Case Control Study Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 7, 2017; 23(37): 6854-6867
Published online Oct 7, 2017. doi: 10.3748/wjg.v23.i37.6854
Association of insertion-deletions polymorphisms with colorectal cancer risk and clinical features
Diego Marques, Layse Raynara Ferreira-Costa, Lorenna Larissa Ferreira-Costa, Raul Hernandes Bortolin, André Ducati Luchessi, Vivian Nogueira Silbiger, Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
Diego Marques, Raul Hernandes Bortolin, André Ducati Luchessi, Vivian Nogueira Silbiger, Programa de Pós-graduação em Ciências Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
Diego Marques, Ândrea Ribeiro-dos-Santos, Sidney Santos, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66055-080, Pará, Brazil
Romualdo da Silva Correa, Aline Maciel Pinheiro Borges, Fernanda Ribeiro Ito, Departamento de Cirurgia Oncológica, Liga Norte Riograndense Contra o Câncer, Natal 59040-000, Rio Grande do Norte, Brazil
Carlos Cesar de Oliveira Ramos, Laboratório de Patologia e Citopatologia, Liga Norte Riograndense Contra o Câncer, Natal 59040-000, Rio Grande do Norte, Brazil
André Ducati Luchessi, Vivian Nogueira Silbiger, Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
Ândrea Ribeiro-dos-Santos, Sidney Santos, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil
ORCID number: Diego Marques (0000-0002-7446-6087); Layse Rayane Ferreira-Costa (0000-0001-5870-9686); Lorenna Larissa Ferreira-Costa (0000-0001-5629-4093); Romualdo da Silva Correa (0000-0002-6255-6974); Aline Maciel Pinheiro Borges (0000-0002-4342-7229); Fernanda Ribeiro Ito (0000-0003-1569-5637); Carlos Cesar de Oliveira Ramos (0000-0001-7581-8921); Raul Hernandes Bortolin (0000-0003-3424-3281); André Ducati Luchessi (0000-0002-2758-8102); Ândrea Ribeiro-dos-Santos (0000-0001-7001-1483); Sidney Santos (0000-0002-8622-9417); Vivian Nogueira Silbiger (0000-0002-9252-0278).
Author contributions: Ribeiro-dos-Santos A, Santos S and Silbiger VN designed the research; Correa RS, Borges AMP and Ito FR selected patients and collected clinical data; Ramos CCO collected histopathological data; Marques D, Ferreira-Costa LR and Ferreira-Costa LL collected biological material and performed the assays; Marques D, Bortolin RH and Luchessi AD analyzed the data; Marques D wrote the paper; Bortolin RH, Silbiger VN and Ribeiro-dos-Santos A critically revised the manuscript; Silbiger VN approved final version of the article to be published.
Supported by the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), No. 483031/2013-5; Rede de Pesquisa em Genomica Populacional Humana, No. Biocomputacional/CAPES-051/2013; Fundação de Amparo à Pesquisa do Estado do Pará, No. 155/2014; and Fundação de Amparo à Pesquisa do Estado do Rio Grande do Norte, No. 005/2011.
Institutional review board statement: This study was reviewed and approved by the Liga Norte Riograndense Contra o Câncer Institutional Review Board.
Informed consent statement: All study participants, or their legal guardian, provided informed written consent prior to study enrollment.
Conflict-of-interest statement: No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.
Data sharing statement: Technical appendix, statistical code, and dataset are available from the corresponding author at viviansilbiger@hotmail.com; viviansilbiger@ufrnet.br.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Vivian Nogueira Silbiger, PhD, Professor, Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte, Av. General Gustavo Cordeiro de Faria S/N, Petrópolis, Natal 59012-570, Rio Grande do Norte, Brazil. viviansilbiger@ufrnet.br
Telephone: +55-84-33429807 Fax: +55-84-33429833
Received: April 17, 2017
Peer-review started: April 27, 2017
First decision: June 5, 2017
Revised: June 24, 2017
Accepted: August 15, 2017
Article in press: August 15, 2017
Published online: October 7, 2017

Abstract
AIM

To investigate the association between 16 insertion-deletions (INDEL) polymorphisms, colorectal cancer (CRC) risk and clinical features in an admixed population.

METHODS

One hundred and forty patients with CRC and 140 cancer-free subjects were examined. Genomic DNA was extracted from peripheral blood samples. Polymorphisms and genomic ancestry distribution were assayed by Multiplex-PCR reaction, separated by capillary electrophoresis on the ABI 3130 Genetic Analyzer instrument and analyzed on GeneMapper ID v3.2. Clinicopathological data were obtained by consulting the patients’ clinical charts, intra-operative documentation, and pathology scoring.

RESULTS

Logistic regression analysis showed that polymorphism variations in IL4 gene was associated with increased CRC risk, while TYMS and UCP2 genes were associated with decreased risk. Reference to anatomical localization of tumor Del allele of NFKB1 and CASP8 were associated with more colon related incidents than rectosigmoid. In relation to the INDEL association with tumor node metastasis (TNM) stage risk, the Ins alleles of ACE, HLAG and TP53 (6 bp INDEL) were associated with higher TNM stage. Furthermore, regarding INDEL association with relapse risk, the Ins alleles of ACE, HLAG, and UGT1A1 were associated with early relapse risk, as well as the Del allele of TYMS. Regarding INDEL association with death risk before 10 years, the Ins allele of SGSM3 and UGT1A1 were associated with death risk.

CONCLUSION

The INDEL variations in ACE, UCP2, TYMS, IL4, NFKB1, CASP8, TP53, HLAG, UGT1A1, and SGSM3 were associated with CRC risk and clinical features in an admixed population. These data suggest that this cancer panel might be useful as a complementary tool for better clinical management, and more studies need to be conducted to confirm these findings.

Key Words: Colorectal cancer, Ins-del polymorphism, Admixed population, Potential biomarker, Diagnostic, Risk stratification, Prognostic, Clinical features

Core tip: The insertion-deletions (INDEL) variations in IL4 gene was associated with increased colorectal cancer (CRC) risk, while TYMS and UP2 genes were associated with decreased risk. The Del-alleles of NFKB1 and CASP8 were associated with more colon related incidents than rectosigmoid. The Ins-alleles of ACE, HLAG and TP53 were associated with higher TNM stage. The Ins-allele of ACE, HLAG, and UGT1A1 were associated with early relapse risk, as well as the Del-allele of TYMS. The Ins-alleles of SGSM3 and UGT1A1 were associated with death risk. These data suggest that these INDEL might be useful as a complementary tool for better CRC clinical management.



INTRODUCTION

Colorectal cancer (CRC) is the third most common cancer type in men and the second in women, considering 1477402 new cases in both sexes in 2015[1]. In that same year, Brazil was the tenth country with the highest CRC incidence, with 37167 new cases in both sexes[1], and making matters worse, its incidence and mortality continue to increase in the country.

Both genetic and environmental factors cause CRC[2], especially when combined[3,4]. Interestingly, these factors are ample and they vary pursuant to the cancer geographical regions[5]. However, inherited susceptibility is a major component of CRC predisposition, with an estimated 12%-35% risk attributed to genetic factors[6-8].

In relation to genetic factors, there are several mutations that might occur in human DNA, such as substitution, insertion, and deletion[9]. The second most abundant form of genetic variation in humans, after single nucleotide polymorphisms (SNPs), are the insertion-deletions (INDEL)[10]. INDELs are important because they are common genetic variations within genomes and among different ethnic groups[11,12], that may alter human traits and cause diseases[10,13], including CRC[14], by modifying the coding region[10,13] or mRNA stability[15]. The polymorphisms investigated in this study exhibit common features, given they are all functional polymorphisms that alter the expression of genes participating in metabolic pathways associated with carcinogenesis. Also, these genes are associated with different types of cancer with high incidence in the Brazilian population, such as stomach and CRC.

Furthermore, allele frequency varies among different populations[16], and genomic ancestry distribution may influence cancer development[17,18] by affecting polymorphisms distribution[19,20]. Few studies have been evaluated INDEL association in CRC in admixed population, mainly in Brazil. Thus, the aim of this study was to determine the association between CRC risk and prognostic follow-up with 16 INDELs in genes involved in apoptosis signaling (CASP8), GTPase-activating (SGSM3), steroids metabolism (CYP2E1, CYP19A1, and UGT1A1), immune system (HLAG, IL1A, IL4, and NFKB1), MDM2-P53 pathway (MDM2 and TP53), DNA replication and repair (TYMS and XRCC1) and angiogenesis (UCP2 and ACE) in an admixed population from Rio Grande do Norte state (in the Northeast Region of Brazil).

MATERIALS AND METHODS
A statement of ethics

The protocol used in this study was approved by the Research Ethics Committee of Liga Norte Riograndense Contra o Câncer (Rio Grande do Norte, Brazil) by number 211/211/2011. Moreover, all participants signed a consent form prior to providing a blood sample.

Casuistic distinctions

The patients in the case group (n = 140) were diagnosed with CRC as primary cancer and treated in the Proctologist Clinic and Colorectal Surgery Department of Liga Norte Riograndense Contra o Câncer (Rio Grande do Norte, Brazil). The control subjects were cancer-free blood donors (n = 140) from the hemotherapy service (Hemovida, Rio Grande do Norte, Brazil) and were recruited in 2014.

Both Peripheral blood samples and questionnaire answers were collected from all subjects. The clinicopathological data were obtained by consulting the patients’ clinical charts, intra-operative documentation, and pathology scoring. Furthermore, the CRC patients were followed up to 20 years by medical records.

Definitions

Alcohol consumption was classified as having the habit of alcohol consumption (Yes) or not having the habit of alcohol consumption (No). The subjects who have the habit of consuming alcohol were subcategorized according to consumption frequency (Eventually: ≤ 3 d per month; Frequently: > 3 d per month). The tobacco consumption was classified as have already smoked (Already) or not (Never). The subjects who have already smoked were subcategorized in Former (Stopped smoking for at least 1 year) and Current.

Tumor location was classified as rectosigmoid (sigmoid colon and rectum) and colon (ascending, transverse and descending colon) based on colonoscopy or on radiographic exam. The relapse records were obtained from histological or radiographic exams with subsequent clinical/radiological progression.

Overall survival was defined as the time from the date of surgery to the date of death or the date of the last follow-up of patients who were still alive. The relapse time was defined as the time from the date of surgery to the date of the first local. Patients with no local or distant relapse evidence at the date of death or the date of the last follow-up were censored.

DNA extraction and quantification

Genomic DNA was extracted by using a DNA extraction commercial kit, QIAamp DNA Blood Mini Kit (Qiagen, Hilden, Germany), and quantified with Qubit® 2.0 Fluorometer (Invitrogen, Carlsbad, CA, United States).

Polymorphism selection

Recently, INDELs have been the focus of multiple investigations[21-25]. This type of polymorphism presents interesting features as genetic markers: (1) INDELs are spread throughout the human genome; (2) INDELs derive from a single event (they do not present homoplasy); (3) since the allele frequencies of many INDELs are significantly different in separated populations; (4) small INDELs can be analyzed using short amplicons, which improves the amplification of degraded DNA and facilitates multiplexing; and (5) INDELs can be easily genotyped with a simple dye-labeling electrophoretic approach. Furthermore, all these genes evaluated in the present study show potential activity in pathway and may contribute to the carcinogenesis process (Table 1). Their genetic variations could contribute to: (1) risk of developing CRC; (2) impact on treatment response; or (3) in prognosis.

Table 1 Potential biological effects of insertion-deletions polymorphism selected in this study.
GenedbSNPLocalization1INDELRegionPotential biological effect[84]Potential impact on carcinogenesis
lenghtmRNA splicingmRNA stabilityGene expressionProtein function
ACErs464699417:63488539289IntronXXXAngiogenesis, proliferation, progression and metastases[85]
CASP8rs38341292:2012328096PromoterXApoptosis[86-88]
SGSM3rs5622877122:4041009243'-UTRXXProliferation and apoptosis [83,89-91]
CYP2E1--965'-FlankingXMetabolism of endo- and exogenous[92-97]
CYP19A1rs1157589915:512277493IntronXXXMetabolism of endo- and exogenous[92-97]
HLAGrs3711946296:29830804143'-UTRXXImmune surveillance[98-102]
IL1Ars37835532:11277413843'-UTRXXInduce chronic inflammation and proliferation[103,104]
IL4rs790718785:13268058470IntronXXXImmune surveillance and proliferation[40,39,43,105,106]
MDM2rs373048512:6880706540PromoterXProliferation and apoptosis[107,108]
NFKB1rs283624914:1025009984PromoterXDifferentiation, proliferation and apoptosis [109,110]
TP53rs1787836217:767637216IntronXXXProliferation, apoptosis, repair, differentiation[111-114]
TP53rs1788056017:766816963'-FlankingXXProliferation, apoptosis, repair, differentiation[111-114]
TYMSrs15126436018:67344463'-UTRXXDifferentiation, replication and repair[50,105]
UCP2--453'-UTRXXTumor aggressiveness and metastasis[56]
UGT1A1rs81753472:23376023523'-UTRXXMetabolism of endo- and exogenous[92-97]
XRCC1rs321323919:4357690745'- FlankingXRepair[115-117]
Genotyping of polymorphism

Multiplex PCR was used to simultaneously amplify the 16 investigated markers, as shown in Supplementary Table 1. The amplification was performed on ABI Verity thermocycler (Life Technologies, Foster City, CA, United States). A single multiplex reaction used Master Mix QIAGEN Multiplex PCR kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. The samples were incubated at 95 °C for 15 min, followed by 35 cycles at 94 °C for 45 s, 60 °C for 90 s, and 72 °C for 1 min, with a final extension at 70 °C for 30 min.

For fragment analysis, we used capillary electrophoresis on the ABI 3130 Genetic Analyzer instrument (Life Technologies). 1.0 μL of PCR product was added to 8.5 μL of HI-DI deionized formamide (Life Technologies) and 0.5 μL of GeneScan 500 LIZ pattern size standard (Life Technologies). After data collection, samples were analyzed on the GeneMapper ID v.3.7 software (Life Technologies).

Analysis of genetic ancestry

Genomic ancestry analysis was performed based on the method described by Santos et al[25] using 62 autosomal ancestry informative markers (AIMs). Two multiplex PCR reactions of 20 and 22 markers were performed and amplicons were analyzed by electrophoresis using the ABI Prism 3130 sequencer and GeneMapper ID v.3.2 software. The individual proportions of European, African, and Amerindian genetic ancestries were estimated using STRUCTURE v.2.3.3 software, assuming three parental populations (European, African, and Amerindian).

Statistical analysis

The categorical variables case and control participants were tested by the Chi-squared test. For ancestry index and age at diagnosis variables we used the Mann-Whitney test. Logistic regression analyses between the genotype model and CRC risk were performed by the SNPassoc package v.1.9-2, along with clinical features variables. The association between genotype and free-relapse survival time was evaluated by Kaplan-Meier plots, performed by the survival package v.2.41-3. Log-rank and Wilcoxon tests were used to examine the genetic effect on survival outcomes. The statistical power was estimated by 10000 simulations. All statistical analyses and plotting were performed with R package v.3.1.2[26]. Differences between groups were considered significant at P < 0.05.

RESULTS
Demographic characteristics

We analyzed 140 subjects with CRC and 140 cancer-free individuals. The demographic characteristics of participants were summarized in Table 2, which shows demographic features of the groups. Regarding genomic ancestry, significance was observed with the distribution of African ancestry (P = 0.049), Table 3. However, there was no difference between groups when an analysis of multinomial logistic regression was performed.

Table 2 Participant demographic and clinical characteristics and their stratification by case and control groups.
CharacteristicTotal, n = 280Cases, n = 140Controls, n = 140P value
Age (yr)48 (21-93)59 (23-93)37 (21-81)< 0.001
< 45136 (48.7)23 (16.5)113 (80.7)
≥ 45144 (51.3)116 (83.5)27 (19.3)
Gender< 0.001
Male172 (61.6)62 (44.6)110 (78.6)
Female108 (38.4)78 (55.4)30 (21.4)
Alcohol consumption< 0.001
No180 (65.1)118 (85.5)62 (44.9)
Yes96 (34.9)20 (14.5)76 (55.1)
Eventually56 (20.4)11 (8.0)45 (32.6)
Frequently40 (14.5)9 (6.5)31 (22.5)
Tobacco consumption< 0.001
Never182 (65.4)68 (48.9)114 (82.0)
Already96 (24.6)71 (48.1)25 (18.0)
Former66 (23.8)55 (39.6)11 (7.9)
Current30 (10.8)16 (11.5)14 (10.1)
Table 3 Genetic ancestry distribution between case and control groups.
Genetic ancestry (%)Total, n = 280Cases, n = 140Controls, n = 140OR (95%CI)P value
European65.3 ± 15.564.2 ± 15.666.4 ± 15.30.243
95-8050 (17.9)21 (15.1)29 (20.7)1.0 (Reference)
80-7070 (25.1)33 (23.7)37 (26.4)1.23 (0.59-2.56)0.577
70-6067 (24.0)38 (27.3)29 (20.7)1.81 (0.86-3.80)0.117
60-5047 (16.8)22 (15.8)25 (17.9)1.21 (0.54-2.71)0.634
50-4026 (9.3)14 (10.1)12 (8.6)1.61 (0.62-4.18)0.327
40-3011 (3.9)6 (4.3)5 (3.6)1.66 (0.45-6.16)0.451
30-207 (2.5)5 (3.6)2 (1.4)3.45 (0.61-19.54)0.161
20-101 (0.4)-1 (0.7)-
Amerindian16.2 ± 10.116.0 ± 10.316.3 ± 9.90.645
02-1090 (32.3)45 (32.4)45 (32.1)1.0 (Reference)
10-20113 (40.5)60 (43.2)53 (37.9)1.13 (0.65-1.97)0.661
20-3047 (16.8)18 (12.9)29 (20.7)0.62 (0.30-1.27)0.193
30-4021 (7.5)11 (7.9)10 (7.1)1.10 (0.42-2.85)0.844
40-507 (2.5)4 (2.9)2 (2.1)2.00 (0.35-11.47)0.437
50-601 (0.4)1 (0.7)--
African18.6 ± 12.019.8 ± 12.317.3 ± 11.70.049
02-1081 (29.0)36 (25.9)45 (32.1)1.0 (Reference)
10-2089 (31.9)41 (29.5)48 (34.3)1.07 (0.58-1.95)0.832
20-3066 (23.7)38 (27.3)28 (20.0)1.70 (0.88-3.27)0.114
30-4028 (10.0)15 (10.8)13 (9.3)1.44 (0.61-3.42)0.405
40-508 (2.9)5 (3.6)3 (2.1)2.08 (0.47-9.31)0.337
50-605 (1.8)3 (2.2)2 (1.4)1.87 (0.30-11.83)0.504
60-702 (0.7)1 (0.7)1 (0.7)1.25 (0.07-20.68)0.876
Distribution of genotypes associated with susceptibility to CRC

All INDEL polymorphisms are in Hardy-Weinberg equilibrium (P > 0.05). The genotypic and allelic frequencies of the subjects are presented in the Table 4. Genotypic frequency (P = 0.01) of IL4 gene polymorphism was significantly different between case-controls, and higher frequency of Del allele were observed in cases than in controls.

Table 4 Genotype and allele frequency in percentage of patients with colorectal cancer and controls.
GenedbSNPCase/control (n = 140/140)
Genotype frequency
Allele frequency
HWE
IIIDDDP valueIDP value
ACErs464699421.0/14.349.3/54.329.7/31.40.33545.7/41.454.3/58.60.161
CASP8rs383412934.5/30.046.0/46.419.4/23.60.60557.6/53.242.4/46.80.424
SGSM3rs562287717.2/3.630.2/36.462.6/60.00.27422.3/21.877.7/78.20.415
CYP19A1rs1157589935.8/37.949.6/50.014.6/12.10.8260.3/62.939.7/37.10.402
CYP2E1-0.7/0.717.3/12.982.0/86.40.5889.4/7.190.6/92.60.716
HLAGrs37119462914.7/13.643.4/50.041.9/36.40.53836.4/38.663.6/61.40.514
IL1Ars378355346.0/52.138.8/37.915.1/10.00.36865.5/71.134.5/28.90.348
IL4rs7907187848.9/60.040.3/37.110.8/2.90.01769.1/78.630.9/21.40.223
MDM2rs373048550.4/50.743.2/37.16.5/12.10.21971.9/69.328.1/30.70.132
NFKB1rs2836249138.1/40.746.0/42.115.8/17.10.80661.2/61.838.8/38.20.203
TP53rs178783623.6/1.427.3/32.169.1/66.40.38317.3/17.582.7/82.50.181
TP53rs178805607.2/7.139.6/32.953.2/60.00.48927.0/23.673.0/76.40.297
TYMSrs15126436046.0/42.943.9/42.110.1/15.00.45968.0/63.932.0/36.10.308
UCP2-6.6/9.335.3/44.358.1/46.40.14924.3/31.475.7/68.60.745
UGT1A1rs817534711.5/10.844.6/46.043.9/43.20.78533.8/33.866.2/66.20.735
XRCC1rs321323941.7/42.947.5/45.010.8/12.10.89465.5/65.434.5/34.60.941

The significant logistic regression analyses between case-controls are summarized in Table 5. Del allele polymorphism in IL4 gene (P = 0.0110) was associated with an increased risk of CRC development, while Ins allele in UCP2 (P = 0.0210) was decreased CRC risk. Furthermore, the Del allele in the TYMS (P = 0.0120) gene was associated with decreased CRC risk.

Table 5 The logistic regression analyses between case-control and insertion-deletions polymorphism.
GeneModelOR (95%CI)P-value
IL4Ins/Ins vs Del/Ins + Del/Del2.26 (1.20-4.31)0.0110
TYMSIns/Ins + Del/Ins vs Del/Del0.26 (0.08-0.75)0.0120
UCP2Del/Del vs Del/Ins + Ins/Ins0.48 (0.25-0.90)0.0210
Distribution of genotypes associated with prognostic follow-up in CRC

The baseline characteristics of CRC patients are summarized in Table 6. The follow-up time median was 5.28 years among 78 patients who had complete genotype and follow-up information. The 5-year free-relapse rate was 70% and the 10-year free-relapse rate was 66.4%. The 5-year survival rate was 91.4% and the 10-year survival rate was 87.9%.

Table 6 Clinical characteristics of patients with colorectal cancer at diagnosis and follow-up.
CharacteristicsCases (n = 140)
Tumor localization
Colon25 (17.9)
Rectosigmoid115 (82.1)
Tumor grade
G1, G2130 (92.9)
G3, G410 (7.1)
Depth of invasion
T1, T237 (26.6)
T3, T489 (64.0)
Tx13 (9.4)
Lymph node involvement
N077 (55.4)
N1, N247 (33.8)
Nx15 (10.8)
Distant metastasis
M0109 (78.4)
M113 (9.4)
Mx17 (12.2)
AJCC stage
StageI31 (22.3)
Stage II43 (30.9)
Stage III43 (30.9)
Stage IV15 (10.8)
Unknown7 (5.1)
Relapse, Yes48 (34.5)
Death, Yes18 (12.9)

We also evaluated the genetic impact in the clinical features. The Del allele in NFKB1 and CASP8 were associated with more incidents to colon than rectosigmoid (Table 7). In relation to the INDEL association with TNM stage risk, the Ins alleles of ACE, HLAG and TP53 (6 bp INDEL) were associated with a higher TNM stage (Table 8). Regarding the INDEL association with relapse risk, the Ins alleles of ACE, HLAG, and UGT1A1 were associated with relapse risk, as well as the Del allele of TYMS (Table 9). Moreover, these findings corroborate those observed in the free-relapse survival curve (Figure 1). Regarding INDEL association with death risk, the Ins alleles of SGSM3 and UGT1A1 were associated with death risk (Table 10).

Figure 1
Figure 1 Free-relapse survival of patients with colorectal cancer related with significant insertion-deletions. Logistic regression adjusted for confounders. The analyses and graphic were performed by survival packages, in R statistical software.
Table 7 The significant insertion-deletions associations with anatomic localization.
GeneModelOR (95%CI)P value
CASP8Ins/Ins vs Del/Ins + Del/Del0.28 (0.08-0.97)0.0303
NFKB1Ins/Ins vs Del/Ins+Del/Del0.31 (0.10-0.93)0.0276
Table 8 The significant insertion-deletions associations with tumor node metastasis stage risks.
GeneModelOR (95%CI)P value
ACEDel/Del vs Del/Ins + Ins/Ins2.82 (1.26-6.31)0.0092
HLAGDel/Del + Del/Ins vs Ins/Ins2.74 (1.01-7.42)0.0416
TP53 06 bpDel/Del vs Del/Ins + Ins/Ins2.50 (1.23-5.06)0.0099
Table 9 The significant insertion-deletions associations with relapse risks.
GeneModelTime of follow-upOR (95%CI)P value
ACEDel/Del vs Del/Ins + Ins/Ins2 yr0.32 (0.13-0.77)0.0113
ACEDel/Del vs Del/Ins + Ins/Ins3 yr0.37 (0.15-0.91)0.0298
HLAGDel/Del vs Del/Ins + Ins/Ins2 yr2.75 (1.07-7.08)0.0281
HLAGDel/Del vs Del/Ins + Ins/Ins4 yr2.83 (1.07-7.52)0.0332
HLAGDel/Del vs Del/Ins + Ins/Ins5 yr3.47 (1.20-9.99)0.0194
TYMSIns/Ins vs Del/Ins + Del/Del2 yr3.35 (1.36-8.28)0.0058
TYMSIns/Ins vs Del/Ins + Del/Del3 yr3.42 (1.41-8.28)0.0046
UGT1A1Del/Del vs Del/Ins + Ins/Ins4 yr3.23 (1.27-8.22)0.0116
UGT1A1Del/Del vs Del/Ins + Ins/Ins5 yr3.50 (1.24-9.84)0.0145
Table 10 The significant insertion-deletions associations with death risks.
GeneModelTime of follow-upOR (95%CI)P value
SGSM3Del/Del vs Del/Ins + Ins/Ins6 yr3.61 (1.01-12.92)0.0487
SGSM3Del/Del vs Del/Ins + Ins/Ins7 yr4.60 (1.16-18.23)0.0260
UGT1A1Del/Del vs Del/Ins + Ins/Ins6 yr5.30 (1.43-19.73)0.0084
UGT1A1Del/Del vs Del/Ins + Ins/Ins7 yr4.64 (1.19-18.10)0.0202
UGT1A1Del/Del vs Del/Ins + Ins/Ins8 yr6.50 (1.47-28.80)0.0091
DISCUSSION

Despite the effective strategies for prevention, early detection, and treatment[27-32], there are ethnic differences in the CRC incidence and survival[33,34], specifically in individuals with African American ancestry, who have higher CRC incidence and lower 5-years survival rates than other ethnic groups[33-38].

In this work, we evaluated the association between 16 INDEL [ACE, CASP8, SGSM3, CYP19A1, CYP2E1, HLAG, IL1A, MDM2, NFKB1, TP53 (16 and 6 bp), TYMS, UCP2, XRCC1, IL4 and UGT1A1] and the risk of developing CRC in a Brazilian population, as well as their clinical features. We found significant association between three investigated INDEL polymorphisms and CRC risk, two associated with anatomical localization, three associated with TNM stage, four associated with early relapse risk, and two associated with death risk before 10 years.

Variations in the IL-4 activity or in the IL-4 receptor due to mutations have been associated with cell proliferation and might affect signal transduction pathways in cancer[39]. We evaluated INDEL of 70 bp in intron 3 of the IL4 gene (rs79071878), a variation which may influence the production of this cytokine. The higher IL-4 production may result in diminished cell-mediated immune response, and escape from immune surveillance in the tumor cells. The cell-mediated immune response may be inhibited by downregulating the expression of Th1 cytokines, decreasing the CD8+ T-cell response in the tumor microenvironment[39-41]. Furthermore, this INDEL has been associated with gastric cancer[39] and other immune diseases[42,43]. However, this is the first study indicating an association between this IL4 polymorphism and the risk of developing CRC. Our results indicate that the Del allele in IL4 was associated with the risk of developing CRC.

The TYMS gene plays an essential role in the biosynthesis of the DNA-component thymidylate (dTTP) and is required for DNA replication and repair[44]. The insertion of 6 bp in the 3’-UTR of TYMS primary transcript (rs151264360) may significantly influence gene expression as shown by using a luciferase assay[15]. Mandola et al[15] observed that a Del allele might decrease the TYMS mRNA stability, and the TYMS protein expression. Moreover, Rahman et al[45] showed in vitro that TYMS overexpression might induce the transformation of mammalian cells into a malignant phenotype. Studies indicate that this INDEL is associated with many cancers[46-49], especially colorectal[48]. These results suggested that this INDEL variation might decrease CRC risk, as showed in the present work. However, this finding diverges from data from Mexico[50], in which association was not observed. On the other hand, our results showed that this Del allele was associated with an increase relapse risk.

Uncoupling proteins (UCPs) are a family of mitochondrial proteins, which were originally reported to play essential roles in reducing the reactive oxygen species[51,52]. UCP2 plays a role in carcinogenesis in various tissues, including colon cancer, and regulates the responsiveness of carcinomas to chemotherapy[53-56]. Adaptive mechanisms of cancer cells include resistance to tumor growth inhibition and evasion of apoptosis, and cellular events that are appreciably affected by oxidative stress[57,58]. The UCP2 expression level is significantly higher in colon cancer tissue than in its adjacent tissue and UCP2 may play a role in intestinal epithelial cells from benign to malignant transformation[59]. However, the role of UCP2 in development of colon cancer is unclear. INDEL polymorphism may regulate UCP2 mRNA stability via post-transcriptional modification of UCP2 protein expression[60,61]. Indeed, in the present study was observed that INDEL polymorphism might be associated with colorectal cancer. However, this is the first study indicating an association between this UCP2 polymorphism and the risk of developing CRC.

The renin-angiotensin system (RAS), which regulates systemic blood pressure, also exerts local effects on cell proliferation, apoptosis, inflammation and angiogenesis in different tissues[62]. In addition, there is evidence linking the RAS with tumorigenesis and tumor angiogenesis[63]. The polymorphisms in the various components of the RAS that may possess clinical relevance[62], and the most common polymorphism in the gene encoding angiotensin converting enzyme (ACE) is INDEL of a 287-bp fragment in intron 16 and is responsible for the inter-individual variation in the ACE levels in blood and tissues[64]. The insertion allele in this gene was associated with ACE levels, the rate of disease progression, shorter TTF, and lower circulating levels of ACE[62,65]. This INDEL has been associated with cancer risk susceptibility[66-68], including CRC[65,68], and with response to bevacizumab[62]. Our results indicate that Ins allele was not associated with CRC risk development, as showed by Yang et al’s meta-analysis[69] and Liu et al[70] case-control study (241 cases and 299 control, China). On the other hand, our results also showed that this INDEL was also associated with TNM stage risk and relapse risk.

The HLAG is an important immunomodulatory molecule related to several mechanisms of tolerance[71]. Since the discovery of the HLA-G protein expression in cancer[72], several pieces of evidence have supported a considerable role for HLA-G in tumor cell escape from immuno-surveillance and antitumor immune responses[73]. The 14 bp INDEL (rs371194629) has been suggested to have functional significance. The Ins allele has been shown to be associated with alternative splicing, resulting in deletion of 92 bp in exon 5 from mature mRNA, which then leads to low levels of soluble HLA-G (sHLA-G)[74]. Furthermore, our results indicated that the Ins allele was associated with a higher TNM stage and relapse up to 5 years. These findings suggest that low levels of sHLA-G might influence in poor prognostics.

UGT1 is a family of membrane-bound enzymes involved in the inactivation and elimination of lipophilic molecules through glucorination. Moreover, variants in this gene have been shown to be useful tools to identifying patients more likely to experience severe toxicity related to irinotecan-containg regimens[75]. In particular, INDEL variants in UGT1A1 (rs8175347) were associated with significantly decreased glucuronidation activity, which results in reduced SN-38 clearance[76] and an increased risk of these toxicities in patients homozygous for the Ins allele[75,77-80]. Our results showed that the Ins allele in UGT1A1 was associated with early relapse risk, as well as with death risk prior to 8 years. This genetic variation may identify patients who might benefit from increased irinotecan dosing, as observed by Chen et al[75].

The SGSM3 belongs to a novel protein family consisting of three members and appears to be associated with small G-protein coupled receptor signal transduction pathways, and could control cellular functions by a Ras-mediated signaling pathway[81]. Studies have linked Rab dysfunction to various human diseases including cancer[82,83], and our results have shown that the Ins allele might also be associated with death risk prior to 8 years.

The aims of this study were to determine the association between CRC risk and the clinical features with 16 INDEL in genes involved with carcinogenesis pathways in an admixed population from Brazil. Although we have achieved our goal, there are limitations regarding sample number. We suggest, therefore, that an extensive study should be conducted in the Brazilian population to confirm the findings, as well as in other admixed populations.

In summary, the present work indicates that polymorphisms in ACE (rs4646994), TYMS (rs151264360), UCP2 (45 bp), IL4 (rs79071878), NFKB1 (rs28362491), CASP8 (rs3834129), TP53 (rs17880560), HLAG (rs371194629), UGT1A1 (rs3213239), and SGSM3 (rs56228771) genes were associated with CRC risk and clinical features in an admixed population. These data suggest that this cancer panel might be useful as a complementary tool for better clinical management, and more studies need to be conducted to confirm these findings.

ACKNOWLEDGMENTS

We thank all participants who allowed us to carry out this study. We are also grateful to the students and technicians from LGHM/UFPA/PA, LBBM-LABMULT/UFRN/RN, Laboratory of Pathology and Cytopathology/Liga Norte Riograndense Contra o Câncer/RN and Hemovida/RN. We are also grateful to Philip Barsanti for the invaluable and constructive English review of the manuscript. Furthermore, we are grateful to André Ribeiro-dos-Santos and Ana Paula Schaan for help with statistical revision.

COMMENTS
Background

Colorectal cancer (CRC) is the third most common cancer type in men and the second in women. Despite the effective strategies for prevention, early detection, and treatment, there are ethnic differences in CRC incidence and survival. These variances occur specifically in African Americans, who have higher CRC incidence and lower survival rates than other ethnic groups. Thus, the present study evaluated the association between 16 insertion-deletions (INDEL) polymorphisms with colorectal cancer risk in an admixture population, as well with clinical features.

Research frontiers

The second most abundant form of genetic variation in humans, after single nucleotide polymorphisms (SNPs), are the INDEL. The INDEL understanding is important because they are common genetic variations within genomes, and they may alter human traits and cause diseases, including colorectal cancer, by modifying the coding region or mRNA stability. One of the challenges for genetic polymorphism association studies is the lack of knowledge regarding the frequency of the polymorphism in the targeted population, mainly in admixed populations (e.g. Brazil).

Innovations and breakthroughs

This is the first case-control study to evaluate the association between these 16 INDEL polymorphisms with colorectal risk, clinical features and prognostic follow-up in an admixture population, adopting the methodology that can be easily used to perform multiplexing assays.

Applications

This pilot study is design and findings could be used to determine sample size for a larger randomized controlled study aiming to test the impact of these INDEL polymorphism panel in colorectal risk, clinical features and prognostic follow-up.

Terminology

Ancestry informative marker - In population genetics, an ancestry informative marker (AIM) is a polymorphism that exhibits substantially different frequencies between populations from different geographical regions. A set of many AIMs can be used to estimate the proportion of ancestry of an individual derived from each geographical region.

Peer-review

This is an interesting study aiming to determine the association between CRC risk, and the clinical features with 16 INDEL in genes involved with carcinogenesis pathways in an admixed population from Brazil. The overall structure of the manuscript is complete and conforms to the academic rules.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Gastroenterology and Hepatology

Country of origin: Brazil

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Dai ZJ, Engin AB, Wang YH S- Editor: Gong ZM L- Editor: A E- Editor: Huang Y

References
1.  Ferlay J, Soerjomataram I, Ervik M, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. GLOBOCCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11 [Internet]. Lyon, France: International Angency for Research on Cancer; 2013. Accessed on 2016-2-12.  Available from: http://globocan.iarc.fr/.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Hong Y, Wu G, Li W, Liu D, He K. A comprehensive meta-analysis of genetic associations between five key SNPs and colorectal cancer risk. Oncotarget. 2016;7:73945-73959.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
3.  Zaridze DG. Molecular epidemiology of cancer. Biochemistry (Mosc). 2008;73:532-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
4.  Liu J, He C, Xu Q, Xing C, Yuan Y. NOD2 polymorphisms associated with cancer risk: a meta-analysis. PLoS One. 2014;9:e89340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
5.  Gomez SL, Shariff-Marco S, DeRouen M, Keegan TH, Yen IH, Mujahid M, Satariano WA, Glaser SL. The impact of neighborhood social and built environment factors across the cancer continuum: Current research, methodological considerations, and future directions. Cancer. 2015;121:2314-2330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 213]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
6.  Peters U, Bien S, Zubair N. Genetic architecture of colorectal cancer. Gut. 2015;64:1623-1636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 132]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
7.  Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, Pukkala E, Skytthe A, Hemminki K. Environmental and heritable factors in the causation of cancer--analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med. 2000;343:78-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2809]  [Cited by in F6Publishing: 2651]  [Article Influence: 110.5]  [Reference Citation Analysis (1)]
8.  Czene K, Lichtenstein P, Hemminki K. Environmental and heritable causes of cancer among 9.6 million individuals in the Swedish Family-Cancer Database. Int J Cancer. 2002;99:260-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 361]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
9.  Iengar P. An analysis of substitution, deletion and insertion mutations in cancer genes. Nucleic Acids Res. 2012;40:6401-6413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
10.  Mullaney JM, Mills RE, Pittard WS, Devine SE. Small insertions and deletions (INDELs) in human genomes. Hum Mol Genet. 2010;19:R131-R136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 209]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
11.  Mills RE, Pittard WS, Mullaney JM, Farooq U, Creasy TH, Mahurkar AA, Kemeza DM, Strassler DS, Ponting CP, Webber C. Natural genetic variation caused by small insertions and deletions in the human genome. Genome Res. 2011;21:830-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 165]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
12.  Boschiero C, Gheyas AA, Ralph HK, Eory L, Paton B, Kuo R, Fulton J, Preisinger R, Kaiser P, Burt DW. Detection and characterization of small insertion and deletion genetic variants in modern layer chicken genomes. BMC Genomics. 2015;16:562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
13.  Veerappa AM, Vishweswaraiah S, Lingaiah K, Murthy NM, Suresh RV, Belur K, Ramachandra NB, Tejaswini , Patel NB, Gowda PK. Insertion-deletions burden in copy number polymorphisms of the Tibetan population. Indian J Hum Genet. 2014;20:166-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
14.  Gao X, Zhang S, Zhu Z. Genetic variation of ErbB4 confers risk of colorectal cancer in a Chinese Han population. Cancer Biomark. 2014;14:435-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
15.  Mandola MV, Stoehlmacher J, Zhang W, Groshen S, Yu MC, Iqbal S, Lenz HJ, Ladner RD. A 6 bp polymorphism in the thymidylate synthase gene causes message instability and is associated with decreased intratumoral TS mRNA levels. Pharmacogenetics. 2004;14:319-327.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Hu Z, Li X, Qu X, He Y, Ring BZ, Song E, Su L. Intron 3 16 bp duplication polymorphism of TP53 contributes to cancer susceptibility: a meta-analysis. Carcinogenesis. 2010;31:643-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
17.  Hernandez-Suarez G, Sanabria MC, Serrano M, Herran OF, Perez J, Plata JL, Zabaleta J, Tenesa A. Genetic ancestry is associated with colorectal adenomas and adenocarcinomas in Latino populations. Eur J Hum Genet. 2014;22:1208-1216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
18.  Carvalho DC, Wanderley AV, Amador MA, Fernandes MR, Cavalcante GC, Pantoja KB, Mello FA, de Assumpção PP, Khayat AS, Ribeiro-Dos-Santos Â, Santos S, Dos Santos NP. Amerindian genetic ancestry and INDEL polymorphisms associated with susceptibility of childhood B-cell Leukemia in an admixed population from the Brazilian Amazon. Leuk Res. 2015;39:1239-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
19.  Cassiano GC, Santos EJ, Maia MH, Furini Ada C, Storti-Melo LM, Tomaz FM, Trindade PC, Capobianco MP, Amador MA, Viana GM. Impact of population admixture on the distribution of immune response co-stimulatory genes polymorphisms in a Brazilian population. Hum Immunol. 2015;76:836-842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
20.  Campanella NC, Berardinelli GN, Scapulatempo-Neto C, Viana D, Palmero EI, Pereira R, Reis RM. Optimization of a pentaplex panel for MSI analysis without control DNA in a Brazilian population: correlation with ancestry markers. Eur J Hum Genet. 2014;22:875-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
21.  Bastos-Rodrigues L, Pimenta JR, Pena SD. The genetic structure of human populations studied through short insertion-deletion polymorphisms. Ann Hum Genet. 2006;70:658-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 100]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
22.  Mills RE, Luttig CT, Larkins CE, Beauchamp A, Tsui C, Pittard WS, Devine SE. An initial map of insertion and deletion (INDEL) variation in the human genome. Genome Res. 2006;16:1182-1190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 491]  [Cited by in F6Publishing: 489]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
23.  Ribeiro-Rodrigues EM, dos Santos NP, dos Santos AK, Pereira R, Amorim A, Gusmão L, Zago MA, dos Santos SE. Assessing interethnic admixture using an X-linked insertion-deletion multiplex. Am J Hum Biol. 2009;21:707-709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
24.  Weber JL, David D, Heil J, Fan Y, Zhao C, Marth G. Human diallelic insertion/deletion polymorphisms. Am J Hum Genet. 2002;71:854-862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 245]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
25.  Santos NP, Ribeiro-Rodrigues EM, Ribeiro-Dos-Santos AK, Pereira R, Gusmão L, Amorim A, Guerreiro JF, Zago MA, Matte C, Hutz MH. Assessing individual interethnic admixture and population substructure using a 48-insertion-deletion (INSEL) ancestry-informative marker (AIM) panel. Hum Mutat. 2010;31:184-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 238]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
26.  R Development Core Team. R A language and environment for statistical computing. R Found Stat Comput, Vienna, Austria.  Available from: http://www.r-project.org/.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Schoen RE, Pinsky PF, Weissfeld JL, Yokochi LA, Church T, Laiyemo AO, Bresalier R, Andriole GL, Buys SS, Crawford ED. Colorectal-cancer incidence and mortality with screening flexible sigmoidoscopy. N Engl J Med. 2012;366:2345-2357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 672]  [Cited by in F6Publishing: 697]  [Article Influence: 58.1]  [Reference Citation Analysis (1)]
28.  Mandel JS, Bond JH, Church TR, Snover DC, Bradley GM, Schuman LM, Ederer F. Reducing mortality from colorectal cancer by screening for fecal occult blood. Minnesota Colon Cancer Control Study. N Engl J Med. 1993;328:1365-1371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2183]  [Cited by in F6Publishing: 2118]  [Article Influence: 68.3]  [Reference Citation Analysis (1)]
29.  Nishihara R, Wu K, Lochhead P, Morikawa T, Liao X, Qian ZR, Inamura K, Kim SA, Kuchiba A, Yamauchi M. Long-term colorectal-cancer incidence and mortality after lower endoscopy. N Engl J Med. 2013;369:1095-1105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 968]  [Cited by in F6Publishing: 1047]  [Article Influence: 95.2]  [Reference Citation Analysis (0)]
30.  US Preventive Services Task Force. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. Ann Intern Med. 2008;149:627-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1005]  [Cited by in F6Publishing: 1053]  [Article Influence: 65.8]  [Reference Citation Analysis (1)]
31.  Scambler G, Hopkins A. Generating a model of epileptic stigma: the role of qualitative analysis. Soc Sci Med. 1990;30:1187-1194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
32.  Atkin WS, Edwards R, Kralj-Hans I, Wooldrage K, Hart AR, Northover JM, Parkin DM, Wardle J, Duffy SW, Cuzick J; UK Flexible Sigmoidoscopy Trial Investigators. Once-only flexible sigmoidoscopy screening in prevention of colorectal cancer: a multicentre randomised controlled trial. Lancet. 2010;375:1624-1633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1121]  [Cited by in F6Publishing: 1083]  [Article Influence: 77.4]  [Reference Citation Analysis (0)]
33.  Soneji S, Iyer SS, Armstrong K, Asch DA. Racial disparities in stage-specific colorectal cancer mortality: 1960-2005. Am J Public Health. 2010;100:1912-1916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 83]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
34.  Robbins AS, Siegel RL, Jemal A. Racial disparities in stage-specific colorectal cancer mortality rates from 1985 to 2008. J Clin Oncol. 2012;30:401-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 152]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
35.  Wu BU, Longstreth GF, Ngor EW. Screening colonoscopy versus sigmoidoscopy: implications of a negative examination for cancer prevention and racial disparities in average-risk patients. Gastrointest Endosc. 2014;80:852-861.e1-2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
36.  Corley DA, Jensen CD, Marks AR, Zhao WK, de Boer J, Levin TR, Doubeni C, Fireman BH, Quesenberry CP. Variation of adenoma prevalence by age, sex, race, and colon location in a large population: implications for screening and quality programs. Clin Gastroenterol Hepatol. 2013;11:172-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 166]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
37.  Lieberman DA, Holub JL, Moravec MD, Eisen GM, Peters D, Morris CD. Prevalence of colon polyps detected by colonoscopy screening in asymptomatic black and white patients. JAMA. 2008;300:1417-1422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 160]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
38.  Kupfer SS, Anderson JR, Hooker S, Skol A, Kittles RA, Keku TO, Sandler RS, Ellis NA. Genetic heterogeneity in colorectal cancer associations between African and European americans. Gastroenterology. 2010;139:1677-1685, 1685.e1-1685.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
39.  Bhayal AC, Krishnaveni D, Rao KP, Kumar AR, Jyothy A, Nallari P, Venkateshwari A. Significant Association of Interleukin4 Intron 3 VNTR Polymorphism with Susceptibility to Gastric Cancer in a South Indian Population from Telangana. PLoS One. 2015;10:e0138442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
40.  Nakashima H, Miyake K, Inoue Y, Shimizu S, Akahoshi M, Tanaka Y, Otsuka T, Harada M. Association between IL-4 genotype and IL-4 production in the Japanese population. Genes Immun. 2002;3:107-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 117]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
41.  Gaur P, Singh AK, Shukla NK, Das SN. Inter-relation of Th1, Th2, Th17 and Treg cytokines in oral cancer patients and their clinical significance. Hum Immunol. 2014;75:330-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
42.  Anovazzi G, Kim YJ, Viana AC, Curtis KM, Orrico SR, Cirelli JA, Scarel-Caminaga RM. Polymorphisms and haplotypes in the interleukin-4 gene are associated with chronic periodontitis in a Brazilian population. J Periodontol. 2010;81:392-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
43.  Cabantous S, Poudiougou B, Oumar AA, Traore A, Barry A, Vitte J, Bongrand P, Marquet S, Doumbo O, Dessein AJ. Genetic evidence for the aggravation of Plasmodium falciparum malaria by interleukin 4. J Infect Dis. 2009;200:1530-1539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
44.  Burdelski C, Strauss C, Tsourlakis MC, Kluth M, Hube-Magg C, Melling N, Lebok P, Minner S, Koop C, Graefen M. Overexpression of thymidylate synthase (TYMS) is associated with aggressive tumor features and early PSA recurrence in prostate cancer. Oncotarget. 2015;6:8377-8387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
45.  Rahman L, Voeller D, Rahman M, Lipkowitz S, Allegra C, Barrett JC, Kaye FJ, Zajac-Kaye M. Thymidylate synthase as an oncogene: a novel role for an essential DNA synthesis enzyme. Cancer Cell. 2004;5:341-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 120]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
46.  Guan X, Liu H, Ju J, Li Y, Li P, Wang LE, Brewster AM, Buchholz TA, Arun BK, Wei Q. Genetic variant rs16430 6bp > 0bp at the microRNA-binding site in TYMS and risk of sporadic breast cancer risk in non-Hispanic white women aged ≤ 55 years. Mol Carcinog. 2015;54:281-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
47.  Shen R, Liu H, Wen J, Liu Z, Wang LE, Wang Q, Tan D, Ajani JA, Wei Q. Genetic polymorphisms in the microRNA binding-sites of the thymidylate synthase gene predict risk and survival in gastric cancer. Mol Carcinog. 2015;54:880-888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
48.  Zhou JY, Shi R, Yu HL, Zeng Y, Zheng WL, Ma WL. The association between two polymorphisms in the TS gene and risk of cancer: a systematic review and pooled analysis. Int J Cancer. 2012;131:2103-2116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
49.  Shaw GM, Yang W, Perloff S, Shaw NM, Carmichael SL, Zhu H, Lammer EJ. Thymidylate synthase polymorphisms and risks of human orofacial clefts. Birth Defects Res A Clin Mol Teratol. 2013;97:95-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
50.  Gallegos-Arreola MP, Peralta-Leal V, Morgan-Villela G, Puebla-Pérez AM. [Frequency of TS1494del6 polymorphism in colorectal patients from west of Mexico]. Rev Invest Clin. 2008;60:21-30.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Wang M, Li G, Yang Z, Wang L, Zhang L, Wang T, Zhang Y, Zhang S, Han Y, Jia L. Uncoupling protein 2 downregulation by hypoxia through repression of peroxisome proliferator-activated receptor γ promotes chemoresistance of non-small cell lung cancer. Oncotarget. 2017;8:8083-8094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
52.  Krauss S, Zhang CY, Lowell BB. The mitochondrial uncoupling-protein homologues. Nat Rev Mol Cell Biol. 2005;6:248-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 93]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
53.  Baffy G. Uncoupling protein-2 and cancer. Mitochondrion. 2010;10:243-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 98]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
54.  Esteves P, Pecqueur C, Ransy C, Esnous C, Lenoir V, Bouillaud F, Bulteau AL, Lombès A, Prip-Buus C, Ricquier D. Mitochondrial retrograde signaling mediated by UCP2 inhibits cancer cell proliferation and tumorigenesis. Cancer Res. 2014;74:3971-3982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 57]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
55.  Dalla Pozza E, Fiorini C, Dando I, Menegazzi M, Sgarbossa A, Costanzo C, Palmieri M, Donadelli M. Role of mitochondrial uncoupling protein 2 in cancer cell resistance to gemcitabine. Biochim Biophys Acta. 2012;1823:1856-1863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
56.  Kuai XY, Ji ZY, Zhang HJ. Mitochondrial uncoupling protein 2 expression in colon cancer and its clinical significance. World J Gastroenterol. 2010;16:5773-5778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Benhar M, Engelberg D, Levitzki A. ROS, stress-activated kinases and stress signaling in cancer. EMBO Rep. 2002;3:420-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 475]  [Cited by in F6Publishing: 455]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
58.  Lenaz G. The mitochondrial production of reactive oxygen species: mechanisms and implications in human pathology. IUBMB Life. 2001;52:159-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 387]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
59.  Horimoto M, Resnick MB, Konkin TA, Routhier J, Wands JR, Baffy G. Expression of uncoupling protein-2 in human colon cancer. Clin Cancer Res. 2004;10:6203-6207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 93]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
60.  Lentes KU, Tu N, Chen H, Winnikes U, Reinert I, Marmann G, Pirke KM. Genomic organization and mutational analysis of the human UCP2 gene, a prime candidate gene for human obesity. J Recept Signal Transduct Res. 1999;19:229-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 48]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
61.  Tu N, Chen H, Winnikes U, Reinert I, Marmann G, Pirke KM, Lentes KU. Structural organization and mutational analysis of the human uncoupling protein-2 (hUCP2) gene. Life Sci. 1999;64:PL41-PL50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 10]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
62.  Moreno-Muñoz D, de la Haba-Rodríguez JR, Conde F, López-Sánchez LM, Valverde A, Hernández V, Martínez A, Villar C, Gómez-España A, Porras I. Genetic variants in the renin-angiotensin system predict response to bevacizumab in cancer patients. Eur J Clin Invest. 2015;45:1325-1332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
63.  George AJ, Thomas WG, Hannan RD. The renin-angiotensin system and cancer: old dog, new tricks. Nat Rev Cancer. 2010;10:745-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 364]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
64.  Rodríguez-Pérez JC, Rodríguez-Esparragón F, Hernández-Perera O, Anabitarte A, Losada A, Medina A, Hernández E, Fiuza D, Avalos O, Yunis C. Association of angiotensinogen M235T and A(-6)G gene polymorphisms with coronary heart disease with independence of essential hypertension: the PROCAGENE study. Prospective Cardiac Gene. J Am Coll Cardiol. 2001;37:1536-1542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
65.  van der Knaap R, Siemes C, Coebergh JW, van Duijn CM, Hofman A, Stricker BH. Renin-angiotensin system inhibitors, angiotensin I-converting enzyme gene insertion/deletion polymorphism, and cancer: the Rotterdam Study. Cancer. 2008;112:748-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 102]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
66.  Srivastava K, Srivastava A, Mittal B. Angiotensin I-converting enzyme insertion/deletion polymorphism and increased risk of gall bladder cancer in women. DNA Cell Biol. 2010;29:417-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
67.  de Martino M, Klatte T, Schatzl G, Waldert M, Remzi M, Haitel A, Kramer G, Marberger M. Insertion/deletion polymorphism of angiotensin I-converting enzyme gene is linked with chromophobe renal cell carcinoma. Urology. 2011;77:1005.e9-1005.e13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
68.  Xie Y, You C, Chen J. An updated meta-analysis on association between angiotensin I-converting enzyme gene insertion/deletion polymorphism and cancer risk. Tumour Biol. 2014;35:6567-6579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
69.  Yang H, Cai C, Ye L, Rao Y, Wang Q, Hu D, Huang X. The relationship between angiotensin-converting enzyme gene insertion/deletion polymorphism and digestive cancer risk: Insights from a meta-analysis. J Renin Angiotensin Aldosterone Syst. 2015;16:1306-1313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
70.  Liu SY, Sima X, Wang CH, Gao M. The association between ACE polymorphism and risk of colorectal cancer in a Chinese population. Clin Biochem. 2011;44:1223-1226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
71.  Zambra FM, Biolchi V, de Cerqueira CC, Brum IS, Castelli EC, Chies JA. Immunogenetics of prostate cancer and benign hyperplasia--the potential use of an HLA-G variant as a tag SNP for prostate cancer risk. HLA. 2016;87:79-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
72.  Paul P, Rouas-Freiss N, Khalil-Daher I, Moreau P, Riteau B, Le Gal FA, Avril MF, Dausset J, Guillet JG, Carosella ED. HLA-G expression in melanoma: a way for tumor cells to escape from immunosurveillance. Proc Natl Acad Sci USA. 1998;95:4510-4515.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Amiot L, Ferrone S, Grosse-Wilde H, Seliger B. Biology of HLA-G in cancer: a candidate molecule for therapeutic intervention? Cell Mol Life Sci. 2011;68:417-431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 91]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
74.  Hviid TV, Hylenius S, Rørbye C, Nielsen LG. HLA-G allelic variants are associated with differences in the HLA-G mRNA isoform profile and HLA-G mRNA levels. Immunogenetics. 2003;55:63-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 285]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
75.  Chen S, Laverdiere I, Tourancheau A, Jonker D, Couture F, Cecchin E, Villeneuve L, Harvey M, Court MH, Innocenti F. A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients. Pharmacogenomics J. 2015;15:513-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
76.  Beutler E, Gelbart T, Demina A. Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci USA. 1998;95:8170-8174.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Barbarino JM, Haidar CE, Klein TE, Altman RB. PharmGKB summary: very important pharmacogene information for UGT1A1. Pharmacogenet Genomics. 2014;24:177-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 82]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
78.  Hu ZY, Yu Q, Pei Q, Guo C. Dose-dependent association between UGT1A1*28 genotype and irinotecan-induced neutropenia: low doses also increase risk. Clin Cancer Res. 2010;16:3832-3842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 98]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
79.  Hu ZY, Yu Q, Zhao YS. Dose-dependent association between UGT1A1*28 polymorphism and irinotecan-induced diarrhoea: a meta-analysis. Eur J Cancer. 2010;46:1856-1865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 71]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
80.  Dias MM, McKinnon RA, Sorich MJ. Impact of the UGT1A1*28 allele on response to irinotecan: a systematic review and meta-analysis. Pharmacogenomics. 2012;13:889-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
81.  Yang H, Sasaki T, Minoshima S, Shimizu N. Identification of three novel proteins (SGSM1, 2, 3) which modulate small G protein (RAP and RAB)-mediated signaling pathway. Genomics. 2007;90:249-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
82.  Lee IK, Kim KS, Kim H, Lee JY, Ryu CH, Chun HJ, Lee KU, Lim Y, Kim YH, Huh PW. MAP, a protein interacting with a tumor suppressor, merlin, through the run domain. Biochem Biophys Res Commun. 2004;325:774-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
83.  Wang C, Zhao H, Zhao X, Wan J, Wang D, Bi W, Jiang X, Gao Y. Association between an insertion/deletion polymorphism within 3’UTR of SGSM3 and risk of hepatocellular carcinoma. Tumour Biol. 2014;35:295-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
84.  Savas S, Liu G. Studying genetic variations in cancer prognosis (and risk): a primer for clinicians. Oncologist. 2009;14:657-666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
85.  Röcken C, Lendeckel U, Dierkes J, Westphal S, Carl-McGrath S, Peters B, Krüger S, Malfertheiner P, Roessner A, Ebert MP. The number of lymph node metastases in gastric cancer correlates with the angiotensin I-converting enzyme gene insertion/deletion polymorphism. Clin Cancer Res. 2005;11:2526-2530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 68]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
86.  Tang YI, Liu Y, Zhao W, Yu T, Yu H. Caspase-8 polymorphisms and risk of oral squamous cell carcinoma. Exp Ther Med. 2015;10:2267-2276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
87.  Hengartner MO. The biochemistry of apoptosis. Nature. 2000;407:770-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5259]  [Cited by in F6Publishing: 5104]  [Article Influence: 212.7]  [Reference Citation Analysis (0)]
88.  Siegel RM. Caspases at the crossroads of immune-cell life and death. Nat Rev Immunol. 2006;6:308-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 227]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
89.  Halder G, Johnson RL. Hippo signaling: growth control and beyond. Development. 2011;138:9-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 796]  [Cited by in F6Publishing: 800]  [Article Influence: 61.5]  [Reference Citation Analysis (0)]
90.  Harvey K, Tapon N. The Salvador-Warts-Hippo pathway - an emerging tumour-suppressor network. Nat Rev Cancer. 2007;7:182-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 512]  [Cited by in F6Publishing: 512]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
91.  Pan D. The hippo signaling pathway in development and cancer. Dev Cell. 2010;19:491-505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1640]  [Cited by in F6Publishing: 1741]  [Article Influence: 124.4]  [Reference Citation Analysis (0)]
92.  Na HK, Lee JY. Molecular Basis of Alcohol-Related Gastric and Colon Cancer. Int J Mol Sci. 2017;18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 84]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
93.  Fernandes GM, Russo A, Proença MA, Gazola NF, Rodrigues GH, Biselli-Chicote PM, Silva AE, Netinho JG, Pavarino ÉC, Goloni-Bertollo EM. CYP1A1, CYP2E1 and EPHX1 polymorphisms in sporadic colorectal neoplasms. World J Gastroenterol. 2016;22:9974-9983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
94.  Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat Rev Cancer. 2006;6:947-960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 668]  [Cited by in F6Publishing: 650]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
95.  Pfeifer GP, Denissenko MF, Olivier M, Tretyakova N, Hecht SS, Hainaut P. Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene. 2002;21:7435-7451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 802]  [Cited by in F6Publishing: 729]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
96.  Cury NM, Russo A, Galbiatti AL, Ruiz MT, Raposo LS, Maniglia JV, Pavarino EC, Goloni-Bertollo EM. Polymorphisms of the CYP1A1 and CYP2E1 genes in head and neck squamous cell carcinoma risk. Mol Biol Rep. 2012;39:1055-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 14]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
97.  Lu Y, Zhu X, Zhang C, Jiang K, Huang C, Qin X. Role of CYP2E1 polymorphisms in breast cancer: a systematic review and meta-analysis. Cancer Cell Int. 2017;17:11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
98.  Garziera M, Toffoli G. Inhibition of host immune response in colorectal cancer: human leukocyte antigen-G and beyond. World J Gastroenterol. 2014;20:3778-3794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
99.  Garziera M, Bidoli E, Cecchin E, Mini E, Nobili S, Lonardi S, Buonadonna A, Errante D, Pella N, D’Andrea M. HLA-G 3’UTR Polymorphisms Impact the Prognosis of Stage II-III CRC Patients in Fluoropyrimidine-Based Treatment. PLoS One. 2015;10:e0144000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
100.  Guo ZY, Lv YG, Wang L, Shi SJ, Yang F, Zheng GX, Wen WH, Yang AG. Predictive value of HLA-G and HLA-E in the prognosis of colorectal cancer patients. Cell Immunol. 2015;293:10-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 72]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
101.  Ye SR, Yang H, Li K, Dong DD, Lin XM, Yie SM. Human leukocyte antigen G expression: as a significant prognostic indicator for patients with colorectal cancer. Mod Pathol. 2007;20:375-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 125]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
102.  Zeestraten EC, Reimers MS, Saadatmand S, Goossens-Beumer IJ, Dekker JW, Liefers GJ, van den Elsen PJ, van de Velde CJ, Kuppen PJ. Combined analysis of HLA class I, HLA-E and HLA-G predicts prognosis in colon cancer patients. Br J Cancer. 2014;110:459-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 122]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
103.  Gao Y, He Y, Ding J, Wu K, Hu B, Liu Y, Wu Y, Guo B, Shen Y, Landi D. An insertion/deletion polymorphism at miRNA-122-binding site in the interleukin-1alpha 3’ untranslated region confers risk for hepatocellular carcinoma. Carcinogenesis. 2009;30:2064-2069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 120]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
104.  Trabert B, Pinto L, Hartge P, Kemp T, Black A, Sherman ME, Brinton LA, Pfeiffer RM, Shiels MS, Chaturvedi AK. Pre-diagnostic serum levels of inflammation markers and risk of ovarian cancer in the prostate, lung, colorectal and ovarian cancer (PLCO) screening trial. Gynecol Oncol. 2014;135:297-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
105.  Volonté A, Di Tomaso T, Spinelli M, Todaro M, Sanvito F, Albarello L, Bissolati M, Ghirardelli L, Orsenigo E, Ferrone S. Cancer-initiating cells from colorectal cancer patients escape from T cell-mediated immunosurveillance in vitro through membrane-bound IL-4. J Immunol. 2014;192:523-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 86]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
106.  Koller FL, Hwang DG, Dozier EA, Fingleton B. Epithelial interleukin-4 receptor expression promotes colon tumor growth. Carcinogenesis. 2010;31:1010-1017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 81]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
107.  Brooks CL, Gu W. p53 ubiquitination: Mdm2 and beyond. Mol Cell. 2006;21:307-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 619]  [Cited by in F6Publishing: 645]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
108.  Bi H, Tian T, Zhu L, Zhou H, Hu H, Liu Y, Li X, Hu F, Zhao Y, Wang G. Copy number variation of E3 ubiquitin ligase genes in peripheral blood leukocyte and colorectal cancer. Sci Rep. 2016;6:29869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
109.  Cerhan JR, Liu-Mares W, Fredericksen ZS, Novak AJ, Cunningham JM, Kay NE, Dogan A, Liebow M, Wang AH, Call TG. Genetic variation in tumor necrosis factor and the nuclear factor-kappaB canonical pathway and risk of non-Hodgkin’s lymphoma. Cancer Epidemiol Biomarkers Prev. 2008;17:3161-3169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
110.  Chen F, Castranova V, Shi X, Demers LM. New insights into the role of nuclear factor-kappaB, a ubiquitous transcription factor in the initiation of diseases. Clin Chem. 1999;45:7-17.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  McLure KG, Takagi M, Kastan MB. NAD+ modulates p53 DNA binding specificity and function. Mol Cell Biol. 2004;24:9958-9967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 66]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
112.  Riley T, Sontag E, Chen P, Levine A. Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol. 2008;9:402-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1374]  [Cited by in F6Publishing: 1394]  [Article Influence: 87.1]  [Reference Citation Analysis (0)]
113.  Sun Y, Zheng W, Guo Z, Ju Q, Zhu L, Gao J, Zhou L, Liu F, Xu Y, Zhan Q. A novel TP53 pathway influences the HGS-mediated exosome formation in colorectal cancer. Sci Rep. 2016;6:28083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
114.  Brosh R, Rotter V. When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer. 2009;9:701-713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 811]  [Cited by in F6Publishing: 831]  [Article Influence: 55.4]  [Reference Citation Analysis (0)]
115.  Siewchaisakul P, Suwanrungruang K, Poomphakwaen K, Wiangnon S, Promthet S. Lack of Association between an XRCC1 Gene Polymorphism and Colorectal Cancer Survival in Thailand. Asian Pac J Cancer Prev. 2016;17:2055-2060.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  de Boer JG. Polymorphisms in DNA repair and environmental interactions. Mutat Res. 2002;509:201-210.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Huang Y, Li X, He J, Chen L, Huang H, Liang M, Zhu Q, Huang Y, Wang L, Pan C. Genetic polymorphisms in XRCC1 genes and colorectal cancer susceptibility. World J Surg Oncol. 2015;13:244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]