PIK3CA mutation and methylation influences the outcome of colorectal cancer

  • Authors:
    • Satoru Iida
    • Shunsuke Kato
    • Megumi Ishiguro
    • Takatoshi Matsuyama
    • Toshiaki Ishikawa
    • Hirotoshi Kobayashi
    • Tetsuro Higuchi
    • Hiroyuki Uetake
    • Masayuki Enomoto
    • Kenichi Sugihara
  • View Affiliations

  • Published online on: December 29, 2011     https://doi.org/10.3892/ol.2011.544
  • Pages: 565-570
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) occurs through the accumulation of genetic and epigenetic alterations. The epigenetic abnormalities, in cooperation with genetic alterations, are capable of causing aberrant gene function that results in cancer. In the present study, we examined mutations and methylation status in 164 CRCs to determine whether the combination of genetic and epigenetic alterations may be used to classify CRC patients in relation to their clinicopathological characteristics and outcomes. Mutation analyses for the KRAS and PIK3CA genes were performed using direct sequencing, and the MethyLight method was used to determine the methylation status of BNIP3, p16 and hMLH1. The combination of the KRAS mutation with methylation status did not have any association with clinicopathological characteristics and outcomes. However, patients with the PIK3CA mutation and/or high methylation (2 or 3 methylated genes) had significantly poorer outcomes in disease-specific survival (DSS) compared with those with wild-type PIK3CA and 0 or 1 methylated genes (P=0.0059). Additionally, multivariate analysis revealed that the PIK3CA mutation and/or a high level of methylation predicts a poor DSS independently of clinicopathological characteristics. Our results suggest that a combination of genetic and epigenetic alterations is a potent biomarker for predicting the prognosis of CRC.

Introduction

Colorectal cancer (CRC) is among the most common types of cancer in both males and females and is associated with high mortality, particularly at advanced stages (1). Markers for defining individual risk signatures in CRC patients are of great clinical value, as they may allow for targeted therapies to improve the outcomes of CRC patients.

CRC arises through the accumulation of multiple genetic and epigenetic changes. Somatic mutations in APC, BRAF, KRAS, PIK3CA and TP53, as well as in other genes, have been frequently observed in CRC and are thought to drive colorectal tumorigenesis (2). We previously examined the mutation of genes in the EGFR pathway and found that the PIK3CA mutation is associated with poor prognosis in CRC patients (8,11).

In addition to genetic alterations, epigenetic silencing is a prevalent mechanism by which abnormal gene inactivation may occur in cancer and is involved in initiation and promotion (3). A predominant mode of epigenetic alteration in cancer is gene silencing via the hypermethylation of the CpG island promoter. Hypermethylation results in abnormal silencing of a number of tumor suppressors in numerous human malignancies including CRC (3).

It has been suggested that epigenetic abnormalities are able to cooperate with genetic alterations to cause aberrant gene function and result in cancer (4). Thus, in the present study, we examined KRAS and PIK3CA mutations and the methylation status of BNIP3, p16 and hMLH1 and analyzed the correlation between these molecular alterations and the clinicopathological characteristics of CRC. We aimed to clarify whether a combination of genetic and epigenetic alterations could be used to classify CRC patients in relation to their clinicopathological characteristics and outcomes.

Materials and methods

Patients and tissue samples

After providing informed consent, a total of 165 CRC patients who underwent surgical resection at Tokyo Medical and Dental University Hospital between March 2000 and April 2003 were recruited in this study. This study was approved by the institutional review board of the Tokyo Medical and Dental University. The study population (Table I) consisted of 62 females and 103 males, with a mean age of 64.5±10.3 years (range, 37–89). Sixty-two cancers were located in the proximal site and 103 were located in the distal site, including the rectum. Histological classification and tumor staging were performed according to the International Union Against Cancer Tumor-Node-Metastasis classification guidelines. No patient received preoperative chemotherapy or radiotherapy. Following surgery, patients with stage III CRC received oral or intravenous 5-fluorouracil (FU)-based adjuvant chemotherapy and patients with stage IV tumors received 5-FU-based systemic chemotherapy without radiotherapy. Patients were prospectively followed up following surgery for a median period of 64 months (range, 0.07–107.6). Resected specimens were fixed in 10% neutral buffered formalin and embedded in paraffin. For all cases, archival hematoxylin and eosin (H&E) slides of primary tumors were retrieved and reviewed to confirm pathological features.

Table I

Clinicopathological characteristics according to the mutation and methylation status of each gene.

Table I

Clinicopathological characteristics according to the mutation and methylation status of each gene.

KRASPIK3CABNIP3p16hMLH1





CharacteristicsMut
52 (32%)
Wt
113 (69%)
PMut
20 (12%)
Wt
145 (88%)
PMet
84 (58%)
Unm
61 (42%)
PMet
48 (33%)
Unm
97 (67%)
PMet
30 (21%)
Unm
115 (79%)
P
Age (years)64.6 (37–89)64.6 (41–88)0.89867.8 (54–84)64.1 (37–89)0.14565.0 (37–89)63.8 (41–88)0.61466.2 (48–87)64.0 (37–89)0.29265.7 (37–88)64.4 (41–89)0.496
Gender0.2160.8110.4560.6610.288
 Female23 (37)39 (63)8 (13)54 (87)28 (54)24 (46)17 (31)38 (69)13 (24)42 (76)
 Male29 (28)74 (72)12 (12)91 (88)56 (60)37 (40)31 (34)59 (64)17 (19)73 (81)
Location0.3720.2210.9180.3050.338
 Proximal22 (35)40 (65)10 (16)52 (84)31 (58)22 (42)20 (38)32 (68)13 (25)39 (75)
 Distal30 (29)73 (71)10 (10)93 (90)53 (58)39 (42)28 (30)65 (70)17 (18)76 (82)
Differentiation0.7730.8860.4660.3310.529
 Wel23 (34)44 (66)9 (13)59 (87)32 (53)28 (47)22 (38)36 (62)11 (19)47 (81)
 Mod24 (28)62 (72)10 (12)74 (88)43 (59)30 (41)23 (31)51 (69)15 (20)59 (80)
 Por4 (36)7 (64)1 (8)11 (92)8 (73)3 (27)2 (17)10 (83)4 (33)8 (67)
T0.2230.3460.6410.3710.558
 Tis5 (56)4 (44)0 (0)9 (100)4 (67)2 (33)1 (17)5 (83)0 (0)6 (100)
 T12 (14)12 (86)3 (23)10 (77)8 (67)4 (33)6 (55)5 (45)1 (9)10 (91)
 T26 (33)12 (67)1 (6)17 (94)9 (64)5 (36)6 (43)8 (57)3 (21)11 (79)
 T322 (36)39 (64)6 (10)55 (90)35 (61)22 (39)19 (33)38 (67)14 (25)43 (75)
 T417 (27)47 (73)10 (16)54 (84)28 (50)28 (50)16 (28)41 (72)12 (21)45 (79)
N0.3250.5700.2950.4040.592
 N028 (29)69 (71)13 (13)84 (87)45 (54)38 (46)30 (36)53 (64)16 (19)67 (81)
 N1-324 (36)43 (64)7 (10)60 (90)39 (63)23 (37)18 (30)43 (70)14 (23)47 (77)
M0.2420.0980.2900.5030.0005
 M045 (33)92 (67)14 (10)123 (90)69 (56)54 (44)39 (32)83 (68)19 (16)103 (84)
 M16 (21)22 (79)6 (21)22 (79)15 (68)7 (32)9 (39)14 (61)11 (48)12 (52)

[i] Mut, mutated; Wt, wild-type; Met, methylated; Unm, unmethylated; Wel, well-differentiated; Mod, moderately differentiated; Por, poorly differentiated; TNM classification according to the International Union Against Cancer TNM staging.

DNA extraction

Tissues were cut into 10-μm sections from paraffin-embedded blocks. After specimens were deparaffinized and washed, tumor tissue was manually dissected for comparison with H&E slides. The same amount of dissected tissue was used for each case. Genomic DNA was extracted using standard proteinase K (Invitrogen, Carlsbad, CA, USA) digestion, phenol/chloroform extraction, and ethanol precipitation, as previously described (5).

Mutation analysis

Exon 1 of the KRAS gene and exons 9 and 20 of the PIK3CA gene were selected for mutation analysis, since mutations are known to cluster in these regions (6,7). Primer sequences and PCR conditions have been reported previously (8). Following the purification of PCR products using a Microcon YM-100 Centrifugal Filter (Millipore Corporation, Billerica, MA, USA) and Centri-Sep Columns (Princeton Separations, Adelphia, NJ, USA), direct sequencing was carried out using the Big Dye Terminator Cycle Sequencing kit (Applied Biosystems, Foster City, CA, USA) and analyzed using an Applied Biosystems 3130 Genetic Analyzer (Applied Biosystems). Somatic mutations were further validated by independent PCR amplification and sequencing and matched with normal mucosa.

MethyLight analysis

Sodium bisulfite conversion and DNA recovery was performed using EpiTect Bisulfite (Qiagen, Hilden, Germany). Following sodium bisulfite conversion, genomic DNA was analyzed using the MethyLight technique, a fluorescence-based real time PCR (Q-PCR) assay (9) and the ABI Prism 7300 Real Time PCR system (Taqman; Applied Biosystems). Four sets of primers and probes designed specifically for bisulfite-converted DNA were used. Three sets were used to detect methylation in the gene of interest and the remaining set served as a reference for β-actin (ACTB) to normalize for input DNA. Reference primers and probes were designed in a region of the ACTB gene lacking CpG dinucleotides, thus allowing for equivalent amplification regardless of methylation levels. Primer and probe sequences were previously reported (10,11). SssI-treated HCT-15 DNA was used as a fully methylated positive control (100% methylation ratio). Parallel TaqMan PCR reactions were performed using primers specific for the bisulfite-converted methylated sequence of a particular locus and with ACTB reference primers. In each case, triplicate threshold cycle (Ct) values were obtained and averaged and expression levels were evaluated using the 2−ΔΔCt method. As an internal standard, each sample was normalized to its ACTB content and compared with the gene expression level of SssI-treated HCT-15 DNA as positive controls (calibration sample) as follows: 2ΔΔCt where ΔΔCt=(Ct-target-Ct-reference) treated sample - (Ct-target-Ct-reference) calibrator sample. We defined the percentage of the fully methylated reference (PMR) to be 2ΔΔCt ×100%. To define p16 or hMLH1 methylation status, a PMR cut-off value of 4 was used based on previously validated data (9). Based on the distribution of PMR values in normal colon epithelial tissue (10), a PMR cut-off value of >0 was used to define positive methylation status for BNIP3.

Statistical analysis

Statistical analysis was carried out using StatView Software (version 5.0). To estimate the differences among the groups, the Chi-square test, Fisher’s exact test, Student’s t-test and log-rank test were used where appropriate. The Kaplan-Meier method was used to estimate survival. Survival was calculated beginning from the date of surgery. Prognostic factors were estimated using multivariate analysis and the Cox proportional hazards model. P<0.05 was considered to indicate a statistically significant result.

Results

Mutation of KRAS and PIK3CA and methylation of BNIP3, p16 and hMLH1

KRAS mutations in exon 1 were observed in 32% of cases and PIK3CA mutations in exons 9 and 20 in 12% of cases. Methylation frequencies of the examined genes were 58% for BNIP3, 33% for p16 and 21% for hMLH1. Although hMLH1 methylation was significantly associated with distant metastasis (P=0.0005), other associations were not observed between clinicopathological characteristics and the mutation or methylation status of individual genes (Table I).

Correlation between mutation and methylation

Associations among gene mutations and gene methylations are shown in Table II. The KRAS mutation was significantly associated with BNIP3 (P<0.0001) and p16 methylation (P=0.04) as well as with the number of methylated genes (P=0.0082). p16 methylation was associated with BNIP3 methylation (P<0.0001) and hMLH1 methylation (P=0.03).

Table II

Concordance between the mutation and methylation status of each gene.

Table II

Concordance between the mutation and methylation status of each gene.

PIK3CABNIP3p16hMLH1Methylationa





MutWtMetUnmMetUnmMetUnmHighLow
KRASP=0.918P<0.0001P=0.0402P=0.467P=0.0082
 Mut74539921268392423
 Wt13884552277122772870
PIK3CAP=0.827P=0.982P=0.428P=0.775
 Mut108612513711
 Wt74534285251024582
BNIP3P<0.0001P=0.541
 Met42391964
 Unm4541148
p16P=0.0295
 Met1533
 Unm1581

a Methylation high, 2 or 3 methylated genes; low, 0 or 1 methylated genes.

{ label (or @symbol) needed for fn[@id='tfn3-ol-03-03-0565'] } Mut, mutated; Wt, wild-type; Met, methylated; Unm, unmethylated.

Correlation between molecular markers and outcomes

The correlation between the molecular parameters and disease-specific survival (DSS) were analyzed (Table III). Patients with hMLH1-methylated tumors had significantly shorter DSS compared with those without methylation [hazards ratio (HR), 2.231; P=0.05], but mutation or methylation of other single genes did not affect DSS. Having mutations in both the KRAS and PIK3CA genes did not correlate with DSS. By contrast, when cases were divided into 2 groups, namely a low methylation group (0 or 1 methylated genes) and a high methylation group (2 or 3 methylated genes), the high methylation group had significantly shorter DSS than the low methylation group (HR, 2.681; P=0.01). Integration of the KRAS mutation and methylation status of multiple genes did not reveal an association with DSS. However, CRCs were divided into 4 groups: CRC containing i) PIK3CA mutation and high methylation (5.2%); ii) PIK3CA mutation and low methylation (8.1%); iii) no mutation and high methylation (31.1%); and iv) no mutation and low methylation (55.6%). Patients with the PIK3CA mutation and/or high methylation (groups i, ii and iii) had a significantly poorer outcome in DSS compared with those with wild-type PIK3CA and low methylation (group iv; HR, 2.924; P=0.0087) (P=0.0059, log-rank test; Fig. 1).

Table III

Correlation of molecular parameters with DSS.

Table III

Correlation of molecular parameters with DSS.

HR95% CIP
KRAS(Mutant vs. wild)0.9300.418–2.0710.8590
PIK3CA(Mutant vs. wild)1.4290.540–3.7790.4721
BNIP3(Methylated vs. unmethylated)2.2330.944–5.2840.0675
p16(Methylated vs. unmethylated)1.9500.912–4.1690.0849
hMLH1(Methylated vs. unmethylated)2.2311.000–4.9770.0500
Mutationa(Mutant vs. wild)1.0010.468–2.1390.9983
Methylationb(High vs. low)2.6811.253–5.7470.0110
Mutation/methylation(Mutant and/or high vs. wild and low)1.4990.634–3.5460.3569
KRAS/methylation(Mutant and/or high vs. others)1.6750.766–3.6630.1963
PIK3CA/methylation(Mutant and/or high vs. others)2.9241.312–6.4940.0087

a Mutant, at least one mutation gene; wild, no mutation.

b Methylation high, 2 or 3 methylated genes; low, 0 or 1 methylated genes.

{ label (or @symbol) needed for fn[@id='tfn6-ol-03-03-0565'] } CI, confidence interval; HR, hazards ratio.

Results of the univariate and multivariate analyses using the Cox proportional hazards model are shown in Table IV. Univariate analysis revealed that the PIK3CA mutation and/or high methylation was a significant prognostic factor for DSS, when used with the depth of the tumor, lymph node metastasis, distant metastasis, histological differentiation, lymphatic invasion and vessel invasion. Variables with P<0.05 in the univariate analysis were used for the multivariate analysis. PIK3CA mutation and/or high methylation, as well as distant metastasis and lymphatic invasion, were found to be independent and significant prognostic factors for DSS.

Table IV

Cox proportional hazards model of prognostic factors of DSS.

Table IV

Cox proportional hazards model of prognostic factors of DSS.

Univariate analysisMultivariate analysis


CharacteristicsHR95% CIPHR95% CIP
Gender(Female vs. male)0.4240.171–1.0500.0636
Age(≥65 vs. <65)0.6990.328–1.4870.3526
Location(Distal vs. proximal)0.8390.389–1.8080.6539
T(T1,2 vs. T3,4)8.5471.157–62.500.03542.4510.270–22.200.4253
N(N− vs. N+)3.5591.558–8.1300.00261.3680.504–3.7170.5377
M(M0 vs. M1)7.8743.448–17.86<0.00017.0922.732–18.18<0.0001
Differentiation(Por vs. wel/mod)3.0311.046–8.7810.04110.6110.172–2.1720.4466
Ly(Ly0,1 vs. ly2,3)4.8312.247–10.31<0.00013.8911.379–10.990.0102
v(v0,1 vs. v2,3)4.0491.399–11.760.00991.7610.540–5.7470.3482
PIK3CA/methylation(Mutant and/or high vs. others)2.9241.312–6.4940.00872.7931.220–6.4100.0151

[i] HR, hazards ratio; CI, confidence interval; TNM classification according to the International Union Against Cancer TNM staging; por, poorly differentiated; wel, well-differentiated; mod, moderately differentiated; ly, lymphatic invasion; v, vessel invasion; high methylation, 2 or 3 methylated genes.

Discussion

In this study, we examined mutations in KRAS and PIK3CA, which comprise components of the EGFR pathway, and the methylation status of BNIP3, p16 and hMLH1 in CRCs to evaluate patients with the PIK3CA mutation and/or 2 or more methylations with a poorer prognosis compared with other patients. Our results suggest that a combination of epigenetic and genetic alterations is a potent biomarker for predicting CRC prognosis.

Integrated analysis of epigenetic and genetic alterations is increasingly significant in cancer research. Since CRC develops as a result of accumulating genetic and epigenetic alterations, the combination of genetic and epigenetic profiles could confer differential clinical phenotypes and potential variability in survival. Ward et al reported that although DNA methylation is associated with a worse outcome in CRC patients, this adverse prognostic effect is lost in methylated tumors with microsatellite instability (12). By integrating genetic and epigenetic analyses, Shen et al revealed that colon cancers correspond to 3 molecularly distinct subclasses of disease, each of which is relatively homogeneous (13). In the present study, CRCs were divided into 4 groups according to PIK3CA mutations and the number of methylated genes: CRCs with i) PIK3CA mutation and 2 or 3 methylations (5.2%); ii) PIK3CA mutation and less than 2 methylations (8.1%); iii) no mutation and 2 or 3 methylations (31.1%); and iv) no mutations and less than 2 methylations (55.6%). The last group was associated with better prognosis than the former 3 groups. CRC patients with the PIK3CA mutation and/or more than 1 methylation had worse outcomes compared with the other groups. This finding suggests that mutation analysis and determining methylation status may provide significant predictive information regarding tumor behavior and clinical outcome in CRC patients.

BNIP3 is a pro-apoptotic member of the Bcl-2 family (14). The expression of BNIP3 is induced by hypoxia, such as that which occurs during cardiac ischemia and in the hypoxic regions of tumors, and it acts against pro-survival proteins, including Bcl-2 and Bcl-xl (1518). Transcriptional silencing of the BNIP3 gene has recently been observed in multiple human cancer cell lines, including colorectal, gastric, pancreatic, and hematopoietic tumors (19). Murai et al reported that aberrant hypermethylation of BNIP3 was detected in 66% of primary colorectal and 49% of primary gastric cancers, but not in normal tissue; these authors also revealed that BNIP3 is silenced by DNA methylation (20). Akada et al and Erkan et al revealed that loss of BNIP3 expression correlates with poor prognosis and increased chemoresistance in patients with pancreatic cancer (21,22). Manka et al demonstrated that BNIP3 silencing induces or increases the metastatic growth of breast cancer in distant organs (23). Moreover, our previous study also indicated that BNIP3 methylation is associated with poor clinical outcome and chemoresistance in primary CRC (10). These previous studies suggest that reduced BNIP3 expression contributes to carcinogenesis of the colon and rectum and may be a predictive factor for the prognosis of CRC. Therefore, the BNIP3 gene, as well as the hMLH1 and p16 genes, were tested in our methylated gene sets for predicting the outcome of CRC.

Activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway is thought to be critical in CRC development and clinical behavior (6,8,24,25). PIK3CA encodes the p110α catalytic subunit of PI3K and is mutated in 10–32% of CRCs (6,8,24,2629). PIK3CA mutations elevate kinase activity, thereby activating the PI3K/AKT pathway and contributing to tumorigenesis through decreased apoptosis, loss of contact inhibition and increased tumor invasion (6,30); these factors have been associated with poor outcomes in CRC patients (8,24,31). Although previous studies have investigated the role of the PIK3CA mutation and gene methylation in CRC, the data are inconclusive. Certain studies reported a significantly higher frequency of the PIK3CA mutation in the CpG island methylator phenotype (CIMP)-high tumor compared with CIMP-negative tumors and a significant correlation between the PIK3CA mutation and RASSF2 gene methylation (29,32,33), whereas other studies failed to observe this correlation (11,34). In the present study, although CIMP status was not examined, the PIK3CA mutation did not exhibit an association with the methylation status of the genes examined. Moreover, few studies have described the effect of the combination of the PIK3CA mutation with gene methylation on patient outcome. Ogino et al demonstrated that the effect of the PIK3CA mutation on CRC patient survival is not significantly modified by CIMP (31). Their result was inconsistent with our finding that integration of the PIK3CA mutation and methylation status may be used to predict outcomes for CRC patients. It is possible that the methylated gene set used in this study was different from that used in the Ogino study. The BNIP gene included in our methylated set was not included in CIMP markers used to date. Moreover, there have been no studies regarding the influence of integrating the PIK3CA mutation and methylation status of the BNIP3 gene on outcomes of CRC. It would be useful to examine whether mutating other genes from the EGFR pathway also affects CRC prognosis in cooperation with BNIP3 methylation.

In conclusion, by integrating genetic and epigenetic alterations, we revealed that CRC patients with poor prognosis could be identified. Further studies are necessary to confirm our finding that a combination of the PIK3CA mutation with gene methylation may be useful for predicting outcomes of CRC patients and to elucidate the underlying mechanism behind these findings.

References

1 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics. CA Cancer J Clin. 60:277–300. 2010.

2 

Wood LD, Parsons DW, Jones S, et al: The genomic landscape of human breast and colorectal cancers. Science. 318:1108–1113. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Jones PA and Baylin SB: The fundamental role of epigenetic events in cancer. Nat Rev Genet. 3:415–428. 2002.PubMed/NCBI

4 

Jones PA and Baylin SB: The epigenomics of cancer. Cell. 128:683–692. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Iida S, Akiyama Y, Nakajima T, Ichikawa W, Nihei Z, Sugihara K and Yuasa Y: Alterations and hypermethylation of the p14ARF gene in gastric cancer. Int J Cancer. 87:654–658. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Samuels Y, Wang Z, Bardelli A, et al: High frequency of mutations of the PIK3CA gene in human cancers. Science. 304:5542004. View Article : Google Scholar : PubMed/NCBI

7 

Vogelstein B, Fearon ER, Hamilton SR, et al: Genetic alterations during colorectal-tumor development. N Engl J Med. 319:525–532. 1988. View Article : Google Scholar : PubMed/NCBI

8 

Kato S, Iida S, Higuchi T, et al: PIK3CA mutation is predictive of poor survival in patients with colorectal cancer. Int J Cancer. 121:1771–1778. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Eads CA, Danenberg KD, Kawakami K, et al: MethyLight: a high-throughput assay to measure DNA methylation. Nucleic Acids Res. 28:E32–00. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Shimizu S, Iida S, Ishiguro M, et al: Methylated BNIP3 gene in colorectal cancer prognosis. Oncol Lett. 1:865–872. 2010.PubMed/NCBI

11 

Aoyagi H, Iida S, Uetake H, et al: Effect of classification based on combination of mutation and methylation in colorectal cancer prognosis. Oncol Rep. 25:789–794. 2011.PubMed/NCBI

12 

Ward RL, Cheong K, Ku SL, Meagher A, O’Connor T and Hawkins NJ: Adverse prognostic effect of methylation in colorectal cancer is reversed by microsatellite instability. J Clin Oncol. 21:3729–3736. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Shen L, Toyota M, Kondo Y, et al: Integrated genetic and epigenetic analysis identifies three different subclasses of colon cancer. Proc Natl Acad Sci USA. 104:18654–18659. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Reed JC: Bcl-2 family proteins. Oncogene. 17:3225–3236. 1998. View Article : Google Scholar

15 

Labi V, Erlacher M, Kiessling S and Villunger A: BH3-only proteins in cell death initiation, malignant disease and anticancer therapy. Cell Death Differ. 13:1325–1338. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Strasser A: The role of BH3-only proteins in the immune system. Nat Rev Immunol. 5:189–200. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Willis SN and Adams JM: Life in the balance: how BH3-only proteins induce apoptosis. Curr Opin Cell Biol. 17:617–625. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Vande VC, Cizeau J, Dubik D, et al: BNIP3 and genetic control of necrosis-like cell death through the mitochondrial permeability transition pore. Mol Cell Biol. 20:5454–5468. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Mellor HR and Harris AL: The role of the hypoxia-inducible BH3-only proteins BNIP3 and BNIP3L in cancer. Cancer Metastasis Rev. 26:553–566. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Murai M, Toyota M, Suzuki H, et al: Aberrant methylation and silencing of the BNIP3 gene in colorectal cancer and gastric cancer. Clin Cancer Res. 11:1021–1027. 2005.PubMed/NCBI

21 

Akada M, Crnogorac-Jurcevic T, Lattimore S, et al: Intrinsic chemoresistance to gemcitabine is associated with decreased expression of BNIP3 in pancreatic cancer. Clin Cancer Res. 11:3094–3101. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Erkan M, Kleeff J, Esposito I, et al: Loss of BNIP3 expression is a late event in pancreatic cancer contributing to chemoresistance and worsened prognosis. Oncogene. 24:4421–4432. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Manka D, Spicer Z and Millhorn DE: Bcl-2/adenovirus E1B 19 kDa interacting protein-3 knockdown enables growth of breast cancer metastases in the lung, liver, and bone. Cancer Res. 65:11689–11693. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Barault L, Veyrie N, Jooste V, et al: Mutations in the RAS-MAPK, PI(3)K (phosphatidylinositol-3-OH kinase) signaling network correlate with poor survival in a population-based series of colon cancers. Int J Cancer. 122:2255–2259. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Manning BD and Cantley LC: AKT/PKB signaling: Navigating downstream. Cell. 129:1261–1274. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Velho S, Oliveira C, Ferreira A, et al: The prevalence of PIK3CA mutations in gastric and colon cancer. Eur J Cancer. 41:1649–1654. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Benvenuti S, Frattini M, Arena S, et al: PIK3CA cancer mutations display gender and tissue specificity patterns. Hum Mutat. 29:284–288. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Abubaker J, Bavi P, Al-Harbi S, et al: Clinicopathological analysis of colorectal cancers with PIK3CA mutations in Middle Eastern population. Oncogene. 27:3539–3545. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Nosho K, Kawasaki T, Ohnishi M, et al: PIK3CA mutation in colorectal cancer: relationship with genetic and epigenetic alterations. Neoplasia. 10:534–541. 2008.PubMed/NCBI

30 

Samuels Y, Diaz LA Jr, Schmidt-Kittler O, et al: Mutant PIK3CA promotes cell growth and invasion of human cancer cells. Cancer Cell. 7:561–573. 2005. View Article : Google Scholar

31 

Ogino S, Nosho K, Kirkner GJ, et al: PIK3CA mutation is associated with poor prognosis among patients with curatively resected colon cancer. J Clin Oncol. 27:1477–1484. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Nosho K, Yamamoto H, Takahashi T, et al: Genetic and epigenetic profiling in early colorectal tumors and prediction of invasive potential in pT1 (early invasive) colorectal cancers. Carcinogenesis. 28:1364–1370. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Whitehall V, Rickman C, Bond C, et al: Oncogenic PIK3CA mutations in colorectal cancers and polyps. Int J Cancer. 2011.(Epub ahead of print). View Article : Google Scholar

34 

Bruin SC, He Y, Mikolajewska-Hanclich I, et al: Molecular alterations associated with liver metastases development in colorectal cancer patients. Br J Cancer. 105:281–287. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2012
Volume 3 Issue 3

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
Iida S, Kato S, Ishiguro M, Matsuyama T, Ishikawa T, Kobayashi H, Higuchi T, Uetake H, Enomoto M, Sugihara K, Sugihara K, et al: PIK3CA mutation and methylation influences the outcome of colorectal cancer. Oncol Lett 3: 565-570, 2012
APA
Iida, S., Kato, S., Ishiguro, M., Matsuyama, T., Ishikawa, T., Kobayashi, H. ... Sugihara, K. (2012). PIK3CA mutation and methylation influences the outcome of colorectal cancer. Oncology Letters, 3, 565-570. https://doi.org/10.3892/ol.2011.544
MLA
Iida, S., Kato, S., Ishiguro, M., Matsuyama, T., Ishikawa, T., Kobayashi, H., Higuchi, T., Uetake, H., Enomoto, M., Sugihara, K."PIK3CA mutation and methylation influences the outcome of colorectal cancer". Oncology Letters 3.3 (2012): 565-570.
Chicago
Iida, S., Kato, S., Ishiguro, M., Matsuyama, T., Ishikawa, T., Kobayashi, H., Higuchi, T., Uetake, H., Enomoto, M., Sugihara, K."PIK3CA mutation and methylation influences the outcome of colorectal cancer". Oncology Letters 3, no. 3 (2012): 565-570. https://doi.org/10.3892/ol.2011.544