Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 1.
Published in final edited form as: Pharmacogenet Genomics. 2015 Jan;25(1):30–37. doi: 10.1097/FPC.0000000000000101

Genetic variants within obesity-related genes are associated with tumor recurrence in patients with stages II/III colon cancer

Ana Sebio 1, Armin Gerger 1,3, Satoshi Matsusaka 1, Dongyun Yang 1, Wu Zhang 1, Stefan Stremitzer 1, Sebastian Stintizing 1, Yu Sunakawa 1, Shinichi Yamauchi 1, Yan Ning 1, Yoshiya Fujimoto 4, Masashi Ueno 4, Heinz-Josef Lenz 1,2
PMCID: PMC4260998  NIHMSID: NIHMS633658  PMID: 25379721

Abstract

Objective

Obesity is an established risk factor for colorectal cancer (CRC) incidence and it is also linked to CRC recurrence and survival. Polymorphisms located in obesity-related genes are associated with increased risk of developing several cancer types including colorectal cancer. We evaluated whether SNPs in obesity-related genes may predict tumor recurrence in colon cancer patients.

Methods

Genotypes were obtained from germline DNA from 207 patients with stage II or III colon cancer at the Norris Comprehensive Cancer Center. Nine polymorphisms in eight obesity-related genes (PPAR, LEP, NFKB, CD36, DRG1, NGAL, REGIA and DSCR1) were evaluated. The primary endpoint of the study was 3-year recurrence rate. Positive associations were also tested in an independent Japanese cohort of 350 stage III CRC patients.

Results

In univariate analysis, for PPAR rs1801282, patients with a CC genotype had significantly lower recurrence probability (29± 4% standard error, SE) compared to patients with a CG genotype (48% ± 8% SE), HR: 1.77; 95%CI, 1.01-3.10; p=0.040. For DSCR1 rs6517239, patients with an AA genotype had higher recurrence probability than patients carrying at least one allele G (37% ± 4% SE vs 15% ± 6% SE), HR: 0.51, 95% CI, 0.27-0.94; p=0.027. This association was stronger in the patients bearing a left-sided tumor (HR: 0.34; 95%CI, 0.13-0.88; p=0.018). In the Japanese cohort no associations were found.

Conclusion

This hypothesis generating study suggests a potential influence of polymorphisms within obesity-related genes in the recurrence probability of colon cancer. These interesting results should be further evaluated.

Keywords: colon cancer, polymorphisms, obesity, recurrence, adjuvant

Introduction

Recurrence of colon cancer after surgery occurs in approximately 30-45% of the patients with stages II and III [1]. Adjuvant 5-fluorouracil (5-FU) has demonstrated to reduce recurrence rates, and the addition of oxaliplatin to 5-FU can further diminish the risk of relapse in stage III colon cancer patients [2, 3]. Despite this reduction, the majority of patients are treated unnecessarily due to the lack of predictive factors to determine which patients will benefit from adjuvant treatment. Therefore, it is of high importance to identify biomarkers that will allow the selection of those patients benefiting from adjuvant treatment in order to avoid unnecessary toxicities and increased costs.

The mechanisms responsible for relapse after treatment are not completely understood. Many factors and processes such as dormancy, epithelial mesenchymal transition, immunosuppression and inflammation appear to impact on the probability of recurrence. Obesity has also been linked to colon cancer recurrence and survival as well as to colon cancer risk [4, 5]. Regarding recurrence and cancer specific survival, several studies have shown an increased risk of recurrence and a decrease in survival in obese patients. A study including 4,288 patients with Dukes B and C colon cancer reported an increased recurrence rate and shorter overall survival as well as colon cancer specific survival for patients with BMI ≥ 35 kg/m2 [6]. More recently, it was suggested that overweight pre-diagnosis of colon cancer but not post-diagnosis was associated with an increased risk of cancer specific mortality [7]. It has also been pointed out by Meyerhardt et al. that this risk may be gender-specific, as this group reported a significant increase in mortality among obese women with stages II and III colon cancer treated with 5-FU based adjuvant chemotherapy [8]. However, negative results regarding the relationship between obesity and cancer risk and survival have also been published. In a study that included more than 1,000 patients with stage III colon cancer enrolled in a randomized adjuvant chemotherapy trial, no differences were found regarding recurrence or death based on a high BMI index [9].

The rationale for the relationship between obesity and the increased risk of colorectal cancer, cancer recurrence and decreased survival is not fully elucidated. The biological mechanisms underlying this association include several processes such as inflammation, modulation of the energy balance, as well as growth factor and hormones signaling pathways, which influence tumorigenesis and cancer progression. Fat tissue is an endocrine organ that produces the polypeptide hormones adipokines. The adipokine leptin, which is directly associated with the amount of adipose tissue, has been reported to induce cancer progression by activation of PI3K, MAPK, I-CAM1 and STAT3 pathways [10, 11]. Specifically in colon cancer, leptin promotes motility and invasion by activating PI3K in colon cancer cell lines [12]. A higher percentage of adipose tissue is also associated with increased levels of interleukins and TNF-α, hence a chronic state of inflammation [13]. Inflammatory cytokines contribute to carcinogenesis through activation of the NF-κβ signaling cascade responsible for inducing expression of genes associated to cell proliferation, apoptosis, metastases and angiogenesis [14].

Interestingly, in normal weight population, fat tissue potentiates an anti-inflammatory environment through the nuclear transcription factors peroxisome proliferator activated receptors (PPAR). PPAR-gamma antagonizes inflammation through the inhibition of NF-κβ activity as well as the attenuation of expression of various cytokines involved in cancer promotion and progression such as IL-1, IL-8 and TNF-α [15, 16]. Particularly in colorectal cancer, PPAR-gamma regulates the transcription of several genes implicated in carcinogenesis such as developmentally-regulated GTP-binding protein (DRG-1), regenerating gene IA (REGIA), neutrophil gelatinase-associated lipocalin (NGAL) or down syndrome critical region 1 gene (DSCR1) [17-20].

Based on the lack of factors to predict colon cancer recurrence and the implication of obesity in the risk and prognosis of this cancer, the aim of this study was to investigate polymorphisms located in obesity-related genes as biomarkers to predict recurrence in stages II and III colon cancer.

Material and Methods

Study population

A total of 207 patients with high-risk stage II and stage III colon cancer treated with 5-FU based adjuvant chemotherapy at the Norris Comprehensive Cancer Center or at the Los Angeles County Hospital, University of Southern California (USC), Los Angeles, USA were analyzed. As an exploratory cohort, a total of 350 Japanese patients with stage III cancer treated with adjuvant FOLFOX at the Cancer Institute Hospital (CIH), Tokyo, Japan were evaluated. Baseline characteristics are shown in table 1.

Table 1.

Baseline characteristics and treatment of USC and Japanese cohorts

USC* (n=207) Japanese (n=350)
n % n %
Age, years
    <55 74 35.8 70 20.0
    55-64 75 36.2 101 28.9
    ≥65 58 28.0 179 51.1
Sex
    Female 96 46.4 175 50.0
    Male 111 53.6 175 50.0
T
    T1 2 1.0 0
    T2 12 5.8 5 1.4
    T3 164 79.2 232 66.3
    T4 25 12.1 113 32.3
    Tx 4 1.9
N
    Negative 97 46.9 0
    N1 61 29.5 229 65.4
    N2 49 23.7 121 34.6
Grade
    Well 11 5.3 75 21.4
    Moderate 132 63.8 236 67.4
    Poor/undifferentiated 47 22.7 24 6.9
    Missing 17 8.2 15 4.3
Stage
    II 97 46.9 0
    III 110 53.1 229 65.4
    IIIC 121 34.6
N of resected lymph nodes
    ≤12 60 29.0 47 13.5
    >12 129 62.3 302 86.5
    Missing 18 8.7 1
Tumor side
    Right 98 47.3 110 31.4
    Left 101 48.8 238 68.0
    Left and right 3 1.5 2 0.6
    Missing 5 2.4
Adjuvant treatment
    Fluoropyrimidines 130 62.8 206 58.9
    5-FU/LV/Oxaliplatin 58 28.0 68 19.4
    5-FU/LV/Irinotecan 19 9.2 0
    None 76 21.7
Ethnicity
    Asian 28 13.5 350 100.0
    African American 14 6.8
    Caucasian 110 53.1
    Hispanic 55 26.6

Follow up consisted of physical examination, blood test including carcinoembryonic antigen and CT scan every 3 months for the first two years and every 6 months thereafter. All patients participating in the study signed informed consent for the collection of samples as well as for the analysis of the molecular correlates. The present study was approved by the Institutional Review Boards of the University of Southern California and the Cancer Institute Hospital in Japan.

Selected polymorphisms and genotyping

Nine common polymorphisms located in eight obesity-related genes were selected. The polymorphisms selected had a reported functionality (NCBI Pubmed) or were considered potentially functional (F-SNP and SNP Info NIH databases). The minimum minor allele frequency (MAF) cut off for the selection was 0.1 for Caucasian population. Table 2 shows the characteristics of the selected polymorphisms.

Table 2.

Characteristics of the selected polymorphisms

GENE rs number Change/Position Function/Association MAF
NFκβ rs28362491 -/ATTG ins upstream Altered mRNA expression41 0.46
Leptin rs7799039 A>G upstream Influence on leptin expression42 0.50
CD36 rs3211882 -/TGT intron NA 0.41
DRG1 rs5749272 T>C downstream NA 0.47
NGAL rs12006030 G>C intron NA 0.2
PPARγ rs1801282 C>G Pro12Ala Reduced transactivation activity26 0.08
rs3856806 C>T His447His Higher leptin levels43 0.11
REG1A rs10165462 T>C 5’UTR NA 0.36
DSCR1 rs6517239 A>G intron NA 0.14

Abbreviations: MAF: minor allele frequency; NFκβ: nuclear factor κβ; DRG1: developmentally-regulated GTP-binding protein; NGAL: neutrophil gelatinase-associated lipocalin; PPARγ: peroxisome proliferator activated receptor gamma; REG1A: regenerating gene IA DSCR1: Down syndrome critical region 1.

DNA was extracted from peripheral blood in the majority of the patients. In cases where peripheral blood was not available, DNA was extracted from formalin-fixed, paraffin-embedded tissue. Most of the samples were tested using PCR- based restriction fragment length polymorphism (PCR-RFLP) analysis. In cases where no matching enzyme was found, samples were analyzed by direct DNA Sanger sequencing. To ensure quality of the results, 5% of the samples for each SNP analyzed by PCR-RFLP were re-tested by direct DNA sequencing.

Statistical analysis

The endpoints of the study were time to recurrence (TTR) and 3-year recurrence rate. TTR was calculated from the diagnosis (USC) or surgical (Japanese) date to the day of first documented recurrence. TTR was censored at the time of last follow-up or death if patients were relapse-free. With 207 patients available for analysis, this study had an 80% power to detect a minimum hazard ratio (HR) of 1.92-2.16 for TTR in the dominant model considering a MAF of 0.1-0.5 and 2.03-3.24 in the recessive model considering a MAF of 0.25-0.5. The association of the SNP with the time to recurrence (TTR) was also analyzed using Kaplan-Meier curves and log-rank test. For multivariable analysis a Cox regression analysis was fitted adjusting by stage, type of adjuvant chemotherapy and stratified by race.

χ2 test was used to analyze deviations from Hardy-Weinberg equilibrium in each ethnic group. Chi-square test was used to analyze the polymorphisms distribution as well as the baseline patients’ characteristics. The inheritance model of these polymorphisms is not known; therefore we assumed a co-dominant, additive, dominant or recessive model wherever appropriate.

SAS 9.4 (SAS Institute, Cary, NC, USA) was used to perform all the analyses. Case-wise deletion was applied when patients with missing SNPs were excluded in the analyses. All tests were 2-sided at a significance level of 0.05. P values were not adjusted for multiple hypothesis testing.

Results

The median follow-up of this series was 3.9 years (range from 0.4 to 16.8). During this follow-up period, ninety patients (38.4%) presented tumor recurrence and the probability of 3-year recurrence was 33% (± 4% SE). The median overall survival of this series has not been reached. In the Japanese cohort, the median follow up was 5.0 years (range 0.3-8.6) and the 3-year recurrence probability was 29% (± 2% SE).

Genotyping was successful in > 90% of the analyzed samples for each SNP. Quality control re-sampling with DNA Sanger sequencing yielded a concordance of >99%. All the studied polymorphisms but two (rs28362491 and rs12006030) were within the probability limits of the Hardy-Weinberg equilibrium.

Polymorphisms in obesity-related genes and their association with recurrence

Detailed information of the studied SNPs with the 3-year recurrence probability is shown in Table 3.

Table 3.

Association of the analyzed SNPs and the 3-year recurrence probability

Polymorphism N Median TTR (95%CI) 3-year recurrence probability ± SE* HR (95%CI) Univariate P value HR (95%CI) Multivariate P value
NF-κβ rs28362491 0.45 0.55
ATTG/ATTG 64 7.1 (2.7, 11.3+) 0.36±0.06 1 (Reference) 1 (Reference)
-/ATTG 82 12.4+ (4.9, 12.4+) 0.31±0.06 0.71 (0.41, 1.23) 0.77 (0.44, 1.36)
-/- 49 5.2 (3.5, 16.8+) 0.29±0.07 0.91 (0.51, 1.64) 1.05 (0.57, 1.93)
Leptin rs7799039 0.38 0.12
A/A 48 4.9 (3.4, 10.6+) 0.28±0.07 1 (Reference) 1 (Reference)
A/G 93 10.7 (2.7, 16.8+) 0.38±0.06 0.91 (0.52, 1.60) 0.78 (0.43, 1.44)
G/G 47 9.4 (5.4, 12.2+) 0.22±0.07 0.63 (0.32, 1.25) 0.45 (0.20, 0.98)
CD36 rs3211882 0.69 0.87
TGT/TGT 67 5.7 (2.7, 10.7+) 0.38±0.07 1 (Reference) 1 (Reference)
-/TGT 94 10.7 (4.9, 16.8+) 0.30±0.05 0.81 (0.48, 1.36) 0.91 (0.53, 1.57)
-/- 31 9.4 (3.2, 11.3+) 0.25±0.08 0.81 (0.39, 1.70) 0.81 (0.38, 1.76)
DRG1 rs5749272 0.33 0.28
T/T 58 5.4 (2.7, 9.0+) 0.38±0.07 1 (Reference) 1 (Reference)
T/C 79 10.7 (4.8, 11.3+) 0.28±0.05 0.66 (0.37, 1.16) 0.62 (0.35, 1.12)
C/C 48 6.6 (2.7, 16.8+) 0.36±0.08 0.85 (0.46, 1.58) 0.83 (0.44, 1.59)
NGAL rs12006030 0.37 0.59
C/C 121 9.4 (5.7, 16.8+) 0.29±0.05 1 (Reference) 1 (Reference)
C/G 79 5.4 (3.4, 10.9+) 0.35±0.06 1.24 (0.77, 1.98) 1.15 (0.70, 1.90)
PPARrs1801282 0.040 0.080
C/C 168 9.4 (5.7, 12.4+) 0.29±0.04 1 (Reference) 1 (Reference)
C/G 34 3.4 (1.5, 16.8+) 0.48±0.10 1.77 (1.01, 3.10) 1.68 (0.94, 2.99)
PPAR rs3856806
C/C 159 0.83 0.76
C/T 39 6.6 (4.9, 12.4+) 0.32±0.04 1 (Reference) 1 (Reference)
T/T 2 7.1 (2.7, 16.8+) 0.34±0.08 0.94 (0.52, 1.68) 1.10 (0.59, 2.06)
REGIA rs10165462 0.27 0.21
T/T 77 6.6 (3.5, 12.2+) 0.35±0.06 1 (Reference) 1 (Reference)
T/C 100 10.7 (4.9, 16.8+) 0.27±0.05 0.73 (0.44, 1.19) 0.71 (0.43, 1.18)
C/C 21 2.8 (1.4, 6.3+) 0.55±0.15 1.26 (0.55, 2.88) 1.34 (0.56, 3.18)
DSCR1 rs6517239 0.027 0.058
A/A 149 6.6 (4.0, 16.8+) 0.37±0.04 1 (Reference) 1 (Reference)
A/G 47 9.4 (5.7, 10.7+) 0.15±0.06 0.51 (0.27, 0.94) 0.54 (0.29, 1.02)
G/G 4
*

Greenwood SE.

+

Estimates were not reached.

Based on log-rank test in the univariate analysis and based on Wald test within multivariate Cox proportional hazards model adjusting for stage and type of adjuvant therapy and stratified by race.

In the dominant model.

Abbreviations: NF-κβ: nuclear factor kappa-light-chain enhancer of activated B cells; DRG1: developmentally regulated GTP-binding protein; NGAL: neutrophile gelatinase-associated lipocalin; PPAR: peroxisome proliferator activated receptor; REGIA: regenerating gene IA; DSCR1: Down syndrome critical region 1.

PARPγ rs1801282 correlated with the 3-year recurrence probability: patients with a CC genotype had a significantly lower recurrence probability of 29% (±4% standard error, SE) compared to 48% (±8% SE) for patients with a CG genotype with a hazard ratio (HR) of 1.77; 95%CI, 1.01-3.10; p=0.040. In multivariate analysis, this association did not retain significance after adjusting for the relevant clinical parameters (HR: 1.68; 95% CI, 0.94-2.99; p= 0.08). The MAF for this polymorphism in Asian population is only 4%, therefore it was not analyzed in the Japanese cohort. The MAF for this SNP across race groups in the USC population varied significantly (p=0.023), being the variant allele more frequent in among the Hispanic population (data not shown).

DSCR1 rs6517239 was also associated with the 3-year recurrence probability in the dominant model: patients with a homozygous wild-type genotype (AA) had higher 3-year recurrence rate than patients carrying at least one variant allele G (37% ±4% vs 15% ±6%, respectively; HR: 0.51, 95% CI, 0.27-0.94; p=0.027). In multivariate analysis, this polymorphism had a trend towards an association with the recurrence probability (HR: 0.54; 95% CI, 0.29-1.03; p= 0.058).

In the Japanese cohort, the evaluation of DSCR1 rs6517239 and the 3-year recurrence probability did not show any association (p=0.53).

Subgroup analysis by gender and tumor location

Detailed information of these analyses is shown in Table 4. Evaluation of the selected polymorphisms by tumor location demonstrated a stronger association of the DSCR1 rs6517239 polymorphism with the 3-year recurrence probability. In patients bearing left-sided tumors, those with an AA genotype had a 3-year recurrence probability of 44% (±6% SE) compared to patients with an AG or GG genotypes whose probability of recurrence at 3-years was 11% (±8% SE) (HR: 0.34; 95%CI, 0.13-0.88; p=0.018). This difference retained statistical significance in multivariate analysis (HR: 0.36; 95%CI, 0.14-0.96; p= 0.040). Subgroup analyses in the Japanese cohort for the DSCR1 rs6517239 did not yield any association.

Table 4.

Subgroup analyses. Association of the analyzed SNPs and the 3-year recurrence probability according to tumor location and gender.

Polymorphism N Median TTR (95%CI) 3-year recurrence probability ± SE* HR (95%CI) Univariate P value HR (95%CI) Multivariate P value
LEFT-SIDED TUMORS
NFKBrs28362491 0.34 0.40
ATTG/ATTG 30 1.8 (1.4, 10.7+) 0.52±0.10 1 (Reference) 1 (Reference)
-/ATTG 35 6.6 (2.4, 9.0+) 0.34±0.09 0.59 (0.28, 1.26) 0.60 (0.26, 1.35)
-/- 30 5.2 (3.2, 16.8+) 0.25±0.08 0.65 (0.30, 1.45) 0.64 (0.28, 1.46)
Leptinrs7799039 0.57 0.20
    A/A 28 3.5 (1.5, 7.1) 0.38±0.10 1 (Reference) 1 (Reference)
    A/G 42 16.8+ (2.7, 16.8+) 0.35±0.09 0.68 (0.32, 1.45) 0.53 (0.23, 1.25)
    G/G 21 6.6 (1.6, 8.0+) 0.37±0.11 0.73 (0.30, 1.73) 0.41 (0.14, 1.21)
CD36rs3211882 0.47 0.42
TGT/TGT 34 7.1 (2.3, 10.7+) 0.33±0.09 1 (Reference) 1 (Reference)
-/TGT 39 4.9 (1.5, 16.8+) 0.46±0.09 1.46 (0.71, 3.00) 1.53 (0.73, 3.19)
-/- 21 6.7+ (1.5, 6.7+) 0.26±0.10 0.96 (0.36, 2.57) 0.94 (0.34, 2.58)
DRG1rs5749272 0.53 0.75
    T/T 33 7.1 (1.8, 9.0+) 0.40±0.10 1 (Reference) 1 (Reference)
    T/C 35 7.2+ (3.5, 7.2+) 0.29±0.08 0.80 (0.36, 1.75) 0.75 (0.33, 1.69)
    C/C 26 5.2 (1.4, 16.8+) 0.47±0.11 1.25 (0.57, 2.75) 0.97 (0.42, 2.26)
NGALrs12006030 0.21 0.51
    C/C 64 7.1 (3.5, 16.8+) 0.32±0.07 1 (Reference) 1 (Reference)
    C/G 33 4.9 (1.6, 7.9+) 0.44±0.09 1.51 (0.79, 2.89) 1.26 (0.63, 2.53)
PPARrs1801282 0.26 0.085
    C/C 78 6.6 (3.5, 10.7+) 0.33±0.06 1 (Reference) 1 (Reference)
    C/G 19 2.7 (1.5, 16.8+) 0.52±0.12 1.51 (0.73, 3.10) 2.05 (0.91, 4.65)
PPARrs3856806 0.89 0.20
    C/C 76 5.7 (3.2, 10.7+) 0.36±0.06 1 (Reference) 1 (Reference)
    C/T, T/T 21 4.0 (1.5, 16.8+) 0.40±0.12 1.05 (0.50, 2.22) 1.84 (0.73, 4.64)
REGIArs10165462 0.18 0.20
    T/T 34 3.5 (1.8, 7.1) 0.42±0.10 1 (Reference) 1 (Reference)
    T/C 52 16.8+ (4.0, 16.8+) 0.31±0.07 0.60 (0.30, 1.18) 0.64 (0.31, 1.32)
    C/C 10 1.6 (0.5, 6.3+) 0.58±0.21 1.28 (0.42, 3.93) 1.76 (0.50, 6.20)
DSCR1rs6517239 0.018 0.040
    A/A 73 4.0 (1.8, 16.8+) 0.44±0.06 1 (Reference) 1 (Reference)
    A/G,G/G 24 10.7+ (5.2, 10.7+) 0.11±0.08 0.34 (0.13, 0.88) 0.36 (0.14, 0.96)
FEMALE POPULATION
NFKBrs28362491 0.36 0.48
ATTG/ATTG 27 9.4 (1.5, 9.4) 0.39±0.10 1 (Reference) 1 (Reference)
-/ATTG 40 12.2+ (4.9, 12.2+) 0.24±0.07 0.61 (0.27, 1.39) 0.67 (0.29, 1.57)
-/- 22 5.4 (1.4, 16.8+) 0.38±0.11 1.04 (0.44, 2.45) 1.13 (0.46, 2.78)
Leptinrs7799039 0.24 0.13
    A/A 23 4.9 (1.5, 7.7+) 0.37±0.10 1 (Reference) 1 (Reference)
    A/G 45 5.9 (2.4, 16.8+) 0.40±0.08 1.01 (0.46, 2.21) 0.88 (0.38, 2.04)
    G/G 22 12.2+ (4.8, 12.2+) 0.15±0.08 0.49 (0.17, 1.37) 0.35 (0.11, 1.06)
CD36rs3211882 0.40 0.71
TGT/TGT 26 5.7 (1.8, 7.7+) 0.37±0.11 1 (Reference) 1 (Reference)
-/TGT 51 16.8+ (4.8, 16.8+) 0.30±0.07 0.74 (0.33, 1.68) 1.01 (0.43, 2.40)
-/- 10 5.9 (0.1, 9.4) 0.31±0.15 1.36 (0.48, 3.89) 1.51 (0.49, 4.66)
DRG1rs5749272 0.59 0.42
    T/T 30 5.9 (2.1, 9.0+) 0.34±0.09 1 (Reference) 1 (Reference)
    T/C 35 10.9+ (4.8, 10.9+) 0.26±0.08 0.75 (0.33, 1.74) 0.62 (0.25, 1.50)
    C/C 21 16.8+ (1.4, 16.8+) 0.48±0.12 1.19 (0.50, 2.84) 1.09 (0.43, 2.75)
NGALrs12006030 0.96 0.54
    C/C 59 9.4 (2.8, 16.8+) 0.36±0.07 1 (Reference) 1 (Reference)
    C/G 35 5.9 (4.8, 10.9+) 0.26±0.08 0.98 (0.49, 1.97) 1.27 (0.60, 2.69)
PPARrs1801282 0.022 0.11
    C/C 77 9.4 (5.4, 10.9+) 0.27±0.05 1 (Reference) 1 (Reference)
    C/G 17 2.4 (0.6, 16.8+) 0.64±0.13 2.29 (1.09, 4.82) 1.94 (0.87, 4.34)
PPARrs3856806 0.85 0.99
    C/C 75 5.9 (4.9, 10.9+) 0.30±0.06 1 (Reference) 1 (Reference)
    C/T, T/T 20 16.8+ (1.1, 16.8+) 0.43±0.12 1.08 (0.49, 2.39) 1.00 (0.44, 2.28)
REGIArs10165462 0.64 0.82
    T/T 34 5.9 (1.8, 12.2+) 0.35±0.09 1 (Reference) 1 (Reference)
    T/C 50 16.8+ (4.8, 16.8+) 0.30±0.07 0.76 (0.38, 1.55) 0.88 (0.39, 2.00)
    C/C 8 2.8 (0.5, 4.9+) 0.67±0.25 1.23 (0.35, 4.35) 1.34 (0.36, 5.01)
DSCR1rs6517239 0.079 0.053
    A/A 71 5.4 (2.8, 16.8+) 0.39±0.06 1 (Reference) 1 (Reference)
    A/G,G/G 23 9.4 (5.7, 10.3+) 0.16±0.09 0.44 (0.17, 1.13) 0.37 (0.14, 1.01)
*

Greenwood SE.

+

Estimates were not reached.

Based on log-rank test in the univariate analysis and based on Wald test within multivariate Cox proportional hazards model adjusting for stage and type of adjuvant therapy and stratified by race.

In the dominant model.

Analysis by gender revealed that the association of PARPγ rs1801282 with the 3-year recurrence was due mainly to the female population. Females carrying a CC genotype had lower 3-year recurrence rate (27% ±5% SE) compared to females with a CG genotype (64% ±13% SE) with a HR: 2.29; 95%CI, 1.09-4.82; p= 0.022, whereas no difference was found in the male population subgroup (p=0.53). However, in multivariate analysis this difference did not remain significant (HR: 1.94; 95% CI, 0.87-4.34; p= 0.11).

Discussion

In the present work, polymorphisms within obesity-related genes were associated with the recurrence probability in high-risk stage II and stage III resected colon cancer patients treated with 5-FU-based adjuvant chemotherapy. Presence of at least one variant allele of PPARγ rs1801282, correlated with a higher 3-year recurrence rate, whereas the presence of a least one variant allele of DSCR1 rs6517239 correlated with a lower 3-year recurrence rate. These associations were more pronounced in the female population for PPARγ rs1801282 and in the patients with left-sided tumors for DSCR1 rs6517239.

PPARγ is a critical regulator of metabolism acting through transcriptional factors in adipose tissue. Additionally, PPARγ also acts as a tumor suppressor by binding with retinoid X receptor α and signaling antiangiogenic and antiproliferative pathways [21]. Particularly in colorectal cancer, in vitro data suggest that a perturbation of the correct function of PPARγ promotes colorectal cancer carcinogenesis. Colorectal cancer cell lines treated with PPARγ agonists induce differentiation and growth arrest through PTEN activation and cyclin D suppression [22-24]. Moreover, β-catenin, a highly important protein for colorectal carcinogenesis part of the Wnt signaling, is downregulated by PPARγ [25]. In the PPARγ gene, the variant allele of the PPARγ rs1801282 C>G polymorphism correlates with reduced PPARγ transactivation activity [26]. These in vitro data are consistent with our results as patients bearing a G allele had significantly higher recurrence probability, which suggests a reduction in PPARγ’s tumor suppressor activity for patients harboring the variant allele. This association was stronger in the female population. This fact underlines the gender differences in colorectal cancer and the incompletely clarified relationship between estrogen, obesity and colorectal cancer [27, 28]. This association did not retain significance in the multivariate analysis. The differences in the MAF among the races in the USC population may have influenced these results. Therefore, further analyses in more homogeneous populations are needed to clarify the value of this SNP. To date, no previous works had evaluated this polymorphism as a recurrence biomarker in cancer patients. Nonetheless, epidemiological studies evaluating cancer risk have yielded inconsistent results for PPARγ rs1801282 as it has been associated with a reduced risk of gastric cancer but an increased risk of colorectal cancer [29].

DSCR1 is one of the genes regulated by PPARγ [20]. DSCR1 codifies for calcipressin, a protein able to modulate the calcineurin-NFAT pathway [30]. The calcineurin-NFAT signaling has important roles in physiological processes such as regulating glucagon synthesis in pancreatic cells [31]. Additionally, DSCR1 has also been shown to be involved in many pathological conditions such as Alzheimer’s disease and Down Syndrome [32]. DSCR1 might also play a critical role in cancer development. Epidemiological studies have underlined that Down syndrome’s individuals develop significantly less solid tumors than healthy controls [33]. Presumably, genes located in chromosome 21 are responsible for this fact. DSCR1 is one of the genes located in chromosome 21 and its increased expression due to an extra copy is able to suppress angiogenesis and tumor growth through inhibition of the calcineurin-NFAT signaling [34]. Moreover, DSCR1 expression is induced by VEGF representing a potential angiogenesis regulation mechanism [35]. In this study, the rs6517239 polymorphism within DSCR1 was associated with the recurrence probability and this association was stronger in patients with left side tumors. Growing evidence indicates that left and right side colorectal cancer tumors are different molecular entities [36, 37]. Left-side tumors rely more in growth factor pathways to develop and progress, which may explain the stronger association of DSCR1 rs6517239 in patients with left-sided tumors [38]. Although no functionality has been described for this polymorphism, in silico analysis revealed this SNP affects a DNA binding site [39, 40]. This association was not validated in the Japanese series, despite of the similar minor allele frequencies for this SNP between the cohorts, which might be explained by several reasons. First, the differences in the initial stage that included stages II and III in the American cohort vs only stage III in the Japanese cohort. This fact influenced the adjuvant treatment that was mainly 5-FU monotherapy for the American series compared to FOLFOX in the Japanese. Second, ethnical background might also have played a role due to different obesity rates in these populations, which is explained not only by life style but also by clear genetic differences.

Overall, study provides the first insight on the potential implication of germline variants within obesity-related genes on colon cancer recurrence after curative treatment. Moreover, these data underlines the potential influence of tumor location in the value of these polymorphisms as biomarkers. However, the lack of a more similar validation cohort and adjustment for multiple testing makes this a hypothesis generating study. Although these results are not statistically robust, we believe that these polymorphisms deserve further evaluation in other populations with bigger sample size and in prospective clinical trials.

Supplementary Material

Cover letter

Acknowledgements

The project described was supported in part by award number P30CA014089 from the National Cancer Institute. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institutes of Health.

During the time this work was performed, Ana Sebio was a recipient of a Rio Hortega Contract (CM11/00102) from the Instituto de Salud Carlos III, Spain, and Sebastian Stintzing was a postdoctoral fellow of the German Cancer Aid (Mildred-Scheel-Foundation). Stefan Stremitzer is a recipient of an Erwin Schrödinger Fellowship Grant from the Austrian Science Fund.

Bibliography

  • 1.Edge SBBDRC,CC, Fritz AG, Greene FL, Trotti A. AJCC Cancer Staging Manual. ed 7th Edition Springer; New York: 2010. [Google Scholar]
  • 2.Poplin EA, Benedetti JK, Estes NC, Haller DG, Mayer RJ, Goldberg RM, et al. Phase III Southwest Oncology Group 9415/Intergroup 0153 randomized trial of fluorouracil, leucovorin, and levamisole versus fluorouracil continuous infusion and levamisole for adjuvant treatment of stage III and high-risk stage II colon cancer. J Clin Oncol. 2005;23(9):1819–25. doi: 10.1200/JCO.2005.04.169. [DOI] [PubMed] [Google Scholar]
  • 3.Andre T, Boni C, Navarro M, Tabernero J, Hickish T, Topham C, et al. Improved overall survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III colon cancer in the MOSAIC trial. J Clin Oncol. 2009;27(19):3109–16. doi: 10.1200/JCO.2008.20.6771. [DOI] [PubMed] [Google Scholar]
  • 4.Adams KF, Leitzmann MF, Albanes D, Kipnis V, Mouw T, Hollenbeck A, et al. Body mass and colorectal cancer risk in the NIH-AARP cohort. American journal of epidemiology. 2007;166(1):36–45. doi: 10.1093/aje/kwm049. [DOI] [PubMed] [Google Scholar]
  • 5.Calle EE, Rodriguez C, Walker-Thurmond K, Thun MJ. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 2003;348(17):1625–38. doi: 10.1056/NEJMoa021423. [DOI] [PubMed] [Google Scholar]
  • 6.Dignam JJ, Polite BN, Yothers G, Raich P, Colangelo L, O'Connell MJ, et al. Body mass index and outcomes in patients who receive adjuvant chemotherapy for colon cancer. J Natl Cancer Inst. 2006;98(22):1647–54. doi: 10.1093/jnci/djj442. [DOI] [PubMed] [Google Scholar]
  • 7.Campbell PT, Newton CC, Dehal AN, Jacobs EJ, Patel AV, Gapstur SM. Impact of body mass index on survival after colorectal cancer diagnosis: the Cancer Prevention Study-II Nutrition Cohort. J Clin Oncol. 2012;30(1):42–52. doi: 10.1200/JCO.2011.38.0287. [DOI] [PubMed] [Google Scholar]
  • 8.Meyerhardt JA, Catalano PJ, Haller DG, Mayer RJ, Benson AB, 3rd, Macdonald JS, et al. Influence of body mass index on outcomes and treatment-related toxicity in patients with colon carcinoma. Cancer. 2003;98(3):484–95. doi: 10.1002/cncr.11544. [DOI] [PubMed] [Google Scholar]
  • 9.Meyerhardt JA, Niedzwiecki D, Hollis D, Saltz LB, Mayer RJ, Nelson H, et al. Impact of body mass index and weight change after treatment on cancer recurrence and survival in patients with stage III colon cancer: findings from Cancer and Leukemia Group B 89803. J Clin Oncol. 2008;26(25):4109–15. doi: 10.1200/JCO.2007.15.6687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gao J, Tian J, Lv Y, Shi F, Kong F, Shi H, et al. Leptin induces functional activation of cyclooxygenase-2 through JAK2/STAT3, MAPK/ERK, and PI3K/AKT pathways in human endometrial cancer cells. Cancer Sci. 2009;100(3):389–95. doi: 10.1111/j.1349-7006.2008.01053.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dong Z, Fu S, Xu X, Yang Y, Du L, Li W, et al. Leptin-mediated regulation of ICAM-1 is Rho/ROCK dependent and enhances gastric cancer cell migration. Br J Cancer. 2014;110(7):1801–10. doi: 10.1038/bjc.2014.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jaffe T, Schwartz B. Leptin promotes motility and invasiveness in human colon cancer cells by activating multiple signal-transduction pathways. Int J Cancer. 2008;123(11):2543–56. doi: 10.1002/ijc.23821. [DOI] [PubMed] [Google Scholar]
  • 13.Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell. 2010;140(2):197–208. doi: 10.1016/j.cell.2009.12.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Vaiopoulos AG, Athanasoula K, Papavassiliou AG. NF-kappaB in colorectal cancer. Journal of molecular medicine. 2013;91(9):1029–37. doi: 10.1007/s00109-013-1045-x. [DOI] [PubMed] [Google Scholar]
  • 15.Chung SW, Kang BY, Kim SH, Pak YK, Cho D, Trinchieri G, et al. Oxidized low density lipoprotein inhibits interleukin-12 production in lipopolysaccharide-activated mouse macrophages via direct interactions between peroxisome proliferator-activated receptor-gamma and nuclear factor-kappa B. J Biol Chem. 2000;275(42):32681–7. doi: 10.1074/jbc.M002577200. [DOI] [PubMed] [Google Scholar]
  • 16.Cortez M, Carmo LS, Rogero MM, Borelli P, Fock RA. A high-fat diet increases IL-1, IL-6, and TNF-alpha production by increasing NF-kappaB and attenuating PPAR-gamma expression in bone marrow mesenchymal stem cells. Inflammation. 2013;36(2):379–86. doi: 10.1007/s10753-012-9557-z. [DOI] [PubMed] [Google Scholar]
  • 17.Guan RJ, Ford HL, Fu Y, Li Y, Shaw LM, Pardee AB. Drg-1 as a differentiation-related, putative metastatic suppressor gene in human colon cancer. Cancer Res. 2000;60(3):749–55. [PubMed] [Google Scholar]
  • 18.Rechreche H, Montalto G, Mallo GV, Vasseur S, Marasa L, Soubeyran P, et al. pap, reg Ialpha and reg Ibeta mRNAs are concomitantly up-regulated during human colorectal carcinogenesis. Int J Cancer. 1999;81(5):688–94. doi: 10.1002/(sici)1097-0215(19990531)81:5<688::aid-ijc3>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 19.Nielsen BS, Borregaard N, Bundgaard JR, Timshel S, Sehested M, Kjeldsen L. Induction of NGAL synthesis in epithelial cells of human colorectal neoplasia and inflammatory bowel diseases. Gut. 1996;38(3):414–20. doi: 10.1136/gut.38.3.414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bush CR, Havens JM, Necela BM, Su W, Chen L, Yanagisawa M, et al. Functional genomic analysis reveals cross-talk between peroxisome proliferator-activated receptor gamma and calcium signaling in human colorectal cancer cells. J Biol Chem. 2007;282(32):23387–401. doi: 10.1074/jbc.M702708200. [DOI] [PubMed] [Google Scholar]
  • 21.Ondrey F. Peroxisome proliferator-activated receptor gamma pathway targeting in carcinogenesis: implications for chemoprevention. Clin Cancer Res. 2009;15(1):2–8. doi: 10.1158/1078-0432.CCR-08-0326. [DOI] [PubMed] [Google Scholar]
  • 22.Kato M, Kusumi T, Tsuchida S, Tanaka M, Sasaki M, Kudo H. Induction of differentiation and peroxisome proliferator-activated receptor gamma expression in colon cancer cell lines by troglitazone. J Cancer Res Clin Oncol. 2004;130(2):73–9. doi: 10.1007/s00432-003-0510-2. [DOI] [PubMed] [Google Scholar]
  • 23.Patel L, Pass I, Coxon P, Downes CP, Smith SA, Macphee CH. Tumor suppressor and anti-inflammatory actions of PPARgamma agonists are mediated via upregulation of PTEN. Curr Biol. 2001;11(10):764–8. doi: 10.1016/s0960-9822(01)00225-1. [DOI] [PubMed] [Google Scholar]
  • 24.Shao J, Sheng H, DuBois RN. Peroxisome proliferator-activated receptors modulate K-Ras-mediated transformation of intestinal epithelial cells. Cancer Res. 2002;62(11):3282–8. [PubMed] [Google Scholar]
  • 25.Sharma C, Pradeep A, Wong L, Rana A, Rana B. Peroxisome proliferator-activated receptor gamma activation can regulate beta-catenin levels via a proteasome-mediated and adenomatous polyposis coli-independent pathway. J Biol Chem. 2004;279(34):35583–94. doi: 10.1074/jbc.M403143200. [DOI] [PubMed] [Google Scholar]
  • 26.Deeb SS, Fajas L, Nemoto M, Pihlajamaki J, Mykkanen L, Kuusisto J, et al. A Pro12Ala substitution in PPARgamma2 associated with decreased receptor activity, lower body mass index and improved insulin sensitivity. Nat Genet. 1998;20(3):284–7. doi: 10.1038/3099. [DOI] [PubMed] [Google Scholar]
  • 27.Chen J, Iverson D. Estrogen in obesity-associated colon cancer: friend or foe? Protecting postmenopausal women but promoting late-stage colon cancer. Cancer causes & control : CCC. 2012;23(11):1767–73. doi: 10.1007/s10552-012-0066-z. [DOI] [PubMed] [Google Scholar]
  • 28.Barzi A, Lenz AM, Labonte MJ, Lenz HJ. Molecular pathways: Estrogen pathway in colorectal cancer. Clin Cancer Res. 2013;19(21):5842–8. doi: 10.1158/1078-0432.CCR-13-0325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Xu W, Li Y, Wang X, Chen B, Liu S, Wang Y, et al. PPARgamma polymorphisms and cancer risk: a meta-analysis involving 32,138 subjects. Oncology reports. 2010;24(2):579–85. [PubMed] [Google Scholar]
  • 30.Fuentes JJ, Genesca L, Kingsbury TJ, Cunningham KW, Perez-Riba M, Estivill X, et al. DSCR1, overexpressed in Down syndrome, is an inhibitor of calcineurin-mediated signaling pathways. Hum Mol Genet. 2000;9(11):1681–90. doi: 10.1093/hmg/9.11.1681. [DOI] [PubMed] [Google Scholar]
  • 31.Furstenau U, Schwaninger M, Blume R, Jendrusch EM, Knepel W. Characterization of a novel calcium response element in the glucagon gene. J Biol Chem. 1999;274(9):5851–60. doi: 10.1074/jbc.274.9.5851. [DOI] [PubMed] [Google Scholar]
  • 32.Harris CD, Ermak G, Davies KJ. Multiple roles of the DSCR1 (Adapt78 or RCAN1) gene and its protein product calcipressin 1 (or RCAN1) in disease. Cell Mol Life Sci. 2005;62(21):2477–86. doi: 10.1007/s00018-005-5085-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yang Q, Rasmussen SA, Friedman JM. Mortality associated with Down's syndrome in the USA from 1983 to 1997: a population-based study. Lancet. 2002;359(9311):1019–25. doi: 10.1016/s0140-6736(02)08092-3. [DOI] [PubMed] [Google Scholar]
  • 34.Baek KH, Zaslavsky A, Lynch RC, Britt C, Okada Y, Siarey RJ, et al. Down's syndrome suppression of tumour growth and the role of the calcineurin inhibitor DSCR1. Nature. 2009;459(7250):1126–30. doi: 10.1038/nature08062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yao YG, Duh EJ. VEGF selectively induces Down syndrome critical region 1 gene expression in endothelial cells: a mechanism for feedback regulation of angiogenesis? Biochem Biophys Res Commun. 2004;321(3):648–56. doi: 10.1016/j.bbrc.2004.06.176. [DOI] [PubMed] [Google Scholar]
  • 36.Bauer KM, Hummon AB, Buechler S. Right-side and left-side colon cancer follow different pathways to relapse. Mol Carcinog. 2012;51(5):411–21. doi: 10.1002/mc.20804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhu H, Wu TC, Chen WQ, Zhou LJ, Wu Y, Zeng L, et al. Screening for differentially expressed genes between left- and right-sided colon carcinoma by microarray analysis. Oncology letters. 2013;6(2):353–8. doi: 10.3892/ol.2013.1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bendardaf R, Buhmeida A, Hilska M, Laato M, Syrjanen S, Syrjanen K, et al. VEGF-1 expression in colorectal cancer is associated with disease localization, stage, and long-term disease-specific survival. Anticancer Res. 2008;28(6B):3865–70. [PubMed] [Google Scholar]
  • 39.Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA, Kasowski M, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome research. 2012;22(9):1790–7. doi: 10.1101/gr.137323.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Badis G, Berger MF, Philippakis AA, Talukder S, Gehrke AR, Jaeger SA, et al. Diversity and complexity in DNA recognition by transcription factors. Science. 2009;324(5935):1720–3. doi: 10.1126/science.1162327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Riemann K, Becker L, Struwe H, Rubben H, Eisenhardt A, Siffert W. Insertion/deletion polymorphism in the promoter of NFKB1 as a potential molecular marker for the risk of recurrence in superficial bladder cancer. International journal of clinical pharmacology and therapeutics. 2007;45(8):423–30. doi: 10.5414/cpp45423. [DOI] [PubMed] [Google Scholar]
  • 42.Hoffstedt J, Eriksson P, Mottagui-Tabar S, Arner P. A polymorphism in the leptin promoter region (−2548 G/A) influences gene expression and adipose tissue secretion of leptin. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme. 2002;34(7):355–9. doi: 10.1055/s-2002-33466. [DOI] [PubMed] [Google Scholar]
  • 43.Meirhaeghe A, Fajas L, Helbecque N, Cottel D, Lebel P, Dallongeville J, et al. A genetic polymorphism of the peroxisome proliferator-activated receptor gamma gene influences plasma leptin levels in obese humans. Hum Mol Genet. 1998;7(3):435–40. doi: 10.1093/hmg/7.3.435. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Cover letter

RESOURCES