Open Access

Impact of microsatellite status on chemotherapy for colorectal cancer patients with KRAS or BRAF mutation

  • Authors:
    • Chi‑Jung Huang
    • Shih‑Hung Huang
    • Chih‑Cheng Chien
    • Henry Hsin‑Chung Lee
    • Shung‑Haur Yang
    • Chun‑Chao Chang
    • Chia‑Long Lee
  • View Affiliations

  • Published online on: October 18, 2016     https://doi.org/10.3892/ol.2016.5275
  • Pages: 4427-4434
  • Copyright: © Huang 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

KRAS and BRAF mutations are frequently detected in cases of colorectal cancer (CRC). The microsatellite status of patients with CRC and mutated KRAS/BRAF is important when determining cancer therapy. In the present study, the microsatellite status and genetic polymorphisms of KRAS (codons 12 and 13) and BRAF (V600E) were characterized in CRC tissue. The mismatch repair activity and oncogenic potential of KRAS were assessed by immunoblots from two KRAS‑mutated CRC cell lines, SW480 and HCT116, with different microsatellite statuses, following treatment with 5‑fluorouracil (5‑FU) and oxaliplatin. Of all the 205 patients with CRC enrolled in the present study, 31.2% (64 of 205) had a KRAS or BRAF mutation, and 79.7% (51 of 64) of these patients with a KRAS/BRAF mutation exhibited microsatellite stability (MSS), indicating that microsatellite status is correlated with KRAS/BRAF mutation (P=0.027). A higher proportion (39.0%, 41 of 105) of elderly patients (≥62.6 years) had mutated KRAS or BRAF than younger patients (<62.6 years; 23.0%, 23 of 100; P=0.013). In the subgroup of 154 patients with MSS, patients without the KRAS or BRAF mutation (n=110) had longer disease‑specific survival rates (58.8±9.4%) than patients with KRAS or BRAF mutations (n=44; 50.6±11.0%; P=0.043). Cytoplasmic KRAS levels decreased whereas nuclear MutS protein homolog 2 (MSH2) levels increased slightly in CRC HCT116 cells that were microsatellite instable, following treatment with 76.9 µM 5‑FU for 2 days. In microsatellite stable SW480 cells, MSH2 levels markedly increased in the nucleus following 150 µM oxaliplatin treatment for 3 days. However, no significant change was observed regarding KRAS distribution in these cells. The results of the present study suggest that it is important to identify patients with CRC who may benefit from adjuvant chemotherapy with 5‑FU or oxaliplatin, particularly CRC patients with MSS and mutated KRAS or BRAF, who have poorer overall survival rates than patients with microsatellite instability. Knowledge of the microsatellite status of patients and whether they harbor KRAS or BRAF mutations may enable more effective therapeutic strategies to be developed. Further prospective studies are required to validate the findings of the current study.

Introduction

Colorectal cancer (CRC) is a leading cause of cancer-associated mortality (1). CRC typically develops slowly by the progressive accumulation of genetic mutations, which cause the normal colonic epithelium to transform into adenocarcinoma (2,3).

Defects in the DNA mismatch repair (MMR) system may arise sporadically or in patients with hereditary nonpolyposis colorectal cancer (HNPCC) syndrome, may be inherited in an autosomal-dominant manner (24). Tumors are characterized by the presence of microsatellite instability (MSI), caused either by genetic changes or by attenuating the expression of proteins in the DNA MMR pathway (5,6). Among MMR genes, abnormalities of MutL homolog 1 (MLH1) and MutS protein homolog 2 (MSH2) have been the subject of several studies investigating CRC (7,8), and it has been determined that the microsatellite status of CRC patients should be evaluated prior to chemotherapy (9). Furthermore, genetic studies have demonstrated that mutations of the GTPase oncogenes, KRAS and BRAF in the mitogen-activated protein kinase (MAPK) pathway, also known as the RAS-RAF-extracellular signal-regulated kinase (ERK)-MAPK/ERK kinase pathway, are detected in a high proportion of CRC patients, including those with defective MMR activity (1012). Activation of the MAPK pathway is important in MSI CRC tumorigenesis (13). In addition, analysis of KRAS mutations has demonstrated an association with sporadic CRC (14,15). Elucidation of the microsatellite status of CRC patients may indicate what type of adjuvant chemotherapy is the most beneficial for a particular patient (9). Therefore, knowledge of MMR activity and KRAS/BRAF mutation status may provide further valuable guidance for planning therapeutic strategies (16).

CRC patients with microsatellite stability (MSS) and KRAS/BRAF mutations usually have a poor prognosis (17). Therefore, personalizing treatment based on patient tumor characteristics is advantageous (18,19). However, only a few studies indicate that distinct chemotherapy is appropriate for CRC patients with different microsatellite status, MMR activity and KRAS/BRAF mutation (20). The present study aimed to assess the MSI status of CRC tumors and the presence of KRAS/BRAF mutation in patients with CRC, and to evaluate the outcome of treating cells from two CRC cell lines, HCT116 and SW480, with different microsatellite statuses, with the chemotherapeutic agents 5-fluorouracil (5-FU) and oxaliplatin (2125).

Materials and methods

Patients and general data collection

A total of 205 patients with CRC (121 males and 84 females; mean age, 62.6 years; range, 25.4–90.1 years) from the Gastrointestinal Department of Cathay General Hospital (Taipei, Taiwan) were enrolled from January 2006 to December 2008 in the current study. Survival data were acquired from 176 patients and others were lost to follow-up due to referral. The mean follow-up time was 17.0±15.6 months (median, 10.5 months). Suspicious growths in patient colonic tissues were sampled with small biopsy forceps inserted through a colonoscope. The tissues were formalin-fixed, paraffin-embedded and cut into slices of 4–5-µm thickness for immunohistochemical staining, or immersed in RNAlater® solution (Thermo Fisher Scientific, Inc., Waltham, MA, USA) for genomic DNA preparation, according to the manufacturer's protocol. Presence of distant metastasis was routinely confirmed by abdominal computed tomography. In addition, blood samples were collected from each patient to serve as controls when determining the microsatellite status. The study protocol was approved by the Institutional Review Board of Cathay General Hospital, and informed consent was obtained from all patients prior to obtaining tissue samples.

Colonic cell lines, protein extraction, and western blotting

Cells from the human colorectal carcinoma HCT116 [American Type Culture Collection (ATCC) no. CCL-247; MSI) and SW480 (ATCC no. CCL-228; MSS) cell lines were purchased from the ATCC (Manassas, VA, USA) and maintained as recommended by their guidelines (www.atcc.org) (22,23). All cultured cells used in the current study were washed in ice-cold PBS (pH 7.4), scraped from culture dishes on ice using a plastic cell scraper and collected in 1.5-ml microcentrifuge tubes in 1 ml ice-cold PBS. Following a short centrifugation step (1,000 × g for 3 min at 4°C) to pellet the cells, the supernatants were removed from each sample, and the cell pellets were resuspended in 900 µl ice-cold 0.1% NP-40 (Merck Millipore, Darmstadt, Germany) in PBS. Subsequently, a reagent-based protocol that enabled the stepwise lysis of cells, separation of cytoplasm from intact nuclei and extraction of nuclear proteins was performed, according to the protocol of the NE-PER™ Nuclear and Cytoplasmic Extraction kit (Thermo Fisher Scientific, Inc.). The cytoplasmic (10 µg) and nuclear (10 µg) fractions underwent 10% SDS-PAGE and were subsequently transferred to polyvinylidene difluoride membranes. Membranes were then blocked with 10% skimmed milk and 3% bovine serum albumin in TBS buffer containing Tween-20 [TBST; 20 mM Tris (pH 7.4), 150 mM NaCl and 0.05% Tween 20] for 1 h to prevent non-specific binding of antibodies. Mutated KRAS and MSH2 were immunoblotted with anti-KRAS antibody (1:2,000 in TBST; 05–516; Merck Millipore) and anti-MSH2 antibody (1:500 in TBST; ab52266; Abcam, Cambridge, MA, USA) for 1 h, respectively. In addition, membranes were incubated with an anti-α-tubulin antibody (1:500 in TBST; sc-5286; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) as a cytoplasmic marker, or with anti-lamin A/C antibody (1:500 in TBST; sc-7292; Santa Cruz Biotechnology, Inc.) as nuclear marker, for 1 h each. Protein signals were detected with Western Lightning Chemiluminescence Reagent Plus (PerkinElmer, Inc., Waltham, MA, USA) upon incubation with an appropriate secondary antibody conjugated with peroxidase (1:5,000 in TBST; Dako, Glostrup, Denmark) for 1 h. All incubations were performed at room temperature on a rocking platform and were followed by several washes with TBST buffer to remove the residual solution. Protein bands were quantified by densitometry using image processing AlphaView software version 3.2.2.0 for the FluorChem FC2 system (Cell Biosciences, Inc., Santa Clara, CA, USA). Relative protein levels were calculated and determined by normalizing their expression to that of a-tubulin or lamin A/C.

Immunohistochemical staining for CRC tissues

Sections were dewaxed in xylene and rehydrated through a graded series of ethanol concentrations (100, 95 and 70%) to water. Antigen retrieval was performed by immersing the slides in BD Retrievagen A (pH 6.5 for MLH1 and pH 6.0 for MSH2; BD Biosciences, San Jose, CA, USA) and heating the slides in a microwave oven for 30 min at 95°C. Sections were treated with 3% hydrogen peroxide for 5 min to block endogenous peroxidase activity. Sections were washed in PBS and subsequently placed in 20% normal goat serum (G9023; Sigma-Aldrich; Merck Millipore) in PBS for 20 min to reduce non-specific staining. Sections were then incubated with monoclonal antibodies raised against human (h) MLH1 (1:10; clone G168-15; BD Biosciences) for 1 h 32 min at 42°C or against MSH2 (1:25; clone 25D12; Thermo Fisher Scientific, Inc.) for 1 h at room temperature. Subsequently, visualization was performed using the EnVision+Dual Link system-HRP (Dako) according to the manufacturer's protocol, using 3,3′-diaminobenzidine as a chromogen. Finally, slides were counterstained with hematoxylin solution. Both MLH1 and MSH2 were scored as either negative or positive staining. Tissue specimens were analyzed by two independent pathologists blinded to the conditions.

Determining microsatellite status and detecting mutations of KRAS and BRAF

Colonic tissues pathologically diagnosed as CRC were subjected to the assessment of microsatellite status and identification of hotspot mutations of KRAS and BRAF. Peripheral blood samples from patients diagnosed with CRC were collected in anticoagulant tubes containing EDTA. Genomic DNA was extracted from blood and tissue samples according to a standard protocol (26). To determine the microsatellite status of the enrolled CRC patients, a reference panel of five fluorescent dye-labeled microsatellite primers (BAT25, BAT26, D2S123, D5S346 and D17S250) purchased from Thermo Fisher Scientific, Inc., was used (27). Primer sets for the BAT25, BAT26 and D5S346 loci, and for the D2S123 and D17S250 loci, were respectively combined for the multiplex polymerase chain reaction (PCR) amplifications in a volume of 5.5 µl containing 20.0 ng genomic DNA. Denatured PCR products were analyzed by capillary electrophoresis in an ABI Prism® 3100 Genetic Analyzer (Applied Biosciences; Thermo Fisher Scientific, Inc.). Raw data were collected using Data Collection Software version 1.1 (Applied Biosciences), and marker performance was evaluated using GeneScan® Analysis Software version 3.7 (Applied Biosciences) electropherograms in full-view display for each dye color. For the purpose of prognostic evaluation, microsatellite status was categorized as MSS or MSI according to whether instability was evident for no markers or for ≥1 marker, respectively (28). To detect KRAS mutations at codons 12 and 13, a restriction enzyme-based analysis (BstNI for codon 12 and XcmI for codon 13) following appropriate PCR amplifications was employed, as previously described (29,30). Briefly, the cycling conditions for codon 12 were 35 cycles of amplification (93°C for 35 sec, 56°C for 50 sec and 72°C for 50 sec), and for codon 13 were 40 cycles of amplification (93°C for 60 sec, 52°C for 60 sec and 72°C for 60 sec). The sequences of the PCR primers used were: BstNI forward, 5′-ACTGAATATAAACTTGTGGTAGTTGGACCT-3′ and reverse, 5′-TCATGAAAATGGTCAGAGAAACC-3′; and XcmI, forward, 5′-ACTGAATATAAACTTGTGGTCCATGGA-3′ and reverse, 5′-TATCTGTATCAAAGAATGGTCCTGCACCAG-3′ (30,31). The presence of the BRAF V600E mutation was determined using an allele-specific PCR assay (32). Depending on the genotypes, either allele-specific reaction could amplify the target sequence. The cycling conditions used for PCR were 10 min at 95°C for initial activation and 40 cycles of amplification (92°C for 15 sec and 60°C for 60 sec).

The components of the reaction for the different genotypes were identical except for the respective allele-specific probes, as described below. The primer sequences were: Forward, 5′-CATGAAGACCTCACAGTAAAAATAGGTGAT-3′ and reverse, 5′-TGGGACCCACTCCATCGA-3′. Allele-specific TaqMan® probes (VIC®-labeled reporter T allele, 5′-CTAGCTACAG[T]GAAATC-3′ and 6-carboxyfluorescein-labeled reporter A allele, 5′-TAGCTACAG[A]GAAATC-3′) (Applied Biosciences) and TaqMan Genotyping Master Mix (Applied Biosciences) were used in a 7300 Real-Time PCR system (Applied Biosciences; Thermo Fisher Scientific, Inc.) (32). All identified KRAS/BRAF mutations were then validated using a BigDye® Terminator v3.1 Cycle Sequencing kit (Applied Biosciences; Thermo Fisher Scientific, Inc.).

Statistical analysis

The association between tumor microsatellite status and patient clinicopathological features was analyzed using a χ2 test. Patient survival time was calculated from the date of complete resection of CRC tumors to the date of last follow-up, with the only patient who succumbed to CRC being excluded from counting towards disease-specific survival (DSS). DSS distributions were estimated using a Kaplan-Meier method and compared using a log-rank test. P<0.05 was considered to indicate a statistically significant difference. All calculations were performed using IBM SPSS Statistics for Windows version 22.0 (IBM SPSS, Armonk, NY, USA).

Results

Microsatellite status and mutation rates of KRAS and BRAF

One patient (age, 58.4 years; female) out of the 205 enrolled patients met the clinical diagnostic criteria for HNPCC (33,34). Immunohistochemical results indicated that staining for hMLH1 was negative for this patient (Fig. 1). As presented in Table I, 12.7% of the tumors (26 of 205) exhibited MSI, including the tumor in the patient with HNPCC. The mean age of the 26 patients with MSI (58.2 years; range, 29.0–81.6 years) was lower than that of the other 179 patients with MSS (63.3 years; range: 25.4–90.1 years). Furthermore, a KRAS or BRAF mutation (5 for BRAF V600E, 24 for KRAS codon 12 and 4 for KRAS codon 13) was detected in 31.2% of patients (64 of 205). A total of 79.7% (51 of 64) of patients with KRAS/BRAF mutations had MSS (P=0.027), demonstrating that a correlation exists between MSS and KRAS/BRAF mutations (Table II). When comprehensively considering the microsatellite status of patients and the site of the tumors, tumors at distal/rectal areas were significantly diagnosed in patients with MSS (90.4%, 142 of 157; P=0.015).

Table I.

Clinicopathological characteristics of patients.

Table I.

Clinicopathological characteristics of patients.

FeatureNumber of patientsa
Age, years
  ≤62.6100
  >62.6105
Gender
  Male121
  Female  84
AJCC staging
  I+II  73
  III+IV  88
Microsatellite status
  MSS179
  MSI  26
KRAS/BRAF mutation
  Wild type141
  Mutant  64
Differentiation
  Well/moderate157
  Poor  15
Tumor location
  Right  48
  Left  91
  Rectum  66
Tumor size, cm
  ≤4.3  89
  >4.3  82

a The number of cases that were assessed in each category was dependent on the number of available cases. All information on female patients includes clinical information regarding the aforementioned patient with hereditary nonpolyposis colorectal cancer. AJCC, American joint committee on cancer; MSS, microsatellite stability; MSI, microsatellite instability.

Table II.

High percentage of patients with MSS in different CRC groups.

Table II.

High percentage of patients with MSS in different CRC groups.

FeaturePercentage, % (n)P-value
Microsatellite status vs. KRAS/BRAF mutation
  MSS79.7 (51/64)0.027
  MSI20.3 (13/64)
Microsatellite status vs. distal/rectal sites
  MSS90.4 (142/157)0.015
  MSI9.6 (15/157)

[i] MSS, microsatellite stability; MSI, microsatellite instability.

Significantly different mutation rates of KRAS or BRAF were observed in patients of different ages (P=0.013; Table III). A higher proportion of elderly patients (≥62.6 years) had mutated KRAS or BRAF (39.0%, 41 of 105), whereas only 23.0% (23 of 100) of younger patients (<62.6 years) harbored these mutations. The odds ratio for KRAS or BRAF mutations in the elderly patients was 2.15 (95% confidence interval, 1.17–3.94). In addition, of the 172 patients with available data for tumor differentiation, 15 were diagnosed with poorly differentiated tumors, and 53.3% (n=8) of these patients harbored a KRAS or BRAF mutation (P=0.036).

Table III.

Mutation rate of KRAS/BRAF according to patient's age and tumor differentiation.

Table III.

Mutation rate of KRAS/BRAF according to patient's age and tumor differentiation.

Feature KRAS/BRAF mutant, % (n)P-valueOdds ratio95% CI
Age, yearsa
  ≤62.6     23.0 (23/100)0.0132.151.17–3.94
  >62.6     39.0 (41/105)
Differentiationb
  Well/moderate     27.4 (43/157)0.0363.031.04–8.86
  Poor53.3 (8/15)

a Mean age, 62.6 years.

b The number of assessed cases was dependent on the number of available cases. CI, confidence interval.

Different survival rates according to KRAS or BRAF mutations in patients with MSS

In the present study, survival data were available for 176 patients with CRC (154 patients with MSS and 22 patients with MSI). Patients with MSS tended to have poorer DSS (57.4±7.0%) compared with patients with MSI, although this difference was not significant (P=0.065; Fig. 2). In addition, it has been demonstrated that BRAF and KRAS mutations are frequently associated with CRC and influence prognosis (35). Therefore, the current study assessed the survival rates of patients with MSS according to their different KRAS/RAS mutations. In the subgroup of 154 patients with MSS, patients without KRAS or BRAF mutations (n=110) had better DSS (58.8±9.4%) than those with KRAS or BRAF mutations (n=44; 50.6±11.0%; P=0.043; Fig. 3).

Different responses to 5-FU- or oxaliplatin-based adjuvant treatment in CRC cell lines

Due to the clinical significance of microsatellite status and KRAS/BRAF mutations in CRC, MSS (SW480 cells; Fig. 4) and MSI (HCT116 cells; Fig. 5) cell lines were employed in the present study (22,23). Both SW480 and HCT116 cells had mutated KRAS but wild-type BRAF (36). In SW480 cells treated with 17.5 µM 5-FU for 2 days or 150 mM oxaliplatin for 3 days, the levels of KRAS detected did not significantly differ between the nucleus and the cytoplasm (Fig. 4A and B). However, under the same treatment conditions, the expression of the DNA MMR protein, MSH2, varied in SW480 cells (Fig. 4C). MSH2 expression increased in the cytoplasm and decreased in the nucleus of SW480 cells following treatment with 5-FU (both P<0.001). By contrast, MSH2 expression decreased in the cytoplasm and increased in the nucleus of SW480 cells treated with oxaliplatin (both P<0.001; Fig. 4B and D).

In addition, distinct expression patterns for KRAS and MSH2 in HCT116 cells treated with 76.9 µM 5-FU for 2 days or 1.4 µM oxaliplatin for 3 days were observed (Fig. 5). The levels of cytoplasmic and nuclear KRAS significantly decreased in HCT116 cells following 5-FU treatment (P=0.050 for cytoplasm and P=0.030 for nucleus; Fig. 5A). However, in cells with treated with oxaliplatin, the levels of nuclear KRAS decreased significantly (P<0.001), while the levels of cytoplasmic KRAS increased but not significantly (P=0.216; Fig. 5B). MSH2 expression decreased in the cytoplasm (P<0.001) and increased in the nucleus (P=0.027) of 5-FU-treated HCT116 cells compared with control cells (Fig. 5C). However, oxaliplatin treatment did not increase the expression of MSH2 in the nucleus of HCT116 cells (Fig. 5D).

Discussion

It has previously been demonstrated that the microsatellite status of CRC patients responds to specific chemotherapy (37). Furthermore, in combination with other genes, assessing the mutation status of KRAS or BRAF may provide guidance during the planning of therapeutic strategies (16,25). In the present study, the majority of enrolled patients were MSS, and their colorectal tumors frequently arose in the distal/rectal colon, similarly to the study by Boland and Goel (24). Another previous study indicated that the highest survival rate was detected in patients with MSI CRC, followed by those with MSS CRC, whereas the lowest survival rate was detected in the subgroup of MSS patients with KRAS or BRAF mutations (38). In the present study, KRAS or BRAF mutations were frequently detected in the tumors of elderly patients, as has previously been demonstrated in the feces of CRC patients (39). Thus, it is important to improve the efficacy of CRC treatment if patients have MSS and KRAS/BRAF mutations (35).

It has been demonstrated that patients with different microsatellite statuses respond differently to chemotherapeutics (24). The results of the current study strongly suggest that more complex tumorigenic pathways exist in MSS CRC than in MSI CRC (40). This may explain why MSS CRC patients have a poorer prognosis than MSI patients, even though they do not have unstable microsatellites (35,41). CRC patients with MSS may slowly and progressively accumulate genetic mutations, including BRAF and KRAS mutations, in the MAPK pathway (3,13). Although the current study demonstrated that the mutation rate of KRAS/BRAF was lower in MSS CRC than in MSI CRC (data not shown), such mutations coupled with the MSS phenotype correlate with metastatic CRC and with high mortality rate (42,43). CRC patients with MSS have aberrant activation of the MAPK pathway (12,13,17). MAPK activation, due to mutational KRAS or BRAF hotspots, has been identified in CRC tumorigenesis (17,44). Mutually exclusive KRAS and BRAF mutations provide useful additional risk stratification of CRC to guide the use of chemotherapy (13,45,46). Therefore, in order to perform appropriate chemotherapy, it is important to identify KRAS or BRAF mutations to treat CRC patients with different microsatellite statuses (18,43,44,47,48).

Two first-line chemotherapeutic agents, 5-FU and oxaliplatin, induce a cytotoxic response, and may be used to treat CRC cells through the stable correction of MMR activity (21,24,25). Combined microsatellite and KRAS/BRAF mutation status provides significant prognostic stratification (16). In the current study, oxaliplatin slightly decreased the level of oncogenic KRAS in the cytoplasm while significantly increased the level of MSH2 in the nucleus of SW480 cells. Similarly, 5-FU was able to induce the same changes in HCT116 cells. Previous studies have demonstrated that decreasing the level of KRAS expression in the cytoplasm would reduce its oncogenic potential (49), and that the translocation of MMR proteins into the nucleus may be induced by increased MMR activity (50,51). Furthermore, Ooki et al (16) have determined that 5-FU is an insufficient treatment for CRC patients with MSS and mutated KRAS or BRAF. Together with the results of the current study, this indicates that oxaliplatin may be an efficient chemotherapeutic agent for certain CRC patients with specific microsatellite statuses. Therefore, identifying CRC patients who would benefit from adjuvant chemotherapy with 5-FU or oxaliplatin by determining their microsatellite and KRAS/BRAF mutation statuses is necessary (52,53). This may enable practitioners to employ more intensive chemotherapy or molecular targeting drugs (52,54).

In conclusion, the microsatellite status and the mutation of KRAS or BRAF should be determined prior to therapeutic decision making. For CRC patients with MSI and KRAS/BRAF mutations, 5-FU treatment is recommended. Otherwise, it is better to perform treatment with oxaliplatin for patients with MSS and KRAS/BRAF mutations. The molecular diagnosis for these CRC patients should be individualized following evaluation of the relevant genetic conditions.

Acknowledgements

The present study was supported by grants from the Cathay General Hospital (Taipei, Taiwan; grant nos. CGH-MR-9419 and CGH-MR-9701 awarded to C.-L. L.).

Glossary

Abbreviations

Abbreviations:

CRC

colorectal cancer

MMR

mismatch repair

HNPCC

hereditary nonpolyposis colorectal cancer

MSI

microsatellite instability

MAPK pathway

mitogen-activated protein kinase pathway

MSS

microsatellite stability

DSS

disease-specific survival

References

1 

Daniel CR, Shu X, Ye Y, Ye Y, Gu J, Raju GS, Kopetz S and Wu X: Severe obesity prior to diagnosis limits survival in colorectal cancer patients evaluated at a large cancer centre. Br J Cancer. 114:103–109. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, Nakamura Y, White R, Smits AM and Bos JL: Genetic alterations during colorectal-tumor development. N Engl J Med. 319:525–532. 1988. View Article : Google Scholar : PubMed/NCBI

3 

Jass JR: Colorectal cancer: A multipathway disease. Crit Rev Oncog. 12:273–287. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Peltomäki P: Deficient DNA mismatch repair: A common etiologic factor for colon cancer. Hum Mol Genet. 10:735–740. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Aaltonen LA, Salovaara R, Kristo P, Canzian F, Hemminki A, Peltomäki P, Chadwick RB, Kääriäinen H, Eskelinen M, Järvinen H, et al: Incidence of hereditary nonpolyposis colorectal cancer and the feasibility of molecular screening for the disease. N Engl J Med. 338:1481–1487. 1998. View Article : Google Scholar : PubMed/NCBI

6 

Yurgelun MB, Goel A, Hornick JL, Sen A, Turgeon DK, Ruffin MT IV, Marcon NE, Baron JA, Bresalier RS, Syngal S, et al: Microsatellite instability and DNA mismatch repair protein deficiency in Lynch syndrome colorectal polyps. Cancer Prev Res (Phila). 5:574–582. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Limburg PJ, Harmsen WS, Chen HH, Gallinger S, Haile RW, Baron JA, Casey G, Woods MO, Thibodeau SN and Lindor NM: Prevalence of alterations in DNA mismatch repair genes in patients with young-onset colorectal cancer. Clin Gastroenterol Hepatol. 9:497–502. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Warrier SK, Trainer AH, Lynch AC, Mitchell C, Hiscock R, Sawyer S, Boussioutas A and Heriot AG: Preoperative diagnosis of Lynch syndrome with DNA mismatch repair immunohistochemistry on a diagnostic biopsy. Dis Colon Rectum. 54:1480–1487. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Saridaki Z, Souglakos J and Georgoulias V: Prognostic and predictive significance of MSI in stages II/III colon cancer. World J Gastroenterol. 20:6809–6814. 2014. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Oliveira C, Pinto M, Duval A, Brennetot C, Domingo E, Espín E, Armengol M, Yamamoto H, Hamelin R, Seruca R and Schwartz S Jr: BRAF mutations characterize colon but not gastric cancer with mismatch repair deficiency. Oncogene. 22:9192–9196. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Seruca R, Velho S, Oliveira C, Leite M, Matos P and Jordan P: Unmasking the role of KRAS and BRAF pathways in MSI colorectal tumors. Expert Rev Gastroenterol Hepatol. 3:5–9. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B and Velculescu VE: Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature. 418:9342002. View Article : Google Scholar : PubMed/NCBI

14 

Nagasaka T, Koi M, Kloor M, Gebert J, Vilkin A, Nishida N, Shin SK, Sasamoto H, Tanaka N, Matsubara N, et al: Mutations in both KRAS and BRAF may contribute to the methylator phenotype in colon cancer. Gastroenterology. 134:1950–1960, 1960.e1. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Naguib A, Mitrou PN, Gay LJ, Cooke JC, Luben RN, Ball RY, McTaggart A, Arends MJ and Rodwell SA: Dietary, lifestyle and clinicopathological factors associated with BRAF and K-ras mutations arising in distinct subsets of colorectal cancers in the EPIC Norfolk study. BMC Cancer. 10:992010. View Article : Google Scholar : PubMed/NCBI

16 

Ooki A, Akagi K, Yatsuoka T, Asayama M, Hara H, Takahashi A, Kakuta M, Nishimura Y and Yamaguchi K: Combined microsatellite instability and BRAF gene status as biomarkers for adjuvant chemotherapy in stage III colorectal cancer. J Surg Oncol. 110:982–988. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Oliveira C, Velho S, Moutinho C, Ferreira A, Preto A, Domingo E, Capelinha AF, Duval A, Hamelin R, Machado JC, et al: KRAS and BRAF oncogenic mutations in MSS colorectal carcinoma progression. Oncogene. 26:158–163. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Demes M, Scheil-Bertram S, Bartsch H and Fisseler-Eckhoff A: Signature of microsatellite instability, KRAS and BRAF gene mutations in German patients with locally advanced rectal adenocarcinoma before and after neoadjuvant 5-FU radiochemotherapy. J Gastrointest Oncol. 4:182–192. 2013.PubMed/NCBI

19 

Wong A and Ma BB: Personalizing therapy for colorectal cancer. Clin Gastroenterol Hepatol. 12:139–144. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Cohen R, Svrcek M, Dreyer C, Cervera P, Duval A, Pocard M, Fléjou JF, de Gramont A and André T: New therapeutic opportunities based on DNA mismatch repair and BRAF status in metastatic colorectal cancer. Curr Oncol Rep. 18:182016. View Article : Google Scholar : PubMed/NCBI

21 

Carethers JM, Chauhan DP, Fink D, Nebel S, Bresalier RS, Howell SB and Boland CR: Mismatch repair proficiency and in vitro response to 5-fluorouracil. Gastroenterology. 117:123–131. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Fischer H, Stenling R, Rubio C and Lindblom A: Differential expression of aquaporin 8 in human colonic epithelial cells and colorectal tumors. BMC Physiol. 1:12001. View Article : Google Scholar : PubMed/NCBI

23 

Fujita H, Kato J, Horii J, Harada K, Hiraoka S, Shiraha H, Sakaguchi K and Shiratori Y: Decreased expression of hMLH1 correlates with reduced 5-fluorouracil-mediated apoptosis in colon cancer cells. Oncol Rep. 18:1129–1137. 2007.PubMed/NCBI

24 

Boland CR and Goel A: Microsatellite instability in colorectal cancer. Gastroenterology. 138:2073–2087.e3. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Nöpel-Dünnebacke S, Schulmann K, Reinacher-Schick A, Porschen R, Schmiegel W, Tannapfel A and Graeven U: Prognostic value of microsatellite instability and p53 expression in metastatic colorectal cancer treated with oxaliplatin and fluoropyrimidine-based chemotherapy. Z Gastroenterol. 52:1394–1401. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Huang CJ, Liao HT, Yeh GC and Hung KL: Distribution of HLA-DQB1 alleles in patients with Kleine-Levin syndrome. J Clin Neurosci. 19:628–630. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Loukola A, Eklin K, Laiho P, Salovaara R, Kristo P, Järvinen H, Mecklin JP, Launonen V and Aaltonen LA: Microsatellite marker analysis in screening for hereditary nonpolyposis colorectal cancer (HNPCC). Cancer Res. 61:4545–4549. 2001.PubMed/NCBI

28 

Jensen SA, Vainer B, Kruhøffer M and Sørensen JB: Microsatellite instability in colorectal cancer and association with thymidylate synthase and dihydropyrimidine dehydrogenase expression. BMC Cancer. 9:252009. View Article : Google Scholar : PubMed/NCBI

29 

Schneider M, Scholtka B, Gottschalk U, Faiss S, Schatz D, Berghof-Jäger K and Steinberg P: Detection of up to 65% of precancerous lesions of the human colon and rectum by mutation analysis of APC, K-Ras, B-Raf and CTNNB1. Cancers (Basel). 3:91–105. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Chien CC, Chen SH, Liu CC, Lee CL, Yang RN, Yang SH and Huang CJ: Correlation of K-ras codon 12 mutations in human feces and ages of patients with colorectal cancer (CRC). Transl Res. 149:96–102. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Shahrzad S, Quayle L, Stone C, Plumb C, Shirasawa S, Rak JW and Coomber BL: Ischemia-induced K-ras mutations in human colorectal cancer cells: Role of microenvironmental regulation of MSH2 expression. Cancer Res. 65:8134–8141. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Ewalt M, Nandula S, Phillips A, Alobeid B, Murty VV, Mansukhani MM and Bhagat G: Real-time PCR-based analysis of BRAF V600E mutation in low and intermediate grade lymphomas confirms frequent occurrence in hairy cell leukaemia. Hematol Oncol. 30:190–193. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Deng G, Bell I, Crawley S, Gum J, Terdiman JP, Allen BA, Truta B, Sleisenger MH and Kim YS: BRAF mutation is frequently present in sporadic colorectal cancer with methylated hMLH1, but not in hereditary nonpolyposis colorectal cancer. Clin Cancer Res. 10:191–195. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Banno K, Yanokura M, Kobayashi Y, Kawaguchi M, Nomura H, Hirasawa A, Susumu N and Aoki D: Endometrial cancer as a familial tumor: Pathology and molecular carcinogenesis (review). Curr Genomics. 10:127–132. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Zlobec I, Bihl MP, Schwarb H, Terracciano L and Lugli A: Clinicopathological and protein characterization of BRAF- and K-RAS-mutated colorectal cancer and implications for prognosis. Int J Cancer. 127:367–380. 2010.PubMed/NCBI

36 

Dunn EF, Iida M, Myers RA, Campbell DA, Hintz KA, Armstrong EA, Li C and Wheeler DL: Dasatinib sensitizes KRAS mutant colorectal tumors to cetuximab. Oncogene. 30:561–574. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Hong SP, Min BS, Kim TI, Cheon JH, Kim NK, Kim H and Kim WH: The differential impact of microsatellite instability as a marker of prognosis and tumour response between colon cancer and rectal cancer. Eur J Cancer. 48:1235–1243. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Phipps AI, Limburg PJ, Baron JA, Burnett-Hartman AN, Weisenberger DJ, Laird PW, Sinicrope FA, Rosty C, Buchanan DD, Potter JD and Newcomb PA: Association between molecular subtypes of colorectal cancer and patient survival. Gastroenterology. 148:77–87.e2. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Huang CJ, Yang SH, Huang SM, Lin CM, Chien CC, Chen YC, Lee CL, Wu HH and Chang CC: A predicted protein, KIAA0247, is a cell cycle modulator in colorectal cancer cells under 5-FU treatment. J Transl Med. 9:822011. View Article : Google Scholar : PubMed/NCBI

40 

Figueiredo JC, Lewinger JP, Song C, Campbell PT, Conti DV, Edlund CK, Duggan DJ, Rangrej J, Lemire M, Hudson T, et al: Genotype-environment interactions in microsatellite stable/microsatellite instability-low colorectal cancer: Results from a genome-wide association study. Cancer Epidemiol Biomarkers Prev. 20:758–766. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Markovic S, Antic J, Dragicevic N, Hamelin R and Krivokapic Z: High-frequency microsatellite instability and BRAF mutation (V600E) in unselected Serbian patients with colorectal cancer. J Mol Histol. 43:137–143. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Nash GM, Gimbel M, Cohen AM, Zeng ZS, Ndubuisi MI, Nathanson DR, Ott J, Barany F and Paty PB: KRAS mutation and microsatellite instability: Two genetic markers of early tumor development that influence the prognosis of colorectal cancer. Ann Surg Oncol. 17:416–424. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Qiu J, Compagnone M, Laibe S, Lagarde A, Goncalves A, Turrini O, Xerri L, Monges G and Olschwang S: BRAF p.Val600Glu (V600E) somatic mutation is mainly associated with MSS phenotype in metastatic colorectal cancer. Cancer Genomics Proteomics. 8:15–18. 2011.PubMed/NCBI

44 

Birgisson H, Edlund K, Wallin U, Påhlman L, Kultima HG, Mayrhofer M, Micke P, Isaksson A, Botling J, Glimelius B and Sundström M: Microsatellite instability and mutations in BRAF and KRAS are significant predictors of disseminated disease in colon cancer. BMC Cancer. 15:1252015. View Article : Google Scholar : PubMed/NCBI

45 

Li WQ, Kawakami K, Ruszkiewicz A, Bennett G, Moore J and Iacopetta B: BRAF mutations are associated with distinctive clinical, pathological and molecular features of colorectal cancer independently of microsatellite instability status. Mol Cancer. 5:22006. View Article : Google Scholar : PubMed/NCBI

46 

Hutchins G, Southward K, Handley K, Magill L, Beaumont C, Stahlschmidt J, Richman S, Chambers P, Seymour M, Kerr D, et al: Value of mismatch repair, KRAS, and BRAF mutations in predicting recurrence and benefits from chemotherapy in colorectal cancer. J Clin Oncol. 29:1261–1270. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S and Bardelli A: Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol. 26:5705–5712. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Lech G, Slotwinski R and Krasnodebski IW: The role of tumor markers and biomarkers in colorectal cancer. Neoplasma. 61:1–8. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Janakiraman M, Vakiani E, Zeng Z, Pratilas CA, Taylor BS, Chitale D, Halilovic E, Wilson M, Huberman K, Filho JC Ricarte, et al: Genomic and biological characterization of exon 4 KRAS mutations in human cancer. Cancer Res. 70:5901–5911. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Chun P and Wainberg ZA: Adjuvant chemotherapy for Stage II colon cancer: The role of molecular markers in choosing therapy. Gastrointest Cancer Res. 3:191–196. 2009.PubMed/NCBI

51 

Smith G, Bounds R, Wolf H, Steele RJ, Carey FA and Wolf CR: Activating K-Ras mutations outwith ‘hotspot’ codons in sporadic colorectal tumours-implications for personalised cancer medicine. Br J Cancer. 102:693–703. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Center MM, Jemal A, Smith RA and Ward E: Worldwide variations in colorectal cancer. CA Cancer J Clin. 59:366–378. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Collura A, Lagrange A, Svrcek M, Marisa L, Buhard O, Guilloux A, Wanherdrick K, Dorard C, Taieb A, Saget A, et al: Patients with colorectal tumors with microsatellite instability and large deletions in HSP110 T17 have improved response to 5-fluorouracil-based chemotherapy. Gastroenterology. 146:401–411.e1. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Matsuyama T, Ishikawa T, Mogushi K, Yoshida T, Iida S, Uetake H, Mizushima H, Tanaka H and Sugihara K: MUC12 mRNA expression is an independent marker of prognosis in stage II and stage III colorectal cancer. Int J Cancer. 127:2292–2299. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 12 Issue 6

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
Huang CJ, Huang SH, Chien CC, Lee HH, Yang SH, Chang CC and Lee CL: Impact of microsatellite status on chemotherapy for colorectal cancer patients with KRAS or BRAF mutation. Oncol Lett 12: 4427-4434, 2016
APA
Huang, C., Huang, S., Chien, C., Lee, H.H., Yang, S., Chang, C., & Lee, C. (2016). Impact of microsatellite status on chemotherapy for colorectal cancer patients with KRAS or BRAF mutation. Oncology Letters, 12, 4427-4434. https://doi.org/10.3892/ol.2016.5275
MLA
Huang, C., Huang, S., Chien, C., Lee, H. H., Yang, S., Chang, C., Lee, C."Impact of microsatellite status on chemotherapy for colorectal cancer patients with KRAS or BRAF mutation". Oncology Letters 12.6 (2016): 4427-4434.
Chicago
Huang, C., Huang, S., Chien, C., Lee, H. H., Yang, S., Chang, C., Lee, C."Impact of microsatellite status on chemotherapy for colorectal cancer patients with KRAS or BRAF mutation". Oncology Letters 12, no. 6 (2016): 4427-4434. https://doi.org/10.3892/ol.2016.5275