Open access peer-reviewed chapter

Cytokine Gene Polymorphism and Cancer Risk: A Promising Tool for Individual Susceptibility and Prognostic Implications

Written By

Arshad A. Pandith, Ina Bhat, Sheikh Mansoor, Aabid Koul, Usma Manzoor, Iqra Anwar, Fozia Mohammad, Qurat Ul Aein, Shahid M. Baba and Carmen Vladulescu

Submitted: 25 May 2021 Reviewed: 09 July 2021 Published: 26 December 2021

DOI: 10.5772/intechopen.99363

From the Edited Volume

Genetic Polymorphisms - New Insights

Edited by Mahmut Çalışkan

Chapter metrics overview

312 Chapter Downloads

View Full Metrics

Abstract

Cytokines are potent molecules produced mainly by specific activated immune cells to control inflammatory responses besides other biologic processes. Although active participation of cytokines provides defense against carcinogenesis on the other hand, deregulation at the genetic level influences their activity to promote tumor development. Among many aspects, constitutional polymorphic sequence variations are key factors that derange the cytokine expression to lead an individual’s propensity to risk for different cancers. Cytokine polymorphisms are now believed to alter these critical molecules that have a dual face in carcinogenesis as, when implicated in the activation of the immune response, these molecules check the cancer development while their persistent inflammatory reaction can envisage the development of malignancy and tumor growth. We have given ample evidence of case-control studies in a range of cancers where substantial evidence, as reported in this chapter, links polymorphism of cytokine gene susceptibility with numerous cancers. Cytokine gene polymorphism is vital to be significant bimolecular genetic determinants of susceptibility and prognosis of cancer. A strong need is felt for more case-control association studies in cytokine candidate genes involved in specific pathways for particular cancer in bigger powered sample sizes involving additional variables to disclose their factual risk for cancer.

Keywords

  • cytokines
  • polymorphism
  • malignancy
  • tumor
  • immune response
  • case-control
  • inflammatory reaction

1. Introduction

Cancer is the world’s second leading cause of death and accounts for around 1 in 6 deaths worldwide. A projected 18.1 million new cancer-related cases and 9.6 million cancer-related deaths were recorded in 2018, according to the 2018 GLOBOCAN study. Lung cancer is the most frequently diagnosed cancer for both sexes with 11.6% of the total cases of cancer and is also the leading cause of cancer death with 18.4% of the total cancer deaths. Cancer, a cellular overgrowth disorder, involves cellular transformation, apoptotic dysregulation, uncontrolled proliferation, invasion, angiogenesis, and metastasis [1]. A significant correlation between chronic infection, inflammation, and certain forms of cancer have been suggested in clinical and epidemiological research, with inflammation frequently occurring in and around tumors [2].

Recent studies have shown that chronic inflammation that regulates the microenvironment of the tumor is involved in the initiation of the tumor and is a critical component of tumor promotion and progression [3]. The insistent inflammatory microenvironment contributes to increased promotion of the tumor, accelerated progression, invasion of the surrounding tissues, angiogenesis, and eventually metastasis [4]. The tumor microenvironment has a vast abundance of cytokines as well as other inflammatory mediators which impact immunosuppression, cancer growth, tissue remodeling, and angiogenesis [5]. Cytokines are a heterogeneous group of glycoproteins or small soluble polypeptides (secreted or membrane-bound) chiefly produced by the immune cells which under normal conditions are produced in response to specific stimuli exerting pleiotropic and redundant effects thereby altering the behavior of the identical or different cells by regulating the growth, differentiation, and activation of normal cells especially immune cells [6]. Depending on the microenvironment, cytokines may either have pro- or anti-inflammatory or may even have immunosuppressive activity [7]. In response to both antigen-specific and nonspecific stimuli, the development of multiple cytokines by immune cells plays a crucial role in the outcome of inflammatory immune responses [8]. They are either released in response to a variety of cellular stresses, including carcinogen-induced injury, infection, inflammation, and immunity that inhibits tumor growth and progression, or are host-derived cytokines that cause cancer cells to promote growth, reduce apoptosis, aid invasion and metastasis. The role of cytokines in subsequent settings is to cause a host response aimed at controlling cellular stress and reducing cell damage. Although critical insult control encourages tissue repair, failure to control the injury can lead to the insistent development of cytokines, leading to further aggravation of tissue damage. Depending on several stages of cancer development and progression, host responses to cellular stress may influence it inherently [9]. Therefore, the cytokine mixture produced in the tumor microenvironment plays an important role in the pathogenesis of cancer [10]. The molecular and cellular changes that eventually lead to cancer stimulate changes in the local cytokine environment, stimulate immune cell intrusions, and release additional cytokines that function in an autocrine or paracrine manner [11]. In addition, pro-inflammatory cytokines are associated with anorectic and cachectic disease in patients with progressive cancer, pain in the form of both algia and dynia, toxicity, and resistance to treatment. Physical exercise can, however, alter cytokine levels and decrease fatigue in cancer patients, and may also increase their prognosis [7].

Cytokines provide a key molecular link between inflammation, tumor promotion, and progression which mainly include the interferons (IFNs), the interleukins (ILs), the tumor necrosis factor super family (TNFs), the transforming growth factor super family (TGFs), and Colony Stimulating Factors (CSFs) [12].

Genetic variations in the human genome can govern the risk of cancer development, symptoms, treatment, and its outcome [7]. In recent years, within cytokine gene sequences, many single nucleotide polymorphisms (SNPs) and a small number of microsatellite polymorphisms have been reported, mainly within their promoter regions [13] Indeed the most common variations in the genome are the SNPs [14]. Some suggest that the differences in SNPs may also help explain cancer disparities among the various ethnic groups [15]. Differential levels of gene transcription, including TNF -308 and IL-10-10822 can be associated with some of these polymorphisms [16]. Several genetic studies have attempted to associate these cytokine polymorphisms with cancers; however, the studies are still limited, but increasingly evolving. The link between TNF alpha-SNPs and specific cancers, including oral carcinoma, has been documented in several studies [17] and non-Hodgkin’s lymphoma [18], but most of these associations are refuted by others. IL-1 gene polymorphisms that provide increased expression of this pro-inflammatory cytokine are associated with an increased risk of cancers, mainly of gastric origin [19]. The association between IL-1B/IL-1RN polymorphisms and the development of gastric adenocarcinoma following Helicobacter pylori infection is also well known [20]. In relation to cancer, IL-10 polymorphisms are of particular concern since IL-10 has immunosuppressive and anti-angiogenic properties [8]. In view of this, this chapter highlights the essential role of genetic polymorphisms with respect to a host of cancers and their susceptibility in different geographical regions.

Advertisement

2. Pro-inflammatory cytokine gene polymprphisms

2.1 Solid tumors

2.1.1 Breast cancer

2.1.1.1 Interleukin 6

IL-6 is a major inflammatory pleiotropic cytokine that is considered a main growth-promoting factor [21]. Several IL-6 target genes are involved in the progression of the cell cycle and apoptosis suppression, which emphasizes the significance of IL-6 in tumorigenesis [22]. Many polymorphic IL-6 studies have shown that it is associated with breast cancer risk and prognosis. A study indicates that IL-6 is a predisposing genetic factor that contributes to the prognosis of breast cancer, with a G/C polymorphism associated with high levels of IL-6 production within the promoter region of the IL-6 gene correlating with a worse prognosis. While few studies have been published to date on polymorphisms within the IL-6 gene cluster and breast cancer [23], there is still controversy about the evidence on 174 G/C IL-6 polymorphism in breast cancer. The presence of IL-6 polymorphism was associated with an improved outcome in high-risk breast cancer, reported by DeMichele et al. [24]. Iacopetta et al. [25] found that the IL-6 polymorphism was predictive of the phenotype of aggressive breast cancer

2.1.1.2 Interleukin 10

IL-10 is an immunosuppressive and anti-angiogenic multifunctional cytokine that can have both tumor-promoting and inhibiting properties [26]. The IL-10 gene promoter has been recognized to have a significant number of polymorphisms (primarily SNPs) [27]. As a consequence, promoter polymorphisms have been the most scrutinized, particularly with regard to possible gene transcription and protein production influences. Many studies on the interactions between genotypes of IL-10 and breast cancer have been published. One small breast cancer study confirmed that the low-expression 21082 AA genotype was associated with a high risk of disease [28]. In comparison, a larger case-control study found a correlation between the 2592 AA genotype and the reduced risk of breast cancers [29] but did not associate with any clinical parameters.

2.1.1.3 Interferon-γ

Interferon-γ (IFN-γ) is the cytokine that likely has a vital role in carcinogenesis. Several combined epidemiological studies have shown that IFN-γ can function as a major risk factor for breast cancer development and progression, primarily due to genetic polymorphisms of IFN-γ [30]. Interestingly, genetic polymorphisms in the IFN-γ gene are likely to affect the degree of IFN-γ expression, leading to impaired function or decreased IFN-γ activity; subsequent low IFN-γ expression levels would encourage tumor development, which can contribute to can susceptibility to breast cancer [31]. In order to be more precise, many common IFN-γ gene polymorphisms have been found to increase the risk of breast cancer, such as rs20697 (−1615C/T) and rs2430561 (+874T/A) [30, 32, 33]. Contradictory findings have also been reported recently, although several studies have also suggested that IFN-γ genetic polymorphisms can play a critical role in breast cancer pathogenesis [30, 31]. Gene polymorphisms, with low levels of IFN-γ, can alter the function and expressions of IFN-γ. The rs2430561 A>T polymorphism was revealed by Liu et al. in intron 1 of IFN-γ [34]. The gene can transform its transcription functionally and lead to susceptibility to breast cancer [35]. Another meta-analysis shows that genetic polymorphisms within the IFN-γ gene were significantly associated with an increased risk of breast cancer, especially the polymorphism of 2430561 T>A. No connection was, however, identified between rs2069705 C>T polymorphism and breast cancer susceptibility. Genetic polymorphisms and the risk of breast cancer in Asians, but not among Caucasians, suggest that ethnic differences may affect the susceptibility of individuals to breast cancer [36].

2.1.1.4 Tumor necrosis factor-alpha (TNF-α)

TNF-α is an essential pro-inflammatory cytokine for human cancer growth and progression [37] by stimulating the development of genotoxic molecules (NO, ROS) that can lead to DNA damage and mutations, TNF-α can promote tumor initiation and progression. The increased risk of many cancers, including breast cancer, is associated with genetic polymorphisms that boost TNF-α development [33].

Increasing evidence has shown that a SNP in the promoter region of the TNF-α gene (−308G>A, rs1800629) induces genetic susceptibility in many forms of tumors like BC [38]. The TNF-α-308A allele tends to have a higher constitutive and inducible expression, as the −308G>A mutation affects the AP-22020 consensus binding site [39]. The results on the association of –308 (G/A) TNF-α polymorphism with breast cancer development are rather contradictory [40]. A rise in homozygous (−308AA) TNF-α genotype frequency was significantly associated with the development of breast cancer and poor prognosis in Tunisian women [40]. No link between the homozygous (−308AA) TNF-α genotype and breast cancer risk was identified in Holland. The association of allele (−308A) TNF-α with tumor vascularization has, however, been shown to be [41]. Where the majority of studies found no link between-308 (G/A) TNF-α polymorphism and the occurrence of breast cancer [42, 43]. Two recent meta-analyses have confirmed that the genotypes TNF-α-308GA and AA were significantly associated with a reduced risk of breast cancer in Caucasians [14, 16, 44, 45]. The allele frequencies in the controls of some studies [42, 46] did not show compliance with Hardy-Weinberg (HW) in the meta-analyses. In addition, Yang et al meta-analysis included one study comparing the frequencies of various genotypes of TNF-α-308 polymorphism in patients with benign breast disease and controls, and another study that did not have frequencies for each genotype [14, 16, 44, 45]

2.1.2 Hepato-celllular carcinoma

2.1.2.1 Interleukin-1

IL-1 α and IL-1β are pro-inflammatory and potent cytokines. It has been stated that the genes of this family are highly polymorphic that modulate the expression of IL-1β. IL-1β −511T/C, −31C/T, and +3593C/T are the most studied SNPs of the IL-1 gene family, in addition to 86bp of VNTR in intron 2 of the IL-1RN gene with 5 different alleles [47]. IL-1β-31C/T is associated with increased transcription initiation factor binding that has been shown to participate in hepatic carcinogenesis [48]. In the presence of IL-1β (−511CC and −31TT) genotypes, allele 2 was known to be a risk for HBV-related hepatocellular carcinoma (HCC). The IL-1β-511C allele was found to be an IL-1RN risk with no HCCC risk [49]. On the other hand, there was no connection between the IL-1β-511C allele and HCC in several studies. In the Wang et al. analysis [50], the IL-1β-31TT genotype and the IL-1β-511/−31CT haplotype were associated with an increased risk of HCC-related HCC and the risk factor was not the IL-1RN VNTR polymorphism.

2.1.2.2 Tumor necrosis factor-α

Polymorphisms of TNF-α (−1031T/C, −863C/A, −857C/T, −308G/A and −238G/A) are found to alter TNF-α development [51]. A critical risk factor for HCCC is regarded as TNF-α −308G/A [45], but TNF-α-238G/A is engineered to play a passive role in the risk of HCC [52]. On the other hand, several other studies indicated an increased risk of HCC for the allele TNF-α-308A [51]. The association between TNF-α promoter alleles such as TNF-α-308A and TNF-α-238A and various TNF-α-238A expressions is indicated in several studies [39].

2.1.2.3 Interleukin-6

In order to counteract inflammatory reactions, IL-6 is a key factor in viral infections. In the promoter area, the most studied genetic polymorphisms in the IL-6 gene are located downstream (−597G/A, −572G/C, −174G/C, and −373A/T), and have an effect on IL-6 development levels at the transcriptional stage [52]. These are reported to be associated with chronic hepatitis, where IL-6-572G/C was found to be associated with the risk of HCC-related HBV [53].

2.1.3 Gastric cancer (GC)

2.1.3.1 Interleukin-1Beta

There are three associated genes in the IL1 cluster: IL1-A, IL-1B, and IL-1RA that encode the signal proteins IL-1, IL-1, and their receptor, IL-1RA, respectively. The association between IL-1B and IL-1RA gene polymorphisms and the development of gastric cancer (GC) has been identified in numerous studies [47]. Polymorphisms of IL-1 are located in positions-511 (CT, rs16944), -31 (TC, rs1143627), and 3954 (CT, rs1143634) that affect the expression of IL-1. The majority of studies classify the IL1β-511 polymorphism T allele as normal among Caucasian individuals with non-cardiac GC but preferably for cancer of the intestinal subtype. It is, therefore, feasible to propose this SNP as a possible GC predictive marker. Even after numerous meta-analyses have been carried out, investigations of the IL1B-31 TATA-box polymorphism appear to be controversial. Around 14 studies [54] reported a slight non-significant correlation between the C variant allele and GC risk in comparison with TT homozygotes. Again, in contrast with Hispanic or Caucasian cultures, there was no correlation between Asian groups. A modest increase in the intestinal GC subtype among C allele carriers in Caucasian populations was suggested by histologic stratification, but this statement was not true for the diffuse GC subtype. Wang et al. also found that the +3954 gene polymorphism T allele contributes to the GC risk. A lack of interaction between the +3954 polymorphism and GC was stated by Xue et al. [55]. A limited number of studies were conducted on the IL-1β-+3954 C/T polymorphism [55].

2.1.3.2 Interleukin-2

In the differentiation of CD41-positive T cells into Th1 and Th2 effector subsets, IL-2 effectively controls the immune response and plays an important role. IL-2 leads to the activation and transmission of immune responses that are inflammatory, including H. pylori-induced gastric inflammation. In the promoter region, IL-2, IL-2-330, and -384 have two types of SNPs that affect IL-2 development [56, 57]. Wu et al. [57], reported that the T allele significantly reduced the risk of gastric cardiac cancer with IL-2-330 polymorphism. Another research, on failed to show a significant association with IL-2-330, while Togawa et al. [58] on the other hand, stated in 2005 that the IL-2-330 T/T genotype in Japan increased the risk of gastric atrophy associated with GC. The IL-2-330 polymorphism yields contradictory findings. In addition, for IL-2-384 and +11.4 polymorphisms and GC growth, no significant association has been seen [59].

2.1.3.3 Interleukin-6

IL-6 appears to be involved in gastric oncogenesis, as serum IL-6 levels show an increase in the gastrointestinal cells and mucosa of patients suffering from GC [60]. The data collected indicated that the polymorphism of IL6-174G/C was related to the risk of GC in the West. The suppression of tumor necrosis factor-α and IL-11 could explain the carcinogenic properties of IL-6 [61]. In 2009, Kang et al. showed a strong negative association among HP-positive cases and controls between the IL6-572G/C polymorphism GG genotype and duodenal ulcer risk. The effect of the G allele on the rate of synthesis of proteins has not been determined to date, and the role of this SNP remains unclear.

2.1.3.4 Interleukin-8

IL-8 improves the proliferation and migration of cells and serves as a chemical attractor and mediates chronic inflammatory processes [62]. In contrast, mucosal levels of IL-8 were found to be elevated in GC patients, and the prognosis was significantly lower in patients with high expression of IL-8 compared to patients with moderate levels of this protein [63]. IL-8 can induce Reg protein expression in stomach cells, which intensifies gastric mucosal cell proliferation and may indirectly promote the initiation of GC [64]. In the IL-8 gene locus, fifteen functional polymorphisms occur, and some can alter gene expression [65]. Several IL8- case-control studies on 251A/T (rs4073), IL8 +396T/G (rs2227307), and IL8+ 781C/T (rs2227306) were conducted in which IL8-251A/T, A allele was associated with increased GCC [66]. Kang et al. [67] also found that higher GC risk is correlated with the AA genotype of HP-positive individuals. Due to conflicting findings, the role of IL-8 gene polymorphisms in GC remains unclear. The −251 polymorphism of the gene, however, seems to play a major role in the development of GC and needs further study. In this region, the +396 and +781 SNPs are almost unexplored.

2.1.3.5 Interleukin-17A and 17F

IL-17A and IL-17F, synthesized by activated T cells, are characterized as pro-inflammatory. Five variants of this cytokine are known which differ in properties and sites of expression from the founding member IL-17A. Several studies have shown that IL-17 leads to gastric carcinoma growth and progression [68]. It has recently been suggested that a low IL-17 tumor expression rate can imply a poor prognosis in GC patients. Wu et al. reported that in comparison with the mutant AA genotype, the GA and GG genotypes of +7488 SNP were correlated with an increased GC risk and also reported the absence of a link between IL17-197G/A polymorphism and GC risk [69].

2.1.3.6 Tumor necrosis factor-α

One of the pro-inflammatory cytokines strongly expressed in H. pylori-induced gastritis is TNF-α, a potent gastric acid secretion inhibitor [70, 71]. While the TNF-α promoter has recorded several polymorphisms, most studies have concentrated on the G/A polymorphism at position 307 since most of the other polymorphisms are functionally silent. In patients with polymorphism at position 307 with malignant tumors, multiple studies find a higher concentration of TNF-α [72]. TNF-α is a pro-inflammatory cytokine, which is a mediator of the immune response in H. pylori and shares many biological behaviors with IL-1 [73]. It doubles the risk of gastric cancer without any correlation with the risk of gastric cancer of the esophagus or cardium [74, 75]. Studies done on the effect of TNF-α 307 polymorphisms on the expression of mucosal cytokine and showed no substantial differences in TNF-α level between different allele carriers that indicate that this polymorphism does not affect the development of cytokine. Some race-specific associations have been proposed by another meta-analysis summarizing data on TNF-α, 308 variants, with an increased risk of gastric cancer in various ethnic populations. TNF-α 238 polymorphisms were not substantially associated with the risk of gastric cancer, consistent with previous results [76]. Another research showed that a possible risk factor for gastric cancers is TNF-α 857 T allele.

2.1.4 Prostate cancer

2.1.4.1 Interleukin-1

The relation between the risk of IL-1 family polymorphisms, including IL-1A, IL-1B, and IL-1RN and prostate Cancer (PCa), has been less studied. There was no important correlation between PCa risk and IL-1B/IL-1RN polymorphism in several studies. In a recent meta-analysis, IL-1A, IL-1RN (rs315951 and rs3087263), and IL-1B+3953 (rs1143634) polymorphisms were not significantly correlated with the risk of PCa. In homozygote and recessive models, IL-1B-511 (rs16944) polymorphism was significantly associated with PCa risk, and in the heterozygote model, the allele comparison IL-1B-31 (rs1143627) polymorphism was also marginally significantly associated with PCa risk. Therefore, the meta-analysis indicated that IL-1B-511 (rs16944) and IL-1B-31 (rs1143627) sequence variants were significantly associated with PCa risk. This result presents more new evidence that pro-inflammatory cytokines and inflammation play an important role in the etiology of PCa [77].

2.1.4.2 Interleukin-6

Interleukin 6 (IL 6) plays a crucial role in the inflammatory phase among the cytokines involved in inflammation. Mandić et al., found that IL-6-174 SNP differs between ethnicities and that single polymorphic cytokine variants most likely have little effect on the susceptibility of PCa [78]. The study by Pierce et al. showed that circulating IL- 6 and its gene polymorphism did not affect the risk of PCa [79]. Whereas Mandal et al. had a contrary view [80]. Another meta-analysis of 11 independent studies, including 10,745 cases and 13,473 controls based on several recently published studies which indicated an inconsistent conflicting and inconsistent trends of association between IL- 6 (174 G/C) and PCa. IL- 6 (174 G/C) polymorphism has been found not to be a risk factor for prostate cancer in the general population [81].

2.1.5 Esophageal cancer

2.1.5.1 Interleukin 6

IL-6 (interleukin-6) is a pro-inflammatory peptide that is actively involved in tumorigenesis [82]. The effect of IL-6 and IL-6 receptors (IL-6R) on the prognosis of esophageal cancer (EC) has been identified [83]. Systemic and/or local IL-6 therefore appears to be a central molecule in the stimulation of ESCC progression. Different studies have shown an association between various IL-6 polymorphisms and cancers. Polymorphism of IL-6-634G>C and prognosis after EC esophagectomy revealed that when EC patients were treated surgically, those with the IL-6-634G/G or G/C genotype had a 3-fold poorer prognosis than those with the C/C genotype. In comparison, IL-6R polymorphism and IL-6 tumor expression are not associated with prognosis [84]. With respect to EC, Oka et al. [85] stated that poor survival was associated with high serum IL-6 levels. Buraczyn et al., on the other hand, stated that in the presence of inflammatory stimulation, patients carrying the G allele and to an even greater extent the G/G genotype showed higher IL-6 output at position −634 in the IL-6 promoter region than patients carrying the C allele [86]. Kitamura et al. also reported that the 634 G allele is associated in vitro with increased IL-6 production in peripheral mononuclear blood cells [87]. While IL-6 levels are associated with different cancers, the relationship between IL-6 and EC polymorphisms requires further evaluation because less studies are available [88].

2.1.5.2 Interleukin 12

The pro-inflammatory cytokine family is the Interleukin 12 (IL-12) family, which is essential for host tumor resistance [89]. A few studies have determined whether polymorphisms and serum levels of the IL-12 family (IL-12A gene rs568408, IL-12Bgene rs3212227, IL-27 gene rs153109, rs17855750, rs181206) and its family receptor (IL-12Rb1, 378 C/G) gene are correlated with EC. Studies that revealed polymorphisms of IL-12 rs568408, rs3212227, and IL-12Rb1 gene 378 C/G and serum levels of IL-12p40 and IL-27p28 were significantly associated with the risk of EC [90]. IL-12Rb1 gene Codon 378 C to G causes a transition in amino acid (glycine to arginine), which may further weaken the transcript’s subsequent biological activity and is linked to many malignancies such as leiomyoma [91].

2.1.5.3 Interleukin-18

Interleukin-18 (IL-18) is another cytokine that is mainly involved in the inflammatory immune response and is a potent factor triggering IFN-γ. Genetic IL-18 gene polymorphisms have recently been observed to have a significant effect on the vulnerability of a number of inflammatory diseases and various malignancies, including EC [92]. Different SNPs have been identified in the promoter region of the IL-18 gene and are likely to influence gene activity [93]. In the promoter region of the same gene, three SNPs were found at distinct positions −137, 607, and 656 relatives to the transcriptional start site. A study conducted by Ye et al. in the Chinese population investigated 137 G/C and 607 C/A polymorphisms of the IL-18 gene in EC patients. The 137 GC and CC genotypes were associated with a significantly increased risk of ESCC as compared with the −137 GG genotypes as G to C substitution at position 137 abolishes a histone4 transcription factor-1(H4TF-1) nuclear factor-binding site and hence has an impact on IL-18 activity. Nevertheless, in EC patients, the genotype and allele frequencies of IL-18 promoter 607 C/A polymorphism were not substantially different from those in healthy controls. The results, therefore, indicate that IL-18 137 G/C polymorphism could be used as an ESCCC genetic susceptibility marker [94].

2.1.5.4 Tumor necrosis factor-α

Tumor necrosis factor-alpha (TNF-α) is a pro-inflammatory cytokine that plays a major role in host defense and inflammatory responses but also induces cell death and tissue degradation in some instances [95]. There have been reports of dysregulated expression of TNF-α associated with a number of tumors, including EC, [96]. A number of SNPs of TNF-α gene have been found, which include TNF-α-238 G/A (rs361525), TNF-α-308G/A (rs1800629), TNF-α-857C/T (rs179972), TNF-α-863C/A (rs1800630), TNF-α-509C/T (rs1800469) and TNF-α-1031T/C (rs1799964) [83]. Among these, the most common TNF-α polymorphisms occur at position 308 in the promoter region and are extensively studied, showing a strong association with increased TNF-α production [97]. Many studies have focused on the association between TNF-α-308 G>A (rs1800629) and EC risk [98]. Deans et al. have found that the genotype AA TNF-α 308 is associated with an adverse prognosis of gastroesophageal cancer [99]. In contrast, the results of the Cui et al. study found that TNF-α-308G/A polymorphism was not associated with EC risk [100]. In addition, Guo et al. did not find a significant difference between EC patients and controls in the overall genotypic distribution of TNF-α-308G/A polymorphism [101]. In addition, few meta-analyzed studies report that TNF-α-308 G>A (rs1800629) is poorly associated with an increased risk of EC [102]. Conclusions on the role of TNF-α -308G/A gene polymorphism in the risk of EC have, therefore, been inconsistent.

2.1.5.5 Interferon-γ

The cytokine produced against viral and intracellular bacterial infections in the human body is Interferon-gamma (IF-γ). Many polymorphisms have been investigated mostly in IFN-gamma, which are significantly associated with many complications such as susceptibility to many infections [103]. It has been stated that a risk factor for EC may be the IFN-γ + 874AT genotype. The study by Du et al. has shown that IFN-γ + 874, T allele may predispose for EC [104]. Another research found that an important association existed between the genetic polymorphism of INF-γ 874A>T and infectious complications following esophagectomy in a cohort of EC patients [105].

2.1.6 Pancreatic cancer

2.1.6.1 Interleukin-1 beta

Interleukin 1b (IL1b), a central IL-1b gene-encoded pro-inflammatory cytokine, has been associated with chronic inflammation and plays an important role in inflammatory pancreatic diseases, including pancreatic cancer [106]. In the production of pNETs, the functions of IL-1b −511C/T and +3954 C/T genotypes remain unclear. PNETs are a heterogeneous group with different biology and prognosis of tumors; they can occur as solitary tumors, and up to 15% of pNETS are part of hereditary syndromes. The findings of Maja et al. [107] showed a significant correlation between the IL-1b −511C/T genotype and CTCT −511/+3954 genotype combination and susceptibility to functional pNET growth, and the risk of non-functional pNET development were associated with patients with CTCC −511/+3954 genotype combination. All of these findings indicate IL-1b participation in the growth of pNET [107].

2.1.6.2 Interleukin-2

In T cell-dependent immunity, IL-2 is a potent pro-inflammatory cytokine and a central regulatory cytokine. Several SNPs) have been found to alter IL-2 production in the promoter region and to be associated with inflammation-based cancer [58]. The G-allele in the IL-2 promoter at the −330 position seems to correlate with higher IL-2 output [108]. It has been shown that during the cell cycle, dividing cancer cells release IL-2 in various concentrations and that IL-2 promotes cancer development [109]. Hofsli and collaborators found that GEP-NET carcinogenesis growth factors downregulate the IL-2 receptor, presenting proof of an additional mechanism through which neuroendocrine cancer cell growth is promoted [110]. It was found that G-allele raises the expression of IL-2 at the −330 position and that individuals homozygous for the G-allele have three times higher IL-2 values [108].

2.1.6.3 TNF-α

In the inflammatory etiology of pancreatic cancer, TNF-α plays a very crucial role. The risk factor for various forms of cancer, such as hepatocellular carcinoma, gastric cancer, and breast cancer, has been identified as TNF-α-308 polymorphism [111, 112, 113], while previous studies have reported that polymorphisms in the TNF-A-308 A/G gene are not linked to pancreatic cancer [78].

2.1.7 Bladder cancer

2.1.7.1 Interleukin-6

IL6 is an inflammatory pleiotropic cytokine released by different types of lymphoid/non-lymphoid cells [114]. Immune response, cell survival, proliferation, and apoptosis are critical for [113]. In the IL-6 promoter region, numerous polymorphic variants have been identified that are associated with IL-6 transcription activity [115] which indicates a connection to cancer [116].

IL-6 polymorphism has been shown to modulate changes in its expression to cause cancer risk, including bladder cancer. Association studies of IL-6 gene polymorphisms conducted in India and globally confirm the risk of several CC genotype and C allele cancers, especially BCC allele cancers [117, 118]. Fishman et al. have stated that IL-6-174 G/C polymorphic variation affects transcription and IL-6 protein expression [119]. Conversely, several studies from other parts of the Indian and Caucasian population indicated that conflicting findings were present [120, 121]. The variant genotype IL-6-174 G/C was shown to be significantly associated with an increased risk of BC [122] but no BC risk association was found by other authors [123]. In addition, there was no proven association between IL-6-572 G/C, -596 A/G polymorphisms, and BC risk [118].

2.1.7.2 TNF-α gene polymorphisms

A major inflammatory cytokine is the tumor necrosis factor-alpha (TNF-α) gene, which mediates a connection to all steps involved in tumor growth and development. TNF-α gene polymorphisms and their receptors have conceptualized the understanding of the genetic effects of inflammatory consequences [124]. Polymorphic sequence differences are mainly documented in association studies of various cancers like BC, with some studies refuting and other studies showing the association for cancer risk, mostly single nucleotide changes in TNF-α promoter [125, 126]. This contentious link between TNF-α polymorphisms and UBC is due to the race, sample size, and technical aspects involved. Of the 7 variants of the TNF-α gene studied by Marsh et al., 859T and +488A polymorphisms were significantly linked to BC risk [127]. Another research investigates 3 TNF-al-1031 T>C polymorphism SNPs where high risk was identified for controls of BC cases [128]. Similarly, experiments on TNF-al-308 G>A polymorphism have also reported controversial results [129, 130]. Although TNF-α-308 A variant alleles with a major risk relationship to BC risk were observed by Lima et al. [131].

2.1.8 Gliomas

2.1.8.1 IL-4 gene polymorphism

Research by Brenner et al. found that the polymorphism of IL-4 (rs2243248, −1098T>G) was substantially correlated with the overall risk of glioma [132]. Another study showed that IL-4 induced aberrant Stat3 activation in glioblastoma cells but not in normal human astrocytes, and hypothesized that aberrant Stat3 activation induced by IL-4 could contribute to the pathogenesis of GBM cells [133].

IL4R encodes the interleukin-4 receptor alpha chain that can connect interleukin 4 and interleukin 13 to regulate the development of IgE [134]. In 2013, TianboJin et al. reported that rs1801275 in the IL-4R gene can predict the over-dominant model of Glioma susceptibility by 2.29-fold [135]. Furthermore, another article also stated that rs1801275 could increase the risk of studying glioblastoma [136]. However, an important link between mutant IL-4R alpha rs1801275 and gliomas was not found by Ruan et al. [137].

2.1.8.2 IL-12gene polymorphism

Many SNPS, like 1188A/C, in the IL-12 gene will affect its levels of expression and are associated with several tumors such as nasopharyngeal carcinoma [138] and breast cancer [139]. This SNP might influence the susceptibility of individuals to glioma. Haidar et al. stated that neither mutant (hetero or homozygous) genotypes nor mutant IL-12p40 1188A/C variant allele appears to be associated with gliomas [140]. In line with a meta-analysis review, this outcome [141]. The stratification by cancer type showed that the variant only represents a risk factor for cervical and nasopharyngeal cancer, although these authors found a substantial association of the variant with overall cancer.

2.1.8.3 IL-13 gene polymorphism

IL-13 plays an important role in allergies and is essential for the suppression of tumor immune surveillance through an immune regulatory pathway [142]. SNPs, including rs25041, rs1800925, and rs1295686, found in the IL-13 gene, are closely linked to IL-13 expression. In human malignant glioma cell lines and in primary tumor cell cultures, IL-13 has been shown to be over-expressed [143]. The serum immunoglobulin E (IgE) level could be significantly increased by the rs20541 A allele (glutamine (Gln, Q) form) in different populations [144]. Evidence has also been established by previous research that IgE levels in patients with glioma were lower than in people without glioma [136]. It is very fair to expect the IL-13 gene rs20541 polymorphism to exert some effect on glioma susceptibility due to its important roles in immune surveillance and serum IgE level modulation. A recent meta-analysis review, however, indicates that the IL-13+rs1800925 genotype could be a risk factor for gliomas and that IL-13+rs20541 contributes to cancer [145] (Table 1).

ActivityCancer typeCytokinesPolymorphismsAssociationAlleleReferences
Pro-inflammatorySolid tumorsBreastIL-6−174 G/CHigh grade tumor−174 CC[25]
IL-1021082 G>AWorst prognosis−21082 AA[28]
High risk−21082 AA[29]
IFN-γ+874T/AIncreased risk+874 AA[36]
−1615C/TNo[36]
TNF-α−308G/APoor prognosis−table308GA, AA[40]
No[43]
HCCTNF-α1031T>CNo1031T>C[146]
863C>AYes863C>A[146]
857C>TNo857C>T[146]
308G>AYes308G>A[146]
IFN- γ+874T>ARisk factor+874T>A[147]
IL-18607A>CRisk factor607A>C[148]
137C>GRisk factor137C>G[149]
148G>CRisk factor148G>C[150]
IL-16rs11556218T>GRisk factorrs11556218T>G[151]
rs4072111C>TRisk factorrs4072111C>T[151]
rs4778889Risk factorrs4778889[152]
IL-12Ars3212227A>CRisk factorrs3212227A>C[153]
IL-12Brs2243115T>GRisk factorrs2243115T>G[147]
GastricTNF-αTNF-α 307Race sp. asso.[150]
TNF-α 307, 1031, 863, 857, and 238Not favoring[154]
[75]
TNF-α 307Not favoring[155]
IL-1βIL1β –31 T/CSlight non sigC allele[155]
IL1β –511C/TSignificant associationTT[94]
AssociationT allele[156]
AssociationT allele[55]
IL1β +3954 C/TLack of associationT allele[55]
AssociationT allele[156]
IL-6Il-6 −174 G/CAssociationGG[157]
IL-6 −572 G/CNegative associationGG
IL-6 −597 G/ALack of associationG allele[158]
IL-8IL8 –251A/T,Lack of associationTA[158]
IL8 +396T/G,Lack of associationGG[158]
IL8+781C/TLack of associationCT[157]
IL-2IL-2 -330Sig reduced riskT allele[57]
Non-sig associationT allele[159]
Sig associationTT genotype[58]
IL-2 −384G/T, +114G/TNon-sig association[59]
IL17A and IL17F–197G/ALack of associationAG[4]
+7488A/GAssociationGG[4]
ProstateIL-6IL-6 −174Least association[78]
IL-1IL-6No. assoc.[79]
IL-6Association[80]
IL-6 −174 G/CNo association[81]
IL-1α/IL-1RN/ILβ +3953No association[77]
IL-1β-511/IL-1β-31Association[77]
EsophagealIL-6−634Poor prognosis−634GG[84]
IL-12Ars568408 G>ASusceptibilityrs568408 GA,GG[160]
IL-12Brs3212227A>CSignificant riskrs3212227AC,CC[160]
IL-12Rb1−378Susceptibility−378 GG[160]
IL-18−137 G/CIncreased risk−137 CC,[94]
−607 C/ANo−607 C/A[94]
TNF-α−308Adverse prognosis−308 A/A[99]
IFN-γ+874A>TProtective+874AT,TT[104]
PancreaticIL-1β−511C/T, −511/+3954FavouringCT genotype[106]
IL-2FavouringCT genotype[107]
TNF-α-308IL-2 −330FavouringG allele[108]
A/GTNF-α-308 A/GNot favouringAG[69]
Bladder cancerTNF-α1031T>CNo1031T>C[161]
−857C>TYes−857C>T[161]
−308G>AYes−308G>A[161]
IFN-γIFN-γ +874YesIFN-γ +874[162]
IL-1β−511C>TNo−511C>T[123]
IL1-RNVNTRYes[163]
GliomaIL-4−588C>TSuggestive association−588TT[132]
−1098T>G−1098GT,GG[132]
IL-4R+828A>GOverall risk+828A>G[164]
Overall risk[137]
IL-12−1188A>CNo[140]
IL-13+2044A>GNo susceptibility+2044AA,AG[136]

Table 1.

A meta-data of different pro-inflammatory cytokine polymorphisms and their association in various solid tumors.

2.2 Hematological malignancies

2.2.1 Lymphomas

2.2.1.1 IL-4 and IL-5 gene polymorphisms

In the proliferation of T cells, Il-4 and Il-5 play a key role. The study of the relationship between SNPs was chosen from the main immunological genes of cytokines and NHL in several studies. This represents a pathway-based approach to the investigation of common genetic variants in the cytokine network of Th1 and Th2. Common genetic variants of the Th2 cytokine genes have been observed and are associated with NHL risk. SNPS has been shown to be substantially associated with an increased risk of NHL in the TH2 genes IL-4 (−1098T>G) and IL-5 (−745C>T) [165].

2.2.1.2 IL-6 gene polymorphism

While monocytes are the main source of IL-6, many cells, including dendritic cells, lymphocytes, neutrophils, mast cells, mesenchymal cells, and tumor cells, may produce it. IL-6 rs1800797 (IL-6 rs1800795G>C, rs1800796G>C, rs1800797G>A) was the only SNP to demonstrate substantial survival outcomes, with DLBCL co-dominant model (GG/AG/AA) and recessive model (AA genotype versus combined GG/GA genotype) subjects having worse overall survival. The correlation between polymorphism of the IL-6 gene promoter and the risk of lymphomas, however, shows inconsistent results. A common case-control study conducted by the International Lymphoma Epidemiology (Inter Lymph) consortium in 2006 to examine the relationship between gene polymorphisms and the risk of lymphoma showed that there is no correlation between IL-6 promoter polymorphism (174G>C rs1800795) and the risk of NHL [166].

2.2.1.3 IL-10 gene polymorphism

A number of studies have evaluated the association between specific polymorphisms of IL-10 genes and the risk of non-Hodgkin lymphoma but fewer studies analyzed the impact of IL-10 polymorphisms on the prognosis of patients with DLBCL. There is clear evidence that in hematologic malignancies, immune-regulatory cytokines play a major role. The relationship of seven single nucleotide polymorphisms (SNPs) in two selected cytokines (IL-6 rs1800795 G C, rs1800796 G>C, rs1800797 G>A, IL-10 rs1800871 G A, rs1800872 G>T, rs1800890 A>T, rs1800896 T>C) to risk and overall survival was examined in different studies [167].

2.2.2 Leukemias

2.2.2.1 IL-15, -18 and-1β gene polymorphism in acute lymphoblastic leukemia (ALL)

The proliferation of T, B, and NK cells is encouraged by IL-15, a pro-inflammatory cytokine. Some studies have previously shown that IL-15 can protect hematologic tumors from drug-induced in vitro apoptosis, and high expression of IL-15 is associated with childhood ALL CNS disease. Several minimal residual diseases (MRD)-related IL-15 SNPs have been shown to have a link to increased in vitro transcription/translation efficiency of IL-15. As found by the GWAS scan, there are 5 SNPs in the IL-15 locus that are significantly correlated with childhood ALL therapy response. In the most recent study, polymorphisms of the IL-15 rs10519612 CC genotype have been shown to be associated with adult ALL1 [69]. Polymorphisms of IL-1β (rs16944) and IL-18 (rs1946518) have been shown to predict the prognostic consequences and manage ALL. The impact of the rs1884444 sequence variant on relapse rate and connection of rs10889677 AA genotype with favorable prognostic factors recommend the influence of the investigated SNPs on ALL response to treatment and outcome.

2.2.2.2 Other gene polymorphisms in acute and chronic leukemia (AML and CML) and chronic lymphocytic leukemia

Polymorphisms of IL-1β (rs16944) and IL-18 (rs1946518) have been shown to predict the prognostic consequences and manage ALL care. In cytokine genes, several SNPs have been identified, indicating that certain alleles may lead to alterations in cytokine production [168]. It is, therefore, hypothesized that variants of the cytokine gene can influence the expression of genes and may be associated with leukemia pathogenesis.

The relation between the polymorphism of IFNγ +874T>A (rs2430561) and the risk of CML [169] or earlier, CLL was assessed. Various studies have suggested that the IFNγ+874T>A polymorphism leads to the susceptibility of CML and CLL [168]. No association between TGFβ1 rs1800470 polymorphism and leukemia was identified in earlier studies. Nursal et al., on the other hand, found that variants of the genes TNF-α rs361525, IL-10 (−1082G>A rs1800896, −819C>T rs1800871, −592C>A rs1800872) and TGF-β1 (codon 25) may have a significant association with AML etiopathogenesis [170]. The relation between TNF-α 308G>A, IL-10 (−592T>G, −819T>C, −1082T>C), IFN-γ +874T>A and TGF-β1 (codons 10 and 25) was not identified to confer any risk to CML [169].

2.2.3 Myelomas

2.2.3.1 IL-1gene polymorphism

IL-1 is a potent cytokine that is pro-inflammatory and functions as an endogenous pyrogen. IL-1 has two cytokines formed by two diverse genes, IL-1 alpha, and IL-1β. Cytokine plays a crucial role in the development, differentiation, and function of different immune cells in cells. An important interaction between IL-1 alpha-889C>T and IL-1β-3737C>T and the risk of MM was shown to have an association with polymorphisms of IL-1 [171].

2.2.3.2 IL-6 gene polymorphism

As a proliferative factor for multiple myeloma (MM), IL-6, a cytokine with broad inflammation and immunity functions, is identified. Some myeloma cells and bone marrow stromal cells may produce IL-6 and can restrain apoptosis in myeloma [172]. Previous studies have shown that IL-6 expression can be partly genetically modulated in the promoter region of IL-6 by polymorphisms located at the rs1800795 location. Recent studies have shown that the association of IL-6 rs1800795 polymorphism with MM risk is negligible.

2.2.3.3 TNF-α gene polymorphism

Gene polymorphisms of TNF-α may also be essential for its functional variation. Research has shown that a TNF-α allele (−308) has been expressed at lower levels in MM subjects, indicating that an allele can have a protective effect against disease [173]. However, the GG genotype of TNF-α (−238) was shown to be correlated to early progression in MM in another study [174].

2.2.3.4 IL -12 gene polymorphism

IL-12 is a cytokine that stimulates immunity that is both innate and adaptive. This induces cytotoxicity of Th1 cells and has been shown to have effective immunomodulatory and anti-tumor activities [175]. However, while IL-12 is an inflammatory cytokine, protection from neoplastic disease appears to be the prevalent activity of the cytokine in this case. There is no clear link between IL-12 (rs1801131) (A>C), polymorphism, and the risk of MM in recent studies [176] (Table 2).

ActivityCancers typeCytokinesPolymorphismsAssociationAlleleReferences
Pro-inflammatoryHematological cancersLymphomasHLIL-1+4345 T>GSignificant−TG
IL-6−174 G>CSignificant174 GC[177]
TNF-α−863/308 C>A/G>ANot significant−863/308 CA,GA[177]
NHLIL-6rs1800795 G> CNot significant−174GC[166]
rs1800796 G>CSignificant−598GA
TNF-αrs1800629 G>CSignificant−308GA[157]
TNF-βrs909253 A>GSignificant−252AG[178]
IL-4rs2243250T>GSignificant−1098TG[178]
IL-5rs2069812 C>TNot significant−745CT
LeukemiasALLIL-1βrs16944 G>ASignificantTT[179]
IL-18rs1946518SignificantTT[180]
IL-15rs10519612SignificantCC[181]
IL-6174G/CSignificantrs1800795 GC[182]
IL-4590C/TNot significantrs2243250 CT[170, 183]
AML, CML, CLLIL-1α889C>T-Significant889CT
IL-1β3737C>TSignificant3737CT[184]
IL-2rs1800795 G>CSignificant174/52GC[185]
IL-6rs1801131C>G rs361525G>ANot SignificantCG[184]
TNF-αrs2430561T>ASignificant-308/238 GA[186]
IFN-γ889C>T-Significant+874T TA
MyelomasIL-1 α889C>THighly significantCT[187]
IL-1β3737C>THighly significantCT[187]
IL-6174/52G>CSignificantrs1800795 GC[188]
IL-12rs1801131 A>CNot significantrs1801131AC[185]
TNF-α−308/238 G>ASignificantrs1800629-308/238 GG[189]

Table 2.

A meta-data of different pro-inflammatory cytokine polymorphisms and their association in various liquid cancers.

Advertisement

3. Anti-inflammatory cytokine gene polymprphisms

3.1 Solid tumors

3.1.1 Breast cancer

3.1.1.1 IL-1β gene polymorphism

A variety of cell types, including monocytes, macrophages, and epithelial cells, are formed by interleukin-1β (IL-1β) belonging to the IL-1 family and have multiple biological effects [190]. IL-1β induces the expression of pro-inflammatory genes that may play a key role in the early stages of carcinogenesis, such as cyclooxygenase type 2, inducible nitric oxide synthase, and other cytokines/chemokines. A number of studies have documented the association of IL-1β polymorphisms with the risk of breast cancer, establishing the important role of IL-1β in its development. The findings are contradictory due to the relatively small sample size of the study. Ito et al. [191] first recorded that rs1143627 is significantly correlated with the risk of breast cancer (CC vs. TT: OR = 1.82). The correlation in the Chinese population was followed by another case-control analysis by Liu et al. (CC vs. TT: adjusted OR=1.72). All the studies for rs1143627 included in the meta-analysis were conducted in Asian populations, suggesting that rs1143627 could contribute to the risk of breast cancer, especially in Asians [34].

3.1.1.2 TGFβ -1 gene polymorphism

A multifunctional cytokine that is important for maintaining homeostasis involving bone and muscle differentiation, immune response, and tumor suppression is the transforming growth factor beta-1(TGFβ-1). Increased TGFβ-1 development occurs in different tumor types and is associated with tumor grade severity [192]. There is evidence that when the proliferative inhibition effect of the TGFβ-1 signaling pathway has been overridden by other oncogenic mutations, TGFβ-1 functions as a suppressor of tumor initiation but as a promoter of tumor progression [193]. The 29T/C transformation that generates a Leu10Pro replacement in the TGFβ-1 precursor signal peptide has been reported to be associated with TGFβ-1 secretion of the protein and thus may have altered the risk of breast cancer. In the published clinical trials, Dunning et al. [194] concluded that in studies (20,837 patients and 22,879 controls) from European countries, TGFβ-129T/C T polymorphism was strongly correlated with breast cancer risk (C/C versus T-carrier, 1.21; 95% CI 1.05–1.37); Hishida et al. [195] found that the C/C genotype was substantially correlated with a decreased risk of breast cancer relative to the T/T genotype (OR = 0.45, 0.20–0.98); polymorphisms are not likely to be associated with major increases in the overall risk of breast cancer among Caucasian women in the promoter region of TGFβ-1. Another meta-analysis review indicated that the 29T/C polymorphism did not contribute to breast cancer risk in both recessive, dominant, and other genetic models [196].

3.1.2 Hepato-cellular carcinoma

3.1.2.1 IL-10 gene polymorphism

During chronic viral infections, IL-10, an immune suppressor cytokine, plays a vital role in the weakened host immune response, where its secretion is stated to be genetically regulated [197]. The most studied SNPs that have an influence on IL-10.1 development are IL-10 (−1082G/A, −819T/C, and −592A/C). The reports suggested that −1082 G allele and GCC haplotype are correlated with high IL-10 expression, where less expression association is shown by ATA haplotype. On the other hand, studies have shown that IL-10GCC and IL-10 haplotype AAGCC (−3575, −2763, −1082, −819, and −592) are associated with lower IL-10 haplotype concentration [198].

3.1.2.2 TGF-β1 gene polymorphism

In addition to +869T/C (Codon 10), +915G/C (Codon 25) and +788C/T (codon 263), TGF-β1 polymorphisms are identified in TGF-β1-988C/A, -800G/A, -509C/T and in insertion/deletion [199, 200]. Several SNPs are identified in TGF-β1 (+869T/C and +915G/C) [201] while TGF-β1 +869T allele was found to be correlated with high TGF-β11 expression [202].

3.1.3 Gastric cancer

3.1.3.1 IL-4 gene polymorphism

IL-4 is also recognized as a pro-inflammatory cytokine suppressor and an enhancer of anti-inflammatory cytokine synthesis [203]. The polymorphism of the promoter IL4-590C/T (rs2243250) is widely studied because the mutant T allele has been documented to increase IL-4 expansion compared to the C allele [204]. In contrast with the TT genotype, Wu et al. found an increased risk of developing diffuse form and cardiac GC for the CT/CC genotype in 2003. It is also proposed that lower IL-4 levels in the gastric mucosa favor the growth of GC. The genetic polymorphism IL4-168T/C (rs2070874) tested by Wu et al. [205] stated that a reduced GC risk among the Chinese population was correlated with mutant C allele.

3.1.3.2 IL-10 gene polymorphism

As an immune suppressor and anti-inflammatory mediator, IL-10 typically acts. From the transcriptional start site, there are 3 functional promoters SNPs at-1082 (A to G, rs1800896), -819 (C to T, rs1800871), and -592 (A to C, rs1800872) pairs in the IL10 locus. Studies of IL10-1082A/G, IL10-592A/C, and IL10-819C/T polymorphisms indicate the absence of major discrepancies in the distribution of genotypes between GC patients and healthy controls [206, 207, 208].

3.1.4 Prostate cancer (PCa)

3.1.4.1 IL-10 gene polymorphism

The most studied cytokine for PCa risk is IL-10. Controversial findings have emerged in this regard, where various studies show conflicting results and the latest meta-analysis showed that IL-10 (rs1800871) polymorphism was not correlated with PCa danger [209]. On the contrary, another meta-analysis indicated that polymorphic variants of IL-10 (rs1800871) and IL-10 (rs1800872) reinforce the idea that these may be moderately correlated with advanced PCa and thus affect disease progression [210]. The 2011 study by Shao et al found no substantial evidence suggesting variations in allele frequency or genotype distribution between PCa patients and control subjects for any of the three SNPs IL-10-1082 A>G, -819 C>T and -592 C>A.

3.1.5 Esophageal cancer

3.1.5.1 IL-1β gene polymorphism

In cell proliferation and apoptosis, interleukin 1β (IL1β) is involved which is an essential mediator of inflammatory response. IL1β induces COX-2 expression, inducible synthase nitric oxide, and other cytokines/chemokines, which can function significantly in the early stages of carcinogenesis. Many cancers such as gastric cancer and inflammatory bowel disease are associated with IL1β polymorphisms [211]. One of the polymorphisms associated with IL1β levels in the promoter area is rs16944 G>A [212]. According to Liang et al., IL1ß rs16944 GA variant heterozygotes had a substantially decreased chance of ESCC rather than IL1ß rs16944 AA homozygotes, in line with the Thai population study showing that IL1ß rs16944 GG genotype was found to be more common in patients with gastric cancer [213, 214]. However, minimal studies have been performed that can provide us with clear proof of the functionality in the risk of ESCC of IL1ß rs16944 G>A polymorphism.

3.1.5.2 TGF-β1 gene polymorphism

Two main components of TGF-β signaling that play an important role in carcinogenesis are the transforming growth factor β1 (TGF-β1) and its TGF-β1RII receptor. Several functional polymorphisms were observed in TGF β1 and TGF β1 RII and were correlated with elevated TGF-β1 serum or plasma levels and increased TGFβ1RIII transcription activity. Epidemiological evidence has shown that two transforming growth factor-beta 1 (TGF-β1) gene polymorphisms (namely rs1800468G>A and rs1800471G>C) could be involved in the production of cancer. Their function in the carcinogenic process of esophageal squamous cell carcinoma (ESCC) has, however, been less well described. In recent studies, two polymorphisms of this gene (namely rs1800468G>A and rs1800471G>C) have been shown to be associated with TGF-β1 dysfunction and increased tumor risk [215, 216]. Several evidence indicates that ESCC risk may be increased by the genotypes with rs1800471 C alleles and rs1800471G>C polymorphism may play an important role in ESCCC tumorigenesis [217]. There were some drawbacks to this analysis, however. In contrast to the above findings, several studies have shown that RS#1800468 polymorphism is not associated with an increased risk of ESCC, but with a reduction in the survival of this tumor, consistent with other studies of various cancers [216, 218].

3.1.6 Pancreatic cancer

3.1.6.1 TGF-β1 gene polymorphism

TGF-β functions not only as a powerful epithelial, endothelial and hematopoietic cell proliferation inhibitor but also acts as an effective pro-inflammatory cytokinetic cell proliferator. In cellular proliferation, angiogenesis, differentiation, migration, and apoptosis, TGF-β pathway has important roles. TGF-β expression levels have been reported to correlate with the period of postoperative survival in various malignancies [219]. A study by Zhang et al. [220] showed the TT genotype to be more common in patients with leakage of pancreatic anastomosis in TGF-β. In addition, TT genotype cases had an increased level of bilio-digestive anastomosis leakage.

3.1.7 Bladder cancer

3.1.7.1 IL-6 gene polymorphism

Interleukin-6 (IL6), a pleiotropic inflammatory cytokine released by different types of lymphoid/non-lymphoid cells, is important for immune response, survival of cells, proliferation, and apoptosis [113]. In the IL-6 promoter region, different polymorphic variants have been identified that are associated with IL-6 transcription activity [115], IL-6 influences the release of acute-phase proteins in the acute inflammatory response and regulates the anti-inflammatory cytokines, thereby influencing the strength of inflammatory response. IL-6 is active in multiple cancer growth pathways and promotes neo-angiogenesis. IL-6 polymorphism has been shown to modulate changes in its expression to induce cancer risk, like bladder cancer. Association studies of IL-6 gene polymorphisms conducted in India and globally support the risk of CC genotype and C allele in many cancers, especially BC. Fishman et al. have stated that IL-6-174 G/C polymorphic heterogeneity affects transcription and IL-6 protein expression. The variant genotype of IL-6-174 G/C has been shown to be substantially associated with an increased risk of BC but other authors have not identified any risk association. In addition, there was no proven association between IL-6-572 G/C, -596 A/G polymorphisms, and BC susceptibility [117].

3.1.7.2 IL-4 gene polymorphism

Interleukin-4 (IL-4), an anti-inflammatory cytokine produced mainly by activated CD4+T cells, plays a significant role in the production of Th2 to examine and destroy distorted cells and eradicate extracellular pathogens [221]. The risk conferred by IL-4-590 C/T polymorphism in different cancers has been substantiated by studies from different quarters [222]. In order to modulate the incidence of BC, a polymorphic variance of IL-4-590 C/T was found and another analysis similarly observed a substantial difference in variant allele distribution between cases and controls [223].

3.1.7.3 TGF- β gene polymorphism

TGF-β1 is the most abundant type of TGF-β, which includes numerous polymorphic variants that regulate the expression of TGF-β1 proteins. The risk association of TGF-β1 polymorphisms in a number of cancers has been verified. TGF-β1 was shown to be associated with the possibility of BCC in 41 SNPs. In comparison, 3 polymorphisms in TGF-β1 and 4 in the TGFβR1 gene showed no correlation yet another study reported comparable outcomes of negligible interaction with BC [210]. Gautam et al., on the other hand, observed an important association between TGF-β1 c.29 C/T and the risk of BCF [118].

3.1.8 Gliomas

3.1.8.1 TGF-β gene polymorphism

TGF-β functions as an oncogenic factor that contributes to the growth and invasion of cells and lowers host tumor immune responses [224]. The −509C/T TGF-β1 gene polymorphism can theoretically control the transcription of TGF-β1. The 869T/C polymorphism may have decreased cancer survival in patients carrying the −509C/T T allele and in patients with the 869T/C C allele, as both of these alleles are associated with elevated levels of TGF-β1. In comparison, TT genotype-carrying glioma patients and CC genotype patients have longer average survival. Therefore, in patients with glioma, the TT genotype of the −509C/T polymorphism and the CC genotype of the 869T/C polymorphism have the ability to be used as predictors of improved survival.

3.1.8.2 IL-10 gene polymorphism

IL-10 has pleiotropic effects on inflammation and immunoregulation and can facilitate carcinogenesis [54]. In gliomas, several IL-10 gene polymorphisms have been shown to influence disease susceptibility and severity [134]. The association of the IL-10 rs1800871 C/T genotype with increased survival in low-grade glioma patients was confirmed by Mingjun et al. [225]. Another research found an important protective interaction of variant IL-10 (−1082A/G) G allele inst glioma whereby It has been shown to induce an increase in the development of IL-10 In 2012, Tanikawa et al. [226] noted that high serum IL-10 levels increased tumor-specific immune response and decreased tumor growth. The exact role of IL-10 in glioma is unclear; further studies are needed to elucidate the mechanisms underlying the relationship between IL-10 polymorphisms and the prognosis of glioma patients (Table 3).

ActivityCancer typeCytokinesPolymorphismsAssociationAlleleReferences
Anti-inflammatorySolid tumorsBreastIL-1β−511C>THigh risk−511 CC[23]
TGFβ1Codon 29T>CSignificant risk29TT[194]
No[196]
HCCIL-1β511T>CRisk Factor511T>C[49]
IL1-RNIntron 286bpRisk FactorIntron 286bp[47]
Allele 2 VNTRAllele 2 VNTR[227]
TGF-β1509C>TRisk Factor509C>T[228]
IL-42590C>TRisk Factor2590C>T[229]
233C>TRisk Factor233C>T[230]
IL-6−597G>ANo−597G>A[231, 232]
−572G>CRisk factor−572G>C[54]
−174G>CRisk factor−174G>C[233]
IL-10−1082G>ARisk factor−1082G>A[156]
−819T>CRisk factor−819T>C[156]
GastricIL-4IL-4 –590C/TAssoT allele[234]
IL-4–168T/CNeg assocC allele[234]
IL-10IL-10–1082 A/GLack of assnAA[206]
IL-10–819 C/TLack of assnTC[235]
IL-10–592 A/CLack of assnAC[207]
ProstateIL-10IL-10 (rs1800871)Not favouring[209]
IL-10rs1800871)/(rs1800872)Favouring[210]
IL-10 −1082 A/GNo association[210]
−819 C/TNo association
−592 C/ANo association
EsophagealIL-1βrs16944 G>AProtectivers16944 GA[213]
TGFβ1rs1800468G>A +915G>CSignificant riskrs1800468AA[216]
Significant risk+915 CC[227, 236]
PancreaticTGFβTGF-βAssociationTT genotype[237]
GliomaTGFβ1−509C/T,869T/CBetter prognosis−509TT,869CC[238]
IL-10−819 C>TImproved survival−819 CC[225]
−1082A/GProtective role−1082GG[239]

Table 3.

A meta-data of different anti-inflammatory cytokine polymorphisms and their association in various solid tumors.

3.1.9 Lymphomas

Studies regarding the role of interleukin gene polymorphisms in Hodgkin’s Lymphoma have revealed anti-inflammatory functions of IL-1R and IL-10 [240, 241].

3.1.10 Leukemias

3.1.10.1 IL-10 gene polymorphism in ALL

In cancer growth and progression, interleukin-10, a pleiotropic cytokine serves as an immune stimulant factor. In ALL IL-10 SNP rs1800896 was correlated with the progression of the disease and also affect the cytokine expression. Many experiments, however, studied IL-10 rs100896 T/C polymorphism to determine the relevant relationship with susceptibility and prognosis.

3.1.11 Myelomas

3.1.11.1 IL-1Ra gene polymorphism

In response to similar stimuli that induce IL-1 release, the IL1-Receptor Antagonist (IL-lRa) is developed and released. According to Liang Zheng et al., carriers of IL1B rs16944 GA variant heterozygotes had a slightly reduced chance of multiple myeloma compared to IL-1B rs16944 AA homozygotes, had a slightly reduced [213].

3.1.11.2 IL-10gene polymorphism

IL-10 suppresses immune responses as an immunosuppressive cytokine by functioning simultaneously on the innate and the adaptive immune system. IL-10 can also inhibit pro-inflammatory cytokine secretion, antigen presentation, and cell growth. In the pathogenesis of hematological disorders, both IL-10 and IL-10R SNPs are involved. IL-10-592G/A and IL-10-1,082G>A SNPs, -592(C) or -1,082(G) are considered to be correlated with a strong IL-10 expression and a low -592(A) or -1,082(G) expression (A). However, studies have shown, that the C allele of IL10-592 has no clear interaction with MM [242].

3.1.11.3 TGF-β gene polymorphism

TGF-β is an active regulatory cytokine with divergent hemopoietic cell effects. Usually, TGF-β serves to reduce the release of immunoglobulin by B cells. Studies have shown that TGFβ1 genotypes with rs1800471 C alleles may raise the risk of MM and rs1800471G>C polymorphism may play a significant role in MMM tumorigenesis [217].

Advertisement

4. Conclusion

Germline constitutional mutations or changes in the polymorphic sequence are key instruments used as molecular tools to predict the susceptibility of a person to risk numerous cancers. Cytokines are also thought to be essential molecules that have a dual face due to pro-inflammatory as well as anti-inflammatory mechanisms in carcinogenesis. These molecules regulate the development of cancer when involved in the activation of the immune response, while their recurrent inflammatory reaction will envisage the development of malignancy and tumor formation. Any differences in the polymorphic sequence of cytokine genes cause heterogeneity in their expression to deregulate genetic regulation, which eventually makes a person vulnerable to multiple cancers. As stated in the t segment, considerable evidence that links cytokine gene susceptibility polymorphism with various cancers has materialized their position as important bimolecular genetic cancer susceptibility and prognosis determinants. In studying candidate genes implicated in particular pathways for specific cancer; we conclude there is a desperate need for more case-control association studies involving additional variables haplotypic gene-gene associations and clinical-pathological features to unearth factual association for cancer. Because cytokine polymorphisms play a crucial role in immunological processes in which a rich supply of inflammatory cytokines are produced by various intensities of inflammation and cancer microenvironment, research into genetic polymorphisms in cytokine genes and their reaction to chemo-radiotherapy is believed to assist patients with cancer treatment and management.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144(5):646-674
  2. 2. Crusz SM, Balkwill FR. Inflammation and cancer: Advances and new agents. Nature Reviews. Clinical Oncology. 2015;12:584-596
  3. 3. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers. 2014;6:1670-1690
  4. 4. Wu Y, Zhou B. TNF-α/NF-κB/Snail pathway in cancer cell migration and invasion. British Journal of Cancer. 2010;102:639-644
  5. 5. Allavena P, Mantovani A. Immunology in the clinic review series; focus on cancer: Tumour-associated macrophages: Undisputed stars of the inflammatory tumour microenvironment. Clinical and Experimental Immunology. 2012;167(2):195-205
  6. 6. Smith KA. Toward a molecular understanding of adaptive immunity: A chronology–part II. Frontiers in Immunology. 2012;3:364
  7. 7. Dantzer R, Meagher MW, Cleeland CS. Translational approaches to treatment-induced symptoms in cancer patients. Nature Reviews. Clinical Oncology. 2012;9(7):414
  8. 8. Swiatek BJ. Is interleukin-10 gene polymorphism a predictive marker in HCV infection? Cytokine and Growth Factor Reviews. 2012;23(1–2):47-59
  9. 9. Candido J, Hagemann T. Cancer-related inflammation. Journal of Clinical Immunology. 2013;33(1):79-84
  10. 10. Eil R, Vodnala SK, Clever D, Klebanoff CA, Sukumar M, Pan JH, et al. Ionic immune suppression within the tumour microenvironment limits T cell effector function. Nature. 2016;537(7621):539-543
  11. 11. Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, Pérez-Gracia JL, et al. Cytokines in clinical cancer immunotherapy. British Journal of Cancer. 2019;120(1):6-15
  12. 12. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883-899
  13. 13. Chen Z, Bouamar R, Van Schaik RH, De Fijter JW, Hartmann A, Zeier M, et al. Genetic polymorphisms in IL-2, IL-10, TGF-β1, and IL-2 RB and acute rejection in renal transplant patients. Clinical Transplantation. 2014;28(6):649-655
  14. 14. Yang Y, Luo C, Feng R, Bi S. The TNF-α, IL-1B and IL-10 polymorphisms and risk for hepatocellular carcinoma: A metaanalysis. Journal of Cancer Research and Clinical Oncology. 2011;137(6):947-952
  15. 15. Stowe RP, Peek MK, Cutchin MP, Goodwin JS. Plasma cytokine levels in a population-based study: Relation to age and ethnicity. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences. 2010;65(4):429-433
  16. 16. Chou IC, Lin WD, Wang CH, Tsai CH, Li TC, Tsai FJ. Interleukin (IL)-1β, IL-1 receptor antagonist, IL-6, IL-8, IL-10, and tumor necrosis factor α gene polymorphisms in patients with febrile seizures. Journal of Clinical Laboratory Analysis. 2010;24(3):154-159
  17. 17. Bandil K, Singhal P, Sharma U, Hussain S, Basu S, Parashari A, et al. Impacts of TNF-LTA SNPs/haplotypes and lifestyle factors on oral carcinoma in an Indian population. Molecular Diagnosis & Therapy. 2016;20(5):469-480
  18. 18. Skibola CF, Bracci PM, Nieters A, Brooks-Wilson A, De Sanjosé S, Hughes AM, et al. Tumor necrosis factor (TNF) and lymphotoxin-α (LTA) polymorphisms and risk of non-Hodgkin lymphoma in the InterLymph Consortium. American Journal of Epidemiology. 2010;171(3):267-276
  19. 19. Chiurillo MA. Role of gene polymorphisms in gastric cancer and its precursor lesions: Current knowledge and perspectives in Latin American countries. World Journal of Gastroenterology. 2014;20(16):4503
  20. 20. Hong JB, Zuo W, Wang AJ, Lu NH. Helicobacter pylori infection synergistic with IL-1β gene polymorphisms potentially contributes to the carcinogenesis of gastric cancer. International Journal of Medical Sciences. 2016;13(4):298
  21. 21. Haura EB, Turkson J, Jove R. Mechanisms of disease: Insights into the emerging role of signal transducers and activators of transcription in cancer. Nature Clinical Practice Oncology. 2005;2:315-324
  22. 22. Berger, Franklin G. “The interleukin-6 gene: a susceptibility factor that may contribute to racial and ethnic disparities in breast cancer mortality.” Breast cancer research and treatment. 2004;88(3):281-285.
  23. 23. Ito LS, Iwata H, Hamajima N, Saito T, et al. Significant reduction in breast cancer risk for Japanese women with interleukin 1B–31 CT/TT relative to CC genotype. Japanese Journal of Clinical Oncology. 2002;32:398-402
  24. 24. DeMichele A, Martin AM, Mick R, et al. Interleukin-6-174 G to C polymorphism is associated with improved outcome in high-risk breast cancer. Cancer Research. 2003;63:8051-8056
  25. 25. Iacopetta B, Grieu F, Joseph D. The _174 G/C gene polymorphism in interleukin-6 is associated with an aggressive breast cancer phenotype. British Journal of Cancer. 2004;90:419-422
  26. 26. Blankenstein T. The role of tumor 53 stroma in the interaction between tumor 54 and immune system. Current Opinion in 55 Immunology. 2005;17:180-186
  27. 27. Broser EEG. 2016;85. Available from: http://www.ensembl.org/index.html
  28. 28. Giordani L, Bruzzi P, Lasalandra C, Quaranta M, et al. Polymorphisms of Interleukin-10 and tumour necrosis factor-a gene promoter and breast cancer risk. Clinical Chemistry. 2003;49:1664-1667
  29. 29. Langsenlehner U, Krippl P, Renner W, Yazdani-Biuki B, Eder T, Koppel H, et al. Interleukin-10 promoter polymorphism is associated with decreased breast cancer risk. Breast Cancer Research and Treatment. 2005;90:113-115
  30. 30. Karakus N, Kara N, Ulusoy AN, Ozaslan C, Bek Y. Tumor necrosis factor alpha and beta and interferon gamma gene polymorphisms in Turkish breast cancer patients. DNA and Cell Biology. 2011;30(6):371-377
  31. 31. He JR, Chen LJ, Su Y, Cen YL, Tang LY, Yu DD, et al. Joint effects of Epstein-Barr virus and polymorphisms in interleukin-10 and interferon-gamma on breast cancer risk. The Journal of Infectious Diseases. 2012;205(1):64-71
  32. 32. Erdei E, Kang H, Meisner A, White K, et al. Polymorphisms in cytokine genes and serum cytokine levels among New Mexican women with and without breast cancer. Cytokine. 2010;51(1):18-24
  33. 33. Luo JL, Maeda S, Hsu LC, Yagita H, Karin M. Inhibition of NF-kB in cancer cells converts inflammation-induced tumor growth mediated by TNF-α to TRAIL-mediated tumor regression. Cancer Cell. 2004;6:297-305
  34. 34. Liu J, Zhai X, Jin G, Hu Z, et al. Functional variants in the promoter of interleukin-1beta are associated with an increased risk of breast cancer: A case-control analysis in a Chinese population. International Journal of Cancer. 2006;118:2554-2558
  35. 35. Mi YY, Yu QQ, Xu B, Zhang LF, et al. Interferon gamma +874 T/A polymorphism contributes to cancer susceptibility: A meta-analysis based on 17 case-control studies. Molecular Biology Reports. 2011;38(7):4461-4467
  36. 36. Correlations of IFN-γ genetic polymorphisms with susceptibility to breast cancer: a meta-analysis. Tumour Biol. 2015;36(9):7307. DOI: 10.1007/s13277-015-3699-1. PMID: 26281989
  37. 37. Mocellin S, Rossi CR, Pilati P, Nitti D. Tumor necrosis factor, cancer and anticancer therapy. Cytokine & Growth Factor Reviews. 2005;16:35-53
  38. 38. Neben K et al. Polymorphisms of the tumor necrosis factor-alpha gene promoter predict for outcome after thalidomide therapy in relapsed and refractory multiple myeloma. Blood. 2002;100:2263-2265
  39. 39. Wilson AG, Symons JA, McDowell TL, McDevitt HO, et al. Effects of a polymorphism in the human tumor necrosis factor alpha promoter on transcriptional activation. Proceedings of the National Academy of Sciences of the United States of America. 1997;94:3195-3199
  40. 40. Mestiri S, Bouaouina N, Ahmed SB, et al. Genetic variation in the tumor necrosis factor-alpha promoter region and in the stress protein hsp 70-2: Susceptibility and prognostic implications in breast carcinoma. Cancer. 2001;91(4):672-678
  41. 41. Azmy IA, Balasubramanion SP, Wilson AG, et al. Role of tumor necrosis factor gene polymorphisms (–308 and –238) in breast cancer susceptibility and severity. Breast Cancer Research. 2004;6(4):395-400
  42. 42. Smith KC, Bateman AC, Fussell HM, Howell WM. Cytokine Gene polymorphisms and breast cancer susceptibility and prognosis. European Journal of Immunogenetics. 2004;31(4):167-173
  43. 43. Scola L, Vaglica M, Crivello A, et al. Cytokine gene polymorphisms and breast cancer susceptibility. Annals. New York Academy of Sciences. 2006;1 89:104-109
  44. 44. Wang J et al. Tumour necrosis factor alpha -308G/A polymorphism and risk of the four most frequent cancers: A meta-analysis. International Journal of Immunogenetics. 2011;38:311-320
  45. 45. Jeng JE, Tsai HR, Chuang LY, Tsai JF, et al. Independent and additive interactive effects among tumor necrosis factor-alpha polymorphisms, substance use habits, and chronic hepatitis B and hepatitis C virus infection on risk for hepatocellular carcinoma. Medicine (Baltimore). 2009;88:349-357. DOI: 10.1097/MD.0b013e3181c10477
  46. 46. Skerrett DL, Moore EM, Bernstein DS, Vahdat L. Cytokine genotype polymorphisms in breast carcinoma: Associations of TGF-beta 1 with relapse. Cancer Investigation. 2005;23:208-214
  47. 47. Chen CC, Yang SY, Liu CJ, Lin CL, et al. Association of cytokine and DNA repair gene polymorphisms with hepatitis B-related hepatocellular carcinoma. International Journal of Epidemiology. 2005;34:1310-1318
  48. 48. El-Omar EM, Carrington M, Chow WH, McColl KE, et al. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000;404:398-402
  49. 49. Hirankarn N, Kimkong I, Kummee P, Tangkijvanich P, et al. Interleukin-1beta gene polymorphism associated with hepatocellular carcinoma in hepatitis B virus infection. World Journal of Gastroenterology. 2006;12:776-779
  50. 50. Wang Y, Kato N, Hoshida Y, Yoshida H, et al. Interleukin-1beta gene polymorphisms associated with hepatocellular carcinoma in hepatitis C virus infection. Hepatology. 2003;37:65-71
  51. 51. Akkiz H, Bayram S, Bekar A, Ozdil B, Sümbül AT, et al. G-308A TNF-alpha polymorphism is associated with an increased risk of hepatocellular carcinoma in the Turkish population: Case-control study. Cancer Epidemiology. 2009;33:261-264. DOI: 10.1016/j.canep.2009.06.001
  52. 52. Hösel M, Quasdorff M, Wiegmann K, Webb D, et al. Not interferon, but interleukin-6 controls early gene expression in hepatitis B virus infection. Hepatology. 2009;50:1773-1782. DOI: 10.1002/hep.23226
  53. 53. Ataseven H, Bahcecioglu IH, Kuzu N, Yalniz M, et al. The levels of ghrelin, leptin, TNF-alpha, and IL-6 in liver cirrhosis and hepatocellular carcinoma due to HBV and HDV infection. Mediators of Inflammation. 2006;2006:78380
  54. 54. Park BL, Lee HS, Kim YJ, Kim JY, et al. Association between interleukin 6 promoter variants and chronic hepatitis B progression. Experimental & Molecular Medicine. 2003;35:76-82
  55. 55. Xue H, Lin B, An J, Zhu Y, et al. Interleukin-10–819 promoter polymorphism in association with gastric cancer risk. BMC Cancer. 2012;12:102
  56. 56. Williams TM, Eisenberg L, Burlein JE, Norris CA, Pancer S, Yao D, et al. Two regions within the human IL-2 gene promoter are important for inducible IL-2 expression. The Journal of Immunology. 1988;141(2):662-666
  57. 57. Wu GY, Hasenberg T, Magdeburg R, Bonninghoff R, et al. Association between EGF, TGF-β1, VEGF gene polymorphism and colorectal cancer. World Journal of Surgery. 2009;33(1):124-129
  58. 58. Togawa S, Joh T, Itoh M, Katsuda N, et al. Interleukin-2 gene polymorphisms associated with increased risk of gastric atrophy from Helicobacter pylori infection. Helicobacter. 2005;10:172-178
  59. 59. Savage SA, Abnet CC, Haque K, Mark SD, et al. Polymorphisms in interleukin -2, -6, and -10 are not associated with gastric cardia or esophageal cancer in a high-risk Chinese population. Cancer Epidemiology, Biomarkers & Prevention. 2004;13:1547-1549
  60. 60. Matsuo K, Oka M, Murase K, Soda H, Isomoto H, Takeshima F, et al. Expression of interleukin 6 and its receptor in human gastric and colorectal cancers. Journal of International Medical Research. 2003;31(2):69-75
  61. 61. Putoczki TL, Thiem S, Loving A, Busuttil RA, Wilson NJ, Ziegler PK, et al. Interleukin-11 is the dominant IL-6 family cytokine during gastrointestinal tumorigenesis and can be targeted therapeutically. Cancer Cell. 2013;24(2):257-271
  62. 62. Brat DJ, Bellail AC, Van Meir EG. The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro-Oncology. 2005;7(2):122-133
  63. 63. Kido S, Kitadai Y, Hattori N, Haruma K, Kido T, Ohta M, et al. Interleukin 8 and vascular endothelial growth factor—Prognostic factors in human gastric carcinomas? European Journal of Cancer. 2001;37(12):1482-1487
  64. 64. Yoshino N, Ishihara S, Rumi MAK, Ortega-Cava CF, Yuki T, Kazumori H, et al. Interleukin-8 regulates expression of Reg protein in Helicobacter pylori-infected gastric mucosa. American Journal of Gastroenterology. 2005;100(10):2157-2166
  65. 65. Hull J, Rowlands K, Lockhart E, Sharland M, Moore C, Hanchard N, et al. Haplotype mapping of the bronchiolitis susceptibility locus near IL8. Human Genetics. 2004;114:272-279
  66. 66. Lu Y, Wang ZD, Shen J, Xu YC. Meta-analysis on the relationship between IL8-251 gene polymorphism and gastric cancer. Zhonghua Yu Fang Yi Xue Za Zhi. 2007;41(suppl):39-42
  67. 67. Kang JM, Kim N, Lee DH, Park JH, Lee MK, Kim JS, et al. The effects of genetic polymorphisms of IL-6, IL-8, and IL-10 on Helicobacter pylori-induced gastroduodenal diseases in Korea. Journal of Clinical Gastroenterology. 2009;43:420-428
  68. 68. Zhang B, Rong G, Wei H, Zhang M, Bi J, Ma L, et al. The prevalence of Th17 cells in patients with gastric cancer. Biochemical and Biophysical Research Communications. 2008;374:533-537
  69. 69. Wu GY, Lu Q, Hasenberg T, Niedergethmann M, et al. Association between EGF, TGF-β1, TNF-α gene polymorphisms and cancer of the pancreatic head. Anticancer Research. 2010;30(12):5257-5261
  70. 70. Noach LA, Bosma NB, Jansen J, Hoek FJ, Van Deventer SJH, Tytgat GNJ. Mucosal tumor necrosis factor-or, interleukin-1/3, and interleukin-8 production in patients with Helicobacter pylori infection. Scandinavian Journal of Gastroenterology. 1994;29(5):425-429
  71. 71. Beales ILP, Calam J. Interleukin 1β and tumour necrosis factor α inhibit acid secretion in cultured rabbit parietal cells by multiple pathways. Gut. 1998;42(2):227-234
  72. 72. Forones NM, Mandowsky SV, Lourenço LG. Serum levels of interleukin-2 and tumor necrosis factor-alpha correlate to tumor progression in gastric cancer. Hepato-Gastroenterology. 2001;48(40):1199-1201
  73. 73. Beales IL, Calam J. Pathogenic mechanisms in Helicobacter pylori infection. Hospital Medicine (London, England: 1998). 1998;59(3):186-190
  74. 74. El-Omar EM, Rabkin CS, Gammon MD, Vaughan TL, Risch HA, Schoenberg JB, et al. Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology. 2003;124(5):1193-1201
  75. 75. Rad R, Dossumbekova A, Neu B, Lang R, Bauer S, Saur D, et al. Cytokine gene polymorphisms influence mucosal cytokine expression, gastric inflammation, and host specific colonisation during Helicobacter pylori infection. Gut. 2004;53(8):1082-1089
  76. 76. Gorouhi F, Islami F, Bahrami H, Kamangar F. Tumour-necrosis factor-A polymorphisms and gastric cancer risk: A meta-analysis. British Journal of Cancer. 2008;98(8):1443-1451
  77. 77. Xu H, Ding Q, Jiang HW. Genetic polymorphism of interleukin-1A (IL-1A), IL-1B, and IL-1 receptor antagonist (IL-1RN) and prostate cancer risk. Asian Pacific Journal of Cancer Prevention. 2014;15(20):8741-8747
  78. 78. Mandić S, Sudarević B, Marczi S, et al. Interleukin-6 polymorphism and prostate cancer risk in population of Eastern Croatia. Collegium Antropologicum. 2013;37:907-911
  79. 79. Pierce BL, Biggs ML, DeCambre M, et al. C-reactive protein, interleukin-6, and prostate cancer risk in men aged 65 years and older. Cancer Causes & Control. 2009;20:1193-1203
  80. 80. Mandal S, Abebe F, Chaudhary J. 174G/C polymorphism in the interleukin-6 promoter is differently associated with prostate cancer incidence depending on race. Genetics and Molecular Research. 2014;13:139-151
  81. 81. Yang M, Li C, Li M. Association of interleukin-6 (-174 G/C) polymorphism with the prostate cancer risk: A meta-analysis. Biomedical Reports;2:637-643. DOI: 10.3892/Br.2014.300
  82. 82. Grivennikov S, Karin M. Autocrine IL-6 signaling: A key event in tumorigenesis? Cancer Cell. 2008;13:7-9
  83. 83. Nguyen TN, Baaklini S, Koukouikila-Koussounda F, et al. Association of a functional TNF variant with Plasmodium falciparum parasitaemia in a congolese population. Genes and Immunity. 2017;18:152-157
  84. 84. Satoru M, Toshinobu N, Masatomo M, Yudai H, et al. Interleukin-6-634G >C genetic polymorphism is associated with prognosis following surgery for advanced thoracic esophageal squamous cell carcinoma. Digestive Surgery. 2012, 2012;29:194-201
  85. 85. Oka M, Yamamoto K, Takahashi M, Hakozaki M, et al. Relationship between serum levels of interleukin 6, various disease parameters and malnutrition in patients with esophageal squamous cell carcinoma. Cancer Research. 1996;56:2776-2780
  86. 86. Buraczynska M, Jozwiak L, Ksiazek P, Borowicz E, Mierzicki P. Interleukin-6 gene polymorphism and faster progression to endstage renal failure in chronic glomerulonephritis. Translational Research. 2007;150:101-105
  87. 87. Kitamura A, Hasegawa G, Obayashi H, Kamiuchi K, et al. Interleukin-6 polymorphism (-634C/G) in the promotor region and the progression of diabetic nephropathy in type 2 diabetes. Diabetic Medicine. 2002;19:1000-1005
  88. 88. Sansone P, Storci G, Tavolari S, Guarnieri T, et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. The Journal of Clinical Investigation. 2007;117:3988-4002
  89. 89. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. National Review. 2003;3:133-146
  90. 90. Yi PT, Wan LW, Song YL, Jian Z, et al. Associations between polymorphisms in IL-12A, IL-12B, IL-12Rb1, IL-27 gene and serum levels of IL-12p40, IL-27p28 with esophageal cancer. Journal of Cancer Research and Clinical Oncology. 2012;138:1891-1900
  91. 91. Hsieh YY, Chang CC, Tsai CH, Lin CC, Tsai FJ. Interleukin (IL)-12 receptor beta 1 codon 378 G homozygote and allele, but not IL-1 (beta-511 promoter, 3953 exon 5, receptor antagonist), IL-2 114, IL-4-590 intron 3, IL-8 30-UTR 2767, and IL-18 105, are associated with higher susceptibility to leiomyoma. Fertility and Sterility. 2007;87(4):886-895
  92. 92. Imboden M, Nicod L, Nieters A, Glaus E, et al. The common G-allele of interleukin-18 single-nucleotide polymorphism is a genetic risk factor for atopic asthma. The SAPALDIA Cohort Study. Clinical and Experimental Allergy. 2006;36:211-218
  93. 93. Takagawa T, Tamura K, Takeda N, Tomita T, et al. Association between IL-18 gene promoter polymorphisms and inflammatory bowel disease in a Japanese population. Inflammatory Bowel Diseases. 2005;11:1038-1043
  94. 94. Ye SW, Yan L, Yun GL, Hui T, et al. Interleukin-18 gene promoter polymorphisms and the risk of esophageal squamous cell carcinoma. Acta Oncologica. 2007;46:1090-1096
  95. 95. Watanabe M, Tanaka K, Takizawa T, et al. Characterization of a canine tetranucleotide microsatellite marker located in the first intron of the tumor necrosis factor alpha gene. The Journal of Veterinary Medical Science. 2014;76:119-122
  96. 96. Zhu G, Du Q, Wang X, Tang N, She F, Chen Y. TNF-alpha promotes gallbladder cancer cell growth and invasion through autocrine mechanisms. International Journal of Molecular Medicine. 2014;33:1431-1440
  97. 97. Piotrowski P, Wudarski M, Sowinska A, Olesinska M, Jagodzinski PP. TNF-308 G/A polymorphism and risk of systemic lupus erythematosus in the Polish population. Modern Rheumatology. 2015;25:719-723
  98. 98. Hai-feng W, Hong-bo L, Hare A, Apizi PL, et al. p 21 and TNF-α gene polymorphisms and susceptibility of Xinjiang Kazakh esophageal family. Chinese Journal of Cancer Prevention and Treatment. 2014;21:329-332
  99. 99. Deans C, Rose-Zerilli M, Wigmore S, Ross J, et al. Host cytokine genotype is related to adverse prognosis and systemic inflammation in gastro-oesophageal cancer. Annals of Surgical Oncology. 2007;14:329-339
  100. 100. Cui X-B, Wang D-D, Zhang H-Y, Li T-T, et al. Tumor necrosis factor-α gene 308G/A polymorphism is not associated with esophageal squamous cell carcinoma risk in Kazakh patients. International Journal of Clinical and Experimental Pathology. 2015;8:9293
  101. 101. Guo W, Wang N, Li Y, Zhang JH. Polymorphisms in tumor necrosis factor genes and susceptibility to esophageal squamous cell carcinoma and gastric cardiac adenocarcinoma in a population of high incidence region of North China. Chinese Medical Journal. 2005;118:1870-1878
  102. 102. Luo M, Yang Y, Luo D, Liu L, Zhang Y, Xiao F, ... & Luo Z. (2016). Tumor necrosis factor-alpha promoter polymorphism 308 G/A is not significantly associated with esophageal cancer risk: a meta-analysis. Oncotarget. 2016;7(48):79901.
  103. 103. Filipe SO, Bustamante J, Chapgier A, Vogt G, de BL, Feinberg J, et al. Inborn errors of IL-12/23- and IFN-gamma-mediated immunity: Molecular, cellular, and clinical features. Seminars in Immunology. 2006;18:347-361
  104. 104. Du W, Ye W, Chen M, Li D, et al. Association research between polymorphism of IFN-gamma and IL-10, environmental risk factors, and susceptibility to esophageal cancer. Journal of Hygiene Research. 2013;42(5):770-776
  105. 105. Motoyama S, Miura M, Hinai Y, Maruyama K, Usami S, Nakatsu T, ... & Ogawa J. I. (2009). Interferon-gamma 874A>T genetic polymorphism is associated with infectious complications following surgery in patients with thoracic esophageal cancer. Surgery. 2009;146(5):931-938
  106. 106. Chen G, Hohmeier HE, Newgard CB. Expression of the transcription factor STAT-1a in insulinoma cells protects against cytotoxic effects of multiple cytokines. The Journal of Biological Chemistry. 2001;276:766-772
  107. 107. Berkovic MC, Ivkovic TC, Marout J, Zjacić-Rotkvic V, et al. Interleukin 1β gene single-nucleotide polymorphisms and susceptibility to pancreatic neuroendocrine tumors. DNA and cell biology. 2012;31(4):531-536
  108. 108. Hoffman SC, Stanley EM, Darrin Cox E, Craighead N, et al. Association of cytokine polymorphic inheritance and in vitro cytokine production in anti-CD3/CD28-stimulated peripheral blood lymphocytes. Transplantation. 2001;72:1444-1450
  109. 109. Reichert TE, Watkins S, Stanson J, Johnson JT, et al. Endogenous IL-2 in cancer cells: A marker of cellular proliferation. The Journal of Histochemistry and Cytochemistry. 1998;46:603-612
  110. 110. Hofsli E, Thommesen L, Yadetie F, Langaas M, et al. Identification of novel growth-factor responsive genes in neuroendocrine gastrointestinal tumor cells. British Journal of Cancer. 2005;92:1506-1516
  111. 111. Lu PH, Tang Y, Li C, Shen W, et al. Metaanalysis of association of tumor necrosis factor alpha-308 gene promoter polymorphism with gastric cancer. Chinese Journal of Preventive Medicine. 2010;44(3):209-214
  112. 112. Zhou P, Li JP, Zhang C. Polymorphisms of tumor necrosis factor-alpha and breast cancer risk: Appraisal of a recent metaanalysis. Breast Cancer Research and Treatment. 2011;126(1):253-254; author reply 255-256
  113. 113. Kishimoto T. Interleukin-6: From basic science to medicine—40 years in immunology. Annual Review of Immunology. 2005;23:1-21
  114. 114. Tahara E. Growth factors and oncogenes in human gastrointestinal carcinomas. Journal of Cancer Research and Clinical Oncology. 1990;116(2):121-131
  115. 115. Morgan MD, Harper L, Williams J, Savage C. Anti-neutrophil cytoplasm-associated glomerulonephritis. Journal of the American Society of Nephrology. 2006;17(5):1224-1234
  116. 116. Paule B, Terry S, Kheuang L, Soyeux P, Vacherot F, de la Taille A. The NF-κB/IL-6 pathway in metastatic androgen-independent prostate cancer: New therapeutic approaches? World Journal of Urology. 2007;25(5):477-489
  117. 117. Landi S, Moreno V, Gioia-Patricola L, Guino E, Navarro M, de Oca J, et al. Association of common polymorphisms in inflammatory genes interleukin (IL) 6, IL8, tumor necrosis factor α, NFKB1, and peroxisome proliferator-activated receptor γ with colorectal cancer. Cancer Research. 2003;63(13):3560-3566
  118. 118. Gautam KA, Tripathi M, Sankhwar SN, et al. Functional polymorphisms in the IL6 gene promoter and the risk of urinary bladder cancer in India. Cytokine. 2016;77:152-156
  119. 119. Fishman D, Faulds G, Jeffery R, et al. The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. The Journal of Clinical Investigation. 1998;102:1369-1376
  120. 120. Leibovici D, Grossman HB, Dinney CP, Millikan RE, Lerner S, Wang Y, et al. Polymorphisms in inflammation genes and bladder cancer: From initiation to recurrence, progression, and survival. Journal of Clinical Oncology. 2005;23(24):5746-5756
  121. 121. Aben KK, Witjes JA, Schoenberg MP, Hulsbergen-van de Kaa C, Verbeek AL, Kiemeney LA. Familial aggregation of urothelial cell carcinoma. International Journal of Cancer. 2002;98:274-278. DOI: 10.1002/ijc.10191
  122. 122. Ebadi N, Jahed M, Mivehchi M, et al. Interleukin-12 and interleukin-6 gene polymorphisms and risk of bladder cancer in the Iranian population. Asian Pacific Journal of Cancer Prevention. 2014;1518:7869-7873
  123. 123. Ahirwar D, Kesarwani P, Manchanda PK, et al. Anti- and proinflammatory cytokine gene polymorphism and genetic predisposition: Association with smoking, tumor stage and grade, and bacillus Calmette-Guérin immunotherapy in bladder cancer. Cancer Genetics and Cytogenetics. 2008;184:1-8
  124. 124. Mcdermott MF. TNF and TNFR biology in health and disease. Cellular AND Molecular Biology-Paris-Wegmann. 2001;47(4):619-636
  125. 125. Kohaar I, Tiwari P, Kumar R, Nasare V, Thakur N, Das BC, et al. Association of single nucleotide polymorphisms (SNPs) in TNF-LTA locus with breast cancer risk in Indian population. Breast Cancer Research and Treatment. 2009;114(2):347-355
  126. 126. Sáenz-López P, Carretero R, Cózar JM, Romero JM, Canton J, Vilchez JR, et al. Genetic polymorphisms of RANTES, IL1-A, MCP-1 and TNF-A genes in patients with prostate cancer. BMC Cancer. 2008;8(1):1-8
  127. 127. Marsh H, Haldar N, Bunce M, et al. Polymorphisms in tumour necrosis factor (TNF) are associated with risk of bladder cancer and grade of tumour at presentation. British Journal of Cancer. 2003;89:1096-1101. DOI: 10.1038/sj.bjc.6601165
  128. 128. Ahirwar DK, Mandhani A, Dharaskar A, Kesarwani P, & Mittal RD. Association of tumour necrosis factor-α gene (T-1031C, C-863A, and C-857T) polymorphisms with bladder cancer susceptibility and outcome after bacille Calmette-Guérin immunotherapy. BJU international. 2009;104(6):867-873.
  129. 129. Kim EJ, Jeong P, Quan C, Kim J, Bae SC, Yoon SJ, et al. Genotypes of TNF-α, VEGF, hOGG1, GSTM1, and GSTT1: Useful determinants for clinical outcome of bladder cancer. Urology. 2005;65(1):70-75
  130. 130. Nonomura N, Tokizane T, Nakayama M, Inoue H, Nishimura K, Muramatsu M, et al. Possible correlation between polymorphism in the tumor necrosis factor-beta gene and the clinicopathological features of bladder cancer in Japanese patients. International Journal of Urology. 2006;13(7):971-976
  131. 131. Lima L, Silva J, Amaro T, et al. IL-4 and TNF-α polymorphisms are associated with risk of multiple superficial tumors or carcinoma in situ development. Urologia Internationalis. 2011;87:457-463
  132. 132. Brenner AV, Butler MA, Wang SS, Ruder AM, et al. Single-nucleotide polymorphisms in selected cytokine genes and risk of adult glioma. Carcinogenesis. 2007;28(12):2543-2547
  133. 133. Rahaman SO, Vogelbaum MA, Haque SJ. Aberrant Stat 3 signaling by interleukin-4 in malignant glioma cells: Involvement of IL-13Rα2. Cancer Research. 2005;65:2956
  134. 134. Choi WA, Kang MJ, Kim YJ, Seo JH, et al. Gene-gene interactions between candidate gene polymorphisms are associated with total IgE levels in Korean children with asthma. The Journal of Asthma. 2012;49(3):243-252
  135. 135. Jin T, Li X, Zhang J, Wang H, et al. Genetic association between selected cytokine genes and glioblastoma in the Han Chinese population. BMC Cancer. 2013;13:236
  136. 136. Schwartzbaum J, Ding B, Johannesen TB, Osnes LT, et al. Association between prediagnostic IgE levels and risk of glioma. Journal of the National Cancer Institute. 2012;104:1251-1259
  137. 137. Ruan Z, Zhao Y, Yan L, Chen H, et al. Single nucleotide polymorphisms in IL-4Ra, IL-13 and STAT6 genes occurs in brain glioma. Frontiers in Bioscience (Elite Edition). 2011;3:33
  138. 138. Ben CA, Busson M, Douik H, Boukouaci W, et al. Association of IL-12p40 +1188 A/C polymorphism with nasopharyngeal cancer risk and tumor extension. Tissue Antigens. 2011;78(2):148-151
  139. 139. Kaarvatn MH, Vrbanec J, Kulic A, Knezevic J, et al. Single nucleotide polymorphism in the interleukin 12B gene is associated with risk for breast cancer development. Scandinavian Journal of Immunology. 2012;76(3):329-335
  140. 140. Shamran HA, Ghazi HF, AL-Salman A, et al. Singh single nucleotide polymorphisms in IL-10, IL-12p40, and IL-13 genes and susceptibility to glioma. International Journal of Medical Sciences. 2015;12(10):790-796
  141. 141. Chen H, Cheng S, Wang J, Cao C, et al. Interleukin-12 rs 3212227 polymorphism and cancer risk: A meta-analysis. Molecular Biology Reports. 2012;39(12):10235-10242
  142. 142. Terabe M, Park JM, Berzofsky JA. Role of IL-13 in regulation of anti-tumor immunity and tumor growth. Cancer Immunology, Immunotherapy. 2004;53:79-85
  143. 143. Husain SR, Puri RK. Interleukin-13 receptor-directed cytotoxin for malignant glioma therapy: From bench to bedside. Journal of Neuro-Oncology. 2003;65:37-48
  144. 144. Graves PE, Kabesch M, Halonen M, Holberg CJ, et al. A cluster of seven tightly linked polymorphisms in the IL-13 gene is associated with total serum IgE levels in three populations of white children. The Journal of Allergy and Clinical Immunology. 2000;105:506-513
  145. 145. Su Y, Tang LY, Chen LJ, He JR, et al. Joint effects of febrile acute infection and an interferon-gamma polymorphism on breast cancer risk. PLoS ONE. 2012;7(5):e37275
  146. 146. Wei Y, Liu F, Li B, Chen X, et al. Polymorphisms of tumor necrosis factor-alpha and hepatocellular carcinoma risk: A HuGE systematic review and meta-analysis. Digestive Diseases and Sciences. 2011;56:2227-2236. DOI: 10.1007/s10620-011-1617-y
  147. 147. Nieters A, Yuan JM, Sun CL, Zhang ZQ, et al. Effect of cytokine genotypes on the hepatitis B virus-hepatocellular carcinoma association. Cancer. 2005;103:740-748
  148. 148. Karra VK, Gumma PK, Chowdhury SJ, Ruttala R, et al. IL-18 polymorphisms in hepatitis B virus related liver disease. Cytokine. 2015;73:277-282. DOI: 10.1016/j.cyto.2015.02.015
  149. 149. Zhang PA, Wu JM, Li Y, Yang XS. Association of polymorphisms of interleukin-18 gene promoter region with chronic hepatitis B in Chinese Han population. World Journal of Gastroenterology. 2005;11:1594-1598
  150. 150. Kim YS, Cheong JY, Cho SW, Lee KM, et al. A functional SNP of the Interleukin-18 gene is associated with the presence of hepatocellular carcinoma in hepatitis B virus-infected patients. Digestive Diseases and Sciences. 2009;54:2722-2728. DOI: 10.1007/s10620-009-0970-6
  151. 151. Li S, Deng Y, Chen ZP, Huang S, et al. Genetic polymorphism of interleukin-16 influences susceptibility to HBV-related hepatocellular carcinoma in a Chinese population. Infection, Genetics and Evolution. 2011;11:2083-2088. DOI: 10.1016/j.meegid.2011.09.025
  152. 152. Romani S, Hosseini SM, Mohebbi SR, Kazemian S, Derakhshani S, Khanyaghma M, ... & Zali MR. Interleukin-16 gene polymorphisms are considerable host genetic factors for patients’ susceptibility to chronic hepatitis B infection. Hepatitis research and treatment; 2014.
  153. 153. Saxena R, Chawla YK, Verma I, Kaur J. IL-6(-572/-597) polymorphism and expression in HBV disease chronicity in an Indian population. American Journal of Human Biology. 2014;26:549-555. DOI: 10.1002/ajhb.22562
  154. 154. Lee SG, Kim B, Yook JH, Oh ST, Lee I, Song K. TNF/LTA polymorphisms and risk for gastric cancer/duodenal ulcer in the Korean population. Cytokine. 2004;28(2):75-82
  155. 155. Zabaleta J, Camargo MC, Piazuelo MB, Fontham E, Schneider BG, Sicinschi LA, et al. Association of interleukin-1β gene polymorphisms with precancerous gastric lesions in African Americans and Caucasians. American Journal of Gastroenterology. 2006;101(1):163-171
  156. 156. Wang Y, Lerner S, Leibovici D, et al. Polymorphisms in the inflammatory genes IL-6, IL-8, TNF-α, NFKB1, and PPARG and bladder cancer risk. Proceedings of the American Association for Cancer Research Abstract. 2004;3979
  157. 157. Purdue MP, Lan Q, Kricker A, Grulich AE, Vajdic CM, Turner J, et al. Polymorphisms in immune function genes and risk of non-Hodgkin lymphoma: Findings from the New South Wales non-Hodgkin Lymphoma Study. Carcinogenesis. 2007;28(3):704-712
  158. 158. Sugimoto M, Yamaoka Y, Furuta T. Influence of interleukin polymorphisms on development of gastric cancer and peptic ulcer. World Journal of Gastroenterology: WJG. 2010;16(10):1188
  159. 159. Shin WG, Jang JS, Kim HS, Kim SJ, Kim KH, Jang MK, et al. Polymorphisms of interleukin-1 and interleukin-2 genes in patients with gastric cancer in Korea. Journal of Gastroenterology and Hepatology. 2008;23(10):1567-1573
  160. 160. Tao YP, Wang WL, Li SY, Zhang J, Shi QZ, Zhao F, et al. Associations between polymorphisms in IL-12A, IL-12B, IL-12Rβ1, IL-27 gene and serum levels of IL-12p40, IL-27p28 with esophageal cancer. Journal of Cancer Research and Clinical Oncology. 2012;138(11):1891-1900
  161. 161. Suganuma M, Okabe S, Marino MW, Sakai A, Sueoka E, Fujiki H. Essential role of tumor necrosis factor α (TNF-α) in tumor promotion as revealed by TNF-α-deficient mice. Cancer Research. 1999;59(18):4516-4518
  162. 162. Sethi G, Sung B, Aggarwal BB. TNF: A master switch for inflammation to cancer. Frontiers in Bioscience. 2008;13(2):5094-5107
  163. 163. Konwar R, Gara R, Singh M, Singh V, Chattopadhyay N, Bid HK. Association of interleukin-4 and interleukin-1 receptor antagonist gene polymorphisms and risk of benign prostatic hyperplasia. Urology. 2008;71(5):868-872
  164. 164. Jin T, Li X, Zhang J, Wang H, Geng T, Li G, et al. Genetic association between selected cytokine genes and glioblastoma in the Han Chinese population. BMC Cancer. 2013;13(1):1-6
  165. 165. Hackstein H, Jakobs C, Peric A, Michel G, Baal N, Bein G, Bruene B, Hornung V, Yakut E. “Extracorporeal photopheresis promotes IL-1β production (VAC3P. 1054).” 2015:71-1.
  166. 166. Gu X, Shen Y, Fu L, Zuo HY, Yasen H, He P, et al. Polymorphic variation of inflammation-related genes and risk of non-Hodgkin lymphoma for Uygur and Han Chinese in Xinjiang. Asian Pacific Journal of Cancer Prevention. 2014;15(21):9177-9183
  167. 167. Rothman N, Skibola CF, Wang SS, Morgan G, Lan Q, Smith MT, et al. Genetic variation in TNF and IL10 and risk of non-Hodgkin lymphoma: A report from the InterLymph Consortium. The Lancet Oncology. 2006;7(1):27-38
  168. 168. Hiroki CH, Amarante MK, Petenuci DL, Sakaguchi AY, Trigo FC, Watanabe MAE, et al. IL-10 gene polymorphism and influence of chemotherapy on cytokine plasma levels in childhood acute lymphoblastic leukemia patients: IL-10 polymorphism and plasma levels in leukemia patients. Blood Cells, Molecules, and Diseases. 2015;55(2):168-172
  169. 169. Pehlivan M, Sahin HH, Pehlivan S, Ozdilli K, Kaynar L, Oguz FS, et al. Prognostic importance of single-nucleotide polymorphisms in IL-6, IL-10, TGF-β1, IFN-γ, and TNF-α genes in chronic phase chronic myeloid leukemia. Genetic Testing and Molecular Biomarkers. 2014;18(6):403-409
  170. 170. Nursal AF, Pehlivan M, Sahin HH, Pehlivan S. The associations of IL-6, IFN-γ, TNF-α, IL-10, and TGF-β1 functional variants with acute myeloid leukemia in Turkish patients. Genetic Testing and Molecular Biomarkers. 2016;20(9):544-551. DOI: 10.1089/gtmb.2016.0036
  171. 171. Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, et al. Interleukins, from 1 to 37, and interferon-γ: Receptors, functions, and roles in diseases. Journal of Allergy and Clinical Immunology. 2011;127(3):701-721
  172. 172. Davies FE, Rollinson SJ, Rawstron AC, Roman E, Richards S, Drayson M, et al. High-producer haplotypes of tumor necrosis factor alpha and lymphotoxin alpha are associated with an increased risk of myeloma and have an improved progression-free survival after treatment. Journal of Clinical Oncology. 2000;18(15):2843-2851
  173. 173. Kadar K, Kovacs M, Karadi I, Melegh B, Pocsai Z, Mikala G, et al. Polymorphisms of TNF-alpha and LT-alpha genes in multiple myeloma. Leukemia Research. 2008;32:1499-1504
  174. 174. Juan D, Yuan Z, Zhang C, Hua WF, Baoan J, Hou CJ. Role of the TNF-α promoter polymorphisms for development of multiple myeloma and clinical outcome in thalidomide plus dexamethasone. Leukemia Research. 2010;34:1453-1458
  175. 175. Vignali DAA, Kuchroo VK. IL-12 family cytokines: Immunological playmakers. Nature Immunology. 2012;13:722-728
  176. 176. Martino A, Buda G, Maggini V, Lapi F, Lupia A, Di Bello D, et al. Could age modify the effect of genetic variants in IL6 and TNF α genes in multiple myeloma? Leukemia Research. 2012;36:594-597
  177. 177. Tan, D.E., Foo, J.N., Bei, J.X., Chang, J., Peng, R., Zheng, X., Wei, L., Huang, Y., Lim, W.Y., Li, J. and Cui, Q., 2013. Genome-wide association study of B cell non-Hodgkin lymphoma identifies 3q27 as a susceptibility locus in the Chinese population. Nature genetics. 45(7):804-807.
  178. 178. Zhu B, Xiao C, Zhu B, Zheng Z, Liang J. Little association between the interleukin 10-3575T/A polymorphism and cancer risk: Pooled analysis of 15608 cancer cases and 17539 controls. International Journal of Clinical and Experimental Medicine. 2015;8(8):14335
  179. 179. Zhang C, Han F, Yu J, Hu X, Hua M, Zhong C, et al. Investigation of NF-κB-94ins/del ATTG and CARD8 (rs 2043211) Gene Polymorphism in Acute Lymphoblastic Leukemia. Frontiers in Endocrinology. 2019;10:501
  180. 180. Wang LJ, Chou P, Gonzalez-Ryan L, Huang W, Haut PR, Kletzel M. Evaluation of mixed hematopoietic chimerism in pediatric patients with leukemia after allogeneic stem cell transplantation by quantitative PCR analysis of variable number of tandem repeat and testis determination gene. Bone Marrow Transplantation. 2002;29(1):51-56
  181. 181. Zhang XJ, Yan KL, Wang ZM, Yang S, Zhang GL, Fan X, et al. Polymorphisms in interleukin-15 gene on chromosome 4q31. 2 are associated with psoriasis vulgaris in Chinese population. Journal of Investigative Dermatology. 2007;127(11):2544-2551
  182. 182. Gonzalez B, Flores-A H, Sanchez D, Alaez C, Gorodezky C. IL6 (− 174G/C) SNP but not IL4 (− 590C/T) polymorphisms contributes to the development of acute lymphoblastic leukemia, and is involved in gender specific risk in Mexican children. Human Immunology. 2015;76:94
  183. 183. Mondet, J., Hussein, K., & Mossuz, P. (2015). Circulating cytokine levels as markers of inflammation in Philadelphia negative myeloproliferative neoplasms: diagnostic and prognostic interest. Mediators of inflammation, 2015
  184. 184. Jevtovic-Stoimenov T, Cvetkovic T, Despotovic M, Basic J, Cvetkovic J, Marjanovic G, et al. The influence of TNF alpha-308 G/A polymorphism on oxidative stress in patients with chronic lymphocytic leukemia. Leukemia Research. 2017;54:66-72
  185. 185. Martino A, Campa D, Jurczyszyn A, Martínez-López J, Moreno MJ, Varkonyi J, et al. Genetic variants and multiple myeloma risk: IMMEnSE validation of the best reported associations—An extensive replication of the associations from the candidate gene era. Cancer Epidemiology and Prevention Biomarkers. 2014;23(4):670-674
  186. 186. Wu Z, Sun Y, Zhu S, Tang S, Liu C, & Qin W. Association of Interferon gamma+ 874T/A polymorphism and leukemia risk: a meta-analysis. Medicine. 2016;95(12)
  187. 187. Nass J, Efferth T. Drug targets and resistance mechanisms in multiple myeloma. Cancer Drug Resistance. 2018;1(2):87-117
  188. 188. Peng X, Shi J, Sun W, Ruan X, Guo Y, Zhao L, et al. Genetic polymorphisms of IL-6 promoter in cancer susceptibility and prognosis: A meta-analysis. Oncotarget. 2018;9(15):12351
  189. 189. Basmaci C, Pehlivan M, Tomatir AG, Sever T, Okan V, Yilmaz M, et al. Effects of TNF, NOS3, MDR1 gene polymorphisms on clinical parameters, prognosis and survival of multiple myeloma cases. Asian Pacific Journal of Cancer Prevention. 2016;17(3):1009-1014
  190. 190. Dinarello CA. Biologic basis for interleukin-1 in disease. Blood. 1996;87:2095-2147
  191. 191. Ito LS, Iwata H, Hamajima N, et al. K Significant reduction in breast cancer risk for Japanese women with interleukin 1B-31CT/TTrelative to CC genotype. Japanese Journal of Clinical Oncology. 2002;32:398-402
  192. 192. Alliston T, Choy L, Ducy P, Karsenty G, et al. TGFbeta- induced repression of CBFA1 by Smad 3 decreases cbfa 1 and osteocalcin expression and inhibits osteoblast differentiation. The EMBO Journal. 2001;20(9):2254-2272
  193. 193. Adorno M, Cordenonsi M, Montagner M, Dupont S, et al. A Mutant-p 53/Smad complex opposes p 63 to empower TGFbeta-induced metastasis. Cell. 2009;137(1):87-98
  194. 194. Dunning AM, Ellis PD, McBride S, Kirschenlohr HL, et al. A transforming growth factorbeta 1 signal peptide variant increases secretion in vitro and is associated with increased incidence of invasive breast cancer. Cancer Research. 2003;63(10):2610-2615
  195. 195. Hishida A, Iwata H, Hamajima N, Matsuo K, et al. Transforming growth factor B1 T29C polymorphism and breast cancer risk in Japanese women. Breast Cancer. 2003;10(1):63-69
  196. 196. Yongsheng H, Binghui L, Ji Q, Jingtian X, et al. TGF-b1 29T/C polymorphism and breast cancer risk: A meta-analysis involving 25, 996 subjects. Breast Cancer Research and Treatment. 2010;123:863-868
  197. 197. Turner DM, Williams DM, Sankaran D, Lazarus M, et al. An investigation of polymorphism in the interleukin-10 gene promoter. European Journal of Immunogenetics. 1997;24:1-8
  198. 198. Keijsers V, Verweij CL, Westendorp RGJ, Breedveld FC, et al. IL-10 polymorphisms in relation to production and rheumatoid arthritis. Arthritis and Rheumatism. 1997;40(Suppl 9):S179
  199. 199. Watanabe Y, Kinoshita A, Yamada T, Ohta T, et al. A catalog of 106 single-nucleotide polymorphisms (SNPs) and 11 other types of variations in genes for transforming growth factor-beta 1 (TGF- beta 1) and its signaling pathway. Journal of Human Genetics. 2002;47:478-483
  200. 200. Talaat RM, Dondeti MF, El-Shenawy SZ, Khamiss OA. Transforming growth factor-β 1 gene polymorphism (T29C) in Egyptian patients with hepatitis B virus infection: A preliminary study. Hepatitis Research and Treatment. 2013;2013:293274. DOI: 10.1155/2013/293274
  201. 201. Perrey C, Turner SJ, Pravica V, Howell WM, et al. ARMS-PCR methodologies to determine IL-10, TNF-alpha, TNF- beta and TGF-beta 1 gene polymorphisms. Transplant Immunology. 1999;7:127-128
  202. 202. Yamada Y, Miyauchi A, Goto J, Takagi Y, Okuizumi H, et al. Association of a polymorphism of the transforming growth factor-beta 1 gene with genetic susceptibility to osteoporosis in postmenopausal Japanese women. Journal of Bone and Mineral Research. 1998;13:1569-1576
  203. 203. Opal SM, DePalo VA. Anti-inflammatory cytokines. Chest. 2000;117:1162-1172
  204. 204. Rosenwasser LJ, Klemm DJ, Dresback JK, Inamura H, Mascali JJ, Klinnert M, et al. Promoter polymorphisms in the chromosome 5 gene cluster in asthma and atopy. Clinical and Experimental Allergy. 1995;25(suppl 2):74-78; discussion: 95-6
  205. 205. Wu J, Lu Y, Ding YB, Ke Q, Hu ZB, Yan ZG, et al. Promoter polymorphisms of IL2, IL4, and risk of gastric cancer in a high-risk Chinese population. Molecular Carcinogenesis. 2009;48(626):32
  206. 206. Zhou Y, Li N, Zhuang W, Liu GJ, Wu TX, Yao X, et al. Interleukin-10-1082 promoter polymorphism associated with gastric cancer among Asians. European Journal of Cancer. 2008;44(17):2648-2654
  207. 207. Zhuang W, Wu XT, Zhou Y, Liu L, Liu GJ, Wu TX, et al. Interleukin 10-592 promoter polymorphism associated with gastric cancer among Asians: A meta-analysis of epidemiologic studies. Digestive Diseases and Sciences. 2010;55(6):1525-1532
  208. 208. Chen KF, Li B, Wei YG, Peng CJ. Interleukin-10-819 promoter polymorphism associated with gastric cancer among Asians. The Journal of International Medical Research. 2010;38:1-8
  209. 209. Yu Z, Liu Q, Huang C, et al. The interleukin 10-819C/T polymorphism and cancer risk: A HuGE review and meta-analysis of 73 studies including 15, 942 cases and 22, 336 controls. OMICS. 2013;17:200-214
  210. 210. Shao N, Xu B, Mi YY, Hua LX. IL-10 polymorphisms and prostate cancer risk: A meta-analysis. Prostate Cancer and Prostatic Diseases. 2011;14(2):129-135
  211. 211. He B, Zhang Y, Pan Y, Xu Y, Gu L, Chen L, et al. Interleukin 1 beta (IL1B) promoter polymorphism and cancer risk: Evidence from 47 published studies. Mutagenesis. 2011;26:637-642
  212. 212. Pociot F, Molvig J, Wogensen L, Worsaae H, et al. Taq I polymorphism in the human interleukin-1 beta (IL-1 beta) gene correlates with IL-1 beta secretion in vitro. European Journal of Clinical Investigation. 1992;22:396-402
  213. 213. Liang Z, Jun Y, Liming W, Xu W, et al. Interleukin 1B rs 16944 G>A polymorphism was associated with a decreased risk of esophageal cancer in a Chinese population. Clinical Biochemistry. 2013;46:1469-1473
  214. 214. Yamada S, Matsuhisa T, Makonkawkeyoon L, Chaidatch S, Kato S, Matsukura N. Helicobacter pylori infection in combination with the serum pepsinogen I/II ratio and interleukin-1beta-511 polymorphisms are independent risk factors for gastric cancer in Thais. Journal of Gastroenterology. 2006;41:1169-1177
  215. 215. Mazur G, Bogunia-Kubik K, Wrobel T, Kuliczkowski K, Lange A. TGF-beta 1 gene polymorphisms influence the course of the disease in non-Hodgkin’s lymphoma patients. Cytokine. 2006;33:145-149
  216. 216. Peters CA et al. TGFB1 single nucleotide polymorphisms are associated with adverse quality of life in prostate cancer patients treated with radiotherapy. International Journal of Radiation Oncology, Biology, Physics. 2008;70:752-759
  217. 217. Wei YS, Xu QQ, Wang CF, Pan Y, et al. Genetic variation in transforming growth factor-beta 1 gene associated with increased risk of esophageal squamous cell carcinoma. Tissue Antigens. 2007;70:464-469
  218. 218. Guan X, Zhao H, Niu J, Tan D, et al. Polymorphisms of TGFB1 and VEGF genes and survival of patients with gastric cancer. Journal of Experimental & Clinical Cancer Research. 2009;28:94
  219. 219. Shu XO, Gao YT, Cai Q, Pierce L, Cai H, Ruan ZX, et al. Genetic polymorphisms in the TGF-β1 gene and breast cancer survival: A report from the Shanghai Breast Cancer Study. Cancer Research. 2004;64(3):836-839
  220. 220. Zhang L, Wu G, Herrle F, Niedergethmann M, et al. Single nucleotide polymorphisms of genes for egf, tgf-β and tnf-α in patients with pancreatic carcinoma. Cancer genomics and proteomics. 2012;9:287-296
  221. 221. Muller-Hermelink N, Braumuller H, Pichler B, et al. TNFR1 signalling and IFN-gamma signalling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell. 2008;13:507-518
  222. 222. Gomes M, Coelho A, Araujo A, Teixeira AL, Catarino R, Medeiros R. Influence of functional genetic polymorphism (-590C/T) in non-small cell lung cancer (NSCLC) development: The paradoxal role of IL4. Gene. 2012;504:111-115
  223. 223. Bozdogan ST, Erol B, Dursun A, et al. The IL-1RN and IL-4 gene polymorphisms are potential genetic markers of susceptibility to bladder cancer: A case-control study. World Journal of Urology. 2015;33:389-395
  224. 224. Kaminska B, Kocyk M, Kijewska M. Tgf beta signaling and its role in glioma pathogenesis. Advances in Experimental Medicine and Biology. 2013;986:171-187
  225. 225. Mingjun H, Jieli D, Lihong C, Tingqin H, et al. IL-10 and PRKDC polymorphisms are associated with glioma patient survival. Oncotarget. 2016;7(49):80680-80687
  226. 226. Tanikawa T, Wilke CM, Kryczek I, Chen GY, Kao J, Nunez G, et al. Interleukin-10 ablation promotes tumor development, growth, and metastasis. Cancer Research. 2012;72:420-429
  227. 227. Saxena R, Chawla YK, Verma I, Kaur J. Interleukin-1 polymorphism and expression in hepatitis B virus-mediated disease outcome in India. Journal of Interferon & Cytokine Research. 2013;33:80-89. DOI: 10.1089/jir.2012.0093
  228. 228. Ma J, Liu YC, Fang Y, Cao Y, Liu ZL. TGF-beta 1 polymorphism 509 C> T is associated with an increased risk for hepatocellular carcinoma in HCV-infected patients. Genetics and Molecular Research. 2015;14(2):4461-4468
  229. 229. Lu Y, Wu Z, Peng Q, Ma L, Zhang X, Zhao J, et al. Role of IL-4 gene polymorphisms in HBV-related hepatocellular carcinoma in a Chinese population. PLoS One. 2014;9(10):e110061
  230. 230. Wu, Z., Qin, W., Zeng, J., Huang, C., Lu, Y., & Li, S. (2015). Association between IL-4 polymorphisms and risk of liver disease: an updated meta-analysis. Medicine, 94(35)
  231. 231. Tang S, Yuan Y, He Y, Pan D, Zhang Y, Liu Y, et al. Genetic polymorphism of interleukin-6 influences susceptibility to HBV-related hepatocellular carcinoma in a male Chinese Han population. Human Immunology. 2014;75:297-301. DOI: 10.1016/j.humimm.2014.02.006
  232. 232. Li Y, Du Z, Wang X, Wang G, Li W. Association of IL-6 promoter and receptor polymorphisms with multiple myeloma risk: A systematic review and meta-analysis. Genetic Testing and Molecular Biomarkers. 2016;20(10):587-596
  233. 233. Ognjanovic S, Yuan JM, Chaptman AK, Fan Y, et al. Genetic polymorphisms in the cytokine genes and risk of hepatocellular carcinoma in low-risk non-Asians of USA. Carcinogenesis. 2009;30:758-762. DOI: 10.1093/carcin/bgn286
  234. 234. Wu J, Lu Y, Ding YB, Ke Q, Hu ZB, Yan ZG, et al. Promoter polymorphisms of IL2, IL4, and risk of gastric cancer in a high-risk Chinese population. Molecular Carcinogenesis. 2009;48(7):626-632
  235. 235. Chen GY, Nuñez G. Sterile inflammation: Sensing and reacting to damage. Nature Reviews Immunology. 2010;10(12):826-837
  236. 236. Namkung JH, Lee JE, Kim E, Park GT, Yang HS, Jang HY, et al. An association between IL-9 and IL-9 receptor gene polymorphisms and atopic dermatitis in a Korean population. Journal of Dermatological Science. 2011;62(1):16-21
  237. 237. Zhang, L., Wu, G., Herrle, F., Niedergethmann, M., & Keese, M. (2012). Single nucleotide polymorphisms of genes for EGF, TGF-β and TNF-α in patients with pancreatic carcinoma. Cancer genomics & proteomics. 9(5):287-295
  238. 238. Gonzalez-Zuloeta Ladd AM, Arias-Vasquez A, Siemes C, Coebergh JW, et al. Transforming growth factor beta 1 leu 10pro polymorphism and breast cancer morbidity. European Journal of Cancer. 2007;43:371-374
  239. 239. Schaaf BM, Boehmke F, Esnaashari H, Seitzer U, et al. Pneumococcal septic shock is associated with the interleukin-10-1082 gene promoter polymorphism. American Journal of Respiratory and Critical Care Medicine. 2003;168(4):476-480
  240. 240. Kornman KS. Interleukin 1 genetics, inflammatory mechanisms, and nutrigenetic opportunities to modulate diseases of aging. The American Journal of Clinical Nutrition. 2006;83(2):475S-483S
  241. 241. Schoof N, Franklin J, Fürst R, Zander T, von Bonin F, Peyrade F, et al. Interleukin-10 gene polymorphisms are associated with freedom from treatment failure for patients with Hodgkin lymphoma. The Oncologist. 2013;18:80-89
  242. 242. Li G, Li D. Relationship between IL-10 gene polymorphisms and the risk of non-Hodgkin lymphoma: A meta-analysis. Human Immunology. 2016;77(5):418-425

Written By

Arshad A. Pandith, Ina Bhat, Sheikh Mansoor, Aabid Koul, Usma Manzoor, Iqra Anwar, Fozia Mohammad, Qurat Ul Aein, Shahid M. Baba and Carmen Vladulescu

Submitted: 25 May 2021 Reviewed: 09 July 2021 Published: 26 December 2021