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

Association between TGFBR1 Polymorphisms and Cancer Risk: A Meta-Analysis of 35 Case-Control Studies

  • Yong-qiang Wang ,

    Contributed equally to this work with: Yong-qiang Wang, Xiao-wei Qi

    Affiliation Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China

  • Xiao-wei Qi ,

    Contributed equally to this work with: Yong-qiang Wang, Xiao-wei Qi

    Affiliation Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China

  • Fan Wang,

    Affiliation Department of Oncology, Jiangjin Central Hospital, Jiangjin, Chongqing, China

  • Jun Jiang ,

    qiaonan85@263.net (QG); jcbd@medmail.com.cn (JJ)

    Affiliation Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China

  • Qiao-nan Guo

    qiaonan85@263.net (QG); jcbd@medmail.com.cn (JJ)

    Affiliation Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China

Abstract

Background

Numerous epidemiological studies have evaluated the association between TGFBR1 polymorphisms and the risk of cancer, however, the results remain inconclusive. To derive a more precise estimation of the relation, we conducted a comprehensive meta-analysis of all available case-control studies relating the TGFBR1*6A and IVS7+24G>A polymorphisms of the TGFBR1 gene to the risk of cancer.

Methods

Eligible studies were identified by search of electronic databases. Overall and subgroup analyses were performed. Odds ratio (OR) and 95% confidence interval (CI) were applied to assess the associations between TGFBR1*6A and IVS7+24G>A polymorphisms and cancer risk.

Results

A total of 35 studies were identified, 32 with 19,767 cases and 18,516 controls for TGFBR1*6A polymorphism and 12 with 4,195 cases and 4,383 controls for IVS7+24G>A polymorphism. For TGFBR1*6A, significantly elevated cancer risk was found in all genetic models (dominant OR = 1.11, 95% CI = 1.04∼1.18; recessive: OR = 1.36, 95% CI = 1.11∼1.66; additive: OR = 1.13, 95% CI = 1.05∼1.20). In subgroup analysis based on cancer type, increased cancer risk was found in ovarian and breast cancer. For IVS7+24G>A, significant correlation with overall cancer risk (dominant: OR = 1.39, 95% CI = 1.15∼1.67; recessive: OR = 2.23, 95% CI = 1.26∼3.92; additive: OR = 1.43, 95% CI = 1.14∼1.80) was found, especially in Asian population. In the subgroup analysis stratified by cancer type, significant association was found in breast and colorectal cancer.

Conclusions

Our investigations demonstrate that TGFBR1*6A and IVS7+24G>A polymorphisms of TGFBR1 are associated with the susceptibility of cancer, and further functional research should be performed to explain the inconsistent results in different ethnicities and cancer types.

Introduction

Cancer is a disease resulting from complex interactions between environmental and genetic factors [1][3]. Genetic factors, including the sequence alterations and organization aberrations of the cellular genome that range from single-nucleotide substitutions to gross chromosome, could modulate several important biological progress and alert susceptibility to cancer consequently.

The transforming growth factor-β (TGF-β) signaling pathway has been the focus of extensive research since it was first discovered in 1981 [4], [5]. It has now been well established that this signaling pathway is an important modulator of several biological processes, including cell proliferation, differentiation, migration and apoptosis [6]. Aberrations of the TGF-β signaling pathway are frequently found in many diseases including human cancers in breast, colon, prostate or pancreas [7][10]. As overall TGF-β signaling may be determined by genetic polymorphisms in several TGF-β pathway genes, an increasing number of studies have pointed to the effects of TGF-β pathway gene variants on cancer risk. As the central propagator of TGF-β signaling pathway, TGF-β receptor type I (TGFBR1) has been the hot spot of research.

TGFBR1 gene locates on chromosome 9q22 [11]. Two commonly studied polymorphisms of TGFBR1 gene are TGFBR1*6A (rs1466445), which results from the deletion of three alanines within a nine-alanine (*9A) stretch in exon 1 [12] and IVS7+24G>A (rs334354), which represents a G to A transversion in the +24 position of the donor splice site in intron 7. Although the functional role of IVS7+24G>A is unclear yet, TGFBR1*6A has been suggested to be responsible for efficiency in mediating TGF-β growth inhibitory signals [13]. Therefore, it is biologically reasonable to hypothesize that polymorphisms of TGFBR1 gene may play a functional role in carcinogenesis.

A number of studies have investigated the association between TGFBR1 polymorphisms and cancer risk, but results are somewhat controversial and underpowered. For TGFBR1*6A, a recent meta-analysis in 2010 by Liao et al. [14] found significant association with overall cancer, however, several new papers are further available [15][23]. With respect to IVS7+24G>A polymorphism, only 2 meta-analysis on this issue had ever appeared [24], [25]. Zhang [24] found the IVS7+24G>A carriers had a 76% increase of risk of cancer (OR = 1.76, 95% CI = 1.33∼2.34) with only 440 cases and 706 controls in 3 studies. Meanwhile, Zhang et al. [25] limited the investigation on colorectal cancer and found that there was a significantly increased risk for homozygosity A/A carriers compared to heterozygosity and homozygosity of the allele G carriers (OR = 1.71, 95% CI = 1.17∼2.51). To derive a more precise estimation of the relationship between TGFBR1 polymorphisms and cancer risk, we carried out an updated meta-analysis of all available case–control studies relating the TGFBR1*6A and/or IVS7+24G>A polymorphisms of the TGFBR1 gene to the risk of cancer. To the best of our knowledge, this is the most comprehensive meta-analysis regarding the TGFBR1 polymorphisms and cancer risk.

thumbnail
Table 1. Characteristics of case-control studies included in TGFBR1 TGFBR1*6A polymorphism and cancer risk.

https://doi.org/10.1371/journal.pone.0042899.t001

thumbnail
Table 2. Characteristics of case-control studies included in TGFBR1 IVS7+24G>A polymorphism and cancer risk.

https://doi.org/10.1371/journal.pone.0042899.t002

Materials and Methods

Identification and Eligibility of Relevant Studies

This study was performed according to the proposal of Meta-analysis of Observational Studies in Epidemiology group (MOOSE) [26]. A systematic literature search was performed for articles regarding TGFBR1 SNPs associated with cancer risk. The MEDLINE, Embase, and Chinese National Knowledge Infrastructure (CNKI) were used simultaneously, with the combination of terms “TGFBR1 or transforming growth factor receptor 1 or Type I TGF-beta receptor”, “polymorphism or variant or SNP” and “cancer or neoplasm or carcinoma” (up to May 12, 2012). Reference lists of the identified articles were also examined and the literature retrieval was performed in duplication by two independent reviewers (Yong-qiang Wang and Xiao-wei Qi). Studies that were included in the meta-analysis had to meet all of the following criteria: (1) the publication was a case–control study referring to the association between TGFBR1 polymorphisms (TGFBR1*6A and/or IVS7+24G>A) and cancer, (2) the papers must offer the sample size, distribution of alleles, genotypes or other information that can help us infer the results, (3) when multiple publications reported on the same or overlapping data, we used the most recent or largest population as recommended by Little et al. [27], and (4) publication language was confined to English and Chinese.

thumbnail
Table 3. Pooled analysis of association of TGFBR1 TGFBR1*6A (rs1466445) and cancer risk.

https://doi.org/10.1371/journal.pone.0042899.t003

thumbnail
Table 4. Pooled analysis of association of IVS7+24G>A (rs334354) and cancer risk.

https://doi.org/10.1371/journal.pone.0042899.t004

thumbnail
Figure 2. Forest plot (random effects model) describing the association of the TGFBR1*6A polymorphism with risk of cancer.

The TGFBR1*6A polymorphism was associated with increased risk of cancer in additive model. Each study is shown by the point estimate of the OR (the size of the square is proportional to the weight of each study) and 95% CI for the OR (extending lines).

https://doi.org/10.1371/journal.pone.0042899.g002

Data Extraction

Two investigators (Yong-qiang Wang and Xiao-wei Qi) independently extracted the data from eligible studies selected according to the pre-specified criteria and the results were compared. Disagreements were resolved by discussion or by involving a third reviewer (Qiao-nan Guo). The following information of each study was collected: first author, reference year, name of studies, total number of cases and controls, studied polymorphisms, ethnicity of subjects, source of controls, and distribution of genotypes in case and control groups. For studies with inadequate information, authors were contacted for further support by E-mail if possible.

thumbnail
Figure 3. Forest plot (random effects model) describing the association of the IVS7+24G>A polymorphism with risk of cancer.

The IVS7+24G>A polymorphism was associated with increased cancer risk in additive model.

https://doi.org/10.1371/journal.pone.0042899.g003

Statistical Analysis

Meta-analysis was performed as described previously [28], [29]. Hardy-Weinberg equilibrium (HWE) in the controls for each study was calculated using goodness-of fit test (chi-square or Fisher’s exact test). It was considered statistically significant when P<0.05. Studies deviated from HWE were removed.

thumbnail
Figure 4. Funnel plot analysis (recessive model of TGFBR1*6A polymorphism) to detect publication bias.

Each point represents an individual study for the indicated association. LogOR, natural logarithm of OR. Perpendicular line, mean effect size.

https://doi.org/10.1371/journal.pone.0042899.g004

Crude odds ratios (ORs) with their 95% CIs were used to assess the strength of association between polymorphisms of TGFBR1 and cancer risk. The pooled ORs were performed for dominant model (1∶1+1∶2 vs. 2∶2), recessive model (1∶1 vs. 1∶2+2∶2), additive model (1 vs. 2) respectively. 1 and 2 represent the minor and the major allele respectively. Stratified analysis was also performed by ethnicity and cancer type. Leukemia, lymphoma and MM (multiple myeloma) were merged as hematologic cancer. For ethnicity classification, African, Jews and the ethnicity not stated in original study were merged as others.

Heterogeneity assumption was assessed by chi-based Q-test. The heterogeneity was considered statistically significant if P<0.10 [30]. With lacking of heterogeneity among studies, the pooled OR was calculated by the fixed effects model (Mantel–Haenszel) [31]. Otherwise, the random effects model (DerSimonian and Laird) was used [32], [33]. We also calculated the quantity I2 that represents the percentage of total variation across studies that is a result of heterogeneity rather than chance. Values of less than 25% may be considered “low”, values of about 50% may be considered “moderate”, and values of more than 75% may be considered “high”. A value of 0 (zero) indicates no observed heterogeneity, and larger values show increasing heterogeneity.

thumbnail
Figure 5. Influence analysis of the summary odds ratio coefficients on the association between IVS7+24G>A polymorphism and cancer risk in recessive model.

Results were computed by omitting each study (left column) in turn. Bars, 95% confidence interval.

https://doi.org/10.1371/journal.pone.0042899.g005

Sensitivity analysis was carried out by removing each study at a time to evaluate the stability of the results. Publication bias was analyzed by performing funnel plots qualitatively, and estimated by Begg’s and Egger’s test quantitatively [34], [35].

All statistical analysis was conducted using STATA software (version 11.0; STATA Corporation, College Station, TX). Two-sided P-values<0.05 were considered statistically significant.

Results

Study Characteristics

After comprehensive searching, a total of 186 publications were identified. We reviewed the titles, abstracts and the full texts of all retrieved articles through defined criteria as shown in Figure 1. Finally, the pool of eligible studies included 35 studies [12], [15][23], [36][60], among which 32 with 19,767 cases and 18,516 controls were for TGFBR1*6A polymorphism and 12 with 4,195 cases and 4,383 controls for IVS7+24G>A polymorphism. Each study in one publication was considered as a data set separately for pooling analysis. Table 1 and Table 2 list the main characteristics of these data sets about these two polymorphisms.

Quantitative Synthesis

The main results of this meta-analysis and the heterogeneity test were shown in Table 3 and 4. With respect to TGFBR1*6A polymorphism, a total of 58 data sets in 32 studies were included in this meta-analysis. Of these data sets, 25 were Caucasian, 6 were Asian, 20 were mixed population and 7 were others. Overall, significantly elevated cancer risk was found in all genetic models (dominant model: OR = 1.11, 95% CI = 1.04∼1.18; recessive model: OR = 1.36, 95% CI = 1.11∼1.66; additive model: OR = 1.13, 95% CI = 1.05∼1.20, Figure 2). The heterogeneity was significant in all genetic models except for recessive model (P = 0.34). In the subgroup analysis stratified by ethnicity, significantly increased cancer risk was suggested among mixed ethnicity from US studies (dominant model: OR = 1.15, 95% CI = 1.05∼1.25; recessive model: OR = 1.85, 95% CI = 1.26∼2.72; additive model: OR = 1.22, 95% CI = 1.10∼1.36) but not among Caucasian or Asian population in all genetic models. In the subgroup analysis by cancer type, no significant association with cancer risk was demonstrated in overall population with colorectal, lung, prostate, bladder, hematological and cervical cancer. For ovarian cancer, significantly increased risk was observed in recessive model (OR = 2.30, 95% CI = 1.01∼5.22) and additive model (OR = 1.25, 95% CI = 1.02∼1.52). With respect to breast cancer, significantly increased risk was found only in additive model (OR = 1.15, 95% CI = 1.01∼1.31).

With respect to IVS7+24G>A polymorphism, a total of 12 studies with 13 data sets were included. Of these data sets, 5 were European, 4 were Asian and 4 were from USA with mixed ethnicity. Similar to TGFBR1*6A polymorphism, significantly elevated cancer risk was associated with IVS7+24G>A in all genetic models (dominant model: OR = 1.39, 95% CI = 1.15∼1.67; recessive model: OR = 2.23, 95% CI = 1.26∼3.92; additive model: OR = 1.43, 95% CI = 1.14∼1.80, Figure 3). The heterogeneity was significant in all genetic models (P<0.1). In the subgroup analysis by ethnicity, significantly increased risk was found in Asian population (dominant model: OR = 1.30, 95% CI = 1.12∼1.51; recessive model: OR = 1.58, 95% CI = 1.07∼2.34; additive model: OR = 1.27, 95% CI = 1.09∼1.48) but not in Caucasian in all genetic models. In the subgroup analysis stratified by cancer type, significantly increased risk was detected in all genetic models in breast cancer (dominant model: OR = 1.99, 95% CI = 1.67∼2.37; recessive model: OR = 5.96, 95% CI = 1.59∼22.33; additive model: OR = 2.54, 95% CI = 2.10∼3.08). With respect to colorectal cancer, significant association was found only in recessive model (OR = 1.38; 95% CI = 1.04∼1.84).

Publication Bias and Sensitivity Analysis

The shapes of the funnel plots did not reveal any evidence of obvious asymmetry for TGFBR1*6A polymorphism in all genetic models, except for recessive model (Figure 4). The Begg’s and Egger’s test also suggested the same results (dominant model: PBegg’s = 0.54, PEgger’s = 0.26; recessive model: PBegg’s = 0.00 (7.13×10−4), PEgger’s = 0.00(2.23×10−5); additive model: PBegg’s = 0.52, PEgger’s = 0.13). For IVS7+24G>A polymorphism, publication bias was not ruled out not only through visual inspection of asymmetry in funnel plots but also through statistical evidence of the Begg’s and Egger’s test (dominant model: PBegg’s = 1.00, PEgger’s = 0.87; recessive model: PBegg’s = 0.25, PEgger’s = 0.89; additive model: PBegg’s = 0.36, PEgger’s = 0.58).

Sensitivity analysis, which was performed to assess the publication bias and the influence of each individual study on the pooled OR by sequential removal of individual studies, showed that Song’s study [52] was far from the midcourt line for IVS7+24G>A polymorphism in recessive model (Figure 5). However, the heterogeneity and the pooled OR were not influenced when this article was excluded (data not shown), which indicated that our results were statistically stable.

Discussion

In the present study, we explored the association between the TGFBR1*6A and IVS7+24G>A polymorphisms and cancer risk, involving 35 eligible case–control studies. For TGFBR1*6A polymorphism, 19,767 cases and 18,516 controls were included. We found that individuals with the TGFBR1*6A allele showed an increased risk of cancer. In the stratified analysis by cancer type, significantly elevated risks were more pronounced among ovarian cancer and breast cancer. However, no significant correlation of polymorphism TGFBR1*6A with colorectal cancer was found. These findings, though including the latest publications, were consistent with a recent meta-analysis study conducted by Liao et al. [14]. While according to Colleran’s study [57], TGFBR1*6A is not associated with breast cancer. This discrepancy may be due to data missing of some important studies, which was exclusively elaborated by Zhang et al. [61]. Another meta-analysis performed by Zhang et al. [25] found TGFBR1*6A is statistically associated with an increased colorectal cancer risk in dominant model. One factor that may contribute to the differences is that we excluded Castillejo’s study [62] for HWE deviation and included two latest studies [22], [23]. Moreover, a significantly increased risk was found among mixed ethnicity from US studies but not among Caucasian and Asian, and this was the first study evaluating the relation between TGFBR1 polymorphism and overall cancer risk among different populations.

With respect to IVS7+24G>A polymorphism, a previous meta-analysis conducted by Zhang [24] with only 440 cases and 706 controls found that the IVS7+24G>A carriers had a 76% increase of cancer risk. Another meta-analysis conducted by Zhang et al. [25] found that IVS7+24G>A polymorphism had significant effects on colorectal cancer risk in recessive model. However, there were defects in their meta-analysis [25] for mistaking adenoma cases of Lundin’s study [54] as colorectal cancer cases. For the current meta-analysis, 4,195 cases and 4,383 controls were included. Significant correlation of IVS7+24G>A polymorphism with cancer risk was found in all genetic models. When coming to colorectal cancer, the results were in line with Zhang et al [25]. Besides, we also found strong association between IVS7+24G>A polymorphism and breast cancer risk, indicating that potentially functional IVS7+24G>A polymorphism may play a low penetrance role in development of breast cancer. Significant association was found in Asian but not in Caucasian, suggesting a possible role of ethnic differences in genetic backgrounds and the environment they lived in.

To some extent, limitations of this meta-analysis should be addressed. First, the sample sizes of several included studies [12], [37] were rather small and not adequate enough to detect the possible risk for TGFBR1 polymorphisms. Second, cancer is a complex disease with multifactorial etiology. The gene–environment and gene–gene interactions should be further evaluated. Third, haplotype association analysis is the most powerful method to explore the intrinsic effects of gene, but most of the literatures identified in our present meta-analysis were focused on the relation between the two TGFBR1 SNPs and tumor susceptibility, which made it difficult to investigate the TGFBR1 haplotype effects on carcinogenesis. Last but not least, most of US studies were mixed ethnicity, which made it hard to obtain the effects of specific ethnicity on the associations between TGFBR1 polymorphisms and cancer risk.

In summary, this meta-analysis provided evidence that the TGFBR1*9A/6A polymorphism is associated with overall cancer susceptibility and seem to be more susceptible to ovarian and breast cancer. Meanwhile, IVS7+24G>A polymorphism is also associated with increased overall cancer risk especially in colorectal and breast cancer. More well-designed epidemiological studies on specific ethnicity and cancer types, which were not well covered by existing studies, will be necessary to validate the findings identified in the current meta-analysis. Further studies regarding other SNPs (or haplotypes) in the TGFBR1 gene and cancer risk are also encouraged to better understand the role of TGFBR1 in carcinogenesis.

Acknowledgments

We thank Dr. Castellví-Bel S from Department of Gastroenterology, Hospital Clinic University of Barcelona, Spain, for providing raw data of his research article [19]. We also thank Miss Jun-lan Liu, from Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China, for language editing of the manuscript.

Author Contributions

Conceived and designed the experiments: QG JJ. Performed the experiments: YW XQ FW. Analyzed the data: YW XQ FW. Contributed reagents/materials/analysis tools: YW XQ FW. Wrote the paper: YW XQ QG.

References

  1. 1. Bredberg A (2011) Cancer: More of polygenic disease and less of multiple mutations? A quantitative viewpoint. Cancer 117: 440–445.
  2. 2. Hoeijmakers JHJ (2001) Genome maintenance mechanisms for preventing cancer. Nature 411: 366–374.
  3. 3. Pharoah PDP, Dunning AM, Ponder BAJ, Easton DF (2004) Association studies for finding cancer-susceptibility genetic variants. Nat Rev Cancer 4: 850–860.
  4. 4. Moses HL, Branum EL, Proper JA, Robinson RA (1981) Transforming growth factor production by chemically transformed cells. Cancer Res 41: 2842–2848.
  5. 5. Roberts AB, Anzano MA, Lamb LC (1981) New class of transforming growth factors potentiated by epidermal growth factor: Isolation from non-neoplastic tissues. Proc Natl Acad Sci U S A 78: 5339–5343.
  6. 6. Gordon KJ, Blobe GC (2008) Role of transforming growth factor-(beta) superfamily signaling pathways in human disease. Biochim Biophys Acta 1782: 197–228.
  7. 7. Benson JR (2004) Role of transforming growth factor (beta) in breast carcinogenesis. Lancet Oncol 5: 229–239.
  8. 8. Elliott RL, Blobe GC (2005) Role of transforming growth factor beta in human cancer. J Clin Oncol 23: 2078–2093.
  9. 9. Bierie B, Moses HL (2006) Tumour microenvironment - TGFB: The molecular Jekyll and Hyde of cancer. Nat Rev Cancer 6: 506–520.
  10. 10. Galliher AJ, Neil JR, Schiemann WP (2006) Role of transforming growth factor-beta in cancer progression. Future Oncol 2: 743–763.
  11. 11. Pasche B, Luo Y, Rao PH, Nimer SD, Dmitrovsky E, et al. (1998) Type I transforming growth factor (beta) receptor maps to 9q22 and exhibits a polymorphism and a rare variant within a polyalanine tract. Cancer Res 58: 2727–2732.
  12. 12. Chen T, Jackson C, Costello B, Singer N, Colligan B, et al. (2004) An intronic variant of the TGFBR1 gene is associated with carcinomas of the kidney and bladder. Int J Cancer 112: 420–425.
  13. 13. Pasche B, Knobloch TJ, Bian Y, Liu J, Phukan S, et al. (2005) Somatic acquisition and signaling of TGFBR1*6A in cancer. JAMA 294: 1634–1646.
  14. 14. Liao RY, Mao C, Qiu LX, Ding H, Chen Q, et al. (2010) TGFBR1*6A/9A polymorphism and cancer risk: A meta-analysis of 13,662 cases and 14,147 controls. Mol Biol Rep 37: 3227–3232.
  15. 15. Dai L, Gast A, Horska A, Schrappe M, Bartram CR, et al. (2009) A case-control study of childhood acute lymphoblastic leukaemia and polymorphisms in the TGF-(beta) and receptor genes. Pediatr Blood Cancer 52: 819–823.
  16. 16. Carvajal-Carmona LG, Churchman M, Bonilla C, Walther A, Lefevre JH, et al. (2010) Comprehensive assessment of variation at the transforming growth factor (beta) type 1 receptor locus and colorectal cancer predisposition. Pro Natl Acad Sci U S A 107: 7858–7862.
  17. 17. Forsti A, Li X, Wagner K, Tavelin B, Enquist K, et al. (2010) Polymorphisms in the transforming growth factor beta 1 pathway in relation to colorectal cancer progression. Genes Chromosomes Cancer 49: 270–281.
  18. 18. Hu YS, Pan Y, Li WH, Zhang Y, Li J, et al. (2010) Association between TGFBR1*6A and osteosarcoma: a Chinese case-control study. BMC Cancer 10: 169.
  19. 19. Abuli A, Fernandez-Rozadilla C, Giraldez MD, Muoz J, Gonzalo V, et al. (2011) A two-phase case-control study for colorectal cancer genetic susceptibility: Candidate genes from chromosomal regions 9q22 and 3q22. Br J Cancer 105: 870–875.
  20. 20. Dong ZM (2011) Correlation of TGF-β receptor type 1 gene 6A and Int7G24A polymorphisms with the risk of esophageal squamous cell carcinoma. Carcinogenesis,Teratogenesis & Mutagenesis 23: 16–21.
  21. 21. Guo W, Dong Z, Guo Y, Chen Z, Yang Z, et al. (2011) Association of polymorphisms in transforming growth factor-(beta) receptors with susceptibility to gastric cardia adenocarcinoma. Mol Biol Rep 39: 4301–4309.
  22. 22. Joshi NN, Kale MD, Hake SS, Kannan S (2011) Transforming growth factor (beta) signaling pathway associated gene polymorphisms may explain lower breast cancer risk in western Indian women. PLoS One 6: e21866.
  23. 23. Martinez-Canto A, Castillejo A, Mata-Balaguer T, Castillejo MI, Hernandez-Illan E, et al. (2012) TGFBR1 intralocus epistatic interaction as a risk factor for colorectal cancer. PLoS One 7: e30812.
  24. 24. Zhang HT (2005) Int7G24A variant of the TGFBR1 gene and cancer risk: A meta-analysis of three case-control studies. Lung Cancer 49: 419–420.
  25. 25. Zhang X, Wu L, Sheng Y, Zhou W, Huang Z, et al. (2012) The association of polymorphisms on TGFBR1 and colorectal cancer risk: a meta-analysis. Mol Biol Rep 39: 2567–2574.
  26. 26. Stroup DF, Berlin JA, Morton SC, Olkin I, Williamson GD, et al. (2000) Meta-analysis of observational studies in epidemiology: A proposal for reporting. JAMA 283: 2008–2012.
  27. 27. Little J, Bradley L, Bray MS, Clyne M, Dorman J, et al. (2002) Reporting, appraising, and integrating data on genotype prevalence and gene-disease associations. Am J Epidemiol 156: 300–310.
  28. 28. Qi X, Ma X, Yang X, Fan L, Zhang Y, et al. (2010) Methylenetetrahydrofolate reductase polymorphisms and breast cancer risk: A meta-analysis from 41 studies with 16,480 cases and 22,388 controls. Breast Cancer Res Treat 123: 499–506.
  29. 29. Qi X, Zhang F, Yang X, Fan L, Zhang Y, et al. (2010) Transforming growth factor-(beta)1 polymorphisms and breast cancer risk: A meta-analysis based on 27 case-control studies. Breast Cancer Res Treat 122: 273–279.
  30. 30. Lau J, Ioannidis JP, Schmid CH (1997) Quantitative synthesis in systematic reviews. Ann Intern Med 127: 820–826.
  31. 31. Mantel N, Haenszel W (1959) Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst 22: 719–748.
  32. 32. DerSimonian R, Kacker R (2007) Random-effects model for meta-analysis of clinical trials: An update. Contemp Clin Trials 28: 105–114.
  33. 33. DerSimonian R, Laird N (1986) Meta-analysis in clinical trials. Control Clin Trials 7: 177–188.
  34. 34. Begg CB, Mazumdar M (1994) Operating characteristics of a rank correlation test for publication bias. Biometrics 50: 1088–1101.
  35. 35. Egger M, Smith GD, Schneider M, Minder C (1997) Bias in meta-analysis detected by a simple, graphical test. BMJ 315: 629–634.
  36. 36. Pasche B, Kolachana P, Nafa K, Satagopan J, Chen YG, et al. (1999) T(beta)R-I(6A) is a candidate tumor susceptibility allele. Cancer Res 59: 5678–5682.
  37. 37. Chen T (1999) Structural alterations of transforming growth factor-beta receptor genes in human cervical carcinoma. Int J Cancer 82(1): 43–51.
  38. 38. van Tilborg AA, de Vries A, Zwarthoff EC (2001) The chromosome 9q genes TGFBRI, TSCI, and ZNFI89 are rarely mutated in bladder cancer. J Pathol 194: 76–80.
  39. 39. Stefanovska AM, Efremov GD, Dimovski AJ, Jasar D, Zografski G, et al. (2001) TbetaR-I(6A) polymorphism is not a tumor susceptibility allele in Macedonian colorectal cancer patients. Cancer Res 61: 8351–8352.
  40. 40. Samowitz WS, Curtin K, Leppert MF, Slattery ML (2001) Uncommon TGFBRI allele is not associated with increased susceptibility to colon cancer. Genes Chromosomes Cancer 32: 381–383.
  41. 41. Baxter SW, Choong DYH, Eccles DM, Campbell IG (2002) Transforming growth factor (beta) receptor 1 polyalanine polymorphism and exon 5 mutation analysis in breast and ovarian cancer. Cancer Epidemiol Biomarkers Prev 11: 211–214.
  42. 42. Kaklamani V, Baddi L, Rosman D, Liu J, Ellis N, et al. (2004) No major association between TGFBR1*6A and prostate cancer. BMC Genet 5: 28.
  43. 43. Pasche B, Kaklamani V, Hou N, Young T, Rademaker A, et al. (2004) TGFBR1*6A and cancer: a meta-analysis of 12 case-control studies. J clin oncol 22: 756–758.
  44. 44. Jin Q, Hemminki K, Grzybowska E, Klaes R, Soderberg M, et al. (2004) Polymorphisms and haplotype structures in genes for transforming growth factor (beta)1 and its receptors in familial and unselected breast cancers. Int J Cancer 112: 94–99.
  45. 45. Suarez BK, Pal P, Jin CH, Kaushal R, Sun G, et al. (2005) TGFBR1*6A is not associated with prostate cancer in men of European ancestry. Prostate Cancer Prostatic Dis 8: 50–53.
  46. 46. Spillman MA, Schildkraut JM, Halabi S, Moorman P, Calingaert B, et al. (2005) Transforming growth factor (beta) receptor I polyalanine repeat polymorphism does not increase ovarian cancer risk. Gynecol Oncol 97: 543–549.
  47. 47. Kaklamani VG, Baddi L, Liu J, Rosman D, Phukan S, et al. (2005) Combined genetic assessment of transforming growth factor-(beta) signaling pathway variants may predict breast cancer risk. Cancer Res 65: 3454–3461.
  48. 48. Chen T, Jackson CR, Link A, Markey MP, Colligan BM, et al. (2006) Int7G24A variant of transforming growth factor-(beta) receptor type I is associated with invasive breast cancer. Clinical Cancer Res 12: 392–397.
  49. 49. Feigelson HS, Patel AV, Ryan Diver W, Stevens VL, Thun MJ, et al. (2006) Transforming growth factor (beta) receptor type I and transforming growth factor (beta)1 polymorphisms are not associated with postmenopausal breast cancer. Cancer Epidemiol Biomarkers Prev 15: 1236–1237.
  50. 50. You W, Liu Z, Zhao J, Zheng M, Zheng SY, et al. (2007) No association between TGFBR1*6A and lung cancer. J Thorac Oncol 2: 657–659.
  51. 51. Cox DG, Penney K, Guo Q, Hankinson SE, Hunter DJ (2007) TGFB1 and TGFBR1 polymorphisms and breast cancer risk in the Nurses’ Health Study. BMC Cancer 7: 175.
  52. 52. Song B, Margolin S, Skoglund J, Zhou X, Rantala J, et al. (2007) TGFBR1*6A and Int7G24A variants of transforming growth factor-(beta) receptor 1 in Swedish familial and sporadic breast cancer. Br J Cancer 97: 1175–1179.
  53. 53. Skoglund J, Song B, Dalen J, Dedorson S, Edler D, et al. (2007) Lack of an association between the TGFBR1*6A variant and colorectal cancer risk. Clin Cancer Res 13: 3748–3752.
  54. 54. Skoglund Lundin J, Vandrovcova J, Song B, Zhou X, Zelada-Hedman M, et al. (2009) TGFBR1 variants TGFBR1*6A and Int7G24A are not associated with an increased familial colorectal cancer risk. Br J Cancer 100: 1674–1679.
  55. 55. Castillejo A, Rothman N, Murta-Nascimento C, Malats N, Garcia-Closas M, et al. (2009) TGFB1 and TGFBR1 polymorphic variants in relationship to bladder cancer risk and prognosis. Int J Cancer 124: 608–613.
  56. 56. Jakubowska A, Gronwald J, Menkiszak J, Gorski B, Huzarski T, et al. (2010) BRCA1-associated breast and ovarian cancer risks in Poland: No association with commonly studied polymorphisms. Breast Cancer Res Treat 119: 201–211.
  57. 57. Colleran G, McInerney N, Rowan A, Barclay E, Jones AM, et al. (2010) The TGFBR1*6A/9A polymorphism is not associated with differential risk of breast cancer. Breast Cancer Res Treat 119: 437–442.
  58. 58. Castillejo A, Mata-Balaguer T, Guarinos C, Castillejo MI, Martinez-Canto A, et al. (2009) The Int7G24A variant of transforming growth factor-beta receptor type I is a risk factor for colorectal cancer in the male Spanish population: a case-control study. BMC Cancer 9: 406.
  59. 59. Zhang Y, Liu B, Jin M, Ni Q, Liang X, et al. (2009) Genetic polymorphisms of transforming growth factor-(beta)1 and its receptors and colorectal cancer susceptibility: A population-based case-control study in China. Cancer Lett 275: 102–108.
  60. 60. Hu YS, Pan Y, Li WH, Zhang Y, Li J, et al. (2011) Int7G24A variant of transforming growth factor-beta receptor 1 is associated with osteosarcoma susceptibility in a Chinese population. Med Oncol 28: 622–625.
  61. 61. Zhang Y (2011) Need for clarification of data in the recent meta-analysis about TGFBR1*6A/9A polymorphism and breast cancer risk. Breast Cancer Res Treat 130: 1073–1074.
  62. 62. Castillejo A, Mata-Balaguer T, Montenegro P, Ochoa E, Lazaro R, et al. (2009) The TGFBR1*6A allele is not associated with susceptibility to colorectal cancer in a Spanish population: A case-control study. BMC Cancer 9: 193.