Skip to main content
International Journal of Clinical and Experimental Medicine logoLink to International Journal of Clinical and Experimental Medicine
. 2015 Mar 15;8(3):3241–3257.

Correlation between smoking history and molecular pathways in sporadic colorectal cancer: a meta-analysis

Ke Chen 1,*, Guanggai Xia 2,*, Changhua Zhang 2, Yunwei Sun 1
PMCID: PMC4443048  PMID: 26064214

Abstract

Background: Epidemiological studies have shown that smoking increases the risk for colorectal cancer (CRC). Evidence of the guiding significance of smoking history for molecular classification and molecular targeted anti-tumor therapy is not well established. Aims: To provide indirectly evidence, we conducted a systematic meta-analysis of association between smoking history and different molecular classification. Methods: We searched in multiple databases up to January 2014, and identified 27 eligible studies. All studies were divided into seven groups based on different molecular alteration categories, which are MSI, CIMP, and three molecular pathway-associated gene alterations (APC, KRAS, P53, BRAF mutation, and APC methylation). Crude odds ratios (ORs) and their 95% confidence intervals (CIs) were calculated to evaluate the association. Results: Smoking showed a significantly positive correlation with P53 mutation (exons 4 to 8), BRAF (codon 600) mutation, MSI positivity, and CIMP positivity, with ORs of 1.25 (95% CI: 1.07-1.45), 1.41 (95% CI: 1.18-1.68), 1.28 (95% CI: 1.12-1.47), and 1.23 (95% CI: 1.01-1.50), respectively. However, smoking was not positively correlated with APC (mutation cluster region) and KRAS (codons 12 and 13) mutation in sporadic CRC patients. Conclusions: These findings suggested smoking history occurred with P53 mutation, BRAF mutation, MSI positivity, and CIMP positivity in sporadic CRCs; and could guide those specifically therapeutic designs when molecular classification with genetic test was infeasible. More associated studies should be conducted for strengthening and renewing the current result.

Keywords: Smoking, molecular pathways, sporadic colorectal cancer, genetic, therapy

Introduction

Compared with the traditional treatment, molecular targeted therapies play a dominant position in the clinical practice of colorectal cancer (CRC), with the better understanding of its molecular features. Generally, stepwise accumulated genetic and epigenetic alterations drive the normal glandular epithelium into invasive adenocarcinoma through at least three diametrically exclusive molecular pathways [1]. Furthermore, genetic alterations can be classified as chromosomal instability (CIN) and microsatellite instability (MSI) [2,3]. Meanwhile, the most common epigenetic alteration is the DNA methylation phenotype of CpG islands named as CpG island methylation phenotype (CIMP) [4]. CIN, MSI, and CIMP are the major molecular pathways involved in colorectal carcinogenesis [3].

These alterations inactivate or activate tumor suppressor genes and oncogenes, such as APC, KRAS, P53, and BRAF [5]. KRAS mutation in codons 12 and 13 or BRAF mutation in codon 600 aberrantly regulates the RAS/RAF/MAPK signal pathway and disturbs the balance between cell proliferations. However, KRAS and BRAF mutations exist in CRC separately; BRAF shows high association with CIMP [6]. APC mutation exists in most cases of sporadic CRC, in which mutation cluster regions (MCRs) from codons 1286 to 1513 are the most common sites [7]. Similarly, upregulation of the downstream target gene cyclin-dependent kinase inhibitor P21 leads to alterations in CRC genes, including the tumor suppressor P53, which acts as guardian of the genome and responds to DNA strand breaks [8]. Published data have robustly proved that different molecular characteristics did not respond similar to chemotherapy of patient with CRC [9-11]. The most classical practice was the worse response to cetuximab, a monoclonal antibody that inhibit the epidermal growth factor receptor, due to the KRAS mutation status. Hence, routine molecular examines have been recommended in clinical practice to design specific therapeutic strategies based on molecular classification [12].

The International Agency for Research on Cancer has recently reviewed epidemiological and evidence-based studies of human carcinogens and smoking, and concluded that smoking is a deleterious risk for human health and a cause of colon and ovarian cancers [13]. Animal experiments showed that many carcinogens in cigarette smoking are processed by the metabolic activation pathway and are covalently bound to DNA. The final DNA adducts might lead to miscoding, resulting in permanent mutations of tumor suppressor genes and oncogenes, such as APC, KRAS, and P53 [14]. Several large-scale case-control studies have focused on determining the association between lifestyle and diet and genetic alterations in CRC, but their results were discrepant and ambiguous. Therefore, deriving a comprehensive estimation of this association is important. In this study, we quantitatively assessed the association between smoking history and several major molecular features of sporadic CRC to study its referential value for molecular classification and targeted therapies.

Materials and methods

Literature search strategy

We searched MEDLINE, EMBASE, and Cochrane Library to identify eligible studies until January 2014. Keywords related to smoking (e.g., “smoking” or “cigarette”) in combination with words related to CRC (e.g., “colorectal” combined with “cancer”, “carcinoma”, “tumor” or “neoplasms”) and gene alteration (e.g., “alteration”, “mutation” or “variation”) were used during the search. All relevant reports identified were included with no restriction.

The inclusion criteria were as follows: (1) original studies on sporadic CRC published in English; (2) CRC diagnosis was based on histological or cytological findings; (3) samples for mutation, methylation, or MSI analysis was obtained from biopsy or surgical tumor tissue specimens; (4) repetitive studies were unified based on the largest edition; (5) case-control, nested case-control, or cohort studies.

The exclusion criteria were as follows: (1) reviews, case reports or meeting abstracts were excluded; (2) papers with insufficient or duplicated data were excluded; (3) studies about hereditary colorectal cancers like Lynch syndrome or inflammatory bowel diseases were excluded.

Data extraction

Two authors (KC, GX) independently extracted data of suitable articles and collected the following information: first author (publication year), country, study design, age, sex ratio, histological types, number of smokers, number of mutation/methylation/MSI cases, source and people distribution. Discrepancies were settled by discussion among the four researchers. Cigarette status was classified as nonsmokers and smokers (former or current). Smokers were those who had more than 100 cigarettes in their lifetime regardless of the smoking status at the enrollment time.

Molecular classification

APC status, KRAS (codons 12 and 13), P53 (exon 4 to 8), and BRAF (codon 600) were classified as either mutant or wild type. The difference between MSI-L and MSS is merely quantitative; thus, tumors with more than 30% unstable markers were defined as MSI positive when the MSI status was sorted into three categories (MSI-high, low, and MSS) [15]. Papers with information only about MSI negativity or positivity were pooled directly. Tumors with promoter hypermethylation in at least three genes of the CACNA1G, IGF2, NEUROG1, RUNX3, and SOCS1 gene panel or in at least two genes of another five-gene panel (p16, MLH1, MINT1, MINT2, and MINT31) were defined as CIMP positive as the blurred boundary between CIMP-L and CIMP-0 [3].

Statistical analysis

In this meta-analysis, crude odds ratios (ORs) and their 95% confidence intervals (CIs) were calculated to measure the association between smoking and gene alteration based on the alteration frequencies in sporadic CRC cases. Adjusted ORs from some parts of articles were rejected because different covariates were considered in the multivariate regression model. Heterogeneity was assessed with the Q and I2 statistics. Results with substantial heterogeneity (I2 > 50%) were pooled with a random-effect model (the DerSimonian and Laird method) [16]; otherwise, a fix-effect model (the Mantel-Haenszel method) was applied [17]. For publication bias assessment, both Begg’s and Egger’s funnel plots were used. All statistical analyses were performed using a commercial statistical software package (STATA 12.0; STATA Corporation, College Station, TX, US). Statistical significance was considered at P < 0.05.

Results

Characteristics of studies

A total of 844 items were found after considering the search criteria described above. After carefully reviewing, 27 eligible studies [18-44] were finally included in this analysis (Figure 1). A total of 22 studies described the three molecular pathway-associated gene alterations. Among these studies, 5 investigated APC mutation, 2 described APC methylation, 11 researched KRAS mutation, and 6 investigated P53 and BRAF mutations. Moreover, MSI was analyzed in 9 articles, and the CIMP was found in 4 articles. The characteristics of the included studies are summarized in Table 1.

Figure 1.

Figure 1

Flow chart of the study selection.

Table 1.

Main characteristics of studies included in the meta-analysis

First Author Country Study design Study Period Age Gender (M/F) No. of cases number No. of molecular cases Histological types Source People distribution Molecular features reported
Martinez (1999) USA Case-case 1990-1995 65.7 451/227 723 678 Colorectal adenoma WBF Hospital based KRAS mutation
Lafuente (2000) Spain Case-control 1996-1997 40-90 61/56 247 117 colorecal cancer N/A Hospital based KRAS mutation
Yang (2000) USA Case-control Case-case 1996-1997 > 50 66/95 161 161 Colorectal adenocarcinoma N/A Hospital based MSI mutation
Slattery (2001) USA Case-control Case-case 1991-1994 30-79 821/689 1,510 1,510 Colon carcinoma KPMCP Population based MSI mutation
Slattery (2001) USA Case-control Case-case 1991-1994 30-79 774/654 1,510 1,428 Colon carcinoma KPMCP Population based KRAS mutation
Wu (2001) USA Case-case 1995-1996 N/A 146/130 276 276 Colon adenocarcinoma CAP Population based MSI mutation
Miyaki (2002) Japan Case-case N/A N/A 27/34 61 61 Colon adenocarcinoma N/A Hospital based APC, P53, KRAS mutation
Slattery (2002) USA Case-control 1991-1994 30-79 793/665 1,510 1,458 Colon carcinoma KPMCP Population based P53 mutation
Diergaarde (2003) Netherlands Case-control Case-case 1989-1993 61.7 93/83 176 176 Colon carcinoma N/A Population based APC, P53, KRAS mutation MSI
Luchtenborg (2005) Netherlands Case-subcohort Case-case 1986-1994 55-69 330/264 594 594 colorecal cancer NCLS Population based APC mutation
Huang (2006) TaiWan Case-case 2000-2005 26-95 80/73 153 153 colorecal cancer N/A Hospital based P53 mutation
Sarebo (2006) Norway Case-control Case-case 1999-2001 50-64 82/51 133 133 Colorectal adenoma/carcinoma NORCCAP Population based APC mutation
Wark (2006) Netherlands Case-control Case-case 1997-2000 18-75 292/242 658 534 Colorectal adenoma N/A Hospital based KRAS mutation
Samowitz (2006) USA Case-control Case-case 1991-1994 30-79 717/598 1,315 1143/1271 Colon carcinoma KPMCP Population based BRAF mutation CIMP
Weijenberg (2008) Netherlands Case-subcohort Case-case 1986-1994 55-69 367/281 648 648 colorecal cancer NCLS Population based KRAS mutation
Curtin (2009) USA Case-control Case-case 1997-2001 30-79 Both 750 750 Rectosigmoid junction or rectum cancer KPMCP Population based KRAS, P53 mutation MSI, CIMP
Poynter (2009) USA Sibling Case-control 1998-2005 54.8 821/743 1,564 1,564 colorecal cancer N/A Population based MSI
Limsui (2010) USA Case-case 1986-2004 55-69 0/540, 527, 537 1,233 540/527/537 colorecal cancer IWHS Population based BRAF mutation MSI, CIMP
Park (2010) UK Case-case 1993-1997 45-79 92/93 185 138 colorecal cancer EPIC Population based P53 mutation
Rozek (2010) Northern Israel Case-case 1998-2004 N/A 651/618 1,297 1,269 colorecal cancer MECC Population based BRAF mutation MSI
Gay (2011) UK Case-case 1993-1997 45-79 84/85 185 169 colorecal cancer EPIC Population based MSI
Gay (2012) UK Case-case 1993-1997 45-79 88/87, 90/88 185 175/178 colorecal cancer EPIC Population based APC mutation and methylation
Naghibalhossaini (2012) Southern Iran Case-case 2003-2005 N/A 71/38 112 109 colorecal cancer N/A Hospital based APC methylation
Samadder (2012) USA Case-case 1986-2004 55-69 0/507 507 507 colorecal cancer IWHS Population based KRAS mutation
Burnett-Hartman (2013) USA Case-case 1998-2007 24-80 209/210 419 419 Serrated lesions N/A Population based BRAF mutation CIMP
Sinha (2013) India Case-case N/A 55.4 47/15 62 62 Colorectal adenocarcinoma N/A Hospital based KRAS mutation
Phipps (2013) USA Case-case 2004-2009 N/A 1024/935 1,959 1,959 Colon adenocarcinoma NCCTG Hospital based KRAS, BRAF mutation

Note: Molecular case is the subgroup of CRC patients that participated in the molecular study with smoking information. In the study by Samowitz (2006), the total number of molecular cases for BRAF, MSI and CIMP are 540, 527and 537, respectively. In the study by Limsui (2010), the total number of molecular cases for BRAF and CIMP are 1271 and 1143, respectively. In the study by Gay (2012), the total number of molecular cases for APC mutation and APC methylation are 175 and 178, respectively. WBF, the Wheat Bran Fiber trial; KPMCP, the Kaiser Permanente Medical Care Program; CAP, the los Angeles County cancer center surveillance program; NCLS, the prospective Netherlands Cohort Study; NORCCAP, the Norwegian Colorectal Cancer Prevention study; IWHS, the prospective Iowa Women’s Health Study; EPIC, the European Prospective Investigation of Cancer study; MECC, the Molecular Epidemiology of Colorectal Cancer study; NCCTG, the North Central Cancer Treatment Group; N/A, not available.

Cigarette smoking and gene mutation in CRC

APC mutation was not affected by the smoking habit of CRC patients. The combined OR estimate was 0.79 (95% CI: 0.51-1.20) (Figure 2A) with a statistically significant heterogeneity (I2 = 53.5%, P = 0.072). We analyzed two eligible studies that included 287 patients to completely understand the relationship between APC status and smoking; results showed no statistically significant association between smoking and APC methylation [22,23]. We combined the two OR estimates and found a negative relationship (OR = 0.94, 95% CI: 0.79-1.12) (data not shown).

Figure 2.

Figure 2

Forest plots of association between sporadic CRC patients’ smoking habit and several major molecular features. A. APC mutation; B. KRAS mutation; C. P53 mutation; D. BRAF mutation; E. MSI; F. CIMP.

A total of 6920 CRC patients from 1 case-control and 10 case-case studies were enrolled for KRAS (codons 12 and 13) mutation [18,19,21,29,30,32,36,38,41,43,44]. Only 30.9% cases showed significant association between KRAS mutation and smoking among CRC patients. The results of the remaining studies were the opposite. The pooled OR estimate was 1.17 (95% CI: 0.93-1.49) (Figure 2B) with the random-effect model, suggesting that smoking is not associated with KRAS mutation in CRC. We also combined 4 of these 11 studies that included patients suffering from colon cancer and detected no associations.

Six studies investigated P53 mutation frequency in smokers vs. nonsmokers with 2736 CRC patients [26,29,34,36-38]. Two of these studies found a significant relationship between smoking and P53 mutation. Furthermore, an increased P53 mutation risk was found in patients who had smoking habits when all the six studies were pooled for the meta-analysis (pooled OR = 1.25, 95% CI: 1.07-1.45) (Figure 2C) without significant heterogeneity.

Four studies focusing on BRAF mutation and smoking in CRC patients had diverse conclusions, with OR ranging from 0.69 (95% CI: 0.46-1.02) to 1.72 (95% CI: 1.15-2.57) [18,20,25,27,29,31]. In summary, the results from the combined analyses of all six studies indicate that cigarette smoking has no effect on BRAF mutation among CRC patients (OR = 1.21, 95% CI: 0.89-1.64) (Figure 2D). After revising the articles, we removed the serrated lesion cases and Burnett-Hartman’s result [20], which is different from sporadic CRC to some degree [45]. This revision yielded a summary OR of 1.41 (95% CI: 1.18-1.68) (Figure 3A).

Figure 3.

Figure 3

Meta-analysis of smoking and BRAF mutation or CIMP in sporadic CRC without Burnett-Hartman’s study. A. BRAF mutation; B. CIMP.

Cigarette smoking and MSI in CRC

Ten studies revealed that an association exists between cigarette smoking and MSI in CRC patients [24,25,27-29,31,36,39,40,42]. OR estimates ranged from 0.88 (95% CI: 0.41-1.89) to 3.33 (95% CI: 0.92-12.04). The OR estimates from the studies of Slattery et al. [40] and Limsui et al. [27] reached statistical significance. Figure 2E shows a forest plot for studies that examined the association between tobacco smoking and MSI in CRC patients. A fixed-effect model was used to pool these data by I2 < 50%. Their results indicated that smoking is strongly associated with MSI positivity (pooled OR = 1.28, 95% CI: 1.12-1.47). To exclude the effects produced by gender, we pooled the data by excluding the study (Limsui et al.) where patients were all women and then obtained a combined OR estimate of 1.26 (95% CI: 1.09-1.45) (Figure 4). Moreover, stratification by the tumor histological type revealed that smoking has a greater effect on increasing risk in the CRC group (pooled OR = 1.32, 95% CI: 1.10-1.59) than in the colon cancer group (pooled OR = 1.24, 95% CI: 1.01-1.50) (Figure 5).

Figure 4.

Figure 4

Meta-analysis of smoking and MSI in male sporadic CRC patients.

Figure 5.

Figure 5

Meta-analysis of smoking and MSI subgroups stratified by tumor histological type in sporadic CRC. A. Colorectal tumor; B. colon tumor.

Cigarette smoking and CIMP in CRC

Four studies [20,27,29,31] reported an association between smoking and CIMP in CRC patients, and OR estimates ranged from 0.88 (95% CI: 0.70-1.41) to 1.33 (95% CI: 1.01-1.70). A meta-analysis combining the CIMP OR estimates for CRC patients in these four studies obtained a summary OR estimate of 1.05 (95% CI: 0.79-1.41) with a significant heterogeneity (I2 = 57.7%, P = 0.069). With BRAF mutation, the obtained pooled OR was 1.23 (95% CI: 1.01-1.50) without Burnett-Hartman’s study 20. The results are shown in Figures 2F and 3B.

Assessment of publication bias

Begg’s and Egger’s tests were performed to predict the publication bias; neither of the tests provided significantly statistical publication bias. Nevertheless, some funnel plots seemed slightly asymmetrical. All funnel plots are displayed in Figure 6, and data are illustrated in Table 2.

Figure 6.

Figure 6

Begg’s and Egger’s funnel plots for association between sporadic CRC patients’ smoking habit and several major molecular features. A. APC mutation; B. KRAS mutation; C. P53 mutation; D. BRAF mutation; E. MSI; F. CIMP.

Table 2.

Summary results of association between sporadic CRC patients’ smoking habit and several major molecular features

Gene No. of sudies Effect model OR (95% CI) Heterogeneity Publication bias (P value)

I2 (%) P Begg’s test Egger’s test
APC mutation 5 Random 0.79 (0.51-1.20) 53.5 0.072 0.221 0.485
APC methylation 2 Fixed 0.94 (0.79-1.12) 0 0.660 N/A N/A
KRAS mutation 11 Random 1.17 (0.93-1.49) 71.2 0 0.213 0.2
P53 mutation 6 Fixed 1.25 (1.07-1.45) 9.4 0.356 0.452 0.453
BRAF mutation 6 Random 1.21 (0.89-1.64) 67 0.010 1 0.783
MSI 10 Fixed 1.28 (1.12-1.47) 0 0.855 1 0.711

N/A, not avaliable.

Discussion

This meta-analysis indicated the different correlations between smoking and several critical gene alterations among CRC patients. The results showed higher rates of P53 (exons 4 to 8) mutation, BRAF (codon 600) mutation, MSI positivity, and CIMP positivity in the smoking patients than in the nonsmoking patients. The rest did not show any significant correlations. The complete summary about the OR estimate, heterogeneity, and publication bias is provided in Table 2. All studies did not show any publication bias. The positive results showed minimal heterogeneity, whereas the negative results did not. Eliminating biased influence from the pooled negative results that originated from the discrepancy among these studies was difficult. Inversely, the positive results should be more convincible. To the best of our knowledge, this meta-analysis is the first to explore the key molecular features of sporadic CRC in smokers. Porta et al. [46] researched the relation between cigarette smoking and KRAS mutations in the pancreas, lung, and colorectal adenocarcinomas. Some of their results agreed with the present findings.

In 85% of sporadic CRC patients, mutation events of oncogenes and tumor suppressor genes, such as APC, KRAS, and P53, followed by CIN promote carcinogenesis via the classical adenoma-carcinoma sequence [47]. The well-known APC cluster region mutation and promoter A1 methylation, which lead to APC inactivation and sequentially WNT signal pathway over activity, could be found in aberrant crypt foci (ACF) in the early stage of adenoma-carcinoma sequence. The downstream event is KRAS mutation, which is usually located in codons 12 and 13. Low-grade dysplasia was also observed. P53 (exons 4 to 8) mutation promotes the progression toward malignancy. Several studies focused on the causality between lifestyle and gene mutations in CRC patients. The most common lifestyles studied include smoking, alcohol, meat consumption, and body mass index. The present meta-analysis revealed that smoking habit increases the P53 mutation rate (pooled OR = 1.25, 95% CI: 1.07-1.45) in sporadic CRC patients. However, the effects for APC and KRAS were uncertain. These results suggested that smoking is associated with the malignant transition instead of the early ACF formation and further epithelial dysplasia in sporadic CRC formation.

The germline mutation of MMR systems (MLH1, MSH2, MSH6, or PMS2 gene) and the promoter methylation of MLH1 gene lead to another molecular pathway called MSI. The former is the major genetic mechanism in hereditary nonpolyposis colorectal cancer or Lynch syndrome, which was excluded in our meta-analysis. Nevertheless, the latter makes up the 64% of the MSI pathway in sporadic CRC patients [48]. According to the NCI consensus panel, five robust microsatellite markers (D2S123, D5S346, D17S250, BAT25, and BAT26) had been recommended to standardize the assessment. In addition, researchers might test additional markers to improve accuracy. With the markers described above, MSI could be classified into MSI positive or negative (more than 30% unstable markers were defined as MSI positive). Slattery et al. [40] first reported that smoking significantly contributes to MSI (adjusted OR = 1.50, 95% CI: 1.20-2.00) in colon tumors in a large population-based study of colon adenocarcinoma. However, studies that followed could not strongly support the result, except for the recent one conducted by Limsui et al. [27]. The present meta-analysis found that smoking is strongly associated with MSI positivity (pooled OR = 1.28, 95% CI: 1.12-1.47) with no heterogeneity (I2 = 0, P = 0.855). Sample size, tumor histological type, gender ratio, and mean age might account for the discrepant results among these published studies. Subgroup analysis in CRC and colon cancer showed that rectal cancer suffers from the influence of smoking to MSI. However, the interpretation of our subgroup analysis remains challenging because MSI-positive tumors are usually located in the proximal colon [24].

MSI-positive tumors are manifested differently from MSI-negative sporadic CRCs, especially in terms of anatomical location and tumor’s Dukes stage. MSI-positive tumors are usually located in the proximal colon and have a Dukes stage of A/B; MSI-negative tumors show the opposite property [24,40,49]. Gay et al. [24] found that MLH1 promoter methylation prefers poor differentiation, except for the characteristics showed in MSI positive tumors. Furthermore, we speculated that smoking interacts with sporadic CRC before further metastasis. Former results showing a relationship between smoking and P53 mutation events before the malignant transition strengthened our speculation. The influence of smoking in different stages of carcinogenesis should be validated with more clinical and basic studies. Both types of tumor had some degree of overlaps in clinical features and predisposing factors because two-thirds of MSI were produced by MLH1 promoter methylation as a consequence of CIMP [50,51]. Obviously, data from our study also established that smoking exposure may be the collective and momentous factor for these tumors.

Aside from genetic alterations, epigenetic alterations have also received increasing attention. A subset of sporadic CRCs is accompanied by the CIMP of tumor suppressor genes, such as BRAF mutation at codon 600 [50,51]. Experiments have proven that smoking exposure is associated with CIMP at the tumor suppressor gene p16 promoter in nonsmall cell lung cancer [52,53]. Several clinical studies attempted to discover this relationship in CRC patients. Their results showed no significant associations with a summary OR estimate of 1.05 (95% CI: 0.79-1.41). Moreover, with the associated molecular events, BRAF mutation did not show definite correlation with smoking (pooled OR = 1.21, 95% CI: 0.89-1.64). We speculated that Burnett-Hartman’s study, which focused on the newly founded serrated pathway, might account for this phenomenon. The rare serrated pathway should be classified into hyperplastic polyp, sessile serrated adenoma/polyp, or traditional serrated adenoma according to the WHO recommendation [45]. Hence, we deleted this article and obtained a significantly positive correlation between smokers with CIMP or BRAF mutation and minimal heterogeneity. Nevertheless, only four eligible studies were included, and insufficient cases for the CIMP pooled analysis should be noticed.

Previous studies reported CRC patients harbored MSI-positive and CIMP-negative might not to suffer the benefit of adjuvant 5-FU regimes [54,55]. Recently, Chen’s meta-analysis indicated P53 mutation was associated with improved good and complete response, decreased poor response in neoadjuvant radiation-based treatment [10]. The latest data shows FOLFOXIRI plus bevacizumab might be a reasonable option for the first-line treatment of BRAF mutant metastatic CRC patients [11]. In those studies, high throughput methods such as RELP, ARMS and DMH based on PCR were utilized for molecular alternation detection [12]. However, high cost or low sensitivity always limited their large-scale extension. Reliable and convenient referential factors should be recommended, when those genetic methods are infeasible, aiming to carry out a better molecular classification and furthermore therapeutic design. With support of our study, smoking history might be effective to indicate a worse response to MSI, CIMP, P53, and BRAF specific therapy.

The correlation analysis between smoking and gene alteration has several limitations. First, standardized and precise gene panel for the assessment of MSI and CIMP positivity is unavailable. In CIMP, two sets of marker panels (described above) are accepted [50,56,57]. Both sets certify the correlation between CIMP and MSI positivity and BRAF mutation, whereas the diagnostic value or prognostic power is discriminating among the two panels [58]. This finding emphasizes the need for further subgroup analysis for CIMP detected by different panels with sufficient data. Moreover, in KRAS mutation analysis, substantial heterogeneity (I2 = 71.2%, P = 0.000) was generated. Heterogeneity could not be eliminated when we attempted to remove studies with less than 100 cases or those with a large scope of mutation sites, such as exon 1 (data not shown). Other factors such as quality grade, race, and histological type might contribute to the observed disparities. Apparently, we could not exclude the unsuspected bias from the final result.

In conclusion, results of this meta-analysis suggested that smoking increases the rates of P53 (exons 4 to 8) mutation, BRAF (codon 600) mutation, MSI positivity, and CIMP positivity in sporadic CRC patients. However, it had no obvious correlation with APC (MCR) mutation, APC promoter methylation, and KRAS (codons 12 and 13). MSI, CIMP, and partial CIN pathways suffered the effects of smoking that probably occurred in tumor formation before further metastasis. Efforts are necessary for the clinical molecular screening of P53, BRAF, MSI, and CIMP events among smokers with precancerous lesions and sequential smoking cessation. Besides, smoking histories could be treated as an important referential factor to guide the P53, BRAF, MSI, and CIMP specifically therapeutic design when molecular classification with genetic test is infeasible. More associated studies should be conducted for strengthening and renewing the current result.

Acknowledgements

The work originated from department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University of Medicine. This study was financially supported by the National Science Foundation of China (no. 81072010).

Disclosure of conflict of interest

None.

References

  • 1.Lao VV, Grady WM. Epigenetics and colorectal cancer. Nat Rev Gastroenterol Hepatol. 2011;8:686–700. doi: 10.1038/nrgastro.2011.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bakhoum SF, Compton DA. Chromosomal instability and cancer: a complex relationship with therapeutic potential. J Clin Invest. 2012;122:1138–1143. doi: 10.1172/JCI59954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ogino S, Goel A. Molecular classification and correlates in colorectal cancer. J Mol Diagn. 2008;10:13–27. doi: 10.2353/jmoldx.2008.070082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31:27–36. doi: 10.1093/carcin/bgp220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fearon ER. Molecular genetics of colorectal cancer. Annu Rev Pathol. 2011;6:479–507. doi: 10.1146/annurev-pathol-011110-130235. [DOI] [PubMed] [Google Scholar]
  • 6.Rajagopalan H, Bardelli A, Lengauer C, Kinzler KW, Vogelstein B, Velculescu VE. Tumorigenesis: RAF/RAS oncogenes and mismatch-repair status. Nature. 2002;418:934. doi: 10.1038/418934a. [DOI] [PubMed] [Google Scholar]
  • 7.Segditsas S, Tomlinson I. Colorectal cancer and genetic alterations in the Wnt pathway. Oncogene. 2006;25:7531–7537. doi: 10.1038/sj.onc.1210059. [DOI] [PubMed] [Google Scholar]
  • 8.Vousden KH, Prives C. Blinded by the Light: The Growing Complexity of p53. Cell. 2009;137:413–431. doi: 10.1016/j.cell.2009.04.037. [DOI] [PubMed] [Google Scholar]
  • 9.Lievre A, Bachet JB, Boige V, Cayre A, Le Corre D, Buc E, Ychou M, Bouche O, Landi B, Louvet C, Andre T, Bibeau F, Diebold MD, Rougier P, Ducreux M, Tomasic G, Emile JF, Penault-Llorca F, Laurent-Puig P. KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab. J. Clin. Oncol. 2008;26:374–379. doi: 10.1200/JCO.2007.12.5906. [DOI] [PubMed] [Google Scholar]
  • 10.Chen MB, Wu XY, Yu R, Li C, Wang LQ, Shen W, Lu PH. P53 status as a predictive biomarker for patients receiving neoadjuvant radiation-based treatment: a meta-analysis in rectal cancer. PLoS One. 2012;7:e45388. doi: 10.1371/journal.pone.0045388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Loupakis F, Cremolini C, Salvatore L, Masi G, Sensi E, Schirripa M, Michelucci A, Pfanner E, Brunetti I, Lupi C, Antoniotti C, Bergamo F, Lonardi S, Zagonel V, Simi P, Fontanini G, Falcone A. FOLFOXIRI plus bevacizumab as first-line treatment in BRAF mutant metastatic colorectal cancer. Eur J Cancer. 2014;50:57–63. doi: 10.1016/j.ejca.2013.08.024. [DOI] [PubMed] [Google Scholar]
  • 12.Harada S, Korf BR. Overview of molecular genetic diagnosis. Curr Protoc Hum Genet. 2013 doi: 10.1002/0471142905.hg0901s76. Chapter 9: Unit9 1. [DOI] [PubMed] [Google Scholar]
  • 13.IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Personal habits and indoor combustions. Volume 100 E. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum. 2012;100:1–538. [PMC free article] [PubMed] [Google Scholar]
  • 14.Hecht SS. Tobacco carcinogens, their biomarkers and tobacco-induced cancer. Nat Rev Cancer. 2003;3:733–744. doi: 10.1038/nrc1190. [DOI] [PubMed] [Google Scholar]
  • 15.Tomlinson I, Halford S, Aaltonen L, Hawkins N, Ward R. Does MSI-low exist? J Pathol. 2002;197:6–13. doi: 10.1002/path.1071. [DOI] [PubMed] [Google Scholar]
  • 16.DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–188. doi: 10.1016/0197-2456(86)90046-2. [DOI] [PubMed] [Google Scholar]
  • 17.Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst. 1959;22:719–748. [PubMed] [Google Scholar]
  • 18.Phipps AI, Shi Q, Newcomb PA, Nelson GD, Sargent DJ, Alberts SR, Limburg PJ. Associations between cigarette smoking status and colon cancer prognosis among participants in North Central Cancer Treatment Group Phase III Trial N0147. J. Clin. Oncol. 2013;31:2016–2023. doi: 10.1200/JCO.2012.46.2457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sinha R, Hussain S, Mehrotra R, Kumar RS, Kumar K, Pande P, Doval DC, Basir SF, Bharadwaj M. Kras gene mutation and RASSF1A, FHIT and MGMT gene promoter hypermethylation: indicators of tumor staging and metastasis in adenocarcinomatous sporadic colorectal cancer in Indian population. PLoS One. 2013;8:e60142. doi: 10.1371/journal.pone.0060142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Burnett-Hartman AN, Newcomb PA, Potter JD, Passarelli MN, Phipps AI, Wurscher MA, Grady WM, Zhu LC, Upton MP, Makar KW. Genomic Aberrations Occurring in Subsets of Serrated Colorectal Lesions but not Conventional Adenomas. Cancer Res. 2013;73:2863–2872. doi: 10.1158/0008-5472.CAN-12-3462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Samadder NJ, Vierkant RA, Tillmans LS, Wang AH, Lynch CF, Anderson KE, French AJ, Haile RW, Harnack LJ, Potter JD, Slager SL, Smyrk TC, Thibodeau SN, Cerhan JR, Limburg PJ. Cigarette smoking and colorectal cancer risk by KRAS mutation status among older women. Am J Gastroenterol. 2012;107:782–789. doi: 10.1038/ajg.2012.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Naghibalhossaini F, Zamani M, Mokarram P, Khalili I, Rasti M, Mostafavi-Pour Z. Epigenetic and genetic analysis of WNT signaling pathway in sporadic colorectal cancer patients from Iran. Mol Biol Rep. 2012;39:6171–6178. doi: 10.1007/s11033-011-1434-6. [DOI] [PubMed] [Google Scholar]
  • 23.Gay LJ, Mitrou PN, Keen J, Bowman R, Naguib A, Cooke J, Kuhnle GG, Burns PA, Luben R, Lentjes M, Khaw KT, Ball RY, Ibrahim AE, Arends MJ. Dietary, lifestyle and clinicopathological factors associated with APC mutations and promoter methylation in colorectal cancers from the EPIC-Norfolk study. J Pathol. 2012;228:405–415. doi: 10.1002/path.4085. [DOI] [PubMed] [Google Scholar]
  • 24.Gay LJ, Arends MJ, Mitrou PN, Bowman R, Ibrahim AE, Happerfield L, Luben R, McTaggart A, Ball RY, Rodwell SA. MLH1 promoter methylation, diet, and lifestyle factors in mismatch repair deficient colorectal cancer patients from EPIC-Norfolk. Nutr Cancer. 2011;63:1000–1010. doi: 10.1080/01635581.2011.596987. [DOI] [PubMed] [Google Scholar]
  • 25.Rozek LS, Herron CM, Greenson JK, Moreno V, Capella G, Rennert G, Gruber SB. Smoking, gender, and ethnicity predict somatic BRAF mutations in colorectal cancer. Cancer Epidemiol Biomarkers Prev. 2010;19:838–843. doi: 10.1158/1055-9965.EPI-09-1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Park JY, Mitrou PN, Keen J, Dahm CC, Gay LJ, Luben RN, McTaggart A, Khaw KT, Ball RY, Arends MJ, Rodwell SA. Lifestyle factors and p53 mutation patterns in colorectal cancer patients in the EPIC-Norfolk study. Mutagenesis. 2010;25:351–358. doi: 10.1093/mutage/geq012. [DOI] [PubMed] [Google Scholar]
  • 27.Limsui D, Vierkant RA, Tillmans LS, Wang AH, Weisenberger DJ, Laird PW, Lynch CF, Anderson KE, French AJ, Haile RW, Harnack LJ, Potter JD, Slager SL, Smyrk TC, Thibodeau SN, Cerhan JR, Limburg PJ. Cigarette smoking and colorectal cancer risk by molecularly defined subtypes. J Natl Cancer Inst. 2010;102:1012–1022. doi: 10.1093/jnci/djq201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Poynter JN, Haile RW, Siegmund KD, Campbell PT, Figueiredo JC, Limburg P, Young J, Le Marchand L, Potter JD, Cotterchio M, Casey G, Hopper JL, Jenkins MA, Thibodeau SN, Newcomb PA, Baron JA. Associations between smoking, alcohol consumption, and colorectal cancer, overall and by tumor microsatellite instability status. Cancer Epidemiol Biomarkers Prev. 2009;18:2745–2750. doi: 10.1158/1055-9965.EPI-09-0517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Curtin K, Samowitz WS, Wolff RK, Herrick J, Caan BJ, Slattery ML. Somatic alterations, metabolizing genes and smoking in rectal cancer. Int J Cancer. 2009;125:158–164. doi: 10.1002/ijc.24338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Weijenberg MP, Aardening PW, de Kok TM, de Goeij AF, van den Brandt PA. Cigarette smoking and KRAS oncogene mutations in sporadic colorectal cancer: results from the Netherlands Cohort Study. Mutat Res. 2008;652:54–64. doi: 10.1016/j.mrgentox.2007.12.008. [DOI] [PubMed] [Google Scholar]
  • 31.Samowitz WS, Albertsen H, Sweeney C, Herrick J, Caan BJ, Anderson KE, Wolff RK, Slattery ML. Association of smoking, CpG island methylator phenotype, and V600E BRAF mutations in colon cancer. J Natl Cancer Inst. 2006;98:1731–1738. doi: 10.1093/jnci/djj468. [DOI] [PubMed] [Google Scholar]
  • 32.Wark PA, Van der Kuil W, Ploemacher J, Van Muijen GN, Mulder CJ, Weijenberg MP, Kok FJ, Kampman E. Diet, lifestyle and risk of K-ras mutation-positive and -negative colorectal adenomas. Int J Cancer. 2006;119:398–405. doi: 10.1002/ijc.21839. [DOI] [PubMed] [Google Scholar]
  • 33.Sarebo M, Skjelbred CF, Breistein R, Lothe IM, Hagen PC, Bock G, Hansteen IL, Kure EH. Association between cigarette smoking, APC mutations and the risk of developing sporadic colorectal adenomas and carcinomas. BMC Cancer. 2006;6:71. doi: 10.1186/1471-2407-6-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Huang CC, Cheng YW, Chen MC, Lin YS, Chou MC, Lee H. Different p53 mutation patterns in colorectal tumors from smokers and nonsmokers. Environ Mol Mutagen. 2006;47:527–532. doi: 10.1002/em.20222. [DOI] [PubMed] [Google Scholar]
  • 35.Luchtenborg M, Weijenberg MP, Kampman E, Van Muijen GN, Roemen GMJM, Zeegers MPA, Goldbohm RA, Van’t Veer P, De Goeij AF, Van Den Brandt PA. Cigarette smoking and colorectal cancer: APC mutations, hMLH1 expression, and GSTM1 and GSTT1 polymorphisms. Am J Epidemiol. 2005;161:806–815. doi: 10.1093/aje/kwi114. [DOI] [PubMed] [Google Scholar]
  • 36.Diergaarde B, Vrieling A, van Kraats AA, van Muijen GN, Kok FJ, Kampman E. Cigarette smoking and genetic alterations in sporadic colon carcinomas. Carcinogenesis. 2003;24:565–571. doi: 10.1093/carcin/24.3.565. [DOI] [PubMed] [Google Scholar]
  • 37.Slattery ML, Curtin K, Ma K, Edwards S, Schaffer D, Anderson K, Samowitz W. Diet activity, and lifestyle associations with p53 mutations in colon tumors. Cancer Epidemiol Biomarkers Prev. 2002;11:541–548. [PubMed] [Google Scholar]
  • 38.Miyaki M, Iijima T, Ishii R, Kita Y, Koike M, Kuroki T, Mori T. Increased frequency of p53 mutation in sporadic colorectal cancer from cigarette smokers. Jpn J Clin Oncol. 2002;32:196–201. doi: 10.1093/jjco/hyf047. [DOI] [PubMed] [Google Scholar]
  • 39.Wu AH, Shibata D, Yu MC, Lai MY, Ross RK. Dietary heterocyclic amines and microsatellite instability in colon adenocarcinomas. Carcinogenesis. 2001;22:1681–1684. doi: 10.1093/carcin/22.10.1681. [DOI] [PubMed] [Google Scholar]
  • 40.Slattery ML, Anderson K, Curtin K, Ma KN, Schaffer D, Samowitz W. Dietary intake and microsatellite instability in colon tumors. Int J Cancer. 2001;93:601–607. doi: 10.1002/ijc.1370. [DOI] [PubMed] [Google Scholar]
  • 41.Slattery ML, Anderson K, Curtin K, Ma K, Schaffer D, Edwards S, Samowitz W. Lifestyle factors and Ki-ras mutations in colon cancer tumors. Mutat Res. 2001;483:73–81. doi: 10.1016/s0027-5107(01)00228-7. [DOI] [PubMed] [Google Scholar]
  • 42.Yang P, Cunningham JM, Halling KC, Lesnick TG, Burgart LJ, Wiegert EM, Christensen ER, Lindor NM, Katzmann JA, Thibodeau SN. Higher risk of mismatch repair-deficient colorectal cancer in alpha(1)-antitrypsin deficiency carriers and cigarette smokers. Mol Genet Metab. 2000;71:639–645. doi: 10.1006/mgme.2000.3089. [DOI] [PubMed] [Google Scholar]
  • 43.Lafuente MJ, Casterad X, Trias M, Ascaso C, Molina R, Ballesta A, Zheng S, Wiencke JK, Lafuente A. NAD(P)H:quinone oxidoreductase-dependent risk for colorectal cancer and its association with the presence of K-ras mutations in tumors. Carcinogenesis. 2000;21:1813–1819. doi: 10.1093/carcin/21.10.1813. [DOI] [PubMed] [Google Scholar]
  • 44.Martinez ME, Maltzman T, Marshall JR, Einspahr J, Reid ME, Sampliner R, Ahnen DJ, Hamilton SR, Alberts DS. Risk factors for Ki-ras protooncogene mutation in sporadic colorectal adenomas. Cancer Res. 1999;59:5181–5185. [PubMed] [Google Scholar]
  • 45.Rex DK, Ahnen DJ, Baron JA, Batts KP, Burke CA, Burt RW, Goldblum JR, Guillem JG, Kahi CJ, Kalady MF, O’Brien MJ, Odze RD, Ogino S, Parry S, Snover DC, Torlakovic EE, Wise PE, Young J, Church J. Serrated lesions of the colorectum: review and recommendations from an expert panel. Am J Gastroenterol. 2012;107:1315–1329. doi: 10.1038/ajg.2012.161. quiz 1314, 1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Porta M, Crous-Bou M, Wark PA, Vineis P, Real FX, Malats N, Kampman E. Cigarette smoking and K-ras mutations in pancreas, lung and colorectal adenocarcinomas: etiopathogenic similarities, differences and paradoxes. Mutat Res. 2009;682:83–93. doi: 10.1016/j.mrrev.2009.07.003. [DOI] [PubMed] [Google Scholar]
  • 47.Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology. 2008;135:1079–1099. doi: 10.1053/j.gastro.2008.07.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kuismanen SA, Holmberg MT, Salovaara R, de la Chapelle A, Peltomaki P. Genetic and epigenetic modification of MLH1 accounts for a major share of microsatellite-unstable colorectal cancers. Am J Pathol. 2000;156:1773–1779. doi: 10.1016/S0002-9440(10)65048-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Jass JR. Towards a molecular classification of colorectal cancer. Int J Colorectal Dis. 1999;14:194–200. doi: 10.1007/s003840050211. [DOI] [PubMed] [Google Scholar]
  • 50.Weisenberger DJ, Siegmund KD, Campan M, Young J, Long TI, Faasse MA, Kang GH, Widschwendter M, Weener D, Buchanan D, Koh H, Simms L, Barker M, Leggett B, Levine J, Kim M, French AJ, Thibodeau SN, Jass J, Haile R, Laird PW. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet. 2006;38:787–793. doi: 10.1038/ng1834. [DOI] [PubMed] [Google Scholar]
  • 51.Goel A, Boland CR. Epigenetics of colorectal cancer. Gastroenterology. 2012;143:1442–1460. e1. doi: 10.1053/j.gastro.2012.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kim DH, Nelson HH, Wiencke JK, Zheng S, Christiani DC, Wain JC, Mark EJ, Kelsey KT. p16(INK4a) and histology-specific methylation of CpG islands by exposure to tobacco smoke in non-small cell lung cancer. Cancer Res. 2001;61:3419–3424. [PubMed] [Google Scholar]
  • 53.Jarmalaite S, Kannio A, Anttila S, Lazutka JR, Husgafvel-Pursiainen K. Aberrant p16 promoter methylation in smokers and former smokers with nonsmall cell lung cancer. Int J Cancer. 2003;106:913–918. doi: 10.1002/ijc.11322. [DOI] [PubMed] [Google Scholar]
  • 54.Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE, Tu D, Redston M, Gallinger S. Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med. 2003;349:247–257. doi: 10.1056/NEJMoa022289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Min BH, Bae JM, Lee EJ, Yu HS, Kim YH, Chang DK, Kim HC, Park CK, Lee SH, Kim KM, Kang GH. The CpG island methylator phenotype may confer a survival benefit in patients with stage II or III colorectal carcinomas receiving fluoropyrimidine-based adjuvant chemotherapy. BMC Cancer. 2011;11:344. doi: 10.1186/1471-2407-11-344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A. 1999;96:8681–8686. doi: 10.1073/pnas.96.15.8681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Park SJ, Rashid A, Lee JH, Kim SG, Hamilton SR, Wu TT. Frequent CpG island methylation in serrated adenomas of the colorectum. Am J Pathol. 2003;162:815–822. doi: 10.1016/S0002-9440(10)63878-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lee S, Cho NY, Yoo EJ, Kim JH, Kang GH. CpG island methylator phenotype in colorectal cancers: comparison of the new and classic CpG island methylator phenotype marker panels. Arch Pathol Lab Med. 2008;132:1657–1665. doi: 10.5858/2008-132-1657-CIMPIC. [DOI] [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Medicine are provided here courtesy of e-Century Publishing Corporation

RESOURCES