Full Text Article

Relationship between Genetic Variants of Glucuronidation Pathway and TNF-Α with Increased Risk of Prostate Cancer in Iranian Men

Received Date: February 12, 2022 Accepted Date: March 12, 2022 Published Date: March 14, 2022

doi: 10.17303/jbcg.2022.5.101

Citation: Saeideh Alidoost (2022) Relationship between Genetic Variants of Glucuronidation Pathway and TNF-Α with Increased Risk of Prostate Cancer in Iranian Men. J Bioinfo Comp Genom 5: 1-17

After lung cancer, prostate cancer has the highest mortality rate. Early and accurate diagnosis of this heterogeneous cancer promises more effective treatment. At present biopsy is the only definitive method of diagnosing the disease. Many gene loci are associated with increased susceptibility to this cancer. Here, the relationship between tumor necrosis factor alpha (TNF-α) gene variants and glucuronidation pathway gene variants with increased risk of prostate cancer in a population of Iranian men has been investigated.

Materials and methods: Blood samples were collected from 360 men including 120 healthy, 120 with benign prostate hyperplasia (BPH), and 120 patients with prostate cancer (PCa). DNA was extracted and tested for each variant with specific primers and PCR based methods. Data were analyzed using agarose and polyacrylamide gel electrophoresis, SPSS software, SNP Stats and Student’s t-test.

Results: UGT2B17 and UGT2B15 polymorphisms were associated with BPH in comparison to the control group (P value = <0.0001 and P value = 0.007). The only significant association in the cancer group was between G Score and UGT2B15, so that 90% of patients with PCa and G score less than or equal to 6, had GG genotype (0.01). Other variants had no significant relationship with the cause of the disease. Ins Del G haplotype was more common in BPH compared to the control group (P value = 0.011).

Discussion: Observation of the association of UGT2B17 and UGT2B15 with BPH, and the prevalence of Ins Del G haplotype in BPH compared with healthy individuals, increases the likelihood of these genotypes for prostate hyperplasia. Further studies on the genetic composition of pathways associated with prostate function in larger populations is promising to find possible new biomarkers, map the natural state and progression of cellular changes in favor of prostate cancer.

Keywords: Prostate cancer; Prostate hyperplasia; CNV; polymorphism; SNP

Prostate cancer is the second most common cancer in Iran, after gastric cancer [1]. In the early stages of PCa, symptoms are rarely appearing. There may be no symptoms at the time of diagnosis even in the advanced states [2]. Blood in the urine and semen [3], recurrent pain in the affected area[4], difficulty in urinating[5], sudden or frequent urination [6], unexplained weight loss [7], pain in the pelvis [8], thighs and back [9], pain or abnormal symptoms in the penis [10], poor urination, and bone pain [11] are signs of cancer progression.

While the underlying causes of prostate cancer remain unknown, the risk of developing prostate cancer is increasing [12]. Having one or more risk factors does not mean that one will definitely develop the disease. The risk of PCa in men under the age of 40 is very low [13]. But, the chance of the disease appearance increases rapidly in people over 50 years old [13], while about 6 out of 10 cases of the patients are found in over 65 years old men [14]. Blacks are more likely to develop PCa than men of other races and are more than twice as likely to die from the cancer as white men [15].

Smoking is one of the most effective risk factors for the disease [16]. This cancer seems to be more common in some families, suggesting that in some cases there may be hereditary or genetic factors [16]. A person with a first degree relative with prostate cancer is two to three times as likely to get the disease as other men [17]. People with a strong family history of breast and ovarian cancer are also more likely to develop prostate cancer than people without a family history [18] . A number of genetic changes increase the risk of prostate cancer, but in general they are likely to account for only a small percentage of cases [19]. Screening tests help in diagnosis and treatment the cancer, early or even before symptoms appear [20]. PCa screening is performed based on PSA level measurement, MRI/ ultrasound and/or CT scan, and finally the results of pathology, in Iran. Determining the level of PSA in a blood test is not a specific test for cancer [21]. Even if the serum level of prostate-specific antigen rises above normal, the risk of prostate cancer is not very high are life-threatening or never cause symptoms, but if they are detected by screening tests, the person may be treated for cancer. Complications of prostate biopsy such as false negative results delay treatment [25] and false positive cases undergo more biopsies[26]. The side effects of biopsy are many and also cause anxiety and worry [27]. Abnormal prostate changes do not always indicate prostate cancer. In some cases, an MRI before a biopsy is recommended [28]. Currently the only reliable way to diagnose this disease is a biopsy that shows a person has prostate cancer. Other measures are taken to find out if the cancerous mass has spread from its original location to other parts of the body [29]. CT scan shows the presence of cancer cells in the bone [5]. Post-diagnosis PET scan is used to identify the stage and degree of disease progression and to diagnose relapse after treatment [30]. For some men, immediate treatment is not necessary or may not be appropriate. The therapist can allow the patient to make treatment based on the stage of the cancer. Surgery, radiotherapy, chemotherapy and palliative care are other current treatments [30].

Given the heterogeneous nature of prostate tumors, researchers are trying to find more sensitive and specific tumor markers in the blood to replace solid biopsy with liquid biopsy and to create a chance to differentiate between different stages and grades of disease development by providing specific patterns of genetic changes and gene expression [31]. So far, more than 100 gene loci associated with the disease have been identified [32]. Numerous reports have shown an increased risk of prostate cancer in connection with certain single or combined genetic polymorphisms. The aim of this study was to investigate the relationship between rs1800629 and rs361525 TNF-α gene polymorphisms and gene variants of UGT2B28, UGT2B17 and UGT2B15 genes from glucuronidation pathway with increased risk of prostate cancer in Iranian population.

Sampling

In this case-control study, 120 men with healthy prostate (control), 120 patients with BPH, and 120 patients with prostate cancer (PCa) were selected from the individuals referred to the urology department of Shohadaye Tajrish Hospital in Tehran between 2019 and 2020. Demographic information was recorded using a conscious questionnaire. The PCa group included 50-year-old men with or without a family history of the disease, high PSA, urinary symptoms, digital rectal examination, CT scan, ultrasound, biopsy with a definitive diagnosis of prostate cancer by a urologist, based on pathological tests and the presence of neoplastic tissue at any stage or degree. Also, body mass index, height and weight, smoking / hookah use or any other drug, place of residence, use of other drugs, PSA level were recorded. The BPH group included men with disturbing urinary symptoms, PSA levels above 4, benign prostate swelling as shown by TR biopsy (rectal examination), pathologically negative results, and a detected prostate volume of more than 30 ml by radiology. The control group included healthy individuals who showed urinary symptoms, abdominal pain, and age 31-41 years. In addition to PSA measurements, ultrasound and urine culture data, their clinical and paraclinical examinations confirmed prostate health. They had no previous surgery, chemotherapy, or radiation therapy for prostate cancer, had no family history of BPH / PCa, and was considered healthy.

Informed consent was received from each patient

DNA extraction from peripheral blood

In this study, 5 ml of peripheral blood of each patient was collected in a tube containing EDTA. Blood samples were taken from the control subjects in the same way and then DNA extraction was performed (DiatomTM DNA prep kit and in some samples using salting out method). The PCR conditions were based on previously works [33, 34]. The forward and reverse primers for examining the studied copy number variations and the gene polymorphisms are shown in Table 1.

Enzyme digestion and gel electrophoresis

After the amplification of rs361525 promoter snp and UGT2B15 D85Y polymorphism, the first variation PCR products were digested by MspI and the second polymorphism products were cut by Sau3AI restriction enzymes (Fermentas, Hanover, MD, USA) and separated by polyacrylamide gel electrophoresis (PAGE, 12%).

Amplification of rs1800629

Tetra-ARMS-PCR technique was used to identify rs1800629 polymorphism. Based on this method, a pair of non-specific primers (Forward outer and Reverse outer) that amplify the entire sequence containing this variant and a pair of specific primers (Forward inner and Reverse inner) that identify and amplify a specific sequence of the interest region were used.

PCR based experiments results were the same as our previous work [33, 34]. The expected fragment of PCR for UGT2B15 D85Y polymorphism was a 215 bp band, which after restriction digest, could produce two bands of 28 and 187 bp (figure1). The PCR products with YY genotypes showed unique fragments of 215 bp, those with DD genotypes showed two different fragments with lengths of 187 and 28 bp, and DY genotypes demonstrated three bands of 215, 187, and 28 bp on gel electrophoresis (Figure 1).

The pattern of UGT2B17 PCR fragments on 2.5% agarose gel is shown in Figure 2. Checking for the presence or absence of the gene required two pairs of primers, one pair for exon1 amplification which could create a 173 bp band to confirm the definite presence of the allele and a second pair of primers which could amplify a region of 893 base pairs when the gene was deleted but could produce two fragments of 173 and 893 base pairs in heterozygotes of the deleted allele.

The pattern of UGT2B28 PCR fragments on 2.5% agarose gel is shown in Figure 3. Checking for the presence or absence of the gene required two pairs of primers, one pair for exon1 amplification which could create a 324 bp band to confirm the definite presence of the allele and a second pair of primers which could amplify a region of 450 base pairs in homozygutes of the deletion, but could produce two fragments of 324 and 450 base pairs in heterozygote samples.

The amplification of TNF-α promoter rs361525 polymorphism produced a fragment of 152 bp on 2% agarose gel, while after PCR-RFLP and digestion by MspI restriction enzyme was digested into a 152 bp, a 132bp, and a 20 bp fragments in GA heterozygotes and into two fragments of 132bp and 20 bp in GG homozygotes (Figures 4 and 5).

For rs1800629 PCR products were electrophoresed directly and without enzymatic digestion on 2% agarose gel. Individuals with the G / G genotype had two fragments of 304 and 197 bp, G/A heterozygotes showed to have three-bands of 304, 197, and 162 bp, and two fragments of 304 and 162 bp. were resulted from A / A homozygotes (Figure 6).

Exact test showed a Hardy-Weinberg Equilibrium in the studied alleles of the target population (p value > 0.05) (Table 2). The examined groups were compared in terms of genotypes to determine a possible allelic relationship in the genes (Table 3). There was a codominant, dominant, and/ or recessive, or Log-Additive model of allelic relationship between the variants of UGT2B17 and BPH (p value<0.05). UGT2B17 showed to be associated with BPH in comparison with the control group (P value = <0.0001) (Del versus INS: OR (95% CI) = 2.08 (1.41-3.08). Also, an allelic codominant, dominant, recessive, or Log-Additive model was anticipated for UGT2B15 and BPH (p value < 0.05).

UGT2B15 was associated with BPH in comparison to the control group (P value = 0.007) (G versus T: OR (95% CI) = 1.64 (1.14-2.37). For other examined variants, there was no significant relationship between allelic models and disease etiology (Table 3).

Since, the understudy genes UGT2B28, UGT2B17 and UGT2B15 were located on 4q13.2, different combinations of their alleles were considered as haplotypes. There was a significant relationship between G score and UGT2B15 in PCa group, so that 90% of patients with GG genotype were found among those with G score ≤ 6 group (Table4).

The frequency of each haplotype was compared using SNP Stats in three groups. Genotypes of three UGT2B28- UGT2B17- UGT2B15 genes in BPH group as Ins-Del-G allelic combination was significantly higher than the frequency of this allelic combination in control subjects (p value= 0.011; OR (95%CI) (Table 5). A chi-squared test was used to derive p-Values.

Prostate cancer is the second most common cancer of men in the world including Iran [35-37]. Numerous studies have examined the effect of functional polymorphisms of UGT2B28, UGT2B17 and UGT2B15 enzymes on androgen metabolism [38], including single-nucleotide substitutions (SNPs and variable number of copies (CNV) [33-40]. The copy number variations (CNVs) of UGT2B17 and UGT2B28 genes are an important source of variation in their expression which affects the accumulation of dihydrotestosterone [41]. Genetic studies in recent years have shown that in cancer patients where both alleles of the UGT2B17 gene have been deleted, the amount of 3-alpha-diol glucuronidate in circulation is decreased by 42% [42]. However, Habibi and colleagues did not find any association between the null genotype (del/del) of UGT2B17 and UGT2B28 genes with prostate cancer risk in Iranian PCa patients [33]. The present study showed that UGT2B17 Del allele possibly is associated to the disease development and increase risk of BPH (P value = <0.0001). As we considered, this probable association could be in either mode of codominance, dominance, recessiveness, and/or in log-Additive models which could depend on locus neighboring genes in the form of haplotype.

In vitro studies have identified two enzymes, UGT2B15 and UGT2B17, as the major enzymes involved in glucuronidation of androgens [43]. UGT2B15 protein level decreases in prostate cancer compared to benign hyperplasia and the protein is further reduced in hormone resistant form of prostate cancer and is as low as to be measured in metastasis state [44]. D85Y polymorphism (rs1902023) in UGT2B15 gene has been shown to be significantly correlated with prostate cancer in some studies ]45 ,33[and is a malignant polymorphism at gene codon 85 [33] where thymine replaces guanine base and causes the amino acid aspartic acid to be converted to tyrosine. The occurrence of this substitution in the N-terminal region of the enzyme, affects the protein activity in the second binding domain to the substrate [45]. UGT2B15 D85Y polymorphism does not appear to alter the specificity of substrates, but this polymorphism causes the maximum rate (Vmax) of glucuronidation to almost double and may play a role in individual differences in glucuronidation [46].

Some studies have reported that the TT genotype of UGT2B15 D85Y is twice as active as the GG homozygotes, so it glucuronates the hormone dihydrotestosterone more rapidly, further protecting the prostate against high levels of androgens, thereby reducing the risk of the disease [47]. Lower activity of the enzyme variant increases the risk of cancer due to the accumulation of dihydrotestosterone in the prostate [48, 49]. Hajdinjak and colleagues showed that the frequency of homozygous G is high in patients with prostate cancer and the frequency of homozygous T is high in controls [45]. A 2013 study by Grant et al. [48], confirmed the association of D85Y polymorphism with prostate cancer, and in this study, as in previous studies by MacLeod et al. 2000 [50]; Hajdinjak et al. In 2004, a homozygous form of G was shown to be associated with an increased risk of prostate cancer [45]. However, some studies, such as a 2002 study by Gsur et al., found no association between this polymorphism and an increased risk of prostate cancer [51].The results of current study showed that UGT2B15 is associated with BPH in either models of Codominance, Recessive, and Log-Additive (P value = 0.007) and its D85Y, rs1902023 GG genotype is most frequent in individuals with developing PCa (P value = 0.011). This is consistent with the previous reports we pointed here.

Also, the haplotype Ins Del G of the target genes was more common in BPH compared to the control group, one could anticipate that this achievement is an indication of the effect of the association of these three alleles on the etiology of the disease which in turn introduces this genotype as a probably important marker for predisposition a person to prostate hyperplasia.

Recent reports suggest that genetic polymorphisms of cytokines, including tumor necrosis factor-alpha (TNF-α), a proinflammatory molecule, are associated with increased inflammation, cytokine production, and possibly an increased risk of prostate cancer .]52[ It is shown that promoter polymorphisms in the TNF-α gene can directly affect TNF-α production, thus causing interpersonal differences in the immune response that may affect susceptibility to prostate cancer ]53[, and/or malignant tumors like gastric cancer, breast cancer, and hepatocellular carcinoma .]54-56[ We revealed that rs361525 TNF-α polymorphism is not associated with prostate cancer, but rs1800629 may increase the risk of PCa. Also, at least in this experimental study, there was no individual with AA genotype which shows that either for this particular polymorphism, the population of Iran is not in Hardy-Weinberg Equation or A allele has a low frequency in Iranian population, and probably has low influence on the incidence of the disease.

According to the results of this study and similar studies, finding functional polymorphisms as probable new biomarkers in the pathogenesis of cancers including prostate cancer encourages us to conduct more extensive research in this area with a larger population. A similar study with a larger population could confirm the results of this work.

The authors thank the medical staff who assisted in preparing the samples. We also thank the patients and people who willingly participated in this study by donating their blood samples at the same time as the pain or problems in life.

This project was based on grants numbers 7698 and 15139 that were supported by Shahid Beheshti University of Medical Sciences Research Council.

Authors have no conflict of interests.

  1. Farhood B, G Geraily, A Alizadeh (2018) Incidence and Mortality of Various Cancers in Iran and Compare to Other Countries: A Review Article. Iran J Public Health 47: 309-16.
  2. Beikzadeh B, SA Angaji, M Abolhasani (2020) Association study between common variations in some candidate genes and prostate adenocarcinoma predisposition through multi-stage approach in Iranian population. BMC medical genetics 21: 1-10.
  3. Mou RY, XJ Li (2017) Laboratory techniques for the diagnosis of prostate cancer: An update. Zhonghua Nan Ke Xue 23: 372-75.
  4. Fürst P, S Lundström, P Strang (2020) Methadone in Swedish specialized palliative care-Is it the magic bullet in complex cancer-related pain? PLoS One 15: e0230845.
  5. Pianou NK (2019) More advantages in detecting bone and soft tissue metastases from prostate cancer using (18)F-PSMA PET/CT. Hell J Nucl Med 22: 6-9.
  6. Narayan VM (2020) A critical appraisal of biomarkers in prostate cancer. World J urology 38: 547-54.
  7. Wilson RL (2021) Weight loss for obese prostate cancer patients on androgen deprivation therapy. Medicine and science in sports and exercise 53: 470.
  8. Liu Y (2020) Chronic prostatitis/chronic pelvic pain syndrome and prostate cancer: study of immune cells and cytokines. Fundamental & clinical pharmacology 34: 160-72.
  9. Magee K, DJ Hata, D Meza (2015) Trichosporon asahii Infection in a Patient with Metastatic Prostate Cancer as an Example of an Emerging Fungal Pathogen. Lab Med 46: e74-8.
  10. Brawley S, R Mohan, CD Nein (2018) Localized Prostate Cancer: Treatment Options. Am Fam Physician 97: 798-805.
  11. Jimenez-Andrade, JM (2010) Bone cancer pain. Ann N Y Acad Sci 1198: 173-81.
  12. Junejo NN, SS AlKhateeb (2020) BRCA2 gene mutation and prostate cancer risk. Comprehensive review and update. Saudi Med J 41: 9-17.
  13. Lange SM (2021) Prostate-specific antigen testing among young men: an opportunity to improve value. Cancer Med 10: 2075-9.
  14. Rawla P (2019) Epidemiology of Prostate Cancer. World J Oncol 10: 63-89.
  15. Panigrahi GK (2019) Exosome proteomic analyses identify inflammatory phenotype and novel biomarkers in African American prostate cancer patients. Cancer Med 8: 1110-23.
  16. Pernar CH (2018) The Epidemiology of Prostate Cancer. Cold Spring Harb Perspect Med 8.
  17. Giri VN, JL Beebe-Dimmer (2016) Familial prostate cancer. Semin Oncol 43: 560-65.
  18. Zheng Q (2021) First-degree family history of prostate cancer is associated the risk of breast cancer and ovarian cancer. Medicine (Baltimore) 100: e23816.
  19. Ramanand SG (2020) The landscape of RNA polymerase II-associated chromatin interactions in prostate cancer. J Clin Invest 130: 3987-4005.
  20. Ong XRS (2020) Understanding the diagnosis of prostate cancer. Med J Aust 213: 424-29.
  21. Albertsen PC (2020) Prostate cancer screening and treatment: where have we come from and where are we going? BJU Int 126: 218-24.
  22. Merriel SWD, G Funston, W Hamilton (2018) Prostate Cancer in Primary Care. Adv Ther 35: 1285-94.
  23. Grozescu T, F Popa (2017) Prostate cancer between prognosis and adequate/proper therapy. J Med Life 10: 5-12.
  24. Mangold KN (2021) Feasibility of collecting patient-generated health data to enhance cancer registry surveillance. J Cancer Surviv.
  25. Breeuwsma AJ (2012) Correlation of [11C]choline PET-CT with time to treatment and disease-specific survival in men with recurrent prostate cancer after radical prostatectomy. Q J Nucl Med Mol Imaging 56: 440-6.
  26. Sehn JK (2018) Prostate Cancer Pathology: Recent Updates and Controversies. Mo Med 115: 151-5.
  27. Erim DO (2020) Associations between prostate cancer-related anxiety and health-related quality of life. Cancer Med, 2020. 9: 4467-73.
  28. Barrett T (2019) PI-RADS version 2.1: one small step for prostate MRI. Clin Radiol 74: 841-52.
  29. Huang Y (2020) Chemokine releasing particle implants for trapping circulating prostate cancer cells. Sci Rep 10: 4433.
  30. Evangelista L (2021) PET/MRI in prostate cancer: a systematic review and meta-analysis. Eur J Nucl Med Mol Imaging 48: 859-73.
  31. Puche-Sanz I (2020) Liquid biopsy and prostate cancer. Current evidence applied to clinical practice. Actas Urol Esp (Engl Ed) 44: 139-47.
  32. Qian D (2019) Potentially functional genetic variants in the complement-related immunity gene-set are associated with non-small cell lung cancer survival. Int J Cancer 144: 1867-76.
  33. Habibi M (2017) Genetic variations in UGT2B28, UGT2B17, UGT2B15 genes and the risk of prostate cancer: A case-control study. Gene 634: 47-52.
  34. Alidoost S (2020) Association between tumor necrosis factor-alpha gene rs1800629 (-308G/A) and rs361525 (-238G > A) polymorphisms and prostate cancer risk in an Iranian cohort. Hum Antibodies 28: 65-74.
  35. Nalairndran G (2020) Phosphoinositide-dependent Kinase-1 (PDPK1) regulates serum/glucocorticoid-regulated Kinase 3 (SGK3) for prostate cancer cell survival. J Cell Mol Med 24: 12188-98.
  36. Kacar S (2020) The effects of L-NAME on DU145 human prostate cancer cell line: A cytotoxicity-based study. Hum Exp Toxicol 39: 182-93.
  37. Rezapour A (2021) The economic burden of cancer in iran during 1995-2019: A systematic review. Iranian Journal of Public Health 50: 35-45.
  38. Gauthier-Landry L, A Bélanger, O Barbier (2015) Multiple roles for UDP-glucuronosyltransferase (UGT)2B15 and UGT2B17 enzymes in androgen metabolism and prostate cancer evolution. J Steroid Biochem Mol Biol 2015. 145: 187-92.
  39. Giroux S (2012) UGT2B17 gene deletion associated with an increase in bone mineral density similar to the effect of hormone replacement in postmenopausal women. Osteoporos Int 23: 1163-70.
  40. Nadeau G (2011) Deletions of the androgen-metabolizing UGT2B genes have an effect on circulating steroid levels and biochemical recurrence after radical prostatectomy in localized prostate cancer. J Clin Endocrinol Metab, 2011. 96: E1550-7.
  41. Novillo A (2018) UDP-glucuronosyltransferase genetic variation in North African populations: a comparison with African and European data. Ann Hum Biol 45: 516-23.
  42. Kpoghomou MA (2013) UGT2B17 Polymorphism and Risk of Prostate Cancer: A Meta-Analysis. ISRN Oncol 2013: 465916.
  43. Salhab H, DP Naughton, J Barker, Potential assessment of UGT2B17 inhibition by salicylic acid in human supersomes in vitro. Molecules 26: 4410.
  44. Barnard M (2020) The role of adrenal derived androgens in castration resistant prostate cancer. J Steroid Biochem Mol Biol 197: 105506.
  45. Hajdinjak T, B Zagradišnik (2004) Prostate cancer and polymorphism D85Y in gene for dihydrotestosterone degrading enzyme UGT2B15: Frequency of DD homozygotes increases with Gleason Score. The Prostate, 2004. 59: 436-39.
  46. Troberg J (2019) Glucuronidation activity of individual UDP-glucuronosyltransferases: Comparison of activity and substrate specificity among recombinant human UGT enzymes and differences between dog and human subfamily 1A UGT enzymes. Dissertationes Scholae Doctoralis Ad Sanitatem Investigandam Universitatis Helsinkiensis, 2019.
  47. Thind J (2013) Modulation of UDP Glucuronosyltransferase 2B15 and 2B17 and prostate cancer risk: current perspectives. Advances in Cancer: Research & Treatment, 2013.
  48. Grant DJ (2013) Association of uridine diphosphate-glucuronosyltransferase 2B gene variants with serum glucuronide levels and prostate cancer risk. Genet Test Mol Biomarkers 17: 3-9.
  49. Zhong X (2017) Uridine diphosphate-glucuronosyltransferase 2B15 D85Y gene polymorphism is associated with lower prostate cancer risk: a systematic review and meta-analysis. Oncotarget 8: 52837-45.
  50. MacLeod SL (2000) An allele-specific polymerase chain reaction method for the determination of the D85Y polymorphism in the human UDPGlucuronosyltransferase 2B15 gene in a case-control study of prostate cancer. 2000, Springer.
  51. Gsur A (2002) A polymorphism in the UDP-Glucuronosyltransferase 2B15 gene (D85Y) is not associated with prostate cancer risk. Cancer Epidemiology and Prevention Biomarkers 11: 497-98.
  52. Ma L (2014) Association between tumor necrosis factor-alpha gene polymorphisms and prostate cancer risk: a meta-analysis. Diagnostic pathology 9: 74.
  53. Bidwell J (1999) Cytokine gene polymorphism in human disease: on-line databases. Genes and immunity. 1: 3.
  54. Kroeger KM, KS Carville, LJ Abraham (1997) The− 308 tumor necrosis factor-α promoter polymorphism effects transcription. Molecular immunology 34: 391-9.
  55. Shen C (2011) Polymorphisms of tumor necrosis factor-alpha and breast cancer risk: a meta-analysis. Breast cancer research and treatment 126: 763-770.
  56. Lu P (2010) Meta-analysis of association of tumor necrosis factor alpha-308 gene promoter polymorphism with gastric cancer.
CommentsTable 1 CommentsTable 2 CommentsTable 3 CommentsTable 4 CommentsTable 5
CommentsFigure 1 CommentsFigure 2 CommentsFigure 3 CommentsFigure 4 CommentsFigure 5 CommentsFigure 6