Open Access

High frequency of microsatellite instability and its substantial co‑existence with KRAS and BRAF mutations in Vietnamese patients with colorectal cancer

  • Authors:
    • Ha Thi Nguyen
    • Do Thanh Le
    • Quan Hong Duong
    • Vinay Bharadwaj Tatipamula
    • Bang Van Nguyen
  • View Affiliations

  • Published online on: November 13, 2020     https://doi.org/10.3892/ol.2020.12302
  • Article Number: 41
  • Copyright: © Nguyen 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

Tumor heterogeneity and resistance to chemotherapy have been recognized as two major obstacles in the diagnosis and treatment of colorectal cancer (CRC). Microsatellite instability (MSI) and KRAS and BRAF mutations are common diagnostic factors that have been widely used to classify CRC for therapeutics. In the present study, 151 patients with CRC were analyzed from the two most populous ethnic groups of Vietnam, Kinh and Muong, for their MSI status, frequency of KRAS and BRAF mutations, and their clinical implications. MSI‑high (MSI‑H) was detected in 45.0% (68/151), while mutated KRAS and BRAF were identified in 37.1% (56/151) and 2.6% (4/151) of the cases, respectively. There was a substantial co‑existence of MSI‑H with KRAS (27/56; 48.2%) and BRAF (3/4; 75.0%) mutations. Statistical analysis showed that MSI‑H tumors were significantly associated with colon location (P=0.011) and more advanced T stages (P=0.016). KRAS exon 2 mutations were significantly more likely to be detected in patients who belonged to the Muong ethnic group (P=0.013) or those with no/fewer lymph node metastasis (P=0.048) as compared with their counterparts. In summary, the data revealed typical molecular features of Vietnamese patients with CRC, including a strikingly high rate of MSI‑H and its high co‑existence with KRAS and BRAF mutations, which should be carefully considered in the future therapeutics for this type of cancer.

Introduction

Colorectal cancer (CRC) is currently the second-most lethal cancer globally, with an estimated 880,792 deaths occurring in 2018, accounting for 9.2% of total cancer deaths worldwide (1). In Vietnam, CRC is the fifth most common cancer and the fourth leading cause of cancer death in both sexes combined (2). Generally, colorectal tumorigenesis undergoes a stepwise process of mutations and clonal expansion, subsequently leading to invasive and metastatic tumors (35). There are two major molecular pathways contributing to CRC tumorigenesis, namely chromosomal instability (CIN) and microsatellite instability (MSI) (6).

CIN appears to be the most common type of genetic change in CRC, accounting for 85% of sporadic cases (5). CIN is often associated with copy number variations (CNVs) and/or the mutations in cancer-associated genes, including KRAS and BRAF (7,8). MSI, on the other hand, accounts for 15% of all CRC (9), as a consequence of a post-replicative DNA mismatch repair (MMR) deficiency mostly caused by germline mutations (10) or epigenetic silencing of MMR genes (11). MSI high (MSI-H) CRC tends to be associated with a high frequency of replication errors due to the slippage of DNA polymerase (12). MSI-low (MSI-L) tumors, however, seem to occur through the CIN pathway, similar to microsatellite stable (MSS) tumors (13). MSI-H CRC has largely been recognized as a favorable prognostic factor (1417).

One of the key early events during tumor progression is the acquisition of mutations in KRAS oncogene, leading to the activation of multiple epidermal growth factor receptor (EGFR) signaling pathways, including the RAS-RAF-ERK-MAPK and the PI3K-PTEN-AKT pathways (18). Activation mutations of the KRAS gene have been detected in 30–50% of CRC cases, mostly affecting codons 12 and 13 (19), resulting in a substitution of glycine with cysteine (p.G12C), valine (p.G12V), or aspartic acid (p.G12D, p.G13D) (1921). These mutations diminish the intrinsic GTPase activity of KRAS and confer unresponsiveness to GTPase activating proteins (GAPs), causing the accumulation of KRAS-GTP and constitutively activating its downstream pathways, including cell proliferation and survival (5,22). BRAF is a serine/threonine protein kinase, which is associated with the MEK/ERK signaling pathway (23). Oncogenic mutations of BRAF have been detected in ~5–15% of CRCs (2325), mostly involving a substitution of glutamic acid for valine at codon 600 (p.V600E) (26). Molecularly, BRAF mutations trigger its kinase activity that activates ERK through its effectors MEK1/2, which eventually induces cell proliferation (27). Mutations in KRAS and BRAF have been largely regarded as negative predictors for anti-EGFR therapy among patients with CRC (2833). Detection of KRAS and BRAF mutations, therefore, has been performed in a routine CRC diagnosis as an aid for prognosis and therapeutic options (3436).

Despite its significance, the MSI status and the prevalence of KRAS and BRAF mutations and their associations with the other tumor characteristics remain poorly known for Vietnamese patients with CRC. The present study aimed to determine the MSI status, mutation frequencies of KRAS exon 2 and BRAF exon 15, as well as the associations between MSI status and other clinicopathological features in 151 Vietnamese patients with CRC from two populous ethnic groups, Kinh and Muong.

Materials and methods

Patients and clinicopathological characteristics

Fifty pairs of primary CRC and adjacent normal tissues (at least 3 cm from the tumor) were collected from an unselected consecutive series of patients of the Kinh ethnic group undergoing surgical resection of CRC at Hue Central Hospital (Hue, Vietnam) between February 2017 and May 2018. These samples were stored at −80°C after snap freezing in liquid nitrogen. The primary formalin-fixed, paraffin-embedded (FFPE) colorectal samples (n=101) of Kinh (n=37) and Muong (n=64) ethnic groups, who were surgically treated and histologically diagnosed with CRC between May 2011 and December 2016, were obtained from the pathology archives of the Hoa Binh Hospital (Hoa Binh, Vietnam). None of the patients underwent any other treatments before the surgery. Routine histopathologic staging of the resected specimens was done by an experienced pathologist, and all the cancer specimens contained at least 70% carcinoma cells.

The clinicopathological characteristics of the cohort, including the patient's sex, ethnicity, age at diagnosis, Tumor-Node-Metastasis (TNM) stage, tumor site and tumor grade (level of differentiation), are shown in Table I. The study was approved by the Ethics Committee of Duy Tan University. Written informed consent for the use of resected tissue and clinical data in research was obtained from all patients.

Table I.

Clinicopathological characteristics of patients with colorectal cancer according to microsatellite instability status.

Table I.

Clinicopathological characteristics of patients with colorectal cancer according to microsatellite instability status.

MSI sub-groupMSI-H vs. MSI-L/MSS


CharacteristicsCases, n=151MSI-HMSI-LMSSP-valueMSI-L/MSSP-value
Age, years 0.122a 0.066a
  Mean ± SD59.94±12.3658.35±12.1562.29±13.7960.51±11.51 61.24±12.44
  (range)(23–90)(27–87)(32–90)(23–84) (23–90)
  ≤50  32 (21.2)19 (27.9)  7 (20.6)  6 (12.2) 13 (15.7)
  >50119 (78.8)49 (72.1)27 (79.4)43 (87.8) 70 (84.3)
Sex, n (%) 0.008a 0.170a
  Male  84 (55.6)42 (61.8)11 (32.4)31 (66.3) 42 (50.6)
  Female  67 (44.4)26 (38.2)23 (67.6)18 (36.7) 41 (49.4)
Ethnicity, n (%) 0.957a 0.786a
  Kinh  87 (57.6)40 (58.8)19 (55.9)28 (57.1) 47 (56.6)
  Muong  64 (42.4)28 (41.2)15 (44.1)21 (42.9) 36 (43.4)
Location, n (%) 0.040a 0.011a
  Colon104 (68.9)54 (79.4)20 (58.8)30 (61.2) 50 (60.2)
  Rectum  47 (31.1)14 (20.6)14 (41.2)19 (38.8) 33 (39.8)
TNM stage, n (%) 0.536c 0.772c
  I  7 (4.6)1 (1.5)1 (2.9)  5 (10.2) 6 (7.2)
  II108 (71.5)51 (75.0)25 (73.5)32 (65.3) 57 (68.7)
  III  28 (18.5)12 (17.6)  7 (20.6)  9 (18.4) 16 (19.3)
  IV  1 (0.7)1 (2.9) 1 (1.2)
  Missing  7 (4.6)4 (5.9)3 (6.1) 3 (3.6)
T stage, n (%) 0.056c 0.016c
  T210 (6.6)2 (2.9)2 (5.9)  6 (12.2) 8 (9.6)
  T3  84 (55.6)34 (50.0)23 (67.6)27 (55.1) 50 (60.2)
  T4  57 (37.7)32 (47.1)  9 (26.5)16 (32.7) 25 (30.1)
Lymph node invasion, n (%) 0.962c 0.797c
  N0115 (76.2)52 (76.5)26 (76.5)37 (75.5) 63 (75.9)
  N1  28 (18.5)13 (19.1)  7 (20.6)  8 (16.3) 15 (18.1)
  N2  2 (1.3)2 (4.1) 2 (2.4)
  Missing  6 (4.0)3 (4.4)1 (2.9)2 (4.1) 3 (3.6)
Distant metastasis, n (%) 0.193c 0.364c
  M0145 (96.0)66 (96.0)33 (97.1)46 (93.9) 79 (95.2)
  M1  1 (0.7)1 (2.9) 1 (1.2)
  Missing  5 (3.3)2 (2.9)3 (6.1) 3 (3.6)
Differentiation, n (%) 0.076c 0.103c
  Well  25 (16.6)6 (8.8)  5 (14.7)14 (28.6) 19 (22.9)
  Moderately  17 (11.3)  9 (13.2)  4 (11.8)4 (8.2) 8 (9.6)
  Poorly  5 (3.3)2 (2.9)2 (5.9)1 (2.0) 3 (3.6)
  Missing104 (68.9)51 (75.0)23 (67.6)30 (61.2) 53 (63.9)
BRAF, n (%) 0.350b 0.327b
  Mutant  4 (2.6)3 (4.4)1 (2.9) 1 (1.2)
  Wild-type   147(97.4)65 (95.6)33 (97.1)  49 (100.0) 82 (98.8)
KRAS, n (%) 0.516a 0.546a
  Mutant  56 (37.1)27 (39.7)14 (41.2)15 (30.6) 29 (34.9)
  Wild-type  95 (62.9)41 (60.3)20 (58.8)34 (69.4) 54 (65.1)

{ label (or @symbol) needed for fn[@id='tfn1-ol-0-0-12302'] } CRC, colorectal cancer; SD, standard deviation; MSI-H, high-frequency microsatellite instability; MSI-L, low-frequency microsatellite instability; MSS, microsatellite stable.

a χ2 test

b Fisher's exact test

c Kruskal-Wallis test.

DNA extraction

For the FFPE CRC specimens (n=101), five to eight sections with a thickness of ~10 µm per sample were cut using a rotary microtome (HM 325 Rotary Microtome; Thermo Fisher Scientific, Inc.) and immediately collected into a sterile 2 ml microcentrifuge tube. After xylene/ethanol deparaffinization, samples were lysed with ~500 µl of lysis buffer (10 mM NaCl, 20 mM Tris-HCl (pH 8.0), 1 mM EDTA (pH 8.0), 0.5% SDS and 200 µg/ml proteinase K) at 56°C for ~3 h. DNA was then isolated using a mixture of phenol-chloroform, followed by ethanol precipitation, as previously described (37).

For the fresh-frozen paired samples (n=50), a pea-size tissue sample was cut and collected into a sterile 2 ml microcentrifuge tube containing 500 µl of ice-cold lysis buffer and a 2.5-mm diameter iron ball. The sample was homogenized by a TissueLyser LT (Qiagen Inc.) according to the manufacturer's instructions, and DNA was extracted as aforementioned. DNA samples were then dissolved in Tris-EDTA buffer or ddH2O and kept at −20°C for later use. DNA purity and concentration were assessed by a NanoDrop 2000 Spectrophotometer (ND-2000; Thermo Fisher Scientific, Inc.).

MSI analysis
Multiplex PCR amplification of short tandem repeat (STR) loci

Typing of MSI was performed using the standard Bethesda marker panel (38) and CAT25 (39), as previously described (40) (Table SI). Multiplex PCR reaction was done in a total volume of 25 µl using 12.5 µl Phusion U Multiplex PCR Master mix (Thermo Fisher Scientific, Inc.), 0.4 mM of each primer pairs (Integrated DNA Technologies, Inc.), and 50 ng of genomic DNA (gDNA). For set 1 (BAT25, BAT26 and CAT25), the cycling parameters were as follows: 30 sec at 98°C; 30 cycles for fresh-frozen and 32 cycles for FFPE samples of 10 sec at 98°C, 30 sec at 57°C, 30 sec at 72°C, and a final extension step at 72°C for 7 min. For set 2 (D2S123, D5S346 and D17S250), the annealing temperature was adjusted to 52°C.

Fragment analysis

The PCR products were subjected to the ABI 3500 Genetic Analyzer (Thermo Fisher Scientific, Inc.) for fragment analysis. The sizes and patterns of the PCR products were then resolved using GeneMapper 5.0 software (Thermo Fisher Scientific, Inc.). The occurrence of MSI in a CRC sample was specified by the presence of novel alleles as compared with the corresponding normal sample and/or the appearance of allelic imbalance at heterozygous loci. Tumors were considered as MSI-H if instability was detected in at least two out of six tested markers; as MSI-L if instability was observed in only one of the markers; and as MSS if no instabilities were detected (38,39).

Determination of allelic imbalance

For dinucleotide makers, allelic ratios (R) were calculated based on the fluorescent intensity peak height, as previously reported (41,42). In normal tissues, the peak height ratio of <70% could be considered as evidence of allelic imbalance (AI) or partial loss of heterozygosity (pLOH) (43). In practice, the AI thresholds were generally set from 70% to as low as 59% (42,44). In the present study, the AI thresholds were set at <59% or >125%. For simplicity, the smaller allele (in size) was always made the numerator, and the site with the allelic ratio of >1.7 or <0.8 was recognized as a site of AI or pLOH and will be reckoned as MSI (Fig. S1).

KRAS and BRAF mutation detection
PCR amplification

The amplification mixture consisted of 12.5 µl 2X DreamTaq PCR Master mix (Thermo Fisher Scientific, Inc.), 0.4 mM of each primer pairs (Table SII), and 50–100 ng gDNA. For KRAS, the PCR conditions were 3 min at 95°C, followed by 32 (for fresh-frozen samples) to 35 cycles (for FFPE samples) of 30 sec at 95, 50 and 72°C, sequentially. For BRAF, the annealing temperature was adjusted to 48°C. To avoid a cross-sample contamination, at least one negative control was included for each round of PCR amplification. PCR products were examined by 2% agarose gel electrophoresis to confirm the existence of amplified fragments before sequencing.

Sanger sequencing

All the successfully amplified fragments were subjected to unidirectional sequence analysis based on the Sanger method on an ABI 3500 Genetic Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.) after being purified using the GeneJet PCR Purification kit (Thermo Fisher Scientific, Inc.).

Data analysis

Tumors were classified by MSI or KRAS mutation status. The clinicopathological features were assessed and compared between groups. Anatomic location was distinguished as the colon (from cecum to sigmoid colon) or rectum (rectum and anus). Tumor grade was categorized as: Well-differentiated (G1), moderately differentiated (G2), poorly differentiated (G3), and undifferentiated (G4) tumors. The node metastasis (N) in the TNM staging system was assigned by the number of metastatic lymph nodes as N1 (1–3 affected nodes) and N2 (≥4 affected nodes).

Statistical analysis

SPSS 22.0 (SPSS, Inc.) was used for all statistical analyses. Continuous variables were presented as the mean±standard deviation. The associations between MSI, KRAS and BRAF mutational status and the clinicopathological factors were determined by the χ2, Fisher's exact, or Kruskal-Wallis test. P<0.05 was considered to indicate a statistically significant difference.

Results

Clinicopathological features of patients

Of the 151 patients, 84 (55.6%) were male, and 67 (44.4%) were female; 87 (57.6%) were Kinh, and 64 (42.4%) were Muong ethnicity. There were 104 (68.9%) tumors at the colon and 47 (31.1%) tumors at the rectum. The pathological stage was defined according to the TNM stage classification; 7 patients (4.6%) had stage I, 108 patients (71.5%) had stage II, 28 patients (18.5%) had stage III, 1 patient (0.7%) had stage IV, and 7 patients (4.6%) had an undetermined stage tumor. Tumor grade was available for only 47 patients, of which 25 tumors (53.2%) were well-differentiated, 17 tumors (36.2%) were moderately differentiated, and 5 tumors (10.6%) were poorly differentiated. The patients' age ranged from 23 to 90 years (mean age, 59.94±12.36 years) (Table I).

Large proportion of Vietnamese patients with CRC are classed as MSI-H

All tumors were typed for MSI and classified as MSI-H, MSI-L or MSS based on their number of unstable markers (Fig. 1A-C). Of these 151 patients, there were 68 patients (45.0%) with MSI-H, 34 patients (22.5%) with MSI-L, and 49 patients (32.5%) with MSS (Fig. 1D). The mean age of patients at diagnosis in the MSI-H group (58.35±12.15 years) was lower compared with MSS (60.51±11.51) and MSI-L (62.29±13.79) groups. There was a significant association between MSI status and the patients' sex (P=0.008), and tumor location (P=0.040) (Table I).

MSI-H vs. MSI-L/MSS CRC showed significant differences in the tumor location and T stage. Accordingly, MSI-H tumors were often found to be located at the colon (54/68; 79.4%) compared with MSI-L/MSS groups (50/83; 60.2%) (P=0.011). Furthermore, MSI-H was positively associated with more advanced T stages (P=0.016), especially for T4 stage tumors, which has been detected in 32 out of 68 (47.1%) patients with MSI-H compared with 25 out of 83 (30.1%) patients with MSI-L/MSS (Table I). In parallel, pairwise comparisons between MSI groups also revealed the significant differences (P≤0.05) between MSI-H vs. MSI-L and MSI-H vs. MSS tumors in both tumor location and T stage (Table SIII). Additionally, the results revealed significant differences in sex between MSI-H vs. MSI-L (P=0.005) and MSI-L vs. MSS (P=0.006) tumors (Table SIII). Specifically, male were more often in the MSI-H (42/68, 61.8%) and MSS (31/49, 66.3%) groups compared with the MSI-L tumors (11/34; 32.4%) (Table I). Moreover, the statistical analyses have also uncovered associations between MSI status and the tumor grade and age at onset (Table SIII). Particularly, MSI-H (P=0.027) tumors appeared to be significantly less differentiated and younger onset (P=0.041) compared with MSS tumors (Table SIII).

Taken together, the data revealed that a substantial proportion of Vietnamese patients with CRC were MSI-H. MSI-H tumors exhibited with more advanced T stage tumors and colon-located tumors. Furthermore, MSI-H tumors occurred more in male compared with MSI-L tumors, and less differentiated and younger age at onset compared with MSS tumors. These distinct clinicopathological features of MSI-H tumors specified that the carcinogenic pathway underlying MSI-H tumors might be different from that of MSI-L/MSS tumors.

Diagnostic role of each individual marker

The diagnostic role of microsatellite markers was assessed by analyzing the frequency of instability of these markers on three groups: MSI-H, MSI-L and MSI-H cases with a minimum number (2 out of 6) of unstable markers (MSI-H min, Table SIV). Noticeably, the frequency of instability was similar between mononucleotide and dinucleotide markers within MSI-H group. Of which, D2S123 and BAT26 were the most unstable markers that showed evidence of instability in 73.5 and 70.6% of the MSI-H cases, respectively (Fig. 2A). However, for MSI-L and especially for MSI-H min group, the dinucleotide markers appeared to be much more sensitive in the detection of instability compared with mononucleotide markers. Particularly, while BAT25 and CAT25 did not show any evidence of instability in MSI-L and MSI-H min CRC, the frequency of instability detected in BAT26, D2S123, D5S346 and D17S250 in MSI-L and MSI-H min were 8.8, 32.4, 35.3, 23.5%, and 14.3, 78.6, 50, 57.1%, respectively (Fig. 2B).

KRAS and BRAF mutations and their co-existence with MSI-H

Electropherograms of KRAS and BRAF gene sequences carrying hotspot substitution mutations were illustrated in Fig. S2A and B.

Sequence analysis of KRAS exon 2 showed single point mutations in 56 of 151 tested samples (37.1%) (Fig. 3A), of which 31 tumors (55.4%) had mutations in codon 12 and 25 tumors (44.6%) had mutations in codon 13 (Fig. 3B). The most frequent alteration in codon 12 was c.35G>A (21/31; 67.7%) transition, causing a replacement of glycine with aspartic acid (p.G12D). Two other mutations at codon 12 were c.35G>T (6/31; 19.4%) and c.34G>T (4/31; 12.9%) transversion, leading to the substitution of valine (p.G12V) or cysteine (p.G12C) for glycine, respectively (Fig. 3B). Mutations in codon 13 were exclusively c.38G>A (25/25) transition, resulting in an exchange aspartic acid for glycine (p.G12D) (Fig. 3B).

Four of 151 tumors (2.6%) comprised a point mutation in BRAF exon 15 (Fig. 3A). Three out of four mutations (75.0%) were c.1799T>A transversion replacing glutamic acid for valine in codon 600 (p.V600E). The remained mutation (25.0%) was c.1801A>G transition that switches a lysine to glutamic acid (p.K601E) (Fig. 3C). Noticeably, there was a substantial co-existence of mutant KRAS and MSI-H (27/151, 17.9%) compared with MSI-L (14/151; 9.3%) and MSS (15/151; 9.9%) (Fig. 4A). Within MSI-H group, mutant KRAS and mutant BRAF accounted for 39.7 and 4.4% (Fig. 4B); while within mutated KRAS and mutated BRAF groups, MSI-H accounted for 48.2% (27/56) and 75% (3/4), respectively (Table II).

Table II.

Association of KRAS and BRAF mutational status with clinicopathological features in colorectal cancer.

Table II.

Association of KRAS and BRAF mutational status with clinicopathological features in colorectal cancer.

BRAFKRAS


Clinicopathological features BRAFmt n (%) BRAFwt n (%)P-value KRASmt n (%) KRASwt n (%)P-value
Age, years 1.000b 0.379a
  ≤50  1 (250.0)  31 (21.1) 14 (25.0)18 (18.9)
  >503 (75.0)116 (78.9) 42 (75.0)77 (81.1)
Sex 1.000b 0.465a
  Male2 (50.0)  82 (55.8) 29 (51.8)55 (57.9)
  Female2 (50.0)  65 (44.2) 27 (48.2)40 (42.1)
Ethnicity 0.638b 0.013a
  Kinh3 (75.0)  84 (57.1) 25 (44.6)62 (65.3)
  Muong1 (25.0)  63 (42.9) 31 (55.4)33 (34.7)
Location 1.000b 0.194a
  Colon3 (75.0)101 (68.7) 35 (62.5)69 (72.6)
  Rectum1 (25.0)  46 (31.3) 21 (37.5)26 (27.4)
TNM stage 0.699c 0.086c
  I  7 (4.8) 4 (7.1)3 (3.2)
  II3 (75.0)105 (71.4) 41 (73.2)67 (70.5)
  III1 (25.0)  27 (18.4)   6 (10.7)22 (23.2)
  IV  1 (0.7) 1 (1.8)
  Missing  7 (4.8) 4 (7.1)3 (3.2)
T stage 0.216 >0.999c
  T210 (6.8) 5 (8.9)5 (5.3)
  T3  4 (100.0)  80 (54.4) 29 (51.8)55 (57.9)
  T4  57 (38.8) 22 (39.3)35 (36.8)
Lymph node metastasis 0.843c 0.048c
  N03 (75.0)112 (76.2) 45 (80.4)70 (73.7)
  N11 (25.0)  27 (18.4)   6 (10.7)22 (23.2)
  N2  2 (1.4) 2 (2.1)
  Missing  6 (4.1) 5 (8.9)1 (1.1)
Tumor grade (differentiation) 0.046c 0.114c
  Well  25 (17.0) 44 (78.6)21 (22.1)
  Moderately2 (50.0)  15 (10.2) 4 (7.1)11 (11.6)
  Poorly1 (25.0)  4 (2.7)   6 (10.7)3 (3.2)
  Missing1 (25.0)103 (70.1) 2 (3.6)60 (63.2)
BRAF 0.297b
  Mutant 4 (4.2)
  Wild-type 56 (100)91 (95.8)
MSI 0.151c 0.356c
  High3 (75.0)  65 (44.2) 27 (48.2)41 (43.2)
  Low1 (25.0)  33 (22.4) 14 (25.0)20 (21.1)
  Stable  49 (33.3) 15 (26.8)34 (35.8)

{ label (or @symbol) needed for fn[@id='tfn5-ol-0-0-12302'] } CRC, colorectal cancer; SD, standard deviation; MSI-H, high-frequency microsatellite instability; MSI-L, low-frequency microsatellite; mt, mutant; wt, wild-type.

a χ2 test

b Fisher's exact test

c Kruskal-Wallis test.

The statistical tests disclosed significant differences in the ethnicity and lymph node metastasis rate between KRAS mutant and wild-type tumors. Accordingly, there were significantly more Muong (55.4%) than Kinh patients (44.6%) within the KRAS mutated tumors (P=0.013). Moreover, KRAS mutations were significantly associated with none (N0) or fewer number lymph node metastasis (N1) compared with KRAS wild-type (P=0.048). Lastly, the Kruskal-Wallis test has unveiled that BRAF mutant tumors were significantly less differentiated (P=0.046) compared with BRAF wild-type tumors. KRAS and BRAF mutations were mutually exclusive (Table II).

Discussion

Currently, it is well accepted that certain alterations at the molecular level favor CRC tumorigenesis and are used as prognostic markers. MSI phenotype, for example, was commonly proposed as a favorable prognostic biomarker for CRC (14,15,4547) and mutations in KRAS and BRAF genes were decisively used as predictors of resistance to monoclonal antibodies targeting epidermal growth factor receptors (EGFRs) (4850). Therefore, defining the status of MSI and the hotspot mutations in KRAS and BRAF is of utmost importance to support the prognosis and selection of treatment methods.

Among 151 patients with CRC, 45.0% of patients were classed as MSI-H, 22.5% were classed as MSI-L, and 32.5% as MSS. The proportion of MSI-H in Vietnamese patients with CRC in the present study is similar to that of African American (45%; 10/22) (51), substantially higher than Singaporean (30%; 32/109) patients with CRC (52), and strikingly higher than that of Japanese (4.5–6%) (53,54), Korean (5.5–9%) (55), and Western (10–20%) patients with CRC (56,57). These differences could be due to the genetic basis of the patient cohorts, microsatellite markers used for MSI detection, and/or the interpretation of the results (15,58). Several previous studies have suggested that BAT26 was a fairly good indicator of what would have been seen with the entire panel (16,59). However, in the present study, all six tested markers showed similar sensitivity in detecting MSI-H tumors, with ~70% of the cases, in which BAT26 and D2S123 were the most unstable markers (Fig. 2A). Particularly, in MSI-L and MSI-H min tumors, dinucleotides markers were found to be more sensitive than mononucleotide markers in detecting MSI (Fig. 2B). This finding was in concordance with one previous study in other Asian patients with CRC (52), indicating the significance of these dinucleotide markers in assessing MSI in patients with CRC. On the other hand, CAT25, the additional marker suggested by Findeisen et al (39), did not seem to have added any value to the MSI diagnostics of this set of tumors (Fig. 2B).

Moreover, the current study showed significant associations between MSI-H and colon tumor location, males, younger onset, and more advanced T stages. The association between MSI-H and proximal tumor sites was noted by a number of previous studies (55,60), but contradictory findings that highlighted the association of MSI-H to female (51,61,62) and lower T stages (55,63) were also reported. These results specified that MSI-H tumors possessed distinct clinicopathological features and possibly have been derived from a distinct carcinogenic pathway compared with MSI-L/MSS ones.

The sequencing data disclosed 37.1% patients with CRC carried a KRAS exon 2 mutation, which typically affects codon 12 (55.4%) and codon 13 (44.6%). This observation was in accordance with previous studies (6466), and marginally lower than several other studies (50,53,6769), where KRAS mutations were detected in ~35-37 and 40–42% of the tested patients with CRC, respectively. The present study also presented a significant difference in the frequency of this mutation in two ethnic groups by showing that 48.4% of Muong and 28.7% of Kinh patients with CRC had this mutation, suggesting a possible variation in the frequency and types of KRAS mutations among populations. Moreover, the present data also indicated that mutant KRAS was reversely associated with the rate of lymph node metastasis, which was in accordance with a study in the Chinese population (69). Mutations in KRAS exon 2 have been largely used as a predictive marker of unresponsiveness to EGFR therapies (4850). However, more than 50% of CRC patients with a wild-type KRAS exon 2 are also resistant to this approach, possibly due to other ‘rare mutations’ in other exons of this gene or in other genes of the RAS/RAF family (25,67,6972). These findings indicated the need for expanding the genetic test in CRC patients with a wild-type KRAS exon 2 before anti-EGFR therapy.

BRAF exon 15 mutations, on the other hand, were detected in only 2.6% Vietnamese patients with CRC. This ratio was similar to that of Greek patients with CRC (21), substantially lower than other Western countries (~9%) (25,50), and marginally lower than other Asian populations (~4.5–7%), including Japan (53,66) and China (69,73). In agreement with the previous studies, the present study also presented the mutual exclusivity of BRAF and KRAS mutations (23,66). Remarkably, the substantial rate of co-existence of MSI with KRAS and BRAF mutations found in the present study may indicate the poor prognosis of these cases (16,74). A future study on the associations between overall survival and/or recurrence-free survival and MSI with KRAS and BRAF mutations will gain some insight into this issue.

In summary, the present results revealed the typical molecular features and subgroups of Vietnamese patients with CRC. Particularly, the strikingly high proportion of MSI-H tumors and their substantial co-existence with KRAS and BRAF mutations should be taken into account for future diagnosis and clinical treatments for this type of cancer.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

The authors would like to thank Dr Adam F. Johnson (Institute of Research and Development, Duy Tan University, Danang, Vietnam) for the critical reading of the manuscript.

Funding

This study was supported by the National Foundation for Science and Technology Development (NAFOSTED; grant no. 106-YS.01-2015.12).

Availability of data and materials

Data and materials available on request from the authors

Authors' contributions

HTN conceived the project, conducted most of the experiments and data analysis, and wrote the manuscript. DTL designed/selected primers and prepared figures. QHD and VBT helped with sample preparation and DNA extraction. BVN collected samples and patients' data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Ethics Committee of Duy Tan University (approval no. DTU-IRB/2019.15). Written informed consent for the use of resected tissue and clinical data in research was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The other authors report no conflicts of interest.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Pham T, Bui L, Kim G, Hoang D, Tran T and Hoang M: Cancers in Vietnam-burden and control efforts: a narrative scoping review. Cancer Contr. 26:1073274819863802. 2019.

3 

Fearon ER and Vogelstein B: A genetic model for colorectal tumorigenesis. Cell. 61:759–767. 1990. View Article : Google Scholar : PubMed/NCBI

4 

Vogelstein B and Kinzler KW: Cancer genes and the pathways they control. Nat Med. 10:789–799. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Nguyen HT and Duong HQ: The molecular characteristics of colorectal cancer: Implications for diagnosis and therapy (Review). Oncol Lett. 16:9–18. 2018.PubMed/NCBI

6 

Brenner H, Kloor M and Pox CP: Colorectal cancer. Lancet. 383:1490–1502. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Grady WM and Carethers JM: Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology. 135:1079–1099. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Markowitz SD and Bertagnolli MM: Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med. 361:2449–2460. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Sinicrope FA and Sargent DJ: Molecular pathways: microsatellite instability in colorectal cancer: prognostic, predictive, and therapeutic implications. Clin Cancer Res. 18:1506–1512. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Worthley DL and Leggett BA: Colorectal cancer: Molecular features and clinical opportunities. Clin Biochem Rev. 31:31–38. 2010.PubMed/NCBI

11 

Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH, Widschwendter M, Weener D, Buchanan D, et al: CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 38:787–793. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Abdel-Rahman WM and Peltomäki P: Molecular basis and diagnostics of hereditary colorectal cancers. Ann Med. 36:379–388. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Pawlik TM, Raut CP and Rodriguez-Bigas MA: Colorectal carcinogenesis: MSI-H versus MSI-L. Dis Markers. 20:199–206. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Benatti P, Gafà R, Barana D, Marino M, Scarselli A, Pedroni M, Maestri I, Guerzoni L, Roncucci L, Menigatti M, et al: Microsatellite instability and colorectal cancer prognosis. Clin Cancer Res. 11:8332–8340. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Popat S, Hubner R and Houlston RS: Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol. 23:609–618. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Samowitz WS, Albertsen H, Herrick J, Levin TR, Sweeney C, Murtaugh MA, Wolff RK and Slattery ML: Evaluation of a large, population-based sample supports a CpG island methylator phenotype in colon cancer. Gastroenterology. 129:837–845. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Sinicrope FA and Sargent DJ: Clinical implications of microsatellite instability in sporadic colon cancers. Curr Opin Oncol. 21:369–373. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Wee P and Wang Z: Epidermal growth factor receptor cell proliferation signaling pathways. Cancers (Basel). 9:1–45. 2017.

19 

Fernández-Medarde A and Santos E: Ras in cancer and developmental diseases. Genes Cancer. 2:344–358. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Neumann J, Zeindl-Eberhart E, Kirchner T and Jung A: Frequency and type of KRAS mutations in routine diagnostic analysis of metastatic colorectal cancer. Pathol Res Pract. 205:858–862. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Kosmidou V, Oikonomou E, Vlassi M, Avlonitis S, Katseli A, Tsipras I, Mourtzoukou D, Kontogeorgos G, Zografos G and Pintzas A: Tumor heterogeneity revealed by KRAS, BRAF, and PIK3CA pyrosequencing: KRAS and PIK3CA intratumor mutation profile differences and their therapeutic implications. Hum Mutat. 35:329–340. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Schubbert S, Shannon K and Bollag G: Hyperactive Ras in developmental disorders and cancer. Nat Rev Cancer. 7:295–308. 2007. View Article : Google Scholar : PubMed/NCBI

23 

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

24 

Yuen ST, Davies H, Chan TL, Ho JW, Bignell GR, Cox C, Stephens P, Edkins S, Tsui WW, Chan AS, et al: Similarity of the phenotypic patterns associated with BRAF and KRAS mutations in colorectal neoplasia. Cancer Res. 62:6451–6455. 2002.PubMed/NCBI

25 

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 efficacy 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

26 

Calistri D, Rengucci C, Seymour I, Leonardi E, Truini M, Malacarne D, Castagnola P and Giaretti W: KRAS, p53 and BRAF gene mutations and aneuploidy in sporadic colorectal cancer progression. Cell Oncol. 28:161–166. 2006.PubMed/NCBI

27 

Cantwell-Dorris ER, O'Leary JJ and Sheils OM: BRAFV600E: Implications for carcinogenesis and molecular therapy. Mol Cancer Ther. 10:385–394. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, Juan T, Sikorski R, Suggs S, Radinsky R, et al: Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 26:1626–1634. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S, et al: 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

30 

Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, Simes RJ, Chalchal H, Shapiro JD, Robitaille S, et al: K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 359:1757–1765. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Bardelli A and Siena S: Molecular mechanisms of resistance to cetuximab and panitumumab in colorectal cancer. J Clin Oncol. 28:1254–1261. 2010. View Article : Google Scholar : PubMed/NCBI

32 

van Brummelen EMJ, de Boer A, Beijnen JH and Schellens JHM: BRAF mutations as predictive biomarker for response to anti-EGFR monoclonal antibodies. Oncologist. 22:864–872. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Zhao B, Wang L, Qiu H, Zhang M, Sun L, Peng P, Yu Q and Yuan X: Mechanisms of resistance to anti-EGFR therapy in colorectal cancer. Oncotarget. 8:3980–4000. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Borràs E, Jurado I, Hernan I, Gamundi MJ, Dias M, Martí I, Mañé B, Arcusa A, Agúndez JAG, Blanca M, et al: Clinical pharmacogenomic testing of KRAS, BRAF and EGFR mutations by high resolution melting analysis and ultra-deep pyrosequencing. BMC Cancer. 11:4062011. View Article : Google Scholar : PubMed/NCBI

35 

Mohamed Suhaimi NA, Foong YM, Lee DYS, Phyo WM, Cima I, Lee EXW, Goh WL, Lim WY, Chia KS, Kong SL, et al: Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients. Mol Oncol. 9:850–860. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Sclafani F, Chau I, Cunningham D, Hahne JC, Vlachogiannis G, Eltahir Z, Lampis A, Braconi C, Kalaitzaki E, De Castro DG, et al: KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Sci Rep. 8:14452018. View Article : Google Scholar : PubMed/NCBI

37 

Nguyen HT, Geens M, Mertzanidou A, Jacobs K, Heirman C, Breckpot K and Spits C: Gain of 20q11.21 in human embryonic stem cells improves cell survival by increased expression of Bcl-xL. Mol Hum Reprod. 20:168–177. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, Eshleman JR, Burt RW, Meltzer SJ, Rodriguez-Bigas MA, Fodde R, Ranzani GN and Srivastava S: A National Cancer Institute Workshop on Microsatellite Instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res. 58:5248–5257. 1998.PubMed/NCBI

39 

Findeisen P, Kloor M, Merx S, Sutter C, Woerner SM, Dostmann N, Benner A, Dondog B, Pawlita M, Dippold W, et al: T25 repeat in the 3′ untranslated region of the CASP2 gene: A sensitive and specific marker for microsatellite instability in colorectal cancer. Cancer Res. 65:8072–8078. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Nguyen HT, Markouli C, Geens M, Barbé L, Sermon K and Spits C: Human embryonic stem cells show low-grade microsatellite instability. Mol Hum Reprod. 20:981–989. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Skotheim RI, Diep CB, Kraggerud SM, Jakobsen KS and Lothe RA: Evaluation of loss of heterozygosity/allelic imbalance scoring in tumor DNA. Cancer Genet Cytogenet. 127:64–70. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Heaphy CM, Hines WC, Butler KS, Haaland CM, Heywood G, Fischer EG, Bisoffi M and Griffith JK: Assessment of the frequency of allelic imbalance in human tissue using a multiplex polymerase chain reaction system. J Mol Diagn. 9:266–271. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Gilder JR, Inman K, Shields W and Krane DE: Magnitude-dependent variation in peak height balance at heterozygous STR loci. Int J Legal Med. 125:87–94. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Leclair B, Frégeau CJ, Bowen KL and Fourney RM: Systematic analysis of stutter percentages and allele peak height and peak area ratios at heterozygous STR loci for forensic casework and database samples. J Forensic Sci. 49:968–980. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Sinicrope FA, Foster NR, Thibodeau SN, Marsoni S, Monges G, Labianca R, Kim GP, Yothers G, Allegra C, Moore MJ, et al: DNA mismatch repair status and colon cancer recurrence and survival in clinical trials of 5-fluorouracil-based adjuvant therapy. J Natl Cancer Inst. 103:863–875. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Roth AD, Delorenzi M, Tejpar S, Yan P, Klingbiel D, Fiocca R, d'Ario G, Cisar L, Labianca R, Cunningham D, et al: Integrated analysis of molecular and clinical prognostic factors in stage II/III colon cancer. J Natl Cancer Inst. 104:1635–1646. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Klingbiel D, Saridaki Z, Roth AD, Bosman FT, Delorenzi M and Tejpar S: Prognosis of stage II and III colon cancer treated with adjuvant 5-fluorouracil or FOLFIRI in relation to microsatellite status: Results of the PETACC-3 trial. Ann Oncol. 26:126–132. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Allegra CJ, Jessup JM, Somerfield MR, Hamilton SR, Hammond EH, Hayes DF, McAllister PK, Morton RF and Schilsky RL: American Society of Clinical Oncology provisional clinical opinion: Testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti-epidermal growth factor receptor monoclonal antibody therapy. J Clin Oncol. 27:2091–2096. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Van Cutsem E, Köhne CH, Láng I, Folprecht G, Nowacki MP, Cascinu S, Shchepotin I, Maurel J, Cunningham D, Tejpar S, et al: Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: Updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol. 29:2011–2019. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al: Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 369:1023–1034. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Ashktorab H, Smoot DT, Carethers JM, Rahmanian M, Kittles R, Vosganian G, Doura M, Nidhiry E, Naab T, Momen B, et al: High incidence of microsatellite instability in colorectal cancer from African Americans. Clin Cancer Res. 9:1112–1117. 2003.PubMed/NCBI

52 

Salto-Tellez M, Tan SY, Chiu LL and Koay ESC: Dinucleotide microsatellite repeats are essential for the diagnosis of microsatellite instability in colorectal cancer in Asian patients. World J Gastroenterol. 11:2781–2783. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Asaka S, Arai Y, Nishimura Y, Yamaguchi K, Ishikubo T, Yatsuoka T, Tanaka Y and Akagi K: Microsatellite instability-low colorectal cancer acquires a KRAS mutation during the progression from Dukes' A to Dukes' B. Carcinogenesis. 30:494–499. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Yamada K, Kanazawa S, Koike J, Sugiyama H, Xu C, Funahashi K, Boland CR, Koi M and Hemmi H: Microsatellite instability at tetranucleotide repeats in sporadic colorectal cancer in Japan. Oncol Rep. 23:551–561. 2010.PubMed/NCBI

55 

Kim CG, Ahn JB, Jung M, Beom SH, Kim C, Kim JH, Heo SJ, Park HS, Kim JH, Kim NK, et al: Effects of microsatellite instability on recurrence patterns and outcomes in colorectal cancers. Br J Cancer. 115:25–33. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Woerner SM, Benner A, Sutter C, Schiller M, Yuan YP, Keller G, Bork P, Doeberitz M and Gebert JF: Pathogenesis of DNA repair-deficient cancers: A statistical meta-analysis of putative Real Common Target genes. Oncogene. 22:2226–2235. 2003. View Article : Google Scholar : PubMed/NCBI

57 

Yamamoto H, Adachi Y, Taniguchi H, Kunimoto H, Nosho K, Suzuki H and Shinomura Y: Interrelationship between microsatellite instability and microRNA in gastrointestinal cancer. World J Gastroenterol. 18:2745–2755. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Poynter JN, Siegmund KD, Weisenberger DJ, Long TI, Thibodeau SN, Lindor N, Young J, Jenkins MA, Hopper JL, Baron JA, et al Colon Cancer Family Registry Investigators, : Molecular characterization of MSI-H colorectal cancer by MLHI promoter methylation, immunohistochemistry, and mismatch repair germline mutation screening. Cancer Epidemiol Biomarkers Prev. 17:3208–3215. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Kim GP, Colangelo LH, Wieand HS, Paik S, Kirsch IR, Wolmark N and Allegra CJ; National Cancer Institute, : Prognostic and predictive roles of high-degree microsatellite instability in colon cancer: a National Cancer Institute-National Surgical Adjuvant Breast and Bowel Project Collaborative Study. J Clin Oncol. 25:767–772. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Thibodeau SN, Bren G and Schaid D: Microsatellite instability in cancer of the proximal colon. Science. 260:816–819. 1993. View Article : Google Scholar : PubMed/NCBI

61 

Cho YK, Kim HC, Kim SH, Park JH, Yun HR, Cho YB, Yun SH, Lee WY and Chun HK: Location-related differences in sporadic microsatellite unstable colorectal cancer. Dig Liver Dis. 42:611–615. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Yang Y, Wang G, He J, Ren S, Wu F, Zhang J and Wang F: Gender differences in colorectal cancer survival: A meta-analysis. Int J Cancer. 141:1942–1949. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Gryfe R, Kim H, Hsieh ETK, Aronson MD, Holowaty EJ, Bull SB, Redston M and Gallinger S: Tumor microsatellite instability and clinical outcome in young patients with colorectal cancer. N Engl J Med. 342:69–77. 2000. View Article : Google Scholar : PubMed/NCBI

64 

Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pintér T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 360:1408–1417. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, Dietrich D, Biesmans B, Bodoky G, Barone C, et al: Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: Results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 28:466–474. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Yokota T: Are KRAS/BRAF mutations potent prognostic and/or predictive biomarkers in colorectal cancers? Anticancer Agents Med Chem. 12:163–171. 2012. View Article : Google Scholar : PubMed/NCBI

67 

Vaughn CP, Zobell SD, Furtado LV, Baker CL and Samowitz WS: Frequency of KRAS, BRAF, and NRAS mutations in colorectal cancer. Genes Chromosomes Cancer. 50:307–312. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Peeters M, Kafatos G, Taylor A, Gastanaga VM, Oliner KS, Hechmati G, Terwey JH and van Krieken JH: Prevalence of RAS mutations and individual variation patterns among patients with metastatic colorectal cancer: A pooled analysis of randomised controlled trials. Eur J Cancer. 51:1704–1713. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Guo F, Gong H, Zhao H, Chen J, Zhang Y, Zhang L, Shi X, Zhang A, Jin H, Zhang J, et al: Mutation status and prognostic values of KRAS, NRAS, BRAF and PIK3CA in 353 Chinese colorectal cancer patients. Sci Rep. 8:60762018. View Article : Google Scholar : PubMed/NCBI

70 

Loupakis F, Ruzzo A, Cremolini C, Vincenzi B, Salvatore L, Santini D, Masi G, Stasi I, Canestrari E, Rulli E, et al: KRAS codon 61, 146 and BRAF mutations predict resistance to cetuximab plus irinotecan in KRAS codon 12 and 13 wild-type metastatic colorectal cancer. Br J Cancer. 101:715–721. 2009. View Article : Google Scholar : PubMed/NCBI

71 

Peeters M, Douillard JY, Van Cutsem E, Siena S, Zhang K, Williams R and Wiezorek J: Mutant KRAS codon 12 and 13 alleles in patients with metastatic colorectal cancer: Assessment as prognostic and predictive biomarkers of response to panitumumab. J Clin Oncol. 31:759–765. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Hsu HC, Thiam TK, Lu YJ, Yeh CY, Tsai WS, You JF, Hung HY, Tsai CN, Hsu A, Chen HC, et al: Mutations of KRAS/NRAS/BRAF predict cetuximab resistance in metastatic colorectal cancer patients. Oncotarget. 7:22257–22270. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Li HT, Lu YY, An YX, Wang X and Zhao QC: KRAS, BRAF and PIK3CA mutations in human colorectal cancer: Relationship with metastatic colorectal cancer. Oncol Rep. 25:1691–1697. 2011.PubMed/NCBI

74 

Hu J, Yan WY, Xie L, Cheng L, Yang M, Li L, Shi J, Liu BR and Qian XP: Coexistence of MSI with KRAS mutation is associated with worse prognosis in colorectal cancer. Medicine (Baltimore). 95:e56492016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

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
Nguyen HT, Le DT, Duong QH, Tatipamula VB and Nguyen BV: High frequency of microsatellite instability and its substantial co‑existence with <em>KRAS</em> and <em>BRAF</em> mutations in Vietnamese patients with colorectal cancer. Oncol Lett 21: 41, 2021
APA
Nguyen, H.T., Le, D.T., Duong, Q.H., Tatipamula, V.B., & Nguyen, B.V. (2021). High frequency of microsatellite instability and its substantial co‑existence with <em>KRAS</em> and <em>BRAF</em> mutations in Vietnamese patients with colorectal cancer. Oncology Letters, 21, 41. https://doi.org/10.3892/ol.2020.12302
MLA
Nguyen, H. T., Le, D. T., Duong, Q. H., Tatipamula, V. B., Nguyen, B. V."High frequency of microsatellite instability and its substantial co‑existence with <em>KRAS</em> and <em>BRAF</em> mutations in Vietnamese patients with colorectal cancer". Oncology Letters 21.1 (2021): 41.
Chicago
Nguyen, H. T., Le, D. T., Duong, Q. H., Tatipamula, V. B., Nguyen, B. V."High frequency of microsatellite instability and its substantial co‑existence with <em>KRAS</em> and <em>BRAF</em> mutations in Vietnamese patients with colorectal cancer". Oncology Letters 21, no. 1 (2021): 41. https://doi.org/10.3892/ol.2020.12302