8.74
c.37G > A p.(G13S) 55509530 NS 4 0.36 3.88
c.37G > C p.(G13R) 55502117 NS 1 0.09 0.97
c.38G > C p.(G13A) 55497357 NS 1 0.09 0.97
103 9.25 100.00

KRAS Codon 61 (n = 1113)

c.183A > C p.(Q61H) 55498802 NS 1 0.09 100.00
1 0.09 100.00

KRAS Codon 146 (n = 1113)

c.436G > A p.(A146T) 55501778 NS 1 1.20 100.00
1 0.09 100.00

NRAS Codon 12 (n = 83)

c.34G > T p.(G12C) 54736487 NS 1 1.20 33.33
c.35G > A p.(G12D) 54736383 NS 1 1.20 33.33
c.35G > C p.(G12A) 54736555 NS 1 1.20 33.33
3 3.61 100.00
Total 375 33.69 94.0% 94.0%

aMutations found by Sanger sequencing.

bSynonymous (S) or non-synonymous (NS).

cTotal number of patients harboring each mutation.

dPercentage of each mutation over total samples analyzed by SS.

eCommercial kits numbered as follows: 1, Cobas® KRAS Mutation Test; 2, TheraScreen®: KRAS Mutation Kit. Percentages on bottom represent the proportion of mutations covered by each kit, calculated as the sum of the incidence of all covered mutations divided by the total of mutations in codons 12 and 13.

Of the 83 tumors analyzed for NRAS, the mutations c.34G > T, c.35G > A, and c.35G > C in codon 12 (~4%) were found. None of the kits included in this study include mutation screening in the NRAS gene; the commercial kits included would have reported 6% of false negatives.

There were no significant differences in the frequency of mutations by age or sex.

DISCUSSION

Our findings agree with the previous studies, in which 35-40% incidence has been reported for KRAS mutations4,8,14,19. Nonetheless, recent studies have unveiled the importance and potential risk of synonymous mutations in disease causation, suggesting that these mutations could affect the predictive value of different malignancies20,21. Physiological effects of synonymous mutations could be related to cancer through diverse mechanisms, such as altering splicing, RNA structure and stability, the translational rate, and other unknown mechanisms21. Moreover, according to a study based on a data set of 292,405 missense mutations and 123,193 synonymous mutations identified in the exomes of 3851 cancer samples, synonymous mutations tend to be enriched in oncogenes (but not in tumor suppressor genes) in a cancer type-specific manner and often affect splicing motifs22. This suggests a greater relevance for synonymous mutations in cancer than previously thought, hence highlighting the importance of detecting and reporting these “silent” mutations. The KRAS c.36T > A (p.G12=) and c.39C > T (p.G13=) synonymous mutations found in this study are predicted to be pathogenic by Functional Analysis through Hidden Markov Models (FATHMM) (COSMIC mutation IDs COSV55573999 and COSV55510792, respectively). The first one has been reported as a somatic mutation in the biliary tract, large intestine, and pancreas carcinomas, while the latter has been found in the large intestine, hematopoietic and lymphoid tissue, pancreas, endometrium, and biliary tract carcinomas23.

Mutations c.37G > A (p.G13S) and c.38G > C (p.G13A), despite being in codon 13, are not covered by FDA-approved qPCR-based kits but were found in our study. These are known predictive biomarkers to determine anti-EGFR therapies. The first has been found distributed in the large intestine, thyroid, stomach, pancreas, and lung carcinomas, while the latter, in the large intestine, lung, ovary, pancreas, and thyroid carcinomas (COSMIC mutation IDs COSV55509530 and COSV55497357, respectively).

In exon 3 of KRAS, we found the c.176C > T (p.A59V) mutation that has not been reported in mCRC, but in the breast (accession RCV000119371.1 in NCBI ClinVar database) and thyroid cancers (COSMIC mutation ID COSV55904854). In contrast, the c.183A > C (pQ61H) mutation (also in exon 3 of KRAS) has been found distributed in the large intestine, hematopoietic and lymphoid tissue, lung, and biliary tract cancer lesions (COSMIC mutation ID COSV55498802). Both mutations are predicted to be pathogenic by FATHMM. We also found the c.436G > A (p.A146T) mutation in exon 4 of KRAS, which is predicted as pathogenic by FATHMM and has been reported to be present in the large intestine, hematopoietic and lymphoid tissue, stomach, biliary tract, and pancreas carcinomas. Further research should explore the effect of these mutations on mCRC proliferation and treatment, and whether they imply a clinical risk or interfere with anti-EGFR antibody treatments.

The mutations found in codon 12 of NRAS in this study have been well documented to predict resistance to anti-EGFR therapy24.

It is worth mentioning that all specimens in this study corresponded to Stage IV carcinomas. The previous studies have shown that the frequency of mutations varies according to the stages25, and for a wider analysis, further studies should include samples from all stages.

Despite 6% of false negatives that would be reported if FDA-approved companion diagnostic tests were used, SS has important constraints when compared to these kits: the processing time is much longer in SS due to the additional steps required (amplification, cycle sequencing, purification of products, and capillary electrophoresis) and the time needed to analyze results. In addition, some problems can arise in the process due to degradation of DNA and the presence of inhibitors as a result of formalin fixation and dewaxing, respectively26. Furthermore, the limit of detection of SS is considerably lower than that of kits: a mutation can be detected by SS in a specimen with at least 20% mutant DNA in a background of wild-type DNA, which means ~20-40% heterozygous mutant cells27. On the other hand, the commercial kits here discussed have reported sensitivities of <5% of mutated DNA in a wild-type background. Despite its modest limit of detection and relative complexity, SS is currently one of the most reliable techniques for mutation discovery and validation, especially helpful in molecular genetic studies of genetically poorly characterized populations18. Studies have reported an overall concordance of 97.5% between SS and qPCR for KRAS mutation detection28. Nevertheless, the drawbacks must be considered in the selection of the technique to be used for a certain application. A notable limitation of SS is that large deletion/duplications could not be detected. An alternative for the detection of low-frequency mutations that are not covered by commercial kits could be next-generation sequencing, which can cover the whole sequence of target genes with an even higher sensitivity than SS29,30, although its cost still prevents it from competing with SS in a commercial laboratory.

Similar studies analyzing additional mutations to those found in KRAS codons 12 and 13 in American mCRC patients using SS have reported that 17% of specimens harbor potentially relevant RAS mutations that are missed with tests limited to codons 12 and 13 of KRAS31.

In conclusion, commercial kits based on technologies such as qPCR and STA-FA can be very practical for routine molecular diagnosis given their high sensitivity and efficiency, their mutation coverage leaves out rare mutations from poorly genetically characterized populations. However, kits could be used for clinical investigation or codon-specific research but not for the identification of low-frequency mutations, tracing of unreported mutations, or analyses in new populations. For those purposes, SS or other sequencing techniques are more suitable, especially considering that the life of mCRC patients could depend on the results obtained.

It should be noted that NRAS should also be included in every companion diagnostic for mCRC, at least in the Mexican population, as we found that ~4% of the 83 specimens analyzed for this gene contained a mutation. Given the non-neglectable frequency of KRAS mutations in mCRC revealed here, to understand their contribution to tumor proliferation, further studies should explore their possible association with the outcome of anti-EGFR therapies.

ACKNOWLEDGMENTS

We thank all the current and previous Vitagénesis personnel that made it possible to obtain and process all the specimens included in this study. We also thank all patients’ advisers for participating in this study. This research was supported in part by the National Council on Science and Technology, Mexico (CONACYT) (Grant Nos. 185427, ECO-2015-C01-260826, 294875, and 280114).

SUPPLEMENTARY DATA

Supplementary data are available at Revista de Investigación Clínica online (www.clinicalandtranslationalinvestigation.com). These data are provided by the corresponding author and published online for the benefit of the reader. The contents of supplementary data are the sole responsibility of the authors.

SUPPLEMENTARY MATERIAL

REFERENCES

1. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, et al. Estimating the global cancer incidence and mortality in 2018:GLOBOCAN sources and methods. Int J Cancer. 2015;136:359-86. [ Links ]

2. de la Chapelle A. Genetic predisposition to colorectal cancer. Nat Rev Cancer. 2004;4:769-80. [ Links ]

3. Smith G, Bounds R, Wolf H, Steele RJ, Carey F, Wolf CR. Activating K-ras mutations out with “hotspot“codons in sporadic colorectal tumours:implications for personalised cancer medicine. Br J Cancer. 2010;102:693-703. [ Links ]

4. Martinez-Garza SG, Núñez-Salazar A, Calderon-Garcidueñas AL, Bosques-Padilla FJ, Niderhauser-García A, Barrera-Saldaña HA. Frequency and clinicopathology associations of K-ras mutations in colorectal cancer in a Northeast Mexican population. Dig Dis. 1999;17:225-9. [ Links ]

5. Jiang Y, Kimchi ET, Staveley-O'Carroll KF, Cheng H, Ajani J. Assessment of K-ras mutation:a step toward personalized medicine for patients with colorectal cancer. Cancer. 2009;115:3609-17. [ Links ]

6. Mcneil BC. K-ras mutations are changing practice in advanced colorectal cancer. J Natl Cancer Inst. 2008;100:1667-9. [ Links ]

7. Wójcik P, Kulig J, Oko K, Zazula M, Modzioch L, Niepsuj A, et al. KRAS mutation profile in colorectal carcinoma and novel mutation internal tandem duplication in KRAS. Polish J Pathol. 2008;59:93-6. [ Links ]

8. Tan C, Du X. KRAS mutation testing in metastatic colorectal cancer. World J Gastroenterol. 2012;18:5171-80. [ Links ]

9. Cárdenas-Ramos SG, Alcázar-González G, Reyes-Cortés LM, Torres-Grimaldo AA, Calderón-Garcidueñas AL, Morales-Casas J, et al. The frequency and type of K-RAS mutations in Mexican patients with colorectal cancer. Am J Clin Oncol. 2017;40:274-6. [ Links ]

10. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3:11-22. [ Links ]

11. Sjöblom T, Jones S, Wood L, Parsons DW, Lin J, Barber TD, et al. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314:268-74. [ Links ]

12. Martin TD, Samuel JC, Routh ED, Der CJ, Yeh JJ. Activation and involvement of ral GTPases in colorectal cancer. Cancer Res. 2011;71:206-15. [ Links ]

13. Shi C, Washington K. Molecular testing in colorectal cancer:diagnosis of lynch syndrome and personalized cancer medicine. Am J Clin Pathol. 2012;137:847-59. [ Links ]

14. Vaughn CP, Zobell SD, Furtado LV, Baker CL, Samowitz WS. Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Genes Chromosom Cancer. 2011;50:307-12. [ Links ]

15. Andreyev HJ, Norman R, Cunningham D, Oates JR, Clarke P. Kirsten ras mutations in patients with colorectal cancer:the multicenter “RASCAL“study. J Natl Cancer Inst. 1998;90:675-84. [ Links ]

16. Arrington AK, Heinrich EL, Lee W, Duldulao M, Patel S, Sanchez J, et al. Prognostic and predictive roles of KRAS mutation in colorectal cancer. Int J Mol Sci. 2012;13:12153-68. [ Links ]

17. Khoo C, Rogers TM, Fellowes A, Bell A, Fox S. Molecular methods for somatic mutation testing in lung adenocarcinoma:EGFR and beyond. Transl Lung Cancer Res. 2015;4:126-41. [ Links ]

18. Jancik S, Drabek J, Berkovcova J, Xu Y, Stankova M, Klein J, et al. A comparison of direct sequencing, pyrosequencing, high resolution melting analysis, TheraScreen DxS, and the K-ras StripAssay for detecting KRAS mutations in non small-cell lung carcinomas. J Exp Clin Cancer Res. 2012;31:79. [ Links ]

19. Neumann J, Zeindl-Eberhart E, Kirchner T, Jung A. Frequency and type of KRAS mutations in routine diagnostic analysis of metastatic colorectal cancer. Pathol Res Pract. 2009;205:858-62. [ Links ]

20. Bosch TM, Meijerman I, Beijnen JH, Schellens JH. Genetic polymorphisms of drug-metabolising enzymes and drug transporters in the chemotherapeutic treatment of cancer. Clin Pharmacokinet. 2006;45:253-85. [ Links ]

21. Sauna ZE, Kimchi-Sarfaty C, Ambudkar SV, Gottesman MM. Silent polymorphisms speak:how they affect pharmacogenomics and the treatment of cancer. Should synonymous single-nucleotide polymorphisms be ignored?Cancer Res. 2007;67:9609-12. [ Links ]

22. Supek F, Miñana B, Valcárcel J, Gabaldón T, Lehner B. Synonymous mutations frequently act as driver mutations in human cancers. Cell. 2014;156:1324-35. [ Links ]

23. Dong LQ, Shi Y, Ma LJ, Yang LX, Wang XY, Zhang S, et al. Spatial and temporal clonal evolution of intrahepatic cholangiocarcinoma. J Hepatol. 2018;69:89-98. [ Links ]

24. Therkildsen C, Bergmann TK, Henrichsen-Schnack T, Ladelund S, Nilbert M. The predictive value of KRAS, NRAS, BRAF, PIK3CA and PTEN for anti-EGFR treatment in metastatic colorectal cancer:a systematic review and meta-analysis. Acta Oncol (Madr). 2014;53:852-64. [ Links ]

25. Shen Y, Han X, Wang J, Wang S, Yang H, Lu SH, et al. Prognostic impact of mutation profiling in patients with stage II and III colon cancer. Sci Rep. 2016;6:1-7. [ Links ]

26. Dietrich D, Uhl B, Sailer V, Holmes EE, Jung M, Meller S, et al. Improved PCR performance using template DNA from formalin-fixed and paraffin-embedded tissues by overcoming PCR inhibition. PLoS One. 2013;8:e77771. [ Links ]

27. Ogino S, Kawasaki T, Brahmandam M, Yan L, Cantor M, Nangyal C, et al. Sensitive sequencing method for KRAS mutation detection by pyrosequencing. J Mol Diagn. 2005;7:413-21. [ Links ]

28. Lee S, Brophy VH, Cao J, Velez M, Hoeppner C, Soviero S, et al. Analytical performance of a PCR assay for the detection of KRAS mutations (codons 12/13 and 61) in formalin-fixed paraffin-embedded tissue samples of colorectal carcinoma. Virchows Arch. 2012;460:141-9. [ Links ]

29. Jiang L, Huang J, Morehouse C, Zhu W, Korolevich S, Sui D, et al. Low frequency KRAS mutations in colorectal cancer patients and the presence of multiple mutations in oncogenic drivers in non-small cell lung cancer patients. Cancer Genet. 2013;206:330-9. [ Links ]

30. Tougeron D, Lecomte T, Pagès JC, Villalva C, Collin C, Ferru A, et al. Effect of low-frequency KRAS mutations on the response to anti-EGFR therapy in metastatic colorectal cancer. Ann Oncol. 2013;24:1267-73. [ Links ]

31. Netzel BC, Grebe SK. Companion-diagnostic testing limited to KRAS codons 12 and 13 misses 17% of potentially relevant RAS mutations in colorectal cancer. Clin Chim Acta. 2013;425:1-2. [ Links ]

Received: March 19, 2020; Accepted: May 18, 2020

* Corresponding author: Hugo A. Barrera-Saldaña E-mail: habarrera@gmail.com

Creative Commons License Instituto Nacional de Cardiología Ignacio Chávez. Published by Permanyer. This is an open ccess article under the CC BY-NC-ND license