Skip to main content

Association of VEGF-2549I/D promoter polymorphism with gastrointestinal tract cancer risk: a meta-analysis

Abstract

Background

Gastrointestinal tract (GIT) cancers are complex disorders affecting millions of people worldwide. The vascular endothelial growth factor (VEGF) helps in the development of different GIT cancers by promoting abnormal angiogenesis in cancer cells. The role of VEGF-2549I/D polymorphism in influencing GIT cancer susceptibility has been studied in different populations with inconclusive results. Therefore, the relationship between VEGF-2549I/D polymorphism with GIT susceptibility was studied by performing a meta-analysis study.

Methods

Various online databases were used for identifying the articles. Based on study selection criteria, five studies on different GIT cancers including 1178 patients and 1520 controls were included in the meta-analysis. The accuracy of the study results was determined by performing a trial sequential analysis.

Results

In this study, the VEGF-2549I/D polymorphism did not influence the GIT cancer susceptibility in the overall analysis as well as when stratified according to ethnicity (p > 0.05). Stratification of all the studies based on the different GIT cancers reported an increased susceptibility to gastric cancer under different genetic models including allele contrast (OR = 1.67, CI = 1.294–2.157, p = 0.00008), recessive (OR = 1.68, CI = 1.056–2.660, p = 0.029), dominant (OR = 2.49, CI = 1.617–3.823, p = 0.00003), over-dominant (OR = 1.52, CI = 1.055–2.177, p = 0.025), II vs DD (OR = 2.97, CI = 1.692–5.208, p = 0.00015) and ID vs DD model (OR = 2.35, CI = 1.501–3.669, p = 0.00018).

Conclusion

There was no relationship between VEGF-2549I/D promoter polymorphism and GIT cancer susceptibility in the overall population and also in different ethnic groups. Stratification analysis revealed higher susceptibility towards gastric cancer development with VEGF-2549I/D polymorphism.

Introduction

Cancer is a serious polygenic disease affecting the global population [1]. The development and progression of cancer occur due to the interaction between several environmental stresses, genetic and epigenetic factors [2]. Genetic variation affects the key biological processes involved in oncogenesis [3, 4] and also determines the individual’s susceptibility to cancer development [5]. Genome-wide association studies (GWAS) performed on different populations have confirmed the relationship between several genes and cancer susceptibility [6]. It has been documented in the literature that the genetic variation reported in the angiogenesis-related genes might be responsible for differences in the individual’s susceptibility towards tumour development [7].

The angiogenesis process, a critical hallmark of cancer, helps in the continuous growth and metastasis of the tumour cells [8, 9]. The coordinated functioning of several growth molecules regulates the angiogenesis process [10]. The vascular endothelial growth factor (VEGF), a crucial angiogenic growth mediator, is responsible for the formation of new vasculature, needed for the continuous blood supply to the growing tumour [11, 12]. VEGF, a highly polymorphic gene localised on 6p21.3, encodes the pro-angiogenic VEGF protein [13]. The VEGF harbours several functional polymorphisms in the promoter and UTRs which alter the gene expression levels of VEGF [14,15,16]. One such functional polymorphism is a 18 bp ins/del polymorphism (− 2549 Ins/del), positioned upstream of the VEGF promoter. Ins/del polymorphisms are one of the most frequently reported genetic variations, which affect the functions of the regulatory region of the gene [17, 18]. Ins/del polymorphisms have been reported to alter several complex human traits and contribute to the disease development [17, 19]. VEGF-2549I/D promoter polymorphism (rs35569394) is an important functional polymorphism, known to regulate VEGF production and activity. The functional relevance of VEGF-2549I/D polymorphism has been studied in several studies. It was observed that the VEGF-2549 D allele enhanced the transcriptional activity of the VEGF promoter [20], whereas the VEGF-2549 II genotype increased the VEGF production in the cells of healthy individuals [21]. The VEGF-2549I/D polymorphism by regulating the protein levels might influence susceptibilities to cancer development.

VEGF-2549I/D polymorphism has been studied in different gastrointestinal tract (GIT) cancers including oesophageal [22, 23], gastric [24], hepatocellular [25], gall bladder [26] and colorectal cancer [27] with conflicting results. The association of VEGF-2549I/D polymorphism with therapy response has been studied in oesophageal [23], colorectal [28, 29] and hepatocellular cancer [30]. Other than the GIT cancers, the association of VEGF-2549I/D polymorphism with cancer risk has been reported in renal cell carcinoma [31], bladder [32], urothelial bladder [33], breast [34, 35] and prostate cancer [36].

GIT cancers are among the most prevalent cancers globally [1]. Increased expression of VEGF in tissue and serum has been detected in several GIT cancers including oral [37], gastric [38] and pancreatic [39] cancer. Increased VEGF levels in the patient’s serum samples have been associated with tumour aggressiveness and lower overall survival in various GIT cancers like oesophageal [40], pancreatic [41], hepatocellular [42] and gastric cancer [43].

The case–control studies with small sample sizes give limited statistical power to provide a definitive decision. Therefore, to overcome this problem, a detailed structured meta-analysis study investigating the role of VEGF-2549I/D polymorphism in modulating GIT cancer susceptibility was performed to identify a more accurate conclusion. The present meta-analysis study is the first study examining the relationship between the VEGF-2549I/D polymorphism and GIT cancer risk.

Methods

Study design

This meta-analysis followed the PRISMA guidelines [44] and the study question was prepared according to the PICO criteria [45]. The formulated research study question was whether VEGF-2549I/D polymorphism was associated with GIT cancer risk, by comparing the genotypic and allelic distribution between patients suffering from GIT cancers and age, gender and geographical matched healthy controls.

Literature search strategy

We systematically searched all the published research articles evaluating the role of VEGF -2549I/D polymorphism in influencing GIT cancer susceptibility by extensively searching several databases such as PubMed, Google Scholar and Embase up to December 2023. The following combination of keywords was used for searching: “VEGF” or “VEGFA”, “SNPs”, “cancer”, “oral”, “oesophageal”, “gall bladder”, “hepatocellular”, “gastric”, “colon”, “colorectal”, “pancreatic” and “angiogenesis”. The references cited in the identified published studies were independently screened to identify eligible articles, missed during the initial search.

Inclusion criteria

The main selection criteria for employing the five studies in the meta-analysis were:

  1. 1)

    Case–control studies examining the relationship between VEGF-2549I/D polymorphism with different GIT cancer risk.

  2. 2)

    Study population: Patients suffering from GIT cancers and healthy controls

  3. 3)

    Genotypic distribution in complete agreement with Hardy–Weinberg equilibrium (HWE) in controls in all of these studies.

  4. 4)

    High or medium quality studies, as per the Newcastle–Ottawa Scale assessment

  5. 5)

    Sufficient genotype data available in the studies to calculate the odds ratio, 95%CIs and p values.

Study selection

All the retrieved research studies were reviewed and analysed carefully by the two authors. The titles and abstract of each research article were screened, and studies following the inclusion criteria were included in the meta-analysis. Studies not following the study selection criteria were removed from the analysis. In case of disagreements between the study selection, the decision to include or exclude the study was only made after author discussions. The PRISMA flowchart depicting the procedure for the selection of the studies is demonstrated in Fig. 1.

Fig. 1
figure 1

The PRISMA flowchart of the study selection process

Data extraction

The authors carefully extracted the relevant data from each included study in this meta-analysis. The data included: type of gastrointestinal tract cancer, the total number of study subjects, genotypic distribution of VEGF-2549I/D polymorphism, HWE p value in controls, population and ethnicity of study subjects, main author’s last name along with publishing year and key findings of study.

Quality assessment

The quality of the selected studies was assessed by scoring the studies according to the Newcastle–Ottawa Scale scoring system [46]. Each study was awarded one star under selection (4 items) and exposure (3 items) category, and two stars were given in the comparability category. The studies with an evaluation score of ≥ 7 were considered high-quality studies, 4–6 were considered medium quality, and < 4 were considered low-quality studies.

Statistical analysis

The deviation from the HWE was tested using the chi-squared test among the control subjects (p > 0.05). The combined ORs, 95%CIs and p value < 0.05 were calculated to determine the risk of GIT cancers with VEGF-2549I/D polymorphism under seven genetic models (allelic, dominant, recessive, over-dominant, II versus DD, II versus ID and ID versus DD comparison model).

Heterogeneity analysis

The Q statistic and I2 metric tests were performed to calculate the heterogeneity between the selected studies. The fixed effect model was applied in the absence of inter-study heterogeneity. However, in the presence of inter-study heterogeneity, a random effects model was applied for performing the statistical analysis. Higher I2 values indicated a higher degree of heterogeneity among the studies [47].

Sensitivity analysis

The stability of the results was determined by performing the sensitivity analysis. The analysis was done by sequentially omitting each study and then re-analysing the association with GIT cancer risk to determine the influence of each study on the overall pooled results.

Publication bias

The graphical symmetry of the constructed funnel plots and the probability value for Egger’s test were taken as standards to calculate the presence or absence of bias in the study results.

Trial sequential analysis (TSA)

There are more chances of type I and type II errors in the results prepared through a meta-analysis study; therefore, the TSA was performed. The TSA was done for checking the reliability and robustness of the meta-analysis results. The TSA was performed to estimate the required sample size using the online available TSA software 0.9.5.10 Beta. The study was conducted using assumptions of 95%CI, 10% relative risk reduction (RRR), 5% α level and 80% β value. The crossing of both the TSA monitoring boundaries and required information size (RIS) by the cumulative Z values is indicative of a conclusive decision, requiring no further clinical trials. However, if TSA monitoring boundaries are not crossed before reaching the RIS, it indicates that the sample size is inadequate and more studies are needed for a conclusive decision.

Results

Characteristics of included studies

Five case–control studies were selected for analysis after applying the selection criteria. Out of the five studies, four studies included patients from Asian ethnicity, whereas only one study included patients from Caucasian ethnicity. Among the five included studies, two were reported on oesophageal cancer and one study each was reported on gastric, hepatocellular and colorectal cancer. All the included studies followed the Hardy–Weinberg equilibrium assumption in control subjects (p > 0.05). All of the studies used the direct-polymerase chain reaction (PCR) method for genotyping VEGF-2549I/D polymorphism. The contribution of VEGF-2549I/D polymorphism in influencing cancer risk was reported in oesophageal and gastric cancer; however, no relationship was reported with hepatocellular and colorectal cancer. The important salient features of all selected studies are detailed in Table 1.

Table 1 General characteristics of the studies including in the meta-analysis

Assessment of the quality of selected studies

As per the Newcastle–Ottawa scale, out of five studies (Table 2), four studies had high-quality (evaluation score ≥ 7), whereas one study had medium quality (evaluation score = 4–6).

Table 2 Quality assessment of the included studies according to the Newcastle–Ottawa Scale

Meta-analysis of VEGF-2549I/D polymorphism in GIT cancer

The current meta-analysis included a total of 1178 patients and 1520 controls to calculate pooled odds ratio (OR). The results indicated that VEGF-2549I/D polymorphism was not associated with susceptibility to GIT cancers (p > 0.05) in any of the studied genetic model in the overall analysis (Table 3) and on data stratification based on ethnicity (Table 4). Stratification of the data based on the different GIT cancers revealed an increased gastric cancer risk under allele contrast (OR = 1.67, CI = 1.294–2.157, p = 0.00008), recessive (OR = 1.68, CI = 1.056–2.660, p = 0.029), dominant (OR = 2.49, CI = 1.617–3.823, p = 0.00003), over-dominant (OR = 1.52, CI = 1.055–2.177, p = 0.025), II vs DD (OR = 2.97, CI = 1.692–5.208, p = 0.00015) and ID vs DD (OR = 2.35, CI = 1.501–3.669, p = 0.00018) genetic models (Table 5). All the results were graphically displayed as forest plots (Fig. 2).

Table 3 Association of VEGF-2549I/D polymorphism with GIT risk in the overall population
Table 4 Association of VEGF-2549I/D polymorphism with GIT cancer risk based on ethnicity
Table 5 Association of VEGF-2549I/D polymorphism with GIT cancer risk based on cancer type
Fig. 2
figure 2

Forest plots of VEGF-2549I/D polymorphism under different genetic models A Allele contrast, B Recessive, C Dominant, D Over-dominant, E ID vs DD, F II vs ID and G II vs DD genetic model

Assessment of the publication bias and sensitivity analysis

There was no publication bias observed in our study in any of the studied genetic models as evidenced by the graphically constructed funnel plots (Fig. 3). The Egger’s test p value (p > 0.05) indicated no bias in all studied genetic models. We omitted each study and then re-analysed the results and observed no substantial change in the overall results which further confirms the stability of our results (Fig. 4).

Fig. 3
figure 3

Funnel plots of VEGF-2549I/D polymorphism under different genetic models A Allele contrast, B Recessive, C Dominant, D Over-dominant, E II vs DD, F II vs ID and G ID vs DD genetic model

Fig. 4
figure 4

Sensitivity analysis of VEGF-2549I/D polymorphism under different genetic models A Allele contrast, B Recessive, C Dominant, D Over-dominant, E II vs DD, F II vs ID and G ID vs DD genetic model

Trial sequential analysis

The allelic model was used as an example for performing the TSA. There was no crossing of the cumulative Z-value with the TSA monitoring boundaries before reaching the required information size in this study (Fig. 5). Therefore, no conclusion can be drawn from the meta-analysis study and more clinical studies are required in future to confirm the reliability of the study.

Fig. 5
figure 5

Trial sequential analysis of VEGF-2549I/D polymorphism under allelic model

Assessment of the inter-study heterogeneity

In this meta-analysis study, on account of significant heterogeneity observed among the included studies under the allele contrast, recessive, dominant, II versus DD and ID versus DD genetic model, the random effect model was applied in the statistical analysis. The potential sources of heterogeneity were investigated by performing the subgroup analysis based on ethnicity and different tumour types. After subgroup analysis, significant heterogeneity remained in Asians and in oesophageal cancer subgroup. Other reasons for this heterogeneity could be difference in sample size, gender distributions, lifestyle factors, variations in age of the study participants and different sources of controls.

Discussion

VEGF glycoprotein is considered an important angiogenic activator required for tumour angiogenesis. VEGF promoter harbours several single nucleotide polymorphisms (SNPs) which impact the binding of the transcription factors, alter the VEGF expression and contribute to the disease development [15]. So far, six case–control genetic association studies have investigated the relationship of VEGF-2549I/D polymorphism with GIT cancer susceptibility. It was reported that the VEGF-2549I allele and II genotype conferred increased oesophageal cancer risk in the North-Indian population [22]. In South Indians, individuals carrying VEGF-2549ID and II genotype had an increased susceptibility towards gastric cancer [24] The VEGF-2549 D allele and DD genotype increased the susceptibility towards oesophageal cancer in Chinese patients [23]. However, studies conducted on North Indian gall bladder cancer patients [26], Chinese hepatocellular cancer patients [25] and Swedish colorectal cancer patients [27] reported no role of VEGF-2549I/D polymorphism in determining cancer susceptibility. Apart from GIT cancers, VEGF-2549I/D polymorphism has been reported to be associated with risk of several cancers. In North Indians, the VEGF-2549ID genotype was associated with a reduced risk of bladder cancer [32] and a higher risk of developing prostate cancer [36]. The VEGF-2549II genotype and I allele have been associated with increased risk of breast cancer in North Indians [34]. There was no association of VEGF-2549I/D polymorphism with breast cancer risk in the Iranian population [35]. The VEGF-2549D allele has been associated with a higher risk of developing renal cell carcinoma in Caucasian subjects [31]. Ben Wafi et al. [33] reported no association of VEGF-2549I/D polymorphism with urothelial bladder cancer risk in the Tunisian population. The possible reasons for disagreements in study findings could be differences in the sample size, lifestyle factors and different ethnicities.

The impact of VEGF-2549I/D polymorphism on therapy response has been studied in a few GIT cancers. VEGF-2549 DD genotype was associated with a complete or partial response to chemotherapy in Han-Chinese oesophageal cancer patients [23] and with better treatment response and longer progression-free survival in Caucasian colorectal cancer (CRC) patients treated with FOLFIRI-cetuximab [29]. On the contrary, in Caucasian, African-American and Hispanic metastatic CRC patients treated with fluorouracil, irinotecan and bevacizumab, no association of VEGF-2549I/D polymorphism was reported [28]. VEGF-2549I/D polymorphism was not associated with clinicopathological characteristics and overall survival in Korean HCC patients treated with TACE therapy [30].

The role of VEGF-2549I/D polymorphism might vary in different GIT cancer types as explained by differences in the pathologies behind different GIT cancer development. In this meta-analysis, we did not observe any relationship between VEGF-2549I/D polymorphism and GIT cancer susceptibility in the overall population as well as in the pooled analysis based on ethnicity. However, VEGF-2549I/D polymorphism increased the susceptibility to gastric cancer under allele contrast, recessive, dominant, over-dominant, II versus DD and ID versus DD genetic models when data were stratified on the basis of different GIT cancer types. Meta-analysis is an improved statistical study design in genetic association studies. It resolves the disagreements in different study results by combining the findings of case–control studies with small sample size and thereby increases the statistical power of the meta-analysis [48].

The cancerous cells' interaction with the stromal cells creates a tumour environment required for the metastasis, growth and vascularization of tumours in gastric cancer [49,50,51]. It has been reported that angiogenesis influences tumour progression and prognosis in gastric cancer [52]. The correlation of VEGF protein with increased vascularity [53], increased angiogenesis and gastric tumour progression [54,55,56] has been previously studied. Elevated VEGF expression in tumour has also been correlated with unfavourable prognosis in gastric cancer [53, 57]. Increased VEGF serum levels were associated with distant tissue invasion [58], whereas the higher VEGF plasma levels were associated with increased venous invasion [59] in gastric cancer patients. The VEGF-2549I/D polymorphism might regulate the VEGF expression in gastric cancer patients, ultimately affecting cancer susceptibility and prognosis in gastric cancer patients.

The present meta-analysis has several advantages. It is the first meta-analysis report evaluating the role of VEGF-2549I/D polymorphism in GIT cancer susceptibility. We confirmed the robustness of the results by performing sensitivity analysis and trial sequential analysis. In this study, publication bias was not observed in any genetic model.

However, some limitations are still present in this meta-analysis. It involved only few case–control studies and the total sample size of the study was not enough as reported by the TSA results. We observed significant heterogeneity in the included studies and could not identify the main source of heterogeneity which might affect results interpretation. Lastly, we did not stratify the studies based on confounding factors like age, body mass index, sex, family history or lifestyle factors involved in GIT development because of unavailability of research data and the small sample size of the studies. Several previous published studies have evaluated the risk of bias of each included study in the randomized clinical trials [60,61,62]. In the present analysis, we did not calculate the risk of bias for each case–control study included in the meta-analysis.

In conclusion, VEGF-2549I/D polymorphism was associated with an increased risk of gastric cancer only. GIT cancers are polygenic multifactorial disorders, influenced by several lifestyle factors including tobacco smoking, diet, physical activity, alcohol consumption, habitat and chemical exposure. Therefore, the study of the synergistic effect of the VEGF polymorphisms and the environmental factors is needed in future to explore the potential role of VEGF polymorphisms in influencing GIT cancer susceptibility. Larger case–control studies on diverse populations evaluating the combined effect of multiple genetic variants in angiogenesis pathway-associated genes are also needed to understand the exact biological mechanisms behind the GIT cancers development.

Availability of data and materials

All data relevant to this study have been included in the manuscript or uploaded as supplementary information.

Abbreviations

GIT:

Gastrointestinal tract

VEGF:

Vascular endothelial growth factor

GWAS:

Genome-wide association studies

PRISMA:

Preferred Reporting Items for Systematic Reviews and Meta-Analyses

PICO:

Participant, intervention, comparison and outcome

HWE:

Hardy–Weinberg equilibrium

CIs:

Confidence intervals

TSA:

Trial sequential analysis

RIS:

Required information size

RRR:

Relative risk reduction

Direct-PCR:

Direct-polymerase chain reaction

SNP:

Single nucleotide polymorphism

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A et al (2021) Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 71:209–249

    Article  PubMed  Google Scholar 

  2. Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M et al (2000) Environmental and heritable factors in the causation of cancer–analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med 343:78–85

    Article  CAS  PubMed  Google Scholar 

  3. Chin L, Gray JW (2008) Translating insights from the cancer genome into clinical practice. Nature 452:553–563

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Stratton MR (2011) Exploring the genomes of cancer cells: progress and promise. Science 331:1553–1558

    Article  CAS  PubMed  Google Scholar 

  5. Cardon LR, Bell JI (2001) Association study designs for complex diseases. Nat Rev Genet 2:91–99

    Article  CAS  PubMed  Google Scholar 

  6. Houlston RS, Cheadle J, Dobbins SE, Tenesa A, Jones AM, Howarth K et al (2010) Meta-analysis of three genome-wide association studies identifies susceptibility loci for colorectal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33. Nat Genet 42:973–977

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Balasubramanian SP, Brown NJ, Reed MW (2002) Role of genetic polymorphisms in tumour angiogenesis. Br J Cancer 87:1057–1065

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ferrara N, Gerber HP, LeCouter J (2003) The biology of VEGF and its receptors. Nat Med 9:669–676

    Article  CAS  PubMed  Google Scholar 

  9. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:646–674

    Article  CAS  PubMed  Google Scholar 

  10. Huang Z, Bao SD (2004) Roles of main pro- and anti-angiogenic factors in tumour angiogenesis. World J Gastroenterol 10:463–470

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Dvorak HF (2000) VPF/VEGF and the angiogenic response. Semin Perinatol 24:75–78

    Article  CAS  PubMed  Google Scholar 

  12. Ferrara N (2004) Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev 25:581–611

    Article  CAS  PubMed  Google Scholar 

  13. Vincenti V, Cassano C, Rocchi M, Persico G (1996) Assignment of the vascular endothelial growth factor gene to human chromosome 6p21.3. Circulation 93:1493–1495

    Article  CAS  PubMed  Google Scholar 

  14. Renner W, Kotschan S, Hoffmann C, Obermayer-Pietsch B, Pilger E (2000) A common 936 C/T mutation in the gene for vascular endothelial growth factor is associated with vascular endothelial growth factor plasma levels. J Vasc Res 37:443–448

    Article  CAS  PubMed  Google Scholar 

  15. Watson CJ, Webb NJ, Bottomley MJ, Brenchley PE (2000) Identification of polymorphisms within the vascular endothelial growth factor (VEGF) gene: correlation with variation in VEGF protein production. Cytokine 12:1232–1235

    Article  CAS  PubMed  Google Scholar 

  16. Stevens A, Soden J, Brenchley PE, Ralph S, Ray DW (2003) Haplotype analysis of the polymorphic human vascular endothelial growth factor gene promoter. Cancer Res 63:812–816

    CAS  PubMed  Google Scholar 

  17. Mullaney JM, Mills RE, Pittard WS, Devine SE (2010) Small insertions and deletions (INDELs) in human genomes. Hum Mol Genet 19:R131–R136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Catalán A, Glaser-Schmitt A, Argyridou E, Duchen P, Parsch J (2016) An indel polymorphism in the MtnA 3’ untranslated region is associated with gene expression variation and local adaptation in drosophila melanogaster. PLoS Genet 12:e1005987

    Article  PubMed  PubMed Central  Google Scholar 

  19. Veerappa AM, Vishweswaraiah S, Lingaiah K, Murthy NM, Suresh RV, Belur K et al (2014) Insertion-deletions burden in copy number polymorphisms of the Tibetan population. Indian J Hum Genet 20:166–174

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yang B, Cross DF, Ollerenshaw M, Millward BA, Demaine AG (2003) Polymorphisms of the vascular endothelial growth factor and susceptibility to diabetic microvascular complications in patients with type 1 diabetes mellitus. J Diabetes Complicat 17:1–6

    Article  Google Scholar 

  21. Salvarani C, Boiardi L, Casali B, Olivieri I, Cantini F, Salvi F et al (2004) Vascular endothelial growth factor gene polymorphisms in Behçet’s disease. J Rheumatol 31:1785–1789

    CAS  PubMed  Google Scholar 

  22. Guleria K, Kaur S, Mahajan D, Sambyal V, Sudan M, Uppal MS (2022) Impact of VEGFA promoter polymorphisms on esophageal cancer risk in North-West Indians: a case-control study. Genes Genomics 44:923–936

    Article  CAS  PubMed  Google Scholar 

  23. Wang Z, Li C, Li X, Shi J, Wu W (2023) Effect of vascular endothelial growth factor rs35569394 in esophageal cancer and response to chemotherapy. Biomol Biomed 23:271–276

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Bayal AC, Sultana S, Nallari P, Ananthapur V (2021) Genetic polymorphisms of vascular endothelial growth factor (VEGF)-2549 I/D and+ 405G/C in the susceptibility to gastric cancer. Arch Clin Gastroenterol 7:1–6

    Google Scholar 

  25. He Y, Ni J, Chen S, Jiang Y, Jia S, Gao Y (2010) The vascular endothelial growth factor-2549 insertion/deletion polymorphism is not associated with susceptibility to hepatocellular carcinoma in Chinese. DNA Cell Biol 29:393–396

    Article  CAS  PubMed  Google Scholar 

  26. Mishra K, Behari A, Kapoor VK, Khan MS, Prakash S, Agrawal S (2013) Vascular endothelial growth factor single-nucleotide polymorphism in gallbladder cancer. J Gastroenterol Hepatol 28:1678–1685

    Article  CAS  PubMed  Google Scholar 

  27. Ungerbäck J, Elander N, Dimberg J, Söderkvist P (2009) Analysis of VEGF polymorphisms, tumor expression of VEGF mRNA and colorectal cancer susceptibility in a Swedish population. Mol Med Rep 2:435–439

    PubMed  Google Scholar 

  28. Formica V, Palmirotta R, Del Monte G, Savonarola A, Ludovici G, De Marchis ML et al (2011) Predictive value of VEGF gene polymorphisms for metastatic colorectal cancer patients receiving first-line treatment including fluorouracil, irinotecan, and bevacizumab. Int J Colorectal Dis 26:143–151

    Article  PubMed  Google Scholar 

  29. Rollin J, Payancé A, Gouilleux-Gruart V, Boisdron-Celle M, Azzopardi N, Morel A et al (2015) Significant effect of VEGFA polymorphisms on the clinical outcome of metastatic colorectal cancer patients treated with FOLFIRI-cetuximab. Pharmacogenomics 16:2035–2043

    Article  CAS  PubMed  Google Scholar 

  30. Kong SY, Park JW, Lee JA, Park JE, Park KW, Hong EK et al (2007) Association between vascular endothelial growth factor gene polymorphisms and survival in hepatocellular carcinoma patients. Hepatology 46:446–455

    Article  CAS  PubMed  Google Scholar 

  31. Bruyère F, Hovens CM, Marson MN, d’Arcier BF, Costello AJ, Watier H et al (2010) VEGF polymorphisms are associated with an increasing risk of developing renal cell carcinoma. J Urol 184:1273–1278

    Article  PubMed  Google Scholar 

  32. Jaiswal PK, Tripathi N, Shukla A, Mittal RD (2013) Association of single nucleotide polymorphisms in vascular endothelial growth factor gene with bladder cancer risk. Med Oncol 30:509

    Article  PubMed  Google Scholar 

  33. Ben Wafi S, Kallel A, Ben Fradj MK, Sallemi A, Ben Rhouma S, Ben Halima M et al (2018) Haplotype-based association of Vascular Endothelial Growth Factor gene polymorphisms with urothelial bladder cancer risk in Tunisian population. J Clin Lab Anal 32:e22610

    Article  PubMed  PubMed Central  Google Scholar 

  34. Kapahi R, Guleria K, Sambyal V, Manjari M, Sudan M, Uppal MS et al (2015) Association of VEGF and VEGFR1 polymorphisms with breast cancer risk in North Indians. Tumour Biol 36:4223–4234

    Article  CAS  PubMed  Google Scholar 

  35. Rezaei M, Hashemi M, Sanaei S, Mashhadi MA, Taheri M (2016) Association between vascular endothelial growth factor gene polymorphisms with breast cancer risk in an Iranian population. Breast Cancer (Auckl) 10:85–91

    CAS  PubMed  Google Scholar 

  36. George GP, Mittal RD (2011) Association of polymorphic variants of Vascular Endothelial Growth Factor (VEGF) gene in relation to risk and androgen therapy response in Prostate cancer patients of North India. http://www.ichg2011.org/cgi-bin/showdetail.pl?absno=10216

  37. Johnstone S, Logan RM (2007) Expression of vascular endothelial growth factor (VEGF) in normal oral mucosa, oral dysplasia and oral squamous cell carcinoma. Int J Oral Maxillofac Surg 36:263–266

    Article  CAS  PubMed  Google Scholar 

  38. Macedo F, Ladeira K, Longatto-Filho A, Martins SF (2017) Gastric cancer and angiogenesis: Is VEGF a useful biomarker to assess progression and remission? J Gastric Cancer 17:1–10

    Article  PubMed  PubMed Central  Google Scholar 

  39. Talar-Wojnarowska R, Gasiorowska A, Olakowski M, Lekstan A, Lampe P, Smolarz B et al (2010) Vascular endothelial growth factor (VEGF) genotype and serum concentration in patients with pancreatic adenocarcinoma and chronic pancreatitis. J PhysiolPharmacol 61:711–716

    CAS  Google Scholar 

  40. Shimada H, Takeda A, Nabeya Y, Okazumi SI, Matsubara H, Funami Y et al (2001) Clinical significance of serum vascular endothelial growth factor in esophageal squamous cell carcinoma. Cancer 92:663–669

    Article  CAS  PubMed  Google Scholar 

  41. Karayiannakis AJ, Bolanaki H, Syrigos KN, Asimakopoulos B, Polychronidis A, Anagnostoulis S et al (2003) Serum vascular endothelial growth factor levels in pancreatic cancer patients correlate with advanced and metastatic disease and poor prognosis. Cancer Lett 194:119–124

    Article  CAS  PubMed  Google Scholar 

  42. Yao DF, Wu XH, Zhu Y, Shi GS, Dong ZZ, Yao DB et al (2005) Quantitative analysis of vascular endothelial growth factor, microvascular density and their clinicopathologic features in human hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 4:220–226

    CAS  PubMed  Google Scholar 

  43. Lordache S, Saftoiu A, Georgescu CV, Ramboiu S, Gheonea DI, Filip M et al (2010) Vascular endothelial growth factor expression and microvessel density–two useful tools for the assessment of prognosis and survival in gastric cancer patients. J Gastrointestin Liver Dis 19:135–139

    Google Scholar 

  44. Moher D, Liberati A, Tetzlaff J, Altman DG (2010) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Int J Surg 8:336–341

    Article  PubMed  Google Scholar 

  45. Eriksen MB, Frandsen TF (2018) The impact of patient, intervention, comparison, outcome (PICO) as a search strategy tool on literature search quality: a systematic review. J Med Libr Assoc 1(06):420–431

    Google Scholar 

  46. Lo CK, Mertz D, Loeb M (2014) Newcastle-Ottawa Scale: comparing reviewers’ to authors’ assessments. BMC Med Res Methodol 14:45

    Article  PubMed  PubMed Central  Google Scholar 

  47. Higgins JP, Thompson SG, Deeks JJ, Altman DG (2003) Measuring inconsistency in meta-analyses. BMJ 327:557–560

    Article  PubMed  PubMed Central  Google Scholar 

  48. Conneely KN, Boehnke M (2010) Meta-analysis of genetic association studies and adjustment for multiple testing of correlated SNPs and traits. Genet Epidemiol 34:739–746

    Article  PubMed  PubMed Central  Google Scholar 

  49. Fidler IJ (1990) Host and tumour factors in cancer metastasis. Eur J Clin Invest 20:481–486

    Article  CAS  PubMed  Google Scholar 

  50. Mantovani A, Allavena P, Sica A, Balkwill F (2008) Cancer-related inflammation. Nature 454:436–444

    Article  CAS  PubMed  Google Scholar 

  51. Whiteside TL (2008) The tumor microenvironment and its role in promoting tumor growth. Oncogene 27:5904–5912

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kitadai Y (2010) Angiogenesis and lymphangiogenesis of gastric cancer. J Oncol 2010:468725

    Article  PubMed  PubMed Central  Google Scholar 

  53. Tanigawa N, Amaya H, Matsumura M, Shimomatsuya T (1997) Correlation between expression of vascular endothelial growth factor and tumor vascularity, and patient outcome in human gastric carcinoma. J Clin Oncol 15:826–832

    Article  CAS  PubMed  Google Scholar 

  54. Tanimoto H, Yoshida K, Yokozaki H, Yasui W, Nakayama H, Ito H et al (1991) Expression of basic fibroblast growth factor in human gastric carcinomas. Virchows Arch B Cell PatholIncl Mol Pathol 61:263–267

    Article  CAS  Google Scholar 

  55. Takahashi Y, Cleary KR, Mai M, Kitadai Y, Bucana CD, Ellis LM (1996) Significance of vessel count and vascular endothelial growth factor and its receptor (KDR) in intestinal-type gastric cancer. Clin Cancer Res 2:1679–1684

    CAS  PubMed  Google Scholar 

  56. Yamamoto S, Yasui W, Kitadai Y, Yokozaki H, Haruma K, Kajiyama G et al (1998) Expression of vascular endothelial growth factor in human gastric carcinomas. Pathol Int 48:499–506

    Article  CAS  PubMed  Google Scholar 

  57. Maeda K, Chung YS, Ogawa Y, Takatsuka S, Kang SM, Ogawa M et al (1996) Prognostic value of vascular endothelial growth factor expression in gastric carcinoma. Cancer 77:858–863

    Article  CAS  PubMed  Google Scholar 

  58. Bilgiç CI, Tez M (2015) Serum VEGF levels in gastric cancer patients: correlation with clinicopathological parameters. Turk J Med Sci 45:112–117

    Article  PubMed  Google Scholar 

  59. Ohta M, Konno H, Tanaka T, Baba M, Kamiya K, Syouji T et al (2003) The significance of circulating vascular endothelial growth factor (VEGF) protein in gastric cancer. Cancer Lett 192:215–225

    Article  CAS  PubMed  Google Scholar 

  60. Lin Z, Sui X, Li L, Wang Y, Zhao J (2022) The effect of metformin on low birth weight girls with precocious puberty: a protocol for systematic review and meta-analysis. Medicine 101:e29765

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhao J, Sui X, Shi Q, Su D, Lin Z (2022) Effects of antioxidant intervention in patients with polycystic ovarian syndrome: a systematic review and meta-analysis. Medicine 101:e30006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhao J, Dong L, Lin Z, Sui X, Wang Y, Li L et al (2023) Effects of selenium supplementation on Polycystic Ovarian Syndrome: a systematic review and meta-analysis on randomized clinical trials. BMC EndocrDisord 23:33

    CAS  Google Scholar 

Download references

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

KG and VS designed the study. DM and KG prepared the manuscript. All authors read and approved the manuscript.

Corresponding author

Correspondence to Kamlesh Guleria.

Ethics declarations

Ethics approval

Not applicable.

Informed consent

Not applicable

Competing interests

All the authors declare that they have no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mahajan, D., Sambyal, V. & Guleria, K. Association of VEGF-2549I/D promoter polymorphism with gastrointestinal tract cancer risk: a meta-analysis. Egypt J Med Hum Genet 25, 65 (2024). https://doi.org/10.1186/s43042-024-00535-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43042-024-00535-0

Keywords