Angiogenesis is an important host process that interacts with cancer cells to promote growth, invasion, and metastasis. Numerous therapeutic agents targeting the VEGF pathway have been developed. Host variability in VEGF pathway can influence angiogenesis-dependent signaling, altering sensitivity to antiangiogenic drugs and prognosis. A systematic review and meta-analysis was conducted (May 1990–July 2011). Eligible studies involved cancer patients and compared polymorphisms in the VEGF pathway [VEGF and molecules directly interacting with VEGF: KDR, FLT1, FGF, FGF2, FGFR, NRP1, endostatin (encoded by COL18A1)], and reported one of the following outcomes: overall survival, progression-free survival, time to recurrence, disease-free survival, response rate, or drug toxicity. We identified 48 cancer studies assessing prognosis and 12 cancer studies exploring pharmacogenetics of anti-VEGF therapy across various VEGF pathway polymorphisms. There was marked inter- and intradisease site heterogeneity in the effect of polymorphisms on both outcome and response to therapy. Meta-analyses of 5 VEGF polymorphisms (+936C>T, −460T>C, +405G>C, −1154G>A, and −2578C>A) identified a significant prognostic relationship: VEGF +405G>C variants showed a highly statistically significant improvement in overall survival [HR, 0.74; 95% confidence interval, 0.60–0.91; P = 0.004]. Variants (heterozygotes and/or homozygotes) of VEGF +405G>C were significantly associated with improved survival in a meta-analysis of multiple cancer sites. Clin Cancer Res; 18(17); 4526–37. ©2012 AACR.

Translational Relevance

Antiangiogenic drugs are part of the clinical armamentarium for numerous solid tumors. Results of clinical trials of antiangiogenic agents have shown variable results. The need for predictive and prognostic markers for antiangiogenic therapy is, therefore, acute. Here, we report a systematic review and meta-analysis of single nucleotide polymorphisms in VEGF genes and their related protein receptors. Results showed substantial heterogeneity in the design and results of both predictive and prognostic studies. Our meta-analysis revealed a significant improvement in survival for VEGF +405G>C variants in unselected cancer patients. There did not appear to be an association between germline polymorphisms and benefit or toxicity from antiangiogeneic agent. However, inconsistency in the definition of these endpoints limited our analysis of predictive power. Future prospective studies should extend this research to include pathway analysis, haplotype analysis, and discovery-focused genome-wide association studies, followed by appropriate validation or meta-analytic analyses.

Angiogenesis is a critical component in oncogenesis (1). Tumors can promote angiogenesis through the secretion of proangiogenic factors such as VEGF (and its isoforms) and basic fibroblast growth factor (bFGF) or overexpression of associated receptors: VEGFR, VEGFR2/kinase insert domain receptor (KDR), and NRP2 (2). Tumors use this host-mediated process to meet their metabolic needs, to help remove biologic waste products of rapidly dividing cells, and to provide a mechanism for metastasis.

Angiogenic factors interact with a multitude of pathways, including matrix metalloproteinases, inflammatory cytokines, chemokines (e.g., interleukins and interferons), integrins, nitric oxide, and other endothelial-related proteins. Downstream signaling effectors of angiogenesis receptors also lead to the activation of the phosphoinositide 3-kinase/AKT (PI3K/AKT) and mitogen-activated protein kinase (MAPK) pathways.

The VEGF pathway is predominantly driven by host factors. Host genetic variability in this pathway may, therefore, influence angiogenesis-dependent biologic pathways during cancer development and hence influence outcome and sensitivity to various therapies. Associations between genetic polymorphisms in VEGF and the risk of developing various cancers have been reviewed previously (3). Less is known about the role of potential germline polymorphisms as prognostic markers of clinical outcomes or the potential predictive or pharmacogenetic nature of these polymorphisms in relation to drug efficacy and toxicity.

We conducted a systematic review and meta-analysis of the role of VEGF pathway polymorphisms as markers of clinical outcome and toxicity in cancer patients. The 2 aims were as follows: (i) to investigate polymorphisms as pharmacogenetic predictors of drug efficacy and toxicities and (ii) to investigate polymorphisms as general prognostic indicators of survival, independent of therapy.

Search query

We conducted a literature search of MEDLINE (host: PubMed) for the period of May 1990 through July 2011 using the keywords and MeSH terms: {“angiogenesis,” “VEGF,” “VEGFR1,” “FLT1,” “KDR,” “VEGFR2,” “FGF2,” “FGF,” “FGFR,” “NRP1,” “Endostatin,” “VEGFA,” “VEGFB,” and “Bevacizumab”} and “cancer” and “polymorphism.” We limited searches to English language articles.

Study inclusion and exclusion criteria

Article eligibility was reviewed independently by 2 reviewers (G. Liu, L. Eng). Eligible articles were original peer-reviewed studies focused on genetic variation in the following angiogenesis pathway genes: VEGFA, VEGFB, and FGF2, and/or genes that code for their direct interactors or receptors: fms-related tyrosine kinase 1 (FLT/VEGFR1), KDR (VEGFR2), neuropilin 1 (NRP1), and FGFR. These genes were selected from The Pharmacogenomics Knowledgebase website (4). We also included endostatin, the 20-kDa C-terminal fragment derived from type XVIII collagen that is a broad-spectrum antiangiogenic factor, which can affect both VEGF and FGF2 pathways and is coded by COL18A1. Eligible studies must have reported cancer outcome (i.e., not risk alone), and may have included either/both prognostic and pharmacogenetic assessments. The outcomes evaluated were of any of the following: overall survival (OS), progression-free survival (PFS), time to progression (TTP), time to recurrence (TTR), disease-free survival (DFS), response rate (RR), and association with treatment toxicity. Finally, all studies must have included genotyping of 25 or more and at least one VEGF pathway polymorphism to warrant inclusion.

Duplicate studies were excluded as were those testing solely the association of polymorphisms and cancer risk/susceptibility, tumor staging/grading, or gene expression. We also excluded case reports, opinion pieces (e.g., letters to the editors), and other review papers. We categorized studies as being pharmacogenetic (effect of polymorphism on response/outcome/toxicity to therapy where treatment included antiangiogenic therapy) or prognostic (effect of polymorphism on outcome irrespective of therapy); data were analyzed for various disease sites: breast, colorectal, gastroesophageal, genitourinary, gynecologic, lung, and other cancers.

Data extraction and organization

The following data were extracted from individual publications: title, authors, disease site, country, sample size, and/or actual number of patients genotyped, patient inclusion/exclusion criteria, treatment/intervention details, histologic subtypes, grade, stage, tissue used for genotyping (e.g., whole blood vs. archival normal tissue), and polymorphisms that were genotyped. Associations between polymorphisms and outcome data were recorded as OR, HR, or RR, along with their respective confidence intervals (CI). We noted whether studies were analyzed using only univariate (or crude) analysis or in multivariate (or adjusted) analysis. Haplotype analyses, if conducted, were also recorded. A common problem was the use of different nomenclature and names for the same polymorphism. Thus, all identified polymorphisms sharing the same RefSNP (rs) numbers were renamed to share common names throughout this manuscript [Supplementary Table S1 provides the common and Human Genome Variation Society nomenclature; ref. (5)].

Meta-analysis

Meta-analyses were conducted to evaluate the overall prognostic associations of the most commonly evaluated VEGF polymorphisms when there were an adequate number of individual studies reporting similar outcomes to justify such an analysis. Only studies reporting HR for OS were included. Analyses were undertaken separately for each polymorphism and were conducted in 2 phases. First, study data reporting outcome by wild-type or variant polymorphisms irrespective of zygosity were assessed together (i.e., pooled). Second, data were assessed separately for homozygous and heterozygous genotypes. Differences between homozygous and heterozygous subgroups were assessed using methods described by Deeks and colleagues (6). Data were analyzed using RevMan 5.1 analysis software (The Cochrane Collaboration). Pooled estimates of HR were computed using generic inverse variance and a random-effects model (7, 8). All statistical tests were 2-sided, and statistical significance was defined as P < 0.01, which corrected for multiple statistical testing using the Bonferroni method.

Summary of included and excluded studies

A total of 614 studies were initially identified (Fig. 1). Of these, 186 were duplicate reports/studies, 130 were related to cancer development risk, and 238 further studies were excluded because they did not include at least 25 patients, did not include polymorphisms of interest, or did not assess any pharmacogenetic or prognostic outcomes of interest. Sixty studies met the inclusion criteria and were included in the final analysis.

Figure 1.

Flow chart summarizing the literature search and selection process for inclusion of studies into the systematic review and meta-analysis.

Figure 1.

Flow chart summarizing the literature search and selection process for inclusion of studies into the systematic review and meta-analysis.

Close modal

Most of the included studies had sample sizes of under 150 patients, and were secondary analyses of phase I/II clinical trials. Other characteristics of included studies are shown in Table 1. The majority of trials were prognostic (n = 48), based primarily on data from case series [n = 33 (69%)]. Most pharmacogenetics studies (n = 12) were nested case–control analyses of clinical trials [n = 9 (75%)].

Table 1.

Baseline characteristics of included studies

Prognostic studiesPharmacogenetic studies
Number of studies (%)Number of studies (%)
Total number of studies 48 (100) 12 (100) 
Source of patients   
 Case series 33 (69) 3 (25) 
 Cases from case–control risk studies 11 (23) 0 (0) 
 Secondary analysis of clinical trial data 4 (9) 9 (75) 
 Developed cancer after entering cohort study 1 (2) 0 (0) 
Type of patient   
 Caucasian predominant sample 33 (69) 11 (92) 
 Asian predominant sample 14 (29) 1 (8) 
Location of study   
 North and Central American 14 (29) 5 (42) 
 South American 1 (2) 0 (0) 
 European 18 (4) 6 (50) 
 East Asian 13 (27) 1 (8) 
 South Asian 1 (2) 0 (0) 
 Australian 1 (2) 0 (0) 
Type of sample   
 Blood 38 (79) 11 (92) 
 Frozen normal tissue 2 (4) 0 (0) 
 Frozen tumor tissue 5 (10) 0 (0) 
 FFPE adjacent tissue 4 (8) 0 (0) 
 FFPE tumor tissue 4 (8) 1 (8) 
Disease site   
 Breast 6 (13) 2 (17) 
 Colorectal (GI) 7 (15) 3 (25) 
 Gastroesophageal (upper GI) 9 (19) 1 (8) 
 Genitourinary 4 (8) 0 (0) 
 Gynecologic 8 (17) 2 (17) 
 Lung 4 (8) 0 (0) 
 Other (includes liver, melanoma, AML, CML, NHL, head, and neck) 10 (21) 4 (33) 
Selection of polymorphisms   
 Any VEGF 42 (88) 9 (75) 
VEGF −2578C>A 20 (42) 6 (50) 
 VEGF −1154G>A 13 (27) 5 (42) 
 VEGF −460C>T 19 (40) 6 (50) 
 VEGF +405G>C 33 (69) 9 (75) 
 VEGF +936C>T 31 (65) 9 (75) 
 Any KDR/VEGFR2 or FLT1/VEGFR1 8 (17) 6 (50) 
 Any Endostatin, FGF2 or FGFR 5 (10) 0 (0) 
Type of analysis   
 Univariate analysis only 13 (27) 9 (75) 
  Observational study 12 (25) 1 (8) 
  Clinical trial 1 (2) 8 (67) 
 Multivariate analysis conducted 35 (73) 3 (25) 
  Observational study 32 (67) 2 (17) 
  Clinical trial 3 (6) 1 (8) 
Type of predictive analysis   
 Survival outcomes Not applicable 9 (75) 
 Toxicity  10 (83) 
Prognostic studiesPharmacogenetic studies
Number of studies (%)Number of studies (%)
Total number of studies 48 (100) 12 (100) 
Source of patients   
 Case series 33 (69) 3 (25) 
 Cases from case–control risk studies 11 (23) 0 (0) 
 Secondary analysis of clinical trial data 4 (9) 9 (75) 
 Developed cancer after entering cohort study 1 (2) 0 (0) 
Type of patient   
 Caucasian predominant sample 33 (69) 11 (92) 
 Asian predominant sample 14 (29) 1 (8) 
Location of study   
 North and Central American 14 (29) 5 (42) 
 South American 1 (2) 0 (0) 
 European 18 (4) 6 (50) 
 East Asian 13 (27) 1 (8) 
 South Asian 1 (2) 0 (0) 
 Australian 1 (2) 0 (0) 
Type of sample   
 Blood 38 (79) 11 (92) 
 Frozen normal tissue 2 (4) 0 (0) 
 Frozen tumor tissue 5 (10) 0 (0) 
 FFPE adjacent tissue 4 (8) 0 (0) 
 FFPE tumor tissue 4 (8) 1 (8) 
Disease site   
 Breast 6 (13) 2 (17) 
 Colorectal (GI) 7 (15) 3 (25) 
 Gastroesophageal (upper GI) 9 (19) 1 (8) 
 Genitourinary 4 (8) 0 (0) 
 Gynecologic 8 (17) 2 (17) 
 Lung 4 (8) 0 (0) 
 Other (includes liver, melanoma, AML, CML, NHL, head, and neck) 10 (21) 4 (33) 
Selection of polymorphisms   
 Any VEGF 42 (88) 9 (75) 
VEGF −2578C>A 20 (42) 6 (50) 
 VEGF −1154G>A 13 (27) 5 (42) 
 VEGF −460C>T 19 (40) 6 (50) 
 VEGF +405G>C 33 (69) 9 (75) 
 VEGF +936C>T 31 (65) 9 (75) 
 Any KDR/VEGFR2 or FLT1/VEGFR1 8 (17) 6 (50) 
 Any Endostatin, FGF2 or FGFR 5 (10) 0 (0) 
Type of analysis   
 Univariate analysis only 13 (27) 9 (75) 
  Observational study 12 (25) 1 (8) 
  Clinical trial 1 (2) 8 (67) 
 Multivariate analysis conducted 35 (73) 3 (25) 
  Observational study 32 (67) 2 (17) 
  Clinical trial 3 (6) 1 (8) 
Type of predictive analysis   
 Survival outcomes Not applicable 9 (75) 
 Toxicity  10 (83) 

Abbreviations: AML, acute myeloid leukemia; CML, chronic myeloid leukemia; FFPE, formalin-fixed paraffin-embedded; GI, gastrointestinal; NHL, non-Hodgkin lymphoma.

VEGF pathway polymorphisms as predictive markers of bevacizumab efficacy

Nine studies investigated the predictive effect of polymorphisms on response to bevacizumab in numerous cancer sites (Table 2, VEGF pathway polymorphic predictive associations of survival and response; additional data in Supplementary Table S2; refs. 9–17). Among those investigating bevacizumab response, Schneider and colleagues (N = 341) reported that the C allele of VEGF −2578C>A and G allele of VEGF −1154G>A (alternatively known as −116G>A) were independently associated with poorer OS in patients treated with bevacizumab (9). Similarly, a smaller study investigating 9 polymorphisms, by Formica and colleagues (N = 40), reported the same finding for VEGF −1154G>A with respect to PFS (corrected for multiple comparisons) and also found the C allele for VEGF +405G>C (alternatively known as +634G>C) associated with improved overall radiologic RR (12). Another small study by Argiris and colleagues (N = 28), found the C/− genotypes of VEGF +405G>C significantly associated with OS and TTP in multivariate analysis, but only 1 patient was present in the G/G genotype group (14).

Table 2.

Summary of pharmacogenetic cancer studies involving VEGF pathway: May 1990–July 2011

SiteLast name of first author(reference)Clinical trialDrugsNCountryGenes assessedPolymorphisms assessed, nSignificant results
VEGF pathway polymorphic predictive associations of survival and response 
 Breast Schneider (9) Bevacizumab + paclitaxel vs. paclitaxel 341 United States VEGF KDR VEGF −2578C>A, A associated with improved OS 
        VEGF −1154G>A, A associated with improved OS 
 Breast Eitenne-Grimaldi (10) Bevacizumab +/− taxane 138 France VEGF VEGF +936C>T, T associated with improved TTP 
 CRC Loupakis (11) FOLFIRI + bevacizumab vs. FOLFIRI 111 Italy VEGF VEGF −460C>T, C associated with improved PFS 
 CRC Formica (12) FOLFIRI + bevacizumab 40 Italy VEGF VEGF −1154G>A, A associated with improved PFS 
        VEGF +405G>C, G associated with improved RR 
 CRC Zhang (13) Bevacizumab + cetuximab vs. cetuximab 65 United States VEGF KDR  
      NRP1   
 SCCHN Argiris (14) Pemetrexed and bevacizumab 28 United States VEGF VEGF +405G>C, G associated with improved with improved OS, TTP but only 1 patient carried G/G 
 Ovarian Schulthesis (15) Bevacizumab + cyclophosphamide 70 United States NRP1 VEGF NRP1 3′UTR C>T, T associated with poorer PFS 
      KDR   
 Ovarian Smerdel (16) Bevacizumab 38 Denmark VEGF  
 Mixed Jain (17) Sorafenib + bevacizumab 178 United States KDR  
VEGF pathway polymorphic predictive associations of toxicity 
Breast Schneider (9) Bevacizumab + paclitaxel vs. paclitaxel 341 United States VEGF KDR VEGF +405G>C, G associated with increased HTNVEGF −460C>T, C associated with increased HTN 
Breast Eitenne-Grimaldi (10) Bevacizumab +/− taxane 138 France VEGF VEGF +405G>C, C associated with increased toxicity 
Esophageal Sakaeda (18) 5-FU/cisplatin/radiation 49 Japan VEGF VEGF −460C>T, C associated with increased chelitis 
        VEGF −1154G>A, A associated with increased chelitis and leukopenia 
CRC Loupakis (11) FOLFIRI + bevacizumab vs. FOLFIRI 111 Italy VEGF  
CRC Formica (12) FOLFIRI + bevacizumab 40 Italy VEGF  
SCCHN Argiris (14) Pemetrexed and bevacizumab 28 United States VEGF  
Ovarian Schulthesis (15) Bevacizumab + cyclophosphamide 70 United States NRP1  
      VEGF   
      KDR   
Mixed Jain (17) Sorafenib + bevacizumab 178 United States KDR KDR +1719A>T, T associated with increased toxicity 
Mixed Steeghs (19) Danusertib 63 Netherlands KDR  
      FLT3   
      FLT4   
Mixed Steeghs (20) Telatinib (phase I) 33 Netherlands KDR  
      FLT4   
SiteLast name of first author(reference)Clinical trialDrugsNCountryGenes assessedPolymorphisms assessed, nSignificant results
VEGF pathway polymorphic predictive associations of survival and response 
 Breast Schneider (9) Bevacizumab + paclitaxel vs. paclitaxel 341 United States VEGF KDR VEGF −2578C>A, A associated with improved OS 
        VEGF −1154G>A, A associated with improved OS 
 Breast Eitenne-Grimaldi (10) Bevacizumab +/− taxane 138 France VEGF VEGF +936C>T, T associated with improved TTP 
 CRC Loupakis (11) FOLFIRI + bevacizumab vs. FOLFIRI 111 Italy VEGF VEGF −460C>T, C associated with improved PFS 
 CRC Formica (12) FOLFIRI + bevacizumab 40 Italy VEGF VEGF −1154G>A, A associated with improved PFS 
        VEGF +405G>C, G associated with improved RR 
 CRC Zhang (13) Bevacizumab + cetuximab vs. cetuximab 65 United States VEGF KDR  
      NRP1   
 SCCHN Argiris (14) Pemetrexed and bevacizumab 28 United States VEGF VEGF +405G>C, G associated with improved with improved OS, TTP but only 1 patient carried G/G 
 Ovarian Schulthesis (15) Bevacizumab + cyclophosphamide 70 United States NRP1 VEGF NRP1 3′UTR C>T, T associated with poorer PFS 
      KDR   
 Ovarian Smerdel (16) Bevacizumab 38 Denmark VEGF  
 Mixed Jain (17) Sorafenib + bevacizumab 178 United States KDR  
VEGF pathway polymorphic predictive associations of toxicity 
Breast Schneider (9) Bevacizumab + paclitaxel vs. paclitaxel 341 United States VEGF KDR VEGF +405G>C, G associated with increased HTNVEGF −460C>T, C associated with increased HTN 
Breast Eitenne-Grimaldi (10) Bevacizumab +/− taxane 138 France VEGF VEGF +405G>C, C associated with increased toxicity 
Esophageal Sakaeda (18) 5-FU/cisplatin/radiation 49 Japan VEGF VEGF −460C>T, C associated with increased chelitis 
        VEGF −1154G>A, A associated with increased chelitis and leukopenia 
CRC Loupakis (11) FOLFIRI + bevacizumab vs. FOLFIRI 111 Italy VEGF  
CRC Formica (12) FOLFIRI + bevacizumab 40 Italy VEGF  
SCCHN Argiris (14) Pemetrexed and bevacizumab 28 United States VEGF  
Ovarian Schulthesis (15) Bevacizumab + cyclophosphamide 70 United States NRP1  
      VEGF   
      KDR   
Mixed Jain (17) Sorafenib + bevacizumab 178 United States KDR KDR +1719A>T, T associated with increased toxicity 
Mixed Steeghs (19) Danusertib 63 Netherlands KDR  
      FLT3   
      FLT4   
Mixed Steeghs (20) Telatinib (phase I) 33 Netherlands KDR  
      FLT4   

Abbreviations: CRC, colorectal cancer; FOLFIRI, 5-fluorouracil, folinic acid, irinotecan; 5-FU, 5-fluorouracil; HN, hypertension; N, no; NS, not statistically significant; SCCHN, squamous cell cancer of the head and neck; Y, yes.

In all other studies, associations have yet to be validated: (i) one report by Etienne-Grimaldi and colleagues (N = 138), identified the C allele of VEGF +936C>T associated with a shorter TTP; (ii) Loupakis and colleagues (N = 111), found the C allele of VEGF −460C>T (alternatively known as −1498T>C) associated with improved PFS; and (iii) Schultheis and colleagues (N = 70) reported that the C/T genotype of C>T polymorphism in the 3′ UTR of NRP1 was associated with improved PFS, after adjustment for 30 comparisons (10, 11, 15). In 3 other studies by Jain and colleagues (n = 178), Zhang and colleagues (n = 65; using both univariate and multivariate analyses), and Smerdel and colleagues (n = 38), VEGF, KDR, and NRP1 polymorphisms were found to have no association with survival (13, 16, 17).

VEGF pathway polymorphisms and treatment toxicity

Seven studies investigated bevacizumab toxicity in various cancer sites (9–12, 14, 15, 17). Three additional studies investigated the predictive effect of VEGF polymorphisms on toxicity of non-VEGF targeting drugs (Table 2, VEGF pathway polymorphic predictive associations of toxicity; additional data in Supplementary Table S3; refs. 18–20). The experimental paclitaxel and bevacizumab arm of a breast cancer study (n = 180) identified the C allele in VEGF −460C>T and the G allele in VEGF +405G>C as being associated with a greater frequency of grade 3 or 4 hypertension (9). In contrast, Etienne-Grimaldi and colleagues, found the G allele of VEGF +405G>C associated with decreased toxicity (which included any signs of hypertension, arterial or venous thromboemboli, or hemorrhages; ref. 10). Four smaller studies (N = 28–111) involving various combinations of bevacizumab with chemotherapeutic drugs found no associations of any evaluated polymorphisms with any of the toxicities (11, 12, 14, 15). In Jain and colleagues' investigation of KDR polymorphisms, the A allele in KDR 1718T>A (alternatively known as 1719A>T, Q472H, 1416A>T, or Exon 11T>A) was found associated with increased frequency of hypertension and hand–foot skin reactions in patients treated with sorafenib and bevacizumab (17).

Three other studies investigated the role of VEGF polymorphisms in the toxicities in non-VEGF targeting drugs (18–20). In a 49 patient study focused on drug toxicity of 5-fluorouracil and cisplatin chemotherapy, a multitude of nominally significant VEGF polymorphic associations with various toxicities such as cheilitis, leukopenia, and stomatitis was reported (18). Two studies, 1 investigating danusertib (a multikinase aurora inhibitor) and another on telatinib found no associations between any KDR or FLT polymorphisms and toxicity (19, 20).

VEGF polymorphisms as prognostic biomarkers

Studies that evaluated VEGF pathway polymorphisms as prognostic markers of cancer outcome are described in Supplementary Tables S4–S7 (21–68).

VEGF +936C>T was associated with OS, DFS, or TTR in the 3 largest colorectal cancer studies involving; however, 2 showed improved outcome by the T allele while 1 showed worse outcome by the T allele (27, 29, 32). Among these 3 studies, the 2 showing improved outcome by the T allele involved patients from the United States and Greece and both used blood as their tissue source while the one showing worse outcome was from Korea and used fresh tumor tissue as their tissue source. There was more consistency with VEGF +405G>C, where 2 of these studies showed improved outcome in patients carrying the C allele (a third smaller study reported no significant association; refs. 27, 32). Of the 2 studies showing improved outcome, both included cases from all stages; Dassoulas and colleagues evaluated a Greek population while Kim and colleagues focused on a Korean population that specifically underwent surgical resection.

There was no consistency in the relationship between various VEGF polymorphisms and clinical outcome in gastroesophageal cancers. For example, 1 gastric cancer study (N = 137) found a strong relationship between carrying at least one A allele in COL18A1 (Endostatin) +4349G>A (alternatively known as D104N) and worse outcome [adjusted HR (aHR) for TTR, 2.28; 95% CI, 1.26–4.11; P = 0.004) while a larger 239 patient study in distal esophageal and gastroesophageal junction adenocarcinomas found no relationship (35, 39).

Of 5 breast cancer studies, VEGF polymorphisms, −7C>T and +405G>C, and the KDR polymorphism, +1192A>G, were associated with either OS or DFS in single, unvalidated studies (21, 22, 24). In genitourinary and gynecologic cancers, associations were either of borderline statistical significance or involved crude, unadjusted analyses, with 2 exceptions: VEGF +405C/C genotype was associated with poorer OS in a cervical cancer study (n = 199) from Korea; and minor alleles of VEGFC polymorphisms (rs17697305 and rs1485766) were associated with altered OS outcomes in a single ovarian cancer study that evaluated more than 1,400 polymorphisms across multiple pathways in 325 patients (52, 54). Separate lung cancer studies reported worse survival with VEGF +405G/G, VEGF −1154A/-, and VEGF −460T/T; none being validated (55, 56, 58).

Meta-analyses of prognostic studies

Because of the large number of studies (n = 42) evaluating 5 individual VEGF polymorphisms (VEGF +936C>T, VEGF −460T>C, VEGF −1154G>A, VEGF −2578C>A and VEGF +405G>C; individual details in Supplementary Tables S4–S7), meta-analyses were conducted separately on these 5 polymorphisms. When the homozygous and heterozygous polymorphisms were assessed together (i.e., pooled; Table 3, Combined heterozygosity and homozygosity results), the C allele of VEGF +405G>C was found significantly associated with improved OS (HR, 0.74; 95% CI, 0.60–0.91; P = 0.004). The corresponding forest plot is shown in Table 4, Forest plots for pooled and subgroup analysis of VEGF polymorphisms as prognostic biomarkers for VEGF +405G>C. Among studies in the zygosity-segregated analysis (Table 3, Zygosity-segregated meta-analysis results), the same variant of VEGF +405G>C also showed a statistically significant improvement in OS (HR, 0.79; 95% CI, 0.67–0.94; P = 0.007) that was not different by zygosity (Table 4, Forest plots for pooled and subgroup analysis of VEGF polymorphisms as prognostic biomarkers for VEGF +405G>C). In addition, a nonsignificant trend was identified where VEGF +936C>T variants (regardless of zygosity) were associated with worse OS (HR, 1.38; 95% CI, 0.96–1.97; P = 0.08; Supplementary Fig. S1B). For other polymorphisms, there was no evidence of association with survival in either pooled or zygosity-segregated analyses (Supplementary Figs. S2–S4). Assessment of funnel plots in both pooled and zygosity-segregated analysis revealed no any evidence of publication bias (data not shown).

Table 3.

Meta-analysis results for prognostic significance of specific VEGF polymorphisms

VEGF SNPsPooled HRs from meta-analysis, (95% CI); P value
Combined heterozygosity and homozygosity results
−2578C>A 0.92 (0.65–1.29); P = 0.62 
−1154G>A 1.07 (0.81–1.41); P = 0.63 
−460T>C 1.05 (0.78–1.43); P = 0.73 
+405G>C 0.74 (0.60–0.91); P = 0.004 
+936C>T 1.15 (0.86–1.53); P = 0.35 
Zygosity-segregated meta-analysis results 
VEGF SNPs Comparison Zygosity-specific HRs from meta-analysis, (95% CI); P value Subgroup differences P value Overall pooled HRs from meta-analysis (95% CI) Overall P value 
−2578C>A A/A vs. C/C 0.93 (0.55–1.59); P = 0.79 0.98 0.91 (0.72–1.14) 0.41 
 C/A vs. C/C 0.93 (0.79–1.08); P = 0.33    
−1154G>A A/A vs. G/G 0.93 (0.55–1.58); P = 0.80 0.45 0.98 (0.72–1.33) 0.71 
 G/A vs. G/G 1.18 (0.89–1.56); P = 0.26    
−460T>C C/C vs. T/T 1.10 (0.79–1.53); P = 0.56 0.77 1.06 (0.89–1.27) 0.52 
 T/C vs. T/T 1.04 (0.84–1.29); P = 0.72    
+405G>C C/C vs. G/G 0.78 (0.58–1.04); P = 0.09 0.72 0.79 (0.67–0.94) 0.007 
 G/C vs. G/G 0.83 (0.68–1.00); P = 0.05    
+936C>T T/T vs. C/C 1.72 (0.79–3.75); P = 0.18 0.35 1.38 (0.96–1.97) 0.08 
 C/T vs. C/C 1.13 (0.78–1.64); P = 0.51    
VEGF SNPsPooled HRs from meta-analysis, (95% CI); P value
Combined heterozygosity and homozygosity results
−2578C>A 0.92 (0.65–1.29); P = 0.62 
−1154G>A 1.07 (0.81–1.41); P = 0.63 
−460T>C 1.05 (0.78–1.43); P = 0.73 
+405G>C 0.74 (0.60–0.91); P = 0.004 
+936C>T 1.15 (0.86–1.53); P = 0.35 
Zygosity-segregated meta-analysis results 
VEGF SNPs Comparison Zygosity-specific HRs from meta-analysis, (95% CI); P value Subgroup differences P value Overall pooled HRs from meta-analysis (95% CI) Overall P value 
−2578C>A A/A vs. C/C 0.93 (0.55–1.59); P = 0.79 0.98 0.91 (0.72–1.14) 0.41 
 C/A vs. C/C 0.93 (0.79–1.08); P = 0.33    
−1154G>A A/A vs. G/G 0.93 (0.55–1.58); P = 0.80 0.45 0.98 (0.72–1.33) 0.71 
 G/A vs. G/G 1.18 (0.89–1.56); P = 0.26    
−460T>C C/C vs. T/T 1.10 (0.79–1.53); P = 0.56 0.77 1.06 (0.89–1.27) 0.52 
 T/C vs. T/T 1.04 (0.84–1.29); P = 0.72    
+405G>C C/C vs. G/G 0.78 (0.58–1.04); P = 0.09 0.72 0.79 (0.67–0.94) 0.007 
 G/C vs. G/G 0.83 (0.68–1.00); P = 0.05    
+936C>T T/T vs. C/C 1.72 (0.79–3.75); P = 0.18 0.35 1.38 (0.96–1.97) 0.08 
 C/T vs. C/C 1.13 (0.78–1.64); P = 0.51    

NOTE: Meta-analysis results for 5 commonly evaluated polymorphisms from our systematic review. Results from the pooled meta-analysis, in which studies were compared regardless of zygosity, are shown in the upper part of the table. Results from our zygosity-segregated analysis are shown in the lower part of the table. “Subgroup differences” measured whether there was a significant difference in the overall HRs between the 2 zygosity groups. The “Overall pooled HRs from meta-analysis” and “Overall P value” represent results when both homozygous and heterozygous results were pooled together. SNP, single-nucleotide polymorphism.

Table 4.

Forest plots for pooled and subgroup analysis of VEGF polymorphisms as prognostic biomarkers for VEGF +405G>C

Forest plots for pooled and subgroup analysis of VEGF polymorphisms as prognostic biomarkers for VEGF +405G>C
Forest plots for pooled and subgroup analysis of VEGF polymorphisms as prognostic biomarkers for VEGF +405G>C

Given that 2 of these 5 VEGF polymorphisms (VEGF −460C>T and VEGF −2578C>A) were in strong linkage disequilibrium with each other [(D′ > 0.90) on the basis of HapMap data, and with several other polymorphisms (rs833068 G>A, rs2146323 C>A, and VEGF −7C>T)], a meta-analysis was conducted on this haplotype block, pooling results from all studies involving any of these polymorphisms and assigning association direction on the basis of the most common haplotype (69). No significant association with survival outcome was found.

Antiangiogenic drugs are part of the clinical armamentarium for numerous solid tumors. Results of clinical trials of antiangiogenic agents have shown variable results (70). The need for predictive and prognostic markers for antiangiogenic therapy is, therefore, acute. There were too few studies focusing on the same polymorphism and outcome to assess consistency of results across studies. Of the 2 largest studies, Jain and colleagues pooled data from multiple cancer sites across several phase I and II trials of sorafenib and bevacizumab and found no significant results (17). In contrast, Schneider and colleagues evaluated 341 breast cancer patients treated with paclitaxel with or without bevacizumab and found 2 variants (VEGF −2578C>A and −1154G>A) associated with improved OS, whereas 2 different variants (VEGF +405G>C and −460C>T) were significantly associated with hypertension; the VEGF −1154G>A and −460C>T are, in general, highly linked in the Caucasian population (9). Both the A allele of VEGF −1154G>A, and the C allele of VEGF −460C>T, were associated with improved outcome in 2 separate pharmacogenetics studies (11, 12). However, these relationships await validation. In the future, comprehensive analyses of polymorphisms in either carefully designed observational studies or in larger phase III trials of bevacizumab and other VEGF-targeting agents in ovarian, colorectal, breast, and other cancers could be substantially more useful in delineating the role of these polymorphisms as pharmacogenetic markers.

We also reviewed VEGF pathway polymorphisms serving as general prognostic markers of outcome in patients not treated with VEGF-targeted agents. In this analysis, a large number of prognostic studies across many different cancers including ovarian, cervical, lung, breast, colorectal, gastric, esophageal/gastroesophageal junction, renal, lung, and liver cancers focused on 5 specific VEGF polymorphic variations. A meta-analysis was conducted on these polymorphisms, which identified the variant of VEGF +405G>C showing a highly significant prognostic role in improving the survival of cancer patients. As expected, given the varied cancer sites there was heterogeneity across individual studies (I2 = 72%). However, the use of a pooled analysis and random effects modeling has been particularly useful for identifying significant relationships in the face of heterogeneous data.

VEGF +405G>C is located in the 5′-untranslated region of the VEGF gene and is in modest linkage disequilibrium with VEGF +936C>T (D′ = 0.65–0.74); this may help explain the nonsignificant trend found with VEGF +936C>T (34, 71). Genotype–phenotype analyses suggest that the C allele VEGF +405G>C is associated with decreased promoter activity and decreased expression of VEGF, as the C allele reduces binding of the transcription factor, MZF1, which then subsequently reduces gene expression (72–74). Hence, the C allele VEGF +405G>C may confer a survival advantage by reducing a tumor's ability to stimulate angiogenesis.

The quality of a systematic review and meta-analysis is partly related to the quality of its underlying studies. The included studies have limitations. First, studies were extremely varied in sample size: some studies had as few as 28 patients and the largest involved more than 1,100 patients. Our meta-analyses included weighting on the basis of sample size. Second, most studies used hypothesis-driven candidate approaches, whereby only specific genes and polymorphisms from a selected list were analyzed. This is a common study design and can be most useful for meta-analyses, but suffers from an inability to identify novel biomarkers. Third, statistical significance of the VEGF +405G>C polymorphism for cancer prognosis required the pooling of heterogeneous studies across disease sites that had different underlying populations (i.e., race/ethnicity), study designs, sample sources (i.e., blood vs. tumor-based tissue), and statistical analyses (including whether there was adjustment for multiple comparisons, and using multivariate vs. univariate analyses). However, this heterogeneity has advantages in that the significance of this finding was robust to all these aforementioned issues. Finally, several of the measured phenotypes, such as therapeutic response and drug toxicity, were defined differently across studies, rendering it difficult for cross-comparison, interpretation, and evaluation in pooling or meta-analytic analysis. For example, some studies explored each toxicity separately while other studies analyzed multiple toxicities as a composite measure. Hence, no meta-analyses were conducted for the pharamacogenetic arm of the study.

In conclusion, there have been a large number of published studies exploring the association of germline polymorphisms of the VEGF pathway with cancer outcomes and treatment toxicities. These studies have substantial heterogeneity both in terms of methodology and in terms of effect size. When studies using similar methodology and involving similar outcome were assessed in meta-analyses, the variants of VEGF +405G>C were highly significantly associated with a 26% relative improvement in OS across a variety of tumor types, even after accounting for the number of comparisons conducted. Future prospective studies should extend this research to include pathway analysis, haplotype analysis, and discovery-focused genome-wide association studies, followed by appropriate validation or meta-analytic analyses.

No potential conflicts of interests were disclosed.

Conception and design: L. Eng, A.K. Azad, S. Habbous, A.H. Maitland-van der Zee, E. Amir, G. Liu

Development of methodology: L. Eng, A.K. Azad, E. Amir, G. Liu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Eng, A.K. Azad, H.J. Mackay, G. Liu

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Eng, A.K. Azad, S. Habbous, W. Xu, A.H. Maitland-van der Zee, S. Savas, H.J. Mackay, E. Amir, G. Liu

Writing, review, and/or revision of the manuscript: L. Eng, A.K. Azad, S. Habbous, V. Pang, A.H. Maitland-van der Zee, S. Savas, H.J. Mackay, E. Amir, G. Liu

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L. Eng, A.K. Azad, S. Savas, G. Liu

Study supervision: A.K. Azad, E. Amir, G. Liu

This work was sponsored in part by the Alan B Brown Chair in Molecular Genomics, CCO Chair in Experimental Therapeutics and Population studies, and Posluns Family Foundation.

1.
McDougall
SR
,
Anderson
AR
,
Chaplain
MA
. 
Mathematical modelling of dynamic adaptive tumour-induced angiogenesis: clinical implications and therapeutic targeting strategies
.
J Theor Biol
2006
;
241
:
564
89
.
2.
Ulahannan
SV
,
Brahmer
JR
. 
Antiangiogenic agents in combination with chemotherapy in patients with advanced non-small cell lung cancer
.
Cancer Invest
2011
;
29
:
325
37
.
3.
Jain
L
,
Vargo
CA
,
Danesi
R
,
Sissung
TM
,
Price
DK
,
Venzon
D
, et al
The role of vascular endothelial growth factor SNPs as predictive and prognostic markers for major solid tumors
.
Mol Cancer Ther
2009
;
8
:
2496
508
.
4.
McDonagh
EM
,
Whirl-Carrillo
M
,
Garten
Y
,
Altman
RB
,
Klein
TE
. 
From pharmacogenomic knowledge acquisition to clinical applications: the PharmGKB as a clinical pharmacogenomic biomarker resource
.
Biomark Med
2011
;
5
:
795
806
.
5.
dbSNP home page [homepage on the Internet]
.
Bethesda, MD
:
National Center for Biotechnology Information
; 
2012
.
[Updated 2012 Mar 19; cited June 05, 2012]. Available from
: http://www.ncbi.nlm.nih.gov/projects/SNP/index.html.
6.
Deeks
JJ
. 
Systematic reviews in health care: systematic reviews of evaluations of diagnostic and screening tests
.
BMJ
2001
;
323
:
157
62
.
7.
DerSimonian
R
,
Laird
N
. 
Meta-analysis in clinical trials
.
Control Clin Trials
1986
;
7
:
177
88
.
8.
Deeks
JJ
,
Higgins
JPT
,
Altman
DG
. 
Analysing data and undertaking meta-analyses
.
In
:
Higgins
JPT
,
Green
S
,
editors
.
Cochrane handbook for systematic reviews of interventions
.
Chichester, UK
:
John Wiley & Sons, Ltd
; 
2006
.
p.
243
93
.
9.
Schneider
BP
,
Wang
M
,
Radovich
M
,
Sledge
GW
,
Badve
S
,
Thor
A
, et al
Association of vascular endothelial growth factor and vascular endothelial growth factor receptor-2 genetic polymorphisms with outcome in a trial of paclitaxel compared with paclitaxel plus bevacizumab in advanced breast cancer: ECOG 2100
.
J Clin Oncol
2008
;
26
:
4672
8
.
10.
Etienne-Grimaldi
MC
,
Formento
P
,
Degeorges
A
,
Pierga
JY
,
Delva
R
,
Pivot
X
, et al
Prospective analysis of the impact of VEGF-A gene polymorphisms on the pharmacodynamics of bevacizumab-based therapy in metastatic breast cancer patients
.
Br J Clin Pharmacol
2011
;
71
:
921
8
.
11.
Loupakis
F
,
Ruzzo
A
,
Salvatore
L
,
Cremolini
C
,
Masi
G
,
Frumento
P
, et al
Retrospective exploratory analysis of VEGF polymorphisms in the prediction of benefit from first-line FOLFIRI plus bevacizumab in metastatic colorectal cancer
.
BMC Cancer
2011
;
11
:
247
.
12.
Formica
V
,
Palmirotta
R
,
Del Monte
G
,
Savonarola
A
,
Ludovici
G
,
De Marchis
ML
, et al
Predictive value of VEGF gene polymorphisms for metastatic colorectal cancer patients receiving first-line treatment including fluorouracil, irinotecan, and bevacizumab
.
Int J Colorectal Dis
2011
;
26
:
143
51
.
13.
Zhang
W
,
Azuma
M
,
Lurje
G
,
Gordon
MA
,
Yang
D
,
Pohl
A
, et al
Molecular predictors of combination targeted therapies (cetuximab, bevacizumab) in irinotecan-refractory colorectal cancer (BOND-2 study)
.
Anticancer Res
2010
;
30
:
4209
17
.
14.
Argiris
A
,
Karamouzis
MV
,
Gooding
WE
,
Branstetter
BF
,
Zhong
S
,
Raez
LE
, et al
Phase II trial of pemetrexed and bevacizumab in patients with recurrent or metastatic head and neck cancer
.
J Clin Oncol
2011
;
29
:
1140
5
.
15.
Schultheis
AM
,
Lurje
G
,
Rhodes
KE
,
Zhang
W
,
Yang
D
,
Garcia
AA
, et al
Polymorphisms and clinical outcome in recurrent ovarian cancer treated with cyclophosphamide and bevacizumab
.
Clin Cancer Res
2008
;
14
:
7554
63
.
16.
Smerdel
MP
,
Steffensen
KD
,
Waldstrom
M
,
Brandslund
I
,
Jakobsen
A
. 
The predictive value of serum VEGF in multiresistant ovarian cancer patients treated with bevacizumab
.
Gynecol Oncol
2010
;
118
:
167
71
.
17.
Jain
L
,
Sissung
TM
,
Danesi
R
,
Kohn
EC
,
Dahut
WL
,
Kummar
S
, et al
Hypertension and hand-foot skin reactions related to VEGFR2 genotype and improved clinical outcome following bevacizumab and sorafenib
.
J Exp Clin Cancer Res
2010
;
29
:
95
.
18.
Sakaeda
T
,
Yamamori
M
,
Kuwahara
A
,
Hiroe
S
,
Nakamura
T
,
Okumura
K
, et al
VEGF G-1154A is predictive of severe acute toxicities during chemoradiotherapy for esophageal squamous cell carcinoma in japanese patients
.
Ther Drug Monit
2008
;
30
:
497
503
.
19.
Steeghs
N
,
Mathijssen
RH
,
Wessels
JA
,
de Graan
AJ
,
van der Straaten
T
,
Mariani
M
, et al
Influence of pharmacogenetic variability on the pharmacokinetics and toxicity of the aurora kinase inhibitor danusertib
.
Invest New Drugs
2011
;
29
:
953
62
.
20.
Steeghs
N
,
Gelderblom
H
,
Wessels
J
,
Eskens
FA
,
de Bont
N
,
Nortier
JW
, et al
Pharmacogenetics of telatinib, a VEGFR-2 and VEGFR-3 tyrosine kinase inhibitor, used in patients with solid tumors
.
Invest New Drugs
2011
;
29
:
137
43
.
21.
Balasubramanian
SP
,
Cox
A
,
Cross
SS
,
Higham
SE
,
Brown
NJ
,
Reed
MW
. 
Influence of VEGF-A gene variation and protein levels in breast cancer susceptibility and severity
.
Int J Cancer
2007
;
121
:
1009
16
.
22.
Kidd
LR
,
Brock
GN
,
VanCleave
TT
,
Benford
ML
,
Lavender
NA
,
Kruer
TL
, et al
Angiogenesis-associated sequence variants relative to breast cancer recurrence and survival
.
Cancer Causes Control
2010
;
21
:
1545
57
.
23.
Knechtel
G
,
Hofmann
G
,
Gerger
A
,
Renner
W
,
Langsenlehner
T
,
Szkandera
J
, et al
Analysis of common germline polymorphisms as prognostic factors in patients with lymph node-positive breast cancer
.
J Cancer Res Clin Oncol
2010
;
136
:
1813
9
.
24.
Lu
H
,
Shu
XO
,
Cui
Y
,
Kataoka
N
,
Wen
W
,
Cai
Q
, et al
Association of genetic polymorphisms in the VEGF gene with breast cancer survival
.
Cancer Res
2005
;
65
:
5015
9
.
25.
Forsti
A
,
Jin
Q
,
Altieri
A
,
Johansson
R
,
Wagner
K
,
Enquist
K
, et al
Polymorphisms in the KDR and POSTN genes: association with breast cancer susceptibility and prognosis
.
Breast Cancer Res Treat
2007
;
101
:
83
93
.
26.
Balasubramanian
SP
,
Cross
SS
,
Globe
J
,
Cox
A
,
Brown
NJ
,
Reed
MW
. 
Endostatin gene variation and protein levels in breast cancer susceptibility and severity
.
BMC Cancer
2007
;
7
:
107
.
27.
Dassoulas
K
,
Gazouli
M
,
Rizos
S
,
Theodoropoulos
G
,
Christoni
Z
,
Nikiteas
N
, et al
Common polymorphisms in the vascular endothelial growth factor gene and colorectal cancer development, prognosis, and survival
.
Mol Carcinog
2009
;
48
:
563
9
.
28.
Vidaurreta
M
,
Sanchez-Munoz
R
,
Veganzones
S
,
Rafael
S
,
Gutierrez
M
,
de-la-Orden
V
, et al
Vascular endothelial growth factor gene polymorphisms in patients with colorectal cancer
.
Rev Esp Enferm Dig
2010
;
102
:
20
31
.
29.
Lurje
G
,
Zhang
W
,
Schultheis
AM
,
Yang
D
,
Groshen
S
,
Hendifar
AE
, et al
Polymorphisms in VEGF and IL-8 predict tumor recurrence in stage III colon cancer
.
Ann Oncol
2008
;
19
:
1734
41
.
30.
Hansen
TF
,
Garm Spindler
KL
,
Andersen
RF
,
Lindebjerg
J
,
Brandslund
I
,
Jakobsen
A
. 
The predictive value of genetic variations in the vascular endothelial growth factor A gene in metastatic colorectal cancer
.
Pharmacogenomics J
2011
;
11
:
53
60
.
31.
Zhang
W
,
Gordon
M
,
Press
OA
,
Rhodes
K
,
Vallbohmer
D
,
Yang
DY
, et al
Cyclin D1 and epidermal growth factor polymorphisms associated with survival in patients with advanced colorectal cancer treated with cetuximab
.
Pharmacogenet Genomics
2006
;
16
:
475
83
.
32.
Kim
JG
,
Chae
YS
,
Sohn
SK
,
Cho
YY
,
Moon
JH
,
Park
JY
, et al
Vascular endothelial growth factor gene polymorphisms associated with prognosis for patients with colorectal cancer
.
Clin Cancer Res
2008
;
14
:
62
6
.
33.
Hansen
TF
,
Sorensen
FB
,
Spindler
KL
,
Olsen
DA
,
Andersen
RF
,
Lindebjerg
J
, et al
Microvessel density and the association with single nucleotide polymorphisms of the vascular endothelial growth factor receptor 2 in patients with colorectal cancer
.
Virchows Arch
2010
;
456
:
251
60
.
34.
Bradbury
PA
,
Zhai
R
,
Ma
C
,
Xu
W
,
Hopkins
J
,
Kulke
MJ
, et al
Vascular endothelial growth factor polymorphisms and esophageal cancer prognosis
.
Clin Cancer Res
2009
;
15
:
4680
5
.
35.
Lurje
G
,
Leers
JM
,
Pohl
A
,
Oezcelik
A
,
Zhang
W
,
Ayazi
S
, et al
Genetic variations in angiogenesis pathway genes predict tumor recurrence in localized adenocarcinoma of the esophagus
.
Ann Surg
2010
;
251
:
857
64
.
36.
Lorenzen
S
,
Panzram
B
,
Keller
G
,
Lordick
F
,
Herrmann
K
,
Becker
K
, et al
Association of the VEGF 936C>T polymorphism with FDG uptake, clinical, histopathological, and metabolic response in patients with adenocarcinomas of the esophagogastric junction
.
Mol Imaging Biol
2011
;
13
:
178
86
.
37.
Stocker
G
,
Ott
K
,
Henningsen
N
,
Becker
K
,
Hapfelmeier
A
,
Lordick
F
, et al
CyclinD1 and interleukin-1 receptor antagonist polymorphisms are associated with prognosis in neoadjuvant-treated gastric carcinoma
.
Eur J Cancer
2009
;
45
:
3326
35
.
38.
Guan
X
,
Zhao
H
,
Niu
J
,
Tan
D
,
Ajani
JA
,
Wei
Q
. 
Polymorphisms of TGFB1 and VEGF genes and survival of patients with gastric cancer
.
J Exp Clin Cancer Res
2009
;
28
:
94
.
39.
Lurje
G
,
Husain
H
,
Power
DG
,
Yang
D
,
Groshen
S
,
Pohl
A
, et al
Genetic variations in angiogenesis pathway genes associated with clinical outcome in localized gastric adenocarcinoma
.
Ann Oncol
2010
;
21
:
78
86
.
40.
Tzanakis
N
,
Gazouli
M
,
Rallis
G
,
Giannopoulos
G
,
Papaconstantinou
I
,
Theodoropoulos
G
, et al
Vascular endothelial growth factor polymorphisms in gastric cancer development, prognosis, and survival
.
J Surg Oncol
2006
;
94
:
624
30
.
41.
Al-Moundhri
MS
,
Al-Nabhani
M
,
Burney
IA
,
Al-Farsi
AA
,
Al-Bahrani
B
. 
Gastric cancer risk predisposition and prognostic significance of vascular endothelial growth factor (VEGF) gene polymorphisms–a case-control study in an omani population
.
Mol Carcinog
2009
;
48
:
1170
6
.
42.
Kim
JG
,
Sohn
SK
,
Chae
YS
,
Cho
YY
,
Bae
HI
,
Yan
G
, et al
Vascular endothelial growth factor gene polymorphisms associated with prognosis for patients with gastric cancer
.
Ann Oncol
2007
;
18
:
1030
6
.
43.
Kawai
Y
,
Sakano
S
,
Korenaga
Y
,
Eguchi
S
,
Naito
K
. 
Associations of single nucleotide polymorphisms in the vascular endothelial growth factor gene with the characteristics and prognosis of renal cell carcinomas
.
Eur Urol
2007
;
52
:
1147
55
.
44.
Kim
EJ
,
Jeong
P
,
Quan
C
,
Kim
J
,
Bae
SC
,
Yoon
SJ
, et al
Genotypes of TNF-alpha, VEGF, hOGG1, GSTM1, and GSTT1: useful determinants for clinical outcome of bladder cancer
.
Urology
2005
;
65
:
70
5
.
45.
Mucci
LA
,
Stark
JR
,
Figg
WD
,
Schumacher
F
,
Li
H
,
Abe
M
, et al
Polymorphism in endostatin, an angiogenesis inhibitor, and prostate cancer risk and survival: a prospective study
.
Int J Cancer
2009
;
125
:
1143
6
.
46.
Li
HC
,
Cai
QY
,
Shinohara
ET
,
Cai
H
,
Cao
C
,
Wang
ZF
, et al
Endostatin polymorphism 4349G/A(D104N) is not associated with aggressiveness of disease in prostate [corrected] cancer
.
Dis Markers
2005
;
21
:
37
41
.
47.
Hefler
LA
,
Mustea
A
,
Konsgen
D
,
Concin
N
,
Tanner
B
,
Strick
R
, et al
Vascular endothelial growth factor gene polymorphisms are associated with prognosis in ovarian cancer
.
Clin Cancer Res
2007
;
13
:
898
901
.
48.
Polterauer
S
,
Grimm
C
,
Mustea
A
,
Concin
N
,
Tanner
B
,
Thiel
F
, et al
Vascular endothelial growth factor gene polymorphisms in ovarian cancer
.
Gynecol Oncol
2007
;
105
:
385
9
.
49.
Lose
F
,
Nagle
CM
,
O'Mara
T
,
Batra
J
,
Bolton
KL
,
Song
H
, et al
Vascular endothelial growth factor gene polymorphisms and ovarian cancer survival
.
Gynecol Oncol
2010
;
119
:
479
83
.
50.
Smerdel
MP
,
Waldstrom
M
,
Brandslund
I
,
Steffensen
KD
,
Andersen
RF
,
Jakobsen
A
. 
Prognostic importance of vascular endothelial growth factor-A expression and vascular endothelial growth factor polymorphisms in epithelial ovarian cancer
.
Int J Gynecol Cancer
2009
;
19
:
578
84
.
51.
Steffensen
KD
,
Waldstrom
M
,
Brandslund
I
,
Jakobsen
A
. 
The relationship of VEGF polymorphisms with serum VEGF levels and progression-free survival in patients with epithelial ovarian cancer
.
Gynecol Oncol
2010
;
117
:
109
16
.
52.
Goode
EL
,
Maurer
MJ
,
Sellers
TA
,
Phelan
CM
,
Kalli
KR
,
Fridley
BL
, et al
Inherited determinants of ovarian cancer survival
.
Clin Cancer Res
2010
;
16
:
995
1007
.
53.
Amano
M
,
Yoshida
S
,
Kennedy
S
,
Takemura
N
,
Deguchi
M
,
Ohara
N
, et al
Association study of vascular endothelial growth factor gene polymorphisms in endometrial carcinomas in a japanese population
.
Eur J Gynaecol Oncol
2008
;
29
:
333
7
.
54.
Kim
YH
,
Kim
MA
,
Park
IA
,
Park
WY
,
Kim
JW
,
Kim
SC
, et al
VEGF polymorphisms in early cervical cancer susceptibility, angiogenesis, and survival
.
Gynecol Oncol
2010
;
119
:
232
6
.
55.
Heist
RS
,
Zhai
R
,
Liu
G
,
Zhou
W
,
Lin
X
,
Su
L
, et al
VEGF polymorphisms and survival in early-stage non-small-cell lung cancer
.
J Clin Oncol
2008
;
26
:
856
62
.
56.
Guan
X
,
Yin
M
,
Wei
Q
,
Zhao
H
,
Liu
Z
,
Wang
LE
, et al
Genotypes and haplotypes of the VEGF gene and survival in locally advanced non-small cell lung cancer patients treated with chemoradiotherapy
.
BMC Cancer
2010
;
10
:
431
.
57.
Dong
J
,
Dai
J
,
Shu
Y
,
Pan
S
,
Xu
L
,
Chen
W
, et al
Polymorphisms in EGFR and VEGF contribute to non-small-cell lung cancer survival in a chinese population
.
Carcinogenesis
2010
;
31
:
1080
6
.
58.
Masago
K
,
Fujita
S
,
Kim
YH
,
Hatachi
Y
,
Fukuhara
A
,
Nagai
H
, et al
Effect of vascular endothelial growth factor polymorphisms on survival in advanced-stage non-small-cell lung cancer
.
Cancer Sci
2009
;
100
:
1917
22
.
59.
Kong
SY
,
Park
JW
,
Lee
JA
,
Park
JE
,
Park
KW
,
Hong
EK
, et al
Association between vascular endothelial growth factor gene polymorphisms and survival in hepatocellular carcinoma patients
.
Hepatology
2007
;
46
:
446
55
.
60.
Wu
LM
,
Xie
HY
,
Zhou
L
,
Yang
Z
,
Zhang
F
,
Zheng
SS
. 
A single nucleotide polymorphism in the vascular endothelial growth factor gene is associated with recurrence of hepatocellular carcinoma after transplantation
.
Arch Med Res
2009
;
40
:
565
70
.
61.
Streit
S
,
Mestel
DS
,
Schmidt
M
,
Ullrich
A
,
Berking
C
. 
FGFR4 Arg388 allele correlates with tumour thickness and FGFR4 protein expression with survival of melanoma patients
.
Br J Cancer
2006
;
94
:
1879
86
.
62.
Monzo
M
,
Brunet
S
,
Urbano-Ispizua
A
,
Navarro
A
,
Perea
G
,
Esteve
J
, et al
Genomic polymorphisms provide prognostic information in intermediate-risk acute myeloblastic leukemia
.
Blood
2006
;
107
:
4871
9
.
63.
Kim
DH
,
Lee
NY
,
Lee
MH
,
Sohn
SK
,
Do
YR
,
Park
JY
. 
Vascular endothelial growth factor (VEGF) gene (VEGFA) polymorphism can predict the prognosis in acute myeloid leukaemia patients
.
Br J Haematol
2008
;
140
:
71
9
.
64.
Kim
DH
,
Xu
W
,
Kamel-Reid
S
,
Liu
X
,
Jung
CW
,
Kim
S
, et al
Clinical relevance of vascular endothelial growth factor (VEGFA) and VEGF receptor (VEGFR2) gene polymorphism on the treatment outcome following imatinib therapy
.
Ann Oncol
2010
;
21
:
1179
88
.
65.
Kim
DH
,
Kong
JH
,
Byeun
JY
,
Jung
CW
,
Xu
W
,
Liu
X
, et al
The IFNG (IFN-gamma) genotype predicts cytogenetic and molecular response to imatinib therapy in chronic myeloid leukemia
.
Clin Cancer Res
2010
;
16
:
5339
50
.
66.
Diao
LP
,
Yu
XM
,
Gao
YH
,
Li
Y
,
Liu
HS
,
Liu
LH
, et al
Association of VEGF genetic polymorphisms with the clinical characteristics of non-hodgkin's lymphoma
.
J Cancer Res Clin Oncol
2009
;
135
:
1473
81
.
67.
Ruiz
MT
,
Biselli
PM
,
Maniglia
JV
,
Pavarino-Bertelli
EC
,
Goloni-Bertollo
EM
. 
Genetic variability of vascular endothelial growth factor and prognosis of head and neck cancer in a brazilian population
.
Braz J Med Biol Res
2010
;
43
:
127
33
.
68.
Formento
JL
,
Etienne-Grimaldi
MC
,
Francoual
M
,
Pages
G
,
Onesto
C
,
Formento
P
, et al
Influence of the VEGF-A 936C>T germinal polymorphism on tumoral VEGF expression in head and neck cancer
.
Pharmacogenomics
2009
;
10
:
1277
83
.
69.
International HapMap Consortium
. 
A haplotype map of the human genome
.
Nature
2005
;
437
:
1299
320
.
70.
Ocana
A
,
Amir
E
,
Vera
F
,
Eisenhauer
EA
,
Tannock
IF
. 
Addition of bevacizumab to chemotherapy for treatment of solid tumors: similar results but different conclusions
.
J Clin Oncol
2011
;
29
:
254
6
.
71.
Zhai
R
,
Liu
G
,
Zhou
W
,
Su
L
,
Heist
RS
,
Lynch
TJ
, et al
Vascular endothelial growth factor genotypes, haplotypes, gender, and the risk of non-small cell lung cancer
.
Clin Cancer Res
2008
;
14
:
612
7
.
72.
Hussein
A
,
Askar
E
,
Elsaeid
M
,
Schaefer
F
. 
Functional polymorphisms in transforming growth factor-beta-1 (TGFbeta-1) and vascular endothelial growth factor (VEGF) genes modify risk of renal parenchymal scarring following childhood urinary tract infection
.
Nephrol Dial Transplant
2010
;
25
:
779
85
.
73.
Young
HS
,
Summers
AM
,
Bhushan
M
,
Brenchley
PE
,
Griffiths
CE
. 
Single-nucleotide polymorphisms of vascular endothelial growth factor in psoriasis of early onset
.
J Invest Dermatol
2004
;
122
:
209
15
.
74.
Watson
CJ
,
Webb
NJ
,
Bottomley
MJ
,
Brenchley
PE
. 
Identification of polymorphisms within the vascular endothelial growth factor (VEGF) gene: correlation with variation in VEGF protein production
.
Cytokine
2000
;
12
:
1232
5
.