Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Systematic Review and Meta-Analysis on the Association between IL-1B Polymorphisms and Cancer Risk

  • Jiali Xu ,

    Contributed equally to this work with: Jiali Xu, Zhiqiang Yin

    Affiliation Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

  • Zhiqiang Yin ,

    Contributed equally to this work with: Jiali Xu, Zhiqiang Yin

    Affiliation Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

  • Songyu Cao,

    Affiliation Department of Epidemiology and Biostatistics, MOE Key Laboratory of Modern Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China

  • Wen Gao,

    Affiliation Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

  • Lingxiang Liu,

    Affiliation Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

  • Yongmei Yin,

    Affiliation Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

  • Ping Liu,

    Affiliation Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

  • Yongqian Shu

    shuyongqian@csco.org.cn

    Affiliation Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China

Abstract

Background

Interleukin-1 beta (IL-1β), a pro-inflammatory cytokine, is emerging as a key mediator of carcinogenesis that characterizes host-environment interactions. Epidemiological studies investigating the association between two polymorphisms of IL-1B (−511C/T and +3954C/T) and cancer susceptibility have shown conflicting results. The aim of this study is to derive a more precise estimation of the relationship.

Methods

Related studies were identified through a systematic literature search of PubMed and Web of Science from their inception to September 15, 2012. Summary odds ratios (ORs) and 95% confidence intervals (CIs) for the IL-1B −511C/T and +3954C/T polymorphisms and cancer risk were calculated. Heterogeneity among studies and publication bias were also tested.

Results

The meta-analysis included 91 case-control studies in 85 publications, 81 studies for the −511C/T (19547 cases and 23935 controls) and 26 studies for the +3954C/T polymorphisms (8083 cases and 9183). The pooled results indicated that IL-1B +3954C/T (dominant model: OR = 1.15, 95% CI: 1.01–1.30) was significantly associated with increased overall cancer risk, especially among hospital-based case-control studies (dominant model: OR = 1.30, 95% CI: 1.02–1.66). As for −511C/T, we observed an inverse relationship in cervical cancer (dominant model: OR = 1.74, 95% CI: 1.35–2.23) and hepatocellular carcinoma (dominant model: OR = 0.68, 95% CI: 0.47–0.99). Moreover, −511C/T was associated with risk of specific subtypes of gastric carcinoma.

Conclusion

This meta-analysis suggested that both the IL-1B –511C/T and +3954C/T polymorphisms might modulate cancer susceptibility. Further well-designed studies based on larger sample sizes should be performed to confirm the findings.

Introduction

Cancer is considered to be a complex, multistep and fatal disease that results from interactions between environmental and genetic factors [1]. It is increasingly recognized that inflammation contributes to pathogenesis of many cancers [2], [3]. Chronic inflammation may result in oxidative stress and potentiate tumor promotion and progression [4][7]. Cytokines are glycoproteins or soluble proteins that act as mediators of inflammatory response and are integral to the function of immune cells. The role of cytokines in cancer immunity and carcinogenesis in general has been well established [8], [9]. They are aberrantly produced by tumor cells, macrophages and other phagocytic cells [10], [11]. These cytokines then activate transcription factors such as NF-κB, AP-1 and STAT3, thus inducing genes that stimulate cell proliferation and survival. In addition to enhancing the proliferation of mutated cells, an inflammatory microenvironment can also increase DNA mutation rates [12]. On the other hand, immune cells affect malignant cells through the production of cytokines, chemokines, reactive oxygen and others [11]. Thus, cytokines are particularly important in neoplastic initiation.

Interleukin-1 (IL-1) is a pro-inflammatory cytokine with multiple biological effects [13]. The IL-1 gene family on chromosome 2q13-14 encodes three proteins: IL-1α, IL-1β and their naturally occurring inhibitor IL-1RN. IL-1β, mainly produced by blood monocytes and tissue macrophages, has been implicated in mediating both acute and chronic inflammation [14]. Moreover, its property of stimulating the tumor microenvironment in favor of increased cell proliferation and tissue angiogenesis has been given much attention [15][18]. IL-1β is emerging as a key mediator of carcinogenesis that characterizes host-environment interactions.

The IL-1B gene is highly polymorphic and base transitions between C and T at positions –511 (C-T; dbSNP: rs16944), –31 (T-C; dbSNP: rs1143627) and +3954 (C-T; dbSNP: rs1143634) base pairs from the transcriptional site have been widely reported. In particular, the first two polymorphisms are located in the promoter region, and show high linkage disequilibrium [19], [20]. The IL-1B –31T/C substitution causes disruption of a TATA-box motif and has been found to markedly affect the binding affinity of several transcription factors [19], [21], [22] and thereby affect the transcription activity of IL-1B [21]. The IL-1B +3954 C/T in exon 5 is a synonymous single nucleotide polymorphism (SNP). In vitro studies have shown that both −511T and +3954T are associated with increased IL-1β secretion from lipopolysaccharide (LPS)-induced IL-1β protein secretion [23][25].

It is not surprising that these functionally important polymorphisms in the IL-1B gene might be associated with cancer susceptibility. Recently, a meta-analysis of IL-1B –31T/C polymorphism and cancer risk has suggested that the –31C allele is a low-penetrance protective factor for the development of cancer [26]. Furthermore, numerous epidemiological studies have investigated the association between IL-1B –511C/T and +3954C/T and different cancers, such as gastric, lung and breast cancers. However, the results remain inconsistent and inconclusive. A clearer understanding of the relationship between these two SNPs and cancer susceptibility is of clinical significance. In this report, a meta-analysis was conducted to provide an overview of all the relevant studies and synthesize conclusions on the associations between the IL-1B –511C/T, +3954C/T polymorphisms and cancer susceptibility.

Materials and Methods

Literature Search Strategy

Electronic databases (PubMed and Web of Science) were comprehensively searched using combinations of the terms “interleuk-1/IL-1” or “Interleuk-1B/IL-1B/IL-1 beta”, “polymorphism” and “cancer” or “tumor” (the last search update on September 15, 2012). For each identified study, additional studies were sought from its citations, references and from the database option “Related Articles”.

Inclusion and Exclusion Criteria

Eligible studies were selected according to the following explicit inclusion criteria: (i) case-control study evaluating the association between at least one of the two polymorphisms (IL-1B −511C/T and +3954C/T) and cancer susceptibility, (ii) sufficient genotype data presented to calculated the odds ratios (ORs) and 95% confidence intervals (CIs). Major reasons for exclusion of studies were: (i) only cancer group, (ii) no usable genotype frequency data, (iii) duplicate of earlier publication, (iv) publication not in English.

Data Extraction and Quality Assessment

Two investigators (Jiali Xu and Zhiqiang Yin) extracted information from all eligible publications independently according to the criteria listed above. The following information was gathered from each study: the first author’s name, year of publication, cancer type, country of origin, ethnicity of subjects, source of control, genotyping method, number of cases and controls and genotype frequency (Table S1). The quality assessment of each study was carried out independently by the two investigators using the Newcastle-Ottawa Scale (NOS) [27]. Studies with a score equal to or higher than 5 were considered “high-quality”, whereas those scored less than 4 were considered “low-quality”. All item-specific ambiguities were discussed by investigators’ consultation until consensus was achieved.

Statistical Analysis

We first calculated Hardy–Weinberg equilibrium (HWE) in the controls for each included study using a goodness-of-fit test and for which P<0.05 was considered not compliant with HWE. The strength of the association between the two SNPs (IL-1B −511C/T and +3954C/T) and cancer risk was measured by ORs with 95% CIs. Pooled ORs were obtained from combination of single studies by homozygote comparison (TT vs. CC), heterozygote comparison (CT vs. CC), dominant model (TT+CT vs. CC) and recessive model (CC+CT vs. TT), respectively. The heterogeneity among different studies was checked by the Q-test [28]. If the P value is <0.10, a random effect model with the DerSimonian and Laird method was used to pool the results. Otherwise, a fixed-effect model with the Mantel-Haenszel method was then used [29].

Sensitivity analysis was performed to assess the stability of the results by omitting a single study in this meta-analysis each time to reflect the influence of individual data on the pooled OR. To further explore the potential source of heterogeneity among studies and test the effects of study characteristics on the overall estimates, subgroup analyses and meta regression were performed stratified by cancer types (if one cancer type contained fewer than three individual studies, it was grouped into the “other cancers”), ethnicity (Caucasian, Asian or Others; Others included African and Mixed populations), and source of controls (population-based or hospital-based).

Publication bias was diagnosed with Begg’s funnel plot and Egger’s linear regression method [30]. Asymmetric or incomplete funnel shaped plots and P<0.05 in Egger’s test indicated the presence of potential publication bias. All statistical analyses used STATA 12.0 (STATA Corp, College Station, Texas). Except for heterogeneity statistics (where significance was declared if P<0.10), each P-value <0.05 was considered to be statistically significant. All P-values were 2-sided.

Results

Flow of Included Studies

Figure 1 depicts the results of the literature search. After review of abstracts, 131 full-text potentially relevant publications were retrieved to be assessed for eligibility. Among the 46 full-text articles excluded, one article [20] was updated by a new publication [31], and two case-control studies were considered “low-quality” (score = 3 and 4) using the NOS quality assessment instrument and thus were excluded [32], [33]. By contrast, five articles each mentioned two or more independent case-control studies, and studies included in these articles were treated as separate studies [34][38]. Finally, a total of 91 case-control studies in 85 publications, of which there were 81 studies for the −511C/T polymorphism [19], [31], [35], [37][83],[34], [36], [84][106] and 26 studies for the +3954C/T polymorphism [19], [34], [36], [42], [50], [51], [54], [56], [61], [65], [74], [81], [82], [105], [107][116], were included in the meta-analysis based on our search strategy and eligibility criteria.

Study Characteristics

Detailed characteristics of the aggregated data for 91 case-control studies are summarized in Table S1. The vast majority of the included studies – all except 14 studies for –511C/T and 2 studies for +3954C/T – indicated that the genotype distributions in the controls were consistent with HWE. There were 47 studies of subjects of Asian descent, 32 studies of subjects of Caucasian descent and 12 studies of subjects with ethnicity “Others” (7 mixed-ethnicity studies and 3 African studies). Minor allele frequencies (MAFs) of −511C/T and +3954C/T of controls in different populations are graphed as Figure S1.

Quantitative Data Synthesis

Association of the IL-1B –511C/T polymorphism with cancer susceptibility: Analyses of 81 case-control studies including 19547 cases and 23935 controls were conducted to explore the relationship between −511C/T and cancer risk (Table 1). Overall, no significant association was detected in any genetic model (homozygote comparison: OR = 1.09, 95% CI: 0.96–1.23; dominant model: OR = 1.04, 95% CI: 0.95–1.13). The pooled estimates remained stable when restricted to studies not deviating from HWE. Intriguingly, in the subgroup analysis stratified by cancer type, the IL-1B -511T allele showed evidence of an association with increased cervical cancer risk (dominant model: OR = 1.74, 95% CI: 1.35–2.23), but demonstrated a protective role in the development of hepatocellular carcinoma (dominant model: OR = 0.68, 95% CI: 0.47–0.99). Increased risk was also observed in blood cancers (recessive model: OR = 1.19, 95% CI: 1.04–1.37).

thumbnail
Table 1. Stratification analyses of the IL-1B –511C/T polymorphism on cancer susceptibility.

https://doi.org/10.1371/journal.pone.0063654.t001

The data were additionally stratified for gastric cancer in the dominant model (Table 2). The association became significant when excluded studies not in HWE (OR = 1.16, 95% CI: 1.02–1.32). Statistically significant findings were also found in population-based case-control studies (OR = 1.20, 95% CI: 1.00–1.43) but not in analysis stratified by ethnicities. When gastric carcinoma was classified according to tumor site (cardia or non-cardia) and histopathology subtypes (intestinal or diffuse/mixed), significant associations were detected in non-cardia gastric cancer (OR = 1.57, 95% CI: 1.06–2.31) and intestinal gastric cancer (OR = 1.24, 95% CI: 1.04–1.49).

thumbnail
Table 2. Stratification analyses of the IL-1B –511C/T polymorphism on gastric cancer susceptibility.

https://doi.org/10.1371/journal.pone.0063654.t002

The genotype distribution of −511C/T among cases and infection-match controls was available in 20 studies that investigated gastric cancer infected by Helicobacter pylori (HP). However, there were no significant gene-environment interactions in this case.

Association of the IL-1B +3954C/T polymorphism with cancer susceptibility: The analysis eventually included 26 case-control studies with 8083 cases and 9183 controls for IL-1B +3954C/T. Table 3 presents the main results of the pooled analysis and Figure 2 shows the association between +3954C/T and cancer risk in the form of forest plots. Overall, the results of combined analyses of all studies suggested that the +3954C/T polymorphism was significantly associated with increased cancer susceptibility in the dominant model (OR = 1.15, 95% CI: 1.01–1.30). This association remained consistently strong when limited to studies in HWE (heterozygote comparison: OR = 1.14, 95% CI: 1.00–1.31; dominant model: OR = 1.16, 95% CI: 1.02–1.33). When stratified by source of controls, hospital-based studies exhibited significantly increased risk (dominant model: OR = 1.30, 95% CI: 1.02–1.66).

thumbnail
Figure 2. Forest plots describing the association of the IL-1B +3954C/T polymorphism with cancer susceptibility in the dominant model.

The squares and horizontal lines correspond to the OR and 95% CI for each study. The area of the squares reflects the weight. The diamond represents the summary OR and 95% CI.

https://doi.org/10.1371/journal.pone.0063654.g002

thumbnail
Table 3. Stratification analyses of the IL-1B +3954C/T polymorphism on cancer susceptibility.

https://doi.org/10.1371/journal.pone.0063654.t003

Meta Regression

Since there was significant heterogeneity for IL-1B –511C/T in all genetic models, a univariable regression was conducted to explore the predefined possible source of heterogeneity (Table S2). We identified that it is variability in MAF of this polymorphism across different ethnicities that was a significant source of heterogeneity (dominant model:β coefficient = −1.60 (−2.67 to −0.47), P = 0.006; recessive model: β coefficient = −1.40 (−2.45 to −0.35), P = 0.010), but not cancer type or source of control. Using dominant model, we also did meta regression for +3954C/T (Table S3). However, cancer type (P = 0.816), MAF (P = 0.050) and source of control (P = 0.308) only explained little of the heterogeneity.

Sensitivity Analyses and Publication Bias

The influence of each study on the pooled OR was examined by repeating the meta-analyses while sequentially omitting individual studies. Both sensitivity analyses for −511C/T and +3954C/T indicated that no single study influenced the pooled ORs qualitatively, suggesting that the results of our meta-analyses are robust and stable. The shape of funnel plots was symmetrical for −511C/T, and the Egger’s test P-value were 0.628, 0.788, 0.888 and 0.579 for homozygote comparison, heterozygote comparison, dominant model and recessive model, respectively (Figure 3). As for the +3954C/T polymorphism, symmetrical funnel plots were found in homozygote comparison (P = 0.382) and recessive model (P = 0.509), but not in heterozygote comparison and dominant model (Egger’s test P = 0.026 and 0.020, respectively, Figure 3), implying the existence of publication bias.

thumbnail
Figure 3. Begg’s funnel plots of publication bias.

Each study is represented by a circle, the area of which presents the study’s precision. A, funnel plot for IL-1B −511C/T in the recessive model (Egger’s test P = 0.579). B, funnel plot for IL-1B +3954C/T in the dominant model (Egger’s test P = 0.020).

https://doi.org/10.1371/journal.pone.0063654.g003

The PRISMA Checklist for our study is shown as Table S4 [117].

Discussion

Large sample epidemiological studies of predisposition gene polymorphisms could provide insight into the in vivo association between candidate genes and diseases. The present meta-analysis, including 91 high-quality case-control studies, is the most comprehensive meta-analysis to have evaluated the IL-1B polymorphisms (−511C/T and +3954C/T) and their relationship to cancer susceptibility. Its strength was based on the accumulation of published data providing greater information to detect significant differences. Our results demonstrated that IL-1B +3954C/T was significantly associated with increased overall cancer risk, especially among hospital-based case-control studies. No significant association was observed between −511C/T and overall cancer risk. However, results from subgroup analyses indicated that −511C/T was associated with susceptibility of certain types of cancer. Given the critical roles of IL-1β in inflammation and carcinogenesis, it is possible that both IL-1B −511C/T and +3954C/T polymorphisms may modulate the risk of cancer development.

Cumulative evidence suggests that IL-1β plays an important role in tumorigenesis and development. It is well known that IL-1 expression is elevated in most human cancers. High plasma IL-1β levels are associated with a significantly increased risk of cancer, and tumor patients with high IL-1 expression have worse prognosis than those without [99], [115], [118]. IL-1β promotes invasiveness, including tumor angiogenesis, and also induces immune suppression in the host [16], [17]. Moreover, IL-1β has been found to combine with estrogen receptor (ER)α in breast cancer cells, resulting in transcriptional activation [119]. In pancreatic cancer cells, IL-1β has been shown to mediate adhesion and invasion, as well as modulating chemoresistance by activating the NF- κB and ERK signaling pathways [120][123]. IL-1β can also attenuate interferon-induced antiviral activity and STAT1 activation in the liver, and modulate immune responses in hepatitis virus-related hepatocellular carcinoma [43], [124].

In the subgroup analysis according to cancer type, a significant association was detected between the -511T variant allele polymorphism and increased cervical cancer risk. Interestingly, this allele seemed to be a protective factor for the development of hepatocellular carcinoma. Some possible reasons may explain this discrepancy. First, the relatively small number of eligible studies in each subgroup might induce significant/insignificant association by chance due to insufficient statistical power. Second, there are some conflicting data regarding the effect of the IL-1B −511/−31 haplotype on gene expression. Ex vivo blood stimulation assay suggests that the -511T/−31C haplotype is significantly associated with an increase in LPS-induced IL-1β intracellular secretion [23]. IL-1B -511T/−31C is associated with a high level of IL-1β in the plasma [125]. However, the IL-1B -31 polymorphism involves a TATA-box motif and the -31T allele is associated with a five-fold elevated binding activity with the transcription initiation factor [19], [21], [22]. In vivo study indicates that a disease-related haplotype including −511C/−31T has higher IL-1B mRNA expression in the lungs compared with the non-risk haplotype [115]. In HP-infected gastric cancer patients, the mucosal IL-1B level is higher in −31T carriers than in −31C carriers [57]. It is likely that gene expression in each organ, such as the cervix and liver, is differently regulated. Functional study in specific tissue is required for better understanding of the role of IL-1B genotype in carcinogenesis.

Our results for gastric cancer were partly consistent with results from a previous meta-analysis performed by Xue et al. [126]. Both our meta-analysis and Xue’s demonstrated no association between IL-1B +3954C/T and gastric cancer risk. They found IL-1B −511T carriers were associated with a significantly increased risk of gastric cancer. However, we observed significant associations only among population-based studies. Our work may be interpreted as an update of Xue’s, because another 30 or more studies concerning −511C/T or +3954C/T polymorphisms and gastric susceptibility with inconsistent results have been published in the past 2 years. We also found that −511T carriers are significantly associated with risk of the non-cardia and intestinal types of gastric cancer, whereas association was not detected in HP-positive group nor in the HP-negative group, and this is consistent with Xue’s meta-analysis. Further in-depth research focused on IL-1B –511C/T and specific subtypes of gastric cancer is warranted.

In the stratification analysis of source of control, significantly increased risk between the IL-1B +3954C/T polymorphism and cancer risk was detected among hospital-based studies, but not among population-based studies. This may be due to some selection biases existing in hospital-based studies because such controls might come from a population with a related disease and may not be a representative of the general population, especially when there is a relationship between the investigated genotypes and the disease conditions that hospital-based controls might have. Although it is more convenient to recruit hospital controls, to reduce biases in such genetic association studies it may be preferable to use population controls.

Significant heterogeneity existed among studies for IL–511C/T in all comparisons, and meta regression indicated that MAF in different ethnicities partly accounts for the heterogeneity. Although we observed a wide variation of -511T allele frequency of control resources in different populations, and the MAF might reflect an environmental impact on gene distribution, subgroup analysis stratified by ethnicity did not detect any significant association between –511C/T and cancer risk in Asians nor in Caucasians. Cancer is a multi-factorial disease resulting from complex interactions between environmental and genetic factors [1]. Some other factors may weaken the effect of IL-1B –511C/T on cancer risk in different ethnic groups.

Some limitations of our meta-analysis should be mentioned. Our results were based on unadjusted estimates because of the absence of available information. If more detailed individual data such as age, sex and exposure were available, a more precise analysis would be performed. Additionally, both the asymmetric funnel plot and Egger’s test indicated the existence of publication bias in two comparisons for the IL-1B +3954C/T polymorphism. This may be explained by the restriction of the meta-analysis to studies published in English. In spite of these limitations, our meta-analysis also had some advantages. First, it is the most comprehensive meta-analysis to evaluate the two polymorphisms (−511C/T and +3954C/T) and their relationship to cancer susceptibility. The number of cases and controls through the pooled studies could significantly increase the statistical power of the analysis. Second, all included studies had acceptable quality (scored at least 5).

Conclusions

In conclusion, this meta-analysis indicated that IL-1B +3954C/T was associated with significantly increased overall cancer risk, especially among hospital-based case-control studies. The IL-1B −511T allele showed evidence of an association with increased cervical cancer risk but demonstrated a protective role in the development of hepatocellular carcinoma. Moreover, −511C/T was associated with risk of specific subtypes of gastric carcinoma. Further large-sample research should use standardized unbiased homogeneous cancer patients and well-matched controls to confirm our findings. Additionally, gene-environment interactions should be considered in future studies.

Supporting Information

Figure S1.

Minor allele frequencies of IL-1B −511C/T and +3954C/T polymorphisms among ethnicities of Asian, Caucasian and Others in controls.

https://doi.org/10.1371/journal.pone.0063654.s001

(TIF)

Table S1.

Main characteristics of the selected studies.

https://doi.org/10.1371/journal.pone.0063654.s002

(DOC)

Table S2.

Results of random effect meta-regression for search of source of heterogeneity for IL-1B –511C/T.

https://doi.org/10.1371/journal.pone.0063654.s003

(DOC)

Table S3.

Results of random effect meta-regression for search of source of heterogeneity for IL-1B +3954C/T in the dominant model.

https://doi.org/10.1371/journal.pone.0063654.s004

(DOC)

Table S4.

PRISMA Checklist for current meta-analysis.

https://doi.org/10.1371/journal.pone.0063654.s005

(DOC)

Author Contributions

Conceived and designed the experiments: JX. Performed the experiments: JX ZY. Analyzed the data: JX ZY SC. Wrote the paper: JX. Helped to answer reviewers’ comments: SC WG LL YP PL. Provided experimental guidance and helped to answer reviewers’ comments: YS.

References

  1. 1. Pharoah PD, Dunning AM, Ponder BA, Easton DF (2004) Association studies for finding cancer-susceptibility genetic variants. Nat Rev Cancer 4: 850–860.
  2. 2. Coussens LM, Werb Z (2002) Inflammation and cancer. Nature 420: 860–867.
  3. 3. Schottenfeld D, Beebe-Dimmer J (2006) Chronic inflammation: a common and important factor in the pathogenesis of neoplasia. CA Cancer J Clin 56: 69–83.
  4. 4. Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A (2009) Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30: 1073–1081.
  5. 5. Germano G, Allavena P, Mantovani A (2008) Cytokines as a key component of cancer-related inflammation. Cytokine 43: 374–379.
  6. 6. Mantovani A (2010) Molecular pathways linking inflammation and cancer. Curr Mol Med 10: 369–373.
  7. 7. Mantovani A, Savino B, Locati M, Zammataro L, Allavena P, et al. (2010) The chemokine system in cancer biology and therapy. Cytokine Growth Factor Rev 21: 27–39.
  8. 8. Smyth MJ, Cretney E, Kershaw MH, Hayakawa Y (2004) Cytokines in cancer immunity and immunotherapy. Immunol Rev 202: 275–293.
  9. 9. Seruga B, Zhang H, Bernstein LJ, Tannock IF (2008) Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer 8: 887–899.
  10. 10. Jin P, Panelli MC, Marincola FM, Wang E (2004) Cytokine polymorphism and its possible impact on cancer. Immunol Res 30: 181–190.
  11. 11. Kurzrock R (2001) Cytokine deregulation in cancer. Biomed Pharmacother 55: 543–547.
  12. 12. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell 140: 883–899.
  13. 13. Dinarello CA (1996) Biologic basis for interleukin-1 in disease. Blood 87: 2095–2147.
  14. 14. Bird S, Zou J, Wang T, Munday B, Cunningham C, et al. (2002) Evolution of interleukin-1beta. Cytokine Growth Factor Rev 13: 483–502.
  15. 15. Apte RN, Voronov E (2008) Is interleukin-1 a good or bad ‘guy’ in tumor immunobiology and immunotherapy? Immunol Rev 222: 222–241.
  16. 16. Saijo Y, Tanaka M, Miki M, Usui K, Suzuki T, et al. (2002) Proinflammatory cytokine IL-1 beta promotes tumor growth of Lewis lung carcinoma by induction of angiogenic factors: in vivo analysis of tumor-stromal interaction. J Immunol 169: 469–475.
  17. 17. Song X, Voronov E, Dvorkin T, Fima E, Cagnano E, et al. (2003) Differential effects of IL-1 alpha and IL-1 beta on tumorigenicity patterns and invasiveness. J Immunol 171: 6448–6456.
  18. 18. Voronov E, Shouval DS, Krelin Y, Cagnano E, Benharroch D, et al. (2003) IL-1 is required for tumor invasiveness and angiogenesis. Proc Natl Acad Sci U S A 100: 2645–2650.
  19. 19. El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, et al. (2000) Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature 404: 398–402.
  20. 20. Machado JC, Pharoah P, Sousa S, Carvalho R, Oliveira C, et al. (2001) Interleukin 1B and interleukin 1RN polymorphisms are associated with increased risk of gastric carcinoma. Gastroenterology 121: 823–829.
  21. 21. Chen H, Wilkins LM, Aziz N, Cannings C, Wyllie DH, et al. (2006) Single nucleotide polymorphisms in the human interleukin-1B gene affect transcription according to haplotype context. Hum Mol Genet 15: 519–529.
  22. 22. Lind H, Haugen A, Zienolddiny S (2007) Differential binding of proteins to the IL1B -31 T/C polymorphism in lung epithelial cells. Cytokine 38: 43–48.
  23. 23. Hall SK, Perregaux DG, Gabel CA, Woodworth T, Durham LK, et al. (2004) Correlation of polymorphic variation in the promoter region of the interleukin-1 beta gene with secretion of interleukin-1 beta protein. Arthritis Rheum 50: 1976–1983.
  24. 24. Pociot F, Molvig J, Wogensen L, Worsaae H, Nerup J (1992) A TaqI polymorphism in the human interleukin-1 beta (IL-1 beta) gene correlates with IL-1 beta secretion in vitro. Eur J Clin Invest 22: 396–402.
  25. 25. Hernandez-Guerrero C, Monzon-Bordonaba F, Jimenez-Zamudio L, Ahued-Ahued R, Arechavaleta-Velasco F, et al. (2003) In-vitro secretion of proinflammatory cytokines by human amniochorion carrying hyper-responsive gene polymorphisms of tumour necrosis factor-alpha and interleukin-1beta. Mol Hum Reprod 9: 625–629.
  26. 26. He B, Zhang Y, Pan Y, Xu Y, Gu L, et al. (2011) Interleukin 1 beta (IL1B) promoter polymorphism and cancer risk: evidence from 47 published studies. Mutagenesis 26: 637–642.
  27. 27. Wells GA, Shea B, O’Connell D, Petersen J, Welch V (Accessed 2012 Sep 30.) The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomized studies in meta-analyses. Available: http://wwwohrica/programs/clinical_epidemiology/oxfordhtm.
  28. 28. Handoll HH (2006) Systematic reviews on rehabilitation interventions. Arch Phys Med Rehabil 87: 875.
  29. 29. Higgins JP, Thompson SG (2002) Quantifying heterogeneity in a meta-analysis. Stat Med 21: 1539–1558.
  30. 30. Peters JL, Sutton AJ, Jones DR, Abrams KR, Rushton L (2006) Comparison of two methods to detect publication bias in meta-analysis. Jama 295: 676–680.
  31. 31. Machado JC, Figueiredo C, Canedo P, Pharoah P, Carvalho R, et al. (2003) A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology 125: 364–371.
  32. 32. Gatti LL, Burbano RR, de Assumpcao PP, Smith Mde A, Payao SL (2004) Interleukin-1beta polymorphisms, Helicobacter pylori infection in individuals from Northern Brazil with gastric adenocarcinoma. Clin Exp Med 4: 93–98.
  33. 33. zur Hausen A, Crusius JB, Murillo LS, Alizadeh BZ, Morre SA, et al. (2003) IL-1B promoter polymorphism and Epstein-Barr virus in Dutch patients with gastric carcinoma. Int J Cancer 107: 866–867.
  34. 34. Persson C, Engstrand L, Nyren O, Hansson LE, Enroth H, et al. (2009) Interleukin 1-beta gene polymorphisms and risk of gastric cancer in Sweden. Scand J Gastroenterol 44: 339–345.
  35. 35. Ito H, Kaneko K, Makino R, Konishi K, Kurahashi T, et al. (2007) Interleukin-1beta gene in esophageal, gastric and colorectal carcinomas. Oncol Rep 18: 473–481.
  36. 36. Zabaleta J, Lin HY, Sierra RA, Hall MC, Clark PE, et al. (2008) Interactions of cytokine gene polymorphisms in prostate cancer risk. Carcinogenesis 29: 573–578.
  37. 37. Wu MS, Chen LT, Shun CT, Huang SP, Chiu HM, et al. (2004) Promoter polymorphisms of tumor necrosis factor-alpha are associated with risk of gastric mucosa-associated lymphoid tissue lymphoma. Int J Cancer 110: 695–700.
  38. 38. El-Omar EM, Rabkin CS, Gammon MD, Vaughan TL, Risch HA, et al. (2003) Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology 124: 1193–1201.
  39. 39. Hwang IR, Kodama T, Kikuchi S, Sakai K, Peterson LE, et al. (2002) Effect of interleukin 1 polymorphisms on gastric mucosal interleukin 1beta production in Helicobacter pylori infection. Gastroenterology 123: 1793–1803.
  40. 40. Wang Y, Kato N, Hoshida Y, Yoshida H, Taniguchi H, et al. (2003) Interleukin-1beta gene polymorphisms associated with hepatocellular carcinoma in hepatitis C virus infection. Hepatology 37: 65–71.
  41. 41. Howell WM, Turner SJ, Theaker JM, Bateman AC (2003) Cytokine gene single nucleotide polymorphisms and susceptibility to and prognosis in cutaneous malignant melanoma. Eur J Immunogenet 30: 409–414.
  42. 42. Zeng ZR, Hu PJ, Hu S, Pang RP, Chen MH, et al. (2003) Association of interleukin 1B gene polymorphism and gastric cancers in high and low prevalence regions in China. Gut 52: 1684–1689.
  43. 43. Tanaka Y, Furuta T, Suzuki S, Orito E, Yeo AE, et al. (2003) Impact of interleukin-1beta genetic polymorphisms on the development of hepatitis C virus-related hepatocellular carcinoma in Japan. J Infect Dis 187: 1822–1825.
  44. 44. Zhang Z, Liu W, Jia X, Gao Y, Hemminki K, et al. (2004) Use of pyrosequencing to detect clinically relevant polymorphisms of genes in basal cell carcinoma. Clin Chim Acta 342: 137–143.
  45. 45. Yang J, Hu Z, Xu Y, Shen J, Niu J, et al. (2004) Interleukin-1B gene promoter variants are associated with an increased risk of gastric cancer in a Chinese population. Cancer Lett 215: 191–198.
  46. 46. Chen A, Li CN, Hsu PI, Lai KH, Tseng HH, et al. (2004) Risks of interleukin-1 genetic polymorphisms and Helicobacter pylori infection in the development of gastric cancer. Aliment Pharmacol Ther 20: 203–211.
  47. 47. Zienolddiny S, Ryberg D, Maggini V, Skaug V, Canzian F, et al. (2004) Polymorphisms of the interleukin-1 beta gene are associated with increased risk of non-small cell lung cancer. Int J Cancer 109: 353–356.
  48. 48. Lee KA, Ki CS, Kim HJ, Sohn KM, Kim JW, et al. (2004) Novel interleukin 1beta polymorphism increased the risk of gastric cancer in a Korean population. J Gastroenterol 39: 429–433.
  49. 49. Grimm C, Berger I, Tomovski C, Zeillinger R, Concin N, et al. (2004) A polymorphism of the interleukin-1 receptor antagonist plays a prominent role within the interleukin-1 gene cluster in vulvar carcinogenesis. Gynecol Oncol 92: 936–940.
  50. 50. Hartland S, Newton JL, Griffin SM, Donaldson PT (2004) A functional polymorphism in the interleukin-1 receptor-1 gene is associated with increased risk of Helicobacter pylori infection but not with gastric cancer. Dig Dis Sci 49: 1545–1550.
  51. 51. Glas J, Torok HP, Schneider A, Brunnler G, Kopp R, et al. (2004) Allele 2 of the interleukin-1 receptor antagonist gene is associated with early gastric cancer. J Clin Oncol 22: 4746–4752.
  52. 52. Tsai FJ, Chang CH, Chen CC, Hsia TC, Chen HY, et al. (2005) Interleukin-4 gene intron-3 polymorphism is associated with transitional cell carcinoma of the urinary bladder. BJU Int 95: 432–435.
  53. 53. Sakuma K, Uozaki H, Chong JM, Hironaka M, Sudo M, et al. (2005) Cancer risk to the gastric corpus in Japanese, its correlation with interleukin-1beta gene polymorphism (+3953*T) and Epstein-Barr virus infection. Int J Cancer 115: 93–97.
  54. 54. Alpizar-Alpizar W, Perez-Perez GI, Une C, Cuenca P, Sierra R (2005) Association of interleukin-1B and interleukin-1RN polymorphisms with gastric cancer in a high-risk population of Costa Rica. Clin Exp Med 5: 169–176.
  55. 55. Perri F, Piepoli A, Bonvicini C, Gentile A, Quitadamo M, et al. (2005) Cytokine gene polymorphisms in gastric cancer patients from two Italian areas at high and low cancer prevalence. Cytokine 30: 293–302.
  56. 56. Zhang WH, Wang XL, Zhou J, An LZ, Xie XD (2005) Association of interleukin-1B (IL-1B) gene polymorphisms with risk of gastric cancer in Chinese population. Cytokine 30: 378–381.
  57. 57. Chang YW, Jang JY, Kim NH, Lee JW, Lee HJ, et al. (2005) Interleukin-1B (IL-1B) polymorphisms and gastric mucosal levels of IL-1beta cytokine in Korean patients with gastric cancer. Int J Cancer 114: 465–471.
  58. 58. Lu W, Pan K, Zhang L, Lin D, Miao X, et al. (2005) Genetic polymorphisms of interleukin (IL)-1B, IL-1RN, IL-8, IL-10 and tumor necrosis factor {alpha} and risk of gastric cancer in a Chinese population. Carcinogenesis 26: 631–636.
  59. 59. Ruzzo A, Graziano F, Pizzagalli F, Santini D, Battistelli V, et al. (2005) Interleukin 1B gene (IL-1B) and interleukin 1 receptor antagonist gene (IL-1RN) polymorphisms in Helicobacter pylori-negative gastric cancer of intestinal and diffuse histotype. Ann Oncol 16: 887–892.
  60. 60. Tsai MH, Chen WC, Tsai CH, Hang LW, Tsai FJ (2005) Interleukin-4 gene, but not the interleukin-1 beta gene polymorphism, is associated with oral cancer. J Clin Lab Anal 19: 93–98.
  61. 61. Chen CC, Yang SY, Liu CJ, Lin CL, Liaw YF, et al. (2005) Association of cytokine and DNA repair gene polymorphisms with hepatitis B-related hepatocellular carcinoma. Int J Epidemiol 34: 1310–1318.
  62. 62. Taguchi A, Ohmiya N, Shirai K, Mabuchi N, Itoh A, et al. (2005) Interleukin-8 promoter polymorphism increases the risk of atrophic gastritis and gastric cancer in Japan. Cancer Epidemiol Biomarkers Prev 14: 2487–2493.
  63. 63. Hefler LA, Grimm C, Lantzsch T, Lampe D, Leodolter S, et al. (2005) Interleukin-1 and interleukin-6 gene polymorphisms and the risk of breast cancer in caucasian women. Clin Cancer Res 11: 5718–5721.
  64. 64. Asada M, Yasuda H, Ebihara S, Tomita N, Suzuki S, et al. (2006) Interleukin-1beta gene polymorphisms associated with risk of lung cancer in Japanese. Lung Cancer 54: 261–263.
  65. 65. Balasubramanian SP, Azmy IA, Higham SE, Wilson AG, Cross SS, et al. (2006) Interleukin gene polymorphisms and breast cancer: a case control study and systematic literature review. BMC Cancer 6: 188.
  66. 66. Kim N, Cho SI, Yim JY, Kim JM, Lee DH, et al. (2006) The effects of genetic polymorphisms of IL-1 and TNF-A on Helicobacter pylori-induced gastroduodenal diseases in Korea. Helicobacter 11: 105–112.
  67. 67. Kamangar F, Abnet CC, Hutchinson AA, Newschaffer CJ, Helzlsouer K, et al. (2006) Polymorphisms in inflammation-related genes and risk of gastric cancer (Finland). Cancer Causes Control 17: 117–125.
  68. 68. Starzynska T, Ferenc K, Wex T, Kahne T, Lubinski J, et al. (2006) The association between the interleukin-1 polymorphisms and gastric cancer risk depends on the family history of gastric carcinoma in the study population. Am J Gastroenterol 101: 248–254.
  69. 69. Rothman N, Skibola CF, Wang SS, Morgan G, Lan Q, et al. (2006) Genetic variation in TNF and IL10 and risk of non-Hodgkin lymphoma: a report from the InterLymph Consortium. Lancet Oncol 7: 27–38.
  70. 70. Hirankarn N, Kimkong I, Kummee P, Tangkijvanich P, Poovorawan Y (2006) Interleukin-1beta gene polymorphism associated with hepatocellular carcinoma in hepatitis B virus infection. World J Gastroenterol 12: 776–779.
  71. 71. Ikehara SK, Ikehara Y, Matsuo K, Hirose K, Niwa T, et al. (2006) A polymorphism of C-to-T substitution at -31 IL1B is associated with the risk of advanced gastric adenocarcinoma in a Japanese population. J Hum Genet 51: 927–933.
  72. 72. Yamada S, Matsuhisa T, Makonkawkeyoon L, Chaidatch S, Kato S, et al. (2006) Helicobacter pylori infection in combination with the serum pepsinogen I/II ratio and interleukin-1beta-511 polymorphisms are independent risk factors for gastric cancer in Thais. J Gastroenterol 41: 1169–1177.
  73. 73. Liu J, Zhai X, Jin G, Hu Z, Wang S, et al. (2006) Functional variants in the promoter of interleukin-1beta are associated with an increased risk of breast cancer: a case-control analysis in a Chinese population. Int J Cancer 118: 2554–2558.
  74. 74. Michaud DS, Daugherty SE, Berndt SI, Platz EA, Yeager M, et al. (2006) Genetic polymorphisms of interleukin-1B (IL-1B), IL-6, IL-8, and IL-10 and risk of prostate cancer. Cancer Res 66: 4525–4530.
  75. 75. Sugimoto M, Furuta T, Shirai N, Nakamura A, Xiao F, et al. (2007) Different effects of polymorphisms of tumor necrosis factor-alpha and interleukin-1 beta on development of peptic ulcer and gastric cancer. J Gastroenterol Hepatol 22: 51–59.
  76. 76. Kang S, Kim JW, Park NH, Song YS, Park SY, et al. (2007) Interleukin-1 beta-511 polymorphism and risk of cervical cancer. J Korean Med Sci 22: 110–113.
  77. 77. Sun H, Wang Y, Ma X, Pei F, Sun H, et al. (2007) A method of oligochip for single nucleotide polymorphism genotyping in the promoter region of the interleukin-1 beta gene and its clinical application. Oligonucleotides 17: 336–344.
  78. 78. Seno H, Satoh K, Tsuji S, Shiratsuchi T, Harada Y, et al. (2007) Novel interleukin-4 and interleukin-1 receptor antagonist gene variations associated with non-cardia gastric cancer in Japan: comprehensive analysis of 207 polymorphisms of 11 cytokine genes. J Gastroenterol Hepatol 22: 729–737.
  79. 79. Zumkeller N, Brenner H, Chang-Claude J, Hoffmeister M, Nieters A, et al. (2007) Helicobacter pylori infection, interleukin-1 gene polymorphisms and the risk of colorectal cancer: evidence from a case-control study in Germany. Eur J Cancer 43: 1283–1289.
  80. 80. Li C, Xia HH, Xie W, Hu Z, Ye M, et al. (2007) Association between interleukin-1 gene polymorphisms and Helicobacter pylori infection in gastric carcinogenesis in a Chinese population. J Gastroenterol Hepatol 22: 234–239.
  81. 81. Lee KM, Shen M, Chapman RS, Yeager M, Welch R, et al. (2007) Polymorphisms in immunoregulatory genes, smoky coal exposure and lung cancer risk in Xuan Wei, China. Carcinogenesis 28: 1437–1441.
  82. 82. Abazis-Stamboulieh D, Oikonomou P, Papadoulis N, Panayiotidis P, Vrakidou E, et al. (2007) Association of interleukin-1A, interleukin-1B and interleukin-1 receptor antagonist gene polymorphisms with multiple myeloma. Leuk Lymphoma 48: 2196–2203.
  83. 83. Ennas MG, Moore PS, Zucca M, Angelucci E, Cabras MG, et al. (2008) Interleukin-1B (IL1B) and interleukin-6 (IL6) gene polymorphisms are associated with risk of chronic lymphocytic leukaemia. Hematol Oncol 26: 98–103.
  84. 84. Hoeft B, Becker N, Deeg E, Beckmann L, Nieters A (2008) Joint effect between regular use of non-steroidal anti-inflammatory drugs, variants in inflammatory genes and risk of lymphoma. Cancer Causes Control 19: 163–173.
  85. 85. Upadhyay R, Jain M, Kumar S, Ghoshal UC, Mittal B (2008) Potential influence of interleukin-1 haplotype IL-1 beta-511*T-IL-1RN*1 in conferring low risk to middle third location of esophageal cancer: a case-control study. Hum Immunol 69: 179–186.
  86. 86. Zhu Y, Xu Y, Wei Y, Liang W, Liao M, et al. (2008) Association of IL-1B gene polymorphisms with nasopharyngeal carcinoma in a Chinese population. Clin Oncol (R Coll Radiol) 20: 207–211.
  87. 87. Vishnoi M, Pandey SN, Choudhuri G, Mittal B (2008) IL-1 gene polymorphisms and genetic susceptibility of gallbladder cancer in a north Indian population. Cancer Genet Cytogenet 186: 63–68.
  88. 88. Singh H, Sachan R, Goel H, Mittal B (2008) Genetic variants of interleukin-1RN and interleukin-1beta genes and risk of cervical cancer. Bjog 115: 633–638.
  89. 89. Shin WG, Jang JS, Kim HS, Kim SJ, Kim KH, et al. (2008) Polymorphisms of interleukin-1 and interleukin-2 genes in patients with gastric cancer in Korea. J Gastroenterol Hepatol 23: 1567–1573.
  90. 90. Hamacher R, Diersch S, Scheibel M, Eckel F, Mayr M, et al. (2009) Interleukin 1 beta gene promoter SNPs are associated with risk of pancreatic cancer. Cytokine 46: 182–186.
  91. 91. Ahirwar DK, Agrahari A, Mandhani A, Mittal RD (2009) Cytokine gene polymorphisms are associated with risk of urinary bladder cancer and recurrence after BCG immunotherapy. Biomarkers 14: 213–218.
  92. 92. Kumar S, Kumar A, Dixit VK (2009) Evidences showing association of interleukin-1B polymorphisms with increased risk of gastric cancer in an Indian population. Biochem Biophys Res Commun 387: 456–460.
  93. 93. Gehmert S, Velapatino B, Herrera P, Balqui J, Santivanez L, et al. (2009) Interleukin-1 beta single-nucleotide polymorphism’s C allele is associated with elevated risk of gastric cancer in Helicobacter pylori-infected Peruvians. Am J Trop Med Hyg 81: 804–810.
  94. 94. Melo Barbosa HP, Martins LC, Dos Santos SE, Demachki S, Assumpcao MB, et al. (2009) Interleukin-1 and TNF-alpha polymorphisms and Helicobacter pylori in a Brazilian Amazon population. World J Gastroenterol 15: 1465–1471.
  95. 95. Whiteman DC, Parmar P, Fahey P, Moore SP, Stark M, et al.. (2010) Association of Helicobacter pylori infection with reduced risk for esophageal cancer is independent of environmental and genetic modifiers. Gastroenterology 139: 73–83; quiz e11–72.
  96. 96. Wex T, Leodolter A, Bornschein J, Kuester D, Kahne T, et al. (2010) Interleukin 1 beta (IL1B) gene polymorphisms are not associated with gastric carcinogenesis in Germany. Anticancer Res 30: 505–511.
  97. 97. Yu J, Zeng Z, Wang S, Tian L, Wu J, et al. (2010) IL-1B-511 polymorphism is associated with increased risk of certain subtypes of gastric cancer in Chinese: a case-control study. Am J Gastroenterol 105: 557–564.
  98. 98. Yoo EJ, Park SY, Cho NY, Kim N, Lee HS, et al. (2010) Influence of IL1B polymorphism on CpG island hypermethylation in Helicobacter pylori-infected gastric cancer. Virchows Arch 456: 647–652.
  99. 99. Qian N, Chen X, Han S, Qiang F, Jin G, et al. (2010) Circulating IL-1beta levels, polymorphisms of IL-1B, and risk of cervical cancer in Chinese women. J Cancer Res Clin Oncol 136: 709–716.
  100. 100. Wu KS, Zhou X, Zheng F, Xu XQ, Lin YH, et al. (2010) Influence of interleukin-1 beta genetic polymorphism, smoking and alcohol drinking on the risk of non-small cell lung cancer. Clin Chim Acta 411: 1441–1446.
  101. 101. Al-Tahhan MA, Etewa RL, El Behery MM (2011) Association between circulating interleukin-1 beta (IL-1beta) levels and IL-1beta C-511T polymorphism with cervical cancer risk in Egyptian women. Mol Cell Biochem 353: 159–165.
  102. 102. Lim WY, Chen Y, Ali SM, Chuah KL, Eng P, et al. (2011) Polymorphisms in inflammatory pathway genes, host factors and lung cancer risk in Chinese female never-smokers. Carcinogenesis 32: 522–529.
  103. 103. Rizzato C, Canzian F, Rudnai P, Gurzau E, Stein A, et al. (2011) Interaction between functional polymorphic variants in cytokine genes, established risk factors and susceptibility to basal cell carcinoma of skin. Carcinogenesis 32: 1849–1854.
  104. 104. He BS, Pan YQ, Xu YF, Zhu C, Qu LL, et al. (2011) Polymorphisms in interleukin-1B (IL-1B) and interleukin 1 receptor antagonist (IL-1RN) genes associate with gastric cancer risk in the Chinese population. Dig Dis Sci 56: 2017–2023.
  105. 105. Cigrovski Berkovic M, Catela Ivkovic T, Marout J, Zjacic-Rotkvic V, Kapitanovic S (2012) Interleukin 1beta gene single-nucleotide polymorphisms and susceptibility to pancreatic neuroendocrine tumors. DNA Cell Biol 31: 531–536.
  106. 106. Santos JC, Ladeira MS, Pedrazzoli J Jr, Ribeiro ML (2012) Relationship of IL-1 and TNF-alpha polymorphisms with Helicobacter pylori in gastric diseases in a Brazilian population. Braz J Med Biol Res 45: 811–817.
  107. 107. Zheng C, Huang DR, Bergenbrant S, Sundblad A, Osterborg A, et al. (2000) Interleukin 6, tumour necrosis factor alpha, interleukin 1beta and interleukin 1 receptor antagonist promoter or coding gene polymorphisms in multiple myeloma. Br J Haematol 109: 39–45.
  108. 108. Snoussi K, Strosberg AD, Bouaouina N, Ben Ahmed S, Chouchane L (2005) Genetic variation in pro-inflammatory cytokines (interleukin-1beta, interleukin-1alpha and interleukin-6) associated with the aggressive forms, survival, and relapse prediction of breast carcinoma. Eur Cytokine Netw 16: 253–260.
  109. 109. Palli D, Saieva C, Luzzi I, Masala G, Topa S, et al. (2005) Interleukin-1 gene polymorphisms and gastric cancer risk in a high-risk Italian population. Am J Gastroenterol 100: 1941–1948.
  110. 110. Al-Moundhri MS, Al-Nabhani M, Al-Bahrani B, Burney IA, Al-Madhani A, et al. (2006) Interleukin-1beta gene (IL-1B) and interleukin 1 receptor antagonist gene (IL-1RN) polymorphisms and gastric cancer risk in an Omani Arab population. Gastric Cancer 9: 284–290.
  111. 111. Sicinschi LA, Lopez-Carrillo L, Camargo MC, Correa P, Sierra RA, et al. (2006) Gastric cancer risk in a Mexican population: role of Helicobacter pylori CagA positive infection and polymorphisms in interleukin-1 and -10 genes. Int J Cancer 118: 649–657.
  112. 112. Wang W, Ni K, Zhou G (2007) Association of IL1B polymorphisms with gastric cancer in a Chinese population. Clin Biochem 40: 218–225.
  113. 113. Crusius JB, Canzian F, Capella G, Pena AS, Pera G, et al. (2008) Cytokine gene polymorphisms and the risk of adenocarcinoma of the stomach in the European prospective investigation into cancer and nutrition (EPIC-EURGAST). Ann Oncol 19: 1894–1902.
  114. 114. Ter-Minassian M, Zhai R, Asomaning K, Su L, Zhou W, et al. (2008) Apoptosis gene polymorphisms, age, smoking and the risk of non-small cell lung cancer. Carcinogenesis 29: 2147–2152.
  115. 115. Landvik NE, Hart K, Skaug V, Stangeland LB, Haugen A, et al. (2009) A specific interleukin-1B haplotype correlates with high levels of IL1B mRNA in the lung and increased risk of non-small cell lung cancer. Carcinogenesis 30: 1186–1192.
  116. 116. Kiyohara C, Horiuchi T, Takayama K, Nakanishi Y (2010) IL1B rs1143634 polymorphism, cigarette smoking, alcohol use, and lung cancer risk in a Japanese population. J Thorac Oncol 5: 299–304.
  117. 117. Moher D, Liberati A, Tetzlaff J, Altman DG (2009) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med 6: e1000097.
  118. 118. Lewis AM, Varghese S, Xu H, Alexander HR (2006) Interleukin-1 and cancer progression: the emerging role of interleukin-1 receptor antagonist as a novel therapeutic agent in cancer treatment. J Transl Med 4: 48.
  119. 119. Speirs V, Kerin MJ, Newton CJ, Walton DS, Green AR, et al. (1999) Evidence for transcriptional activation of ERalpha by IL-1beta in breast cancer cells. Int J Oncol 15: 1251–1254.
  120. 120. Muerkoster S, Wegehenkel K, Arlt A, Witt M, Sipos B, et al. (2004) Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin-1beta. Cancer Res 64: 1331–1337.
  121. 121. Stefani AL, Basso D, Panozzo MP, Greco E, Mazza S, et al. (1999) Cytokines modulate MIA PaCa 2 and CAPAN-1 adhesion to extracellular matrix proteins. Pancreas 19: 362–369.
  122. 122. ten Kate M, Hofland LJ, van Koetsveld PM, Jeekel J, van Eijck CH (2006) Pro-inflammatory cytokines affect pancreatic carcinoma cell. Endothelial cell interactions. Jop 7: 454–464.
  123. 123. Angst E, Reber HA, Hines OJ, Eibl G (2008) Mononuclear cell-derived interleukin-1 beta confers chemoresistance in pancreatic cancer cells by upregulation of cyclooxygenase-2. Surgery 144: 57–65.
  124. 124. Tian Z, Shen X, Feng H, Gao B (2000) IL-1 beta attenuates IFN-alpha beta-induced antiviral activity and STAT1 activation in the liver: involvement of proteasome-dependent pathway. J Immunol 165: 3959–3965.
  125. 125. Hulkkonen J, Laippala P, Hurme M (2000) A rare allele combination of the interleukin-1 gene complex is associated with high interleukin-1 beta plasma levels in healthy individuals. Eur Cytokine Netw 11: 251–255.
  126. 126. Xue H, Lin B, Ni P, Xu H, Huang G (2010) Interleukin-1B and interleukin-1 RN polymorphisms and gastric carcinoma risk: a meta-analysis. J Gastroenterol Hepatol 25: 1604–1617.