Open access peer-reviewed chapter

IL10 as Cancer Biomarker

Written By

Maria Teresa Gonzalez-Garza, Delia Elva Cruz-Vega and Carmen Maldonado-Bernal

Submitted: 30 September 2019 Reviewed: 09 December 2019 Published: 08 June 2020

DOI: 10.5772/intechopen.90806

From the Edited Volume

Translational Research in Cancer

Edited by Sivapatham Sundaresan and Yeun-Hwa Gu

Chapter metrics overview

1,063 Chapter Downloads

View Full Metrics

Abstract

Chronic inflammation can trigger events that would induce the malignant transformation of cells and carcinogenesis. Cytokines play a crucial role and can control the development and multiplication of cancerous cells. However, clinical data present controversy about the participation of these proteins in the establishment and development of cancer. Interleukin 10 (IL-10), a potent anti-inflammatory cytokine, has been the subject of multiple studies. Several studies have reported that IL-10 has pro- and antitumor effects. Elevated levels of IL-10 are associated with increased tumor growth with poor prognosis and drug resistance. However, this cytokine has both tumor-promoting and tumor-inhibiting properties. In vitro and in vivo studies report mechanisms by which IL-10 expression downregulates class I, which results in the control of the metastatic disease. IL-10 also inhibits tumorigenesis via downregulation of other cytokines. The variation observed could be the result of concentration ranges of this protein, genetic polymorphism, or both. The value obtained may serve as a biomarker indicative of tumor development and its prognosis.

Keywords

  • interleukin 10
  • IL-10
  • IL10 polymorphism
  • cancer biomarker

1. Introduction

The immune system is responsible for monitoring and getting rid of molecules or cells outside the body that can be potentially malignant. This system is constituted by cells that can act by themselves or the synthesis of molecules capable of inducing the destruction of strange agents or invading cells. In addition, these cells synthesize proteins called cytokines, which are not only capable of destroying invading cells; they activate other cells of the same immune system, which makes the system efficient. In a coordinated way, the synthesis of anti-inflammatory cytokines begins that will stop the inflammatory process and prevent the damage from spreading in healthy tissue. In the case of cancer, the cells will have nonnormal characteristics due to the changes they undergo during their transformation. The immune system works to eliminate these cells; however, for reasons not well established yet, at some point, the immune system fails, and the malignant cells survive and establish [1]. Even when the production of anti-inflammatory cytokines is present, the levels of pro- and anti-inflammatory cytokines have an imbalance that prevents completing the final objective. An increase of anti-inflammatory cytokines, such as interleukin 10 (IL-10), has been associated with a poor prognosis and considered as a biomarker for cancer disease. This chapter presents evidence that shows alterations in the serum levels of IL-10 in cancer patients, the participation of the cancerous tumor in the synthesis of this cytokine, and its possible relationship with polymorphisms in the gene promoter, the gene of this protein.

Advertisement

2. Interleukin 10 (IL-10)

IL-10 is an important regulatory cytokine with a potent anti-inflammatory effect. In humans, it is encoded by the IL10 gene [2]. It presents polymorphisms (SNPs), most of them within the promoter regions, and has been proposed as responsible for the altered function, by dysregulating their expression. Several groups report associations between polymorphism and cancer risk [3, 4, 5, 6, 7]. It is a helical cytokine and exists in solution predominantly as a homodimer, composed of two polypeptide chains of 160 amino acids, each with a molecular weight of 20,6419 kD [8].

IL-10 was initially thought to be produced only by T helper (Th2) cells but is now known to be made by a variety of cell types [9]. It is primarily produced by monocytes upon PD-1 triggering in these cells and, to a lesser extent, lymphocytes type 2 T helper cells (TH2), mast cells, CD4+CD25+Foxp3+ regulatory T cells, and in a specific subset of activated T cells and B cells (Figure 1) [10]. IL-10 expression is minimal in unstimulated tissues and seems to require triggering [11].

Figure 1.

IL-10R homodimers bind to form in a tetrameric heterodimer after IL-10 binds to its receptor α. It downstream signaling through STAT3 until target genes.

Cytokine signals are generated through a receptor complex consisting of two IL-10 receptor-1 and two IL-10 receptor-2 proteins. After IL-10 binding, it induces STAT3 signaling via the phosphorylation of the cytoplasmic tails of IL-10 receptor 1 + IL-10 receptor 2 by JAK1 and Tyk2, respectively (Figure 2) [12, 13].

Figure 2.

After antigenic stimulus on cells from innate and adaptive system, the IL-10 is expressed from these cells. To avoid severe danger from pro-inflammatory cytokines and reactive radicals, IL-10 acts as anti-inflammatory cytokine by blocking its expression.

IL-10 expression is regulated at the transcriptional and posttranscriptional levels. Extensive IL-10 locus remodeling in monocytes upon the stimulation of Toll-like receptor (TLR) or Fc receptor pathways was described. Induction involves ERK1/2, p38, and NF-κB signaling and transcriptional activation via promoter binding of the transcription factors NF-κB and AP-1; other regulation as a complex of multiple transcriptional factors has been described, such as GATA-3, E4BP4, MAF, and Blimp [14, 15]. These reports are allowing the possibility that transcriptional regulatory machinery could be specific to certain cell types [16, 17]. In addition, the regulation of IL-10 production depending on the TLR2 or TLR4-stimulated in BM-derived macrophages presents different stability profiles for the IL10 mRNA [18].

It has the ability to inhibit activation and effector function of T cells, monocytes, and macrophages. It regulates the growth and differentiation of several cells such as B cells, NK cells, cytotoxic, and helper T cells, as well as mast cells, granulocytes, dendritic cells, keratinocytes, and endothelial cells (Figure 1).

Discovered in 1991, IL-10 was initially reported to suppress cytokine secretion, antigen presentation, and CD4+ T cell activation. IL-10 is crucial for controlling these T-cell responses, as IFNγ [19, 20, 21, 22]. Further investigations have shown that IL-10 predominantly inhibits the synthesis of the pro-inflammatory cytokines IL-1, IL12, tumor necrosis factor (TNFα), and gamma Interferon (IFNγ) by stimulated monocytes/macrophages [23, 24, 25]. IL-10 is crucial for controlling these T-cell responses, as IFNγ-secreting and blocking proliferation [22, 26, 27, 28].

The pivotal role of autocrine IL-10 and the interaction with CD28 in the induction of T cell are to initiate peripheral tolerance as an immunoregulatory mechanism controlling antigen-specific T cell responses [21].

In addition, several effects have been reported: it downregulates the expression of Th1 cytokines, MHC class II antigens, dendritic cells, and co-stimulatory molecules on macrophages [29].

Biphasic effects have been reported on B cells. It prevents apoptosis in germinal B cells but induces apoptosis on B-chronic lymphocytic leukemia cells, enhances proliferation induced by CD40L, and promotes differentiation of B cells to become plasma cells secreting IgM, IgG, and IgA immunoglobulins [30, 31, 32, 33, 34].

IL-10 appears to have considerable importance in the development of human cancer and its immune escape. These have suggested that it could serve as a biomarker for prognostic diseases or as a target for treatment. Two factors should be considered: high levels of this cytokine in the system, and genetic polymorphisms.

Advertisement

3. IL-10 serum levels on cancer patients

Considering their possible role in the development and establishment of malignant cells, the first studies conducted to detect serum IL-10 levels in cancer patients, reported a higher concentration than in healthy subjects. In melanoma patients with lymph node metastases, stages III and IV showed significantly high concentration with respect to a healthy subject. In vitro determination performed on the supernatant of primary malignant melanoma cultures, IL-I0 mRNA, and protein was expressed. Later, in a meta-analysis that included melanoma patients, high expression of serous IL-10 leads to an adverse survival and correlated with worse outcomes in cancer patients [35, 36, 37, 38, 39].

On serum samples of 90 patients with gastric cancer, high IL-10 levels were associated with a worse prognosis independent of the gastric-stage patients and pancreatic cancer [40]. Similar observations were reported on another group of patients with a pancreatic and gastric cancer stage (IV). In the same study, patients with colon and renal carcinoma IL-10 levels did not significantly differ from controls [41]. In vitro study, supernatants of pancreatic tumors primary cultures, high concentration of IL-10 was detected [42].

Similar clinical findings on sera and tissues samples from lung cancer patients were report; samples with high levels were found on patients in stages III and IV, and less in stages II and I. The increased IL-10 levels correlate with a poor prognosis. On lung tumor tissues samples from those patients, IL10 concentration showed than the higher expression, presented lower survival rates [43].

Samples from breast cancer from patients, who underwent surgery as well as peritumoral normal breast tissue, were analyzed. Correlation between the IL-10 expressions of breast cancer tissue showed poor prognosis. No IL-10 was detected on normal tissue samples [44]. Determination of IL-10 on patients with breast cancer and early breast cancer showed that it could be usefully associated with other cytokines as biomarkers to discriminating advanced cancer, reported that IL-10 is significantly upregulated. These data could be used discerning between the two stages. [45].

Hodgkin’s disease presented sera elevated IL-10 levels on range 4.5–225.6 pg/ml, suggesting that IL-10 could be an independent prognostic factor and correlates to poor survival [46]. Lymph nodes from pediatric patients with Hodgkin’s disease express high levels of IL-10 mRNA where there were associated with an unfavorable prognosis [47, 48]. For the study of sera levels in 153 patients with Non-Hodgkin’s disease, IL-10 was detectable with a similar frequency in all subtypes and all clinical stages. Nevertheless, high levels correlate with poor prognosis. Patients with stage IV disease and detectable serum IL-10 had a particularly poor prognosis [49, 50]. Additional information on sera levels of IL-10 is shown in Table 1.

DiseasePatients IL-10 levelsHealthy donorReferences
Melanoma15–480 pg/ml<3.0 pg/ml[35]
8.75 pg/ml<3.0 pg/ml[39]
24.3 ng/ml3.4 ng/ml[41]
Gastric cancer6.3 ng/ml3.4 ng/ml[41]
12.5 pg/ml4.0 pg/ml[40]
27.52 pg/ml<12 np/ml[51]
19.6 pg/ml9.2 pg/ml[52]
>21.0 pg/ml<3.0 pg/ml[53]
Pancreatic cancer6.8 ng/ml3.4 ng/ml[41]
>10.0 pg/mlND[54]
>9.8 pg/ml3.0 pg/ml[55]
Colorectal cancer97.36 ng/l24.53 ng/l[56]
16.09 pg/ml5.1 pg/ml[57]
Hepatic cancer12 pg/ml6.3 pg/ml[58]
Hodgkin lymphoma61.5 pg/mlNo detectable[46]
>10 pg/ml7.1 pg/ml[59]
>10 pg/mlND[60]
>10 pg/mlND[61]
26.79 pg/mlND[62]
Non-Hodgkin lymphoma>7.98 pg/ml<5.0 pg/ml[50]
Lung cancer>38.16 pg/ml32.55 pg/ml[43]
21.4 pg/ml9.2 pg/ml[63]
Multiple myeloma201.96 pg/mlND[64]
2.39 ± 0.82 ng/ml(0.34 ± 0.15 ng/ml)[65]
B-cell lymphoma26.0 pg/ml18.0 pg/ml[66]
Chronic lymphocytic leukemia74 pg/ml<13.68 pg/ml[67]

Table 1.

Determination of IL-10 levels in serum from cancer patients.

IL-10 protein or mRNA appears to be an important component of the tumor micro-environment in a range of human cancer types, as renal cell carcinoma [68]. The role of macrophages in the regulation of tumor cell proliferation, invasion, angiogenesis, or immune control does not always have a positive effect; in fact, it has shown a negative effect. The secretion of IL-10 inhibits the inflammatory response [69, 70, 71, 72] and also has been suggested that the detection of IL-10 into and surrounding the tumor may be derived directly from the tumor cells; in vitro studies reveal that melanoma cells themselves are the primary origin of IL-10 in tumor specimens in vivo [35]. Nevertheless, Mocellin et al. report evidence about immunostimulating anticancer properties, suggesting IL-10 over-expression within the tumor micro-environment and it may catalyze cancer immune rejection [73].

IL-10 and IL10R were quantified in sera and surgical specimens, although no significant serum IL-10 elevation was found. On surgical lung tumor cells, IL-10 expression was considered as a prognostic factor on nonsmall-cell lung cancer surgical specimens. In addition to an immunohistochemistry study on human lung surgery, a positive correlation between IL-10 and IL-10 receptor expressions was related to the lung tumor diameter. Interestingly, IL-10R was mainly expressed on the surface of Foxp-3þ T-regulatory lymphocytes infiltrating the tumor [74, 75].

Advertisement

4. IL10 polymorphims related to the development and prognosis of cancer

Single nucleotide polymorphisms (SNPs) has been described on IL10 promoter, where adenine was substituted by guanine at −1082 bp (rs1800896). Thymine was replaced by cytosine at −819 bp (rs1800871) and adenine by cytosine at −592 bp (rs1800872). All of those contribute to the variation in protein expression [76]. The IL10 production have a hereditary component estimated in 75%, suggesting that clinical outcome imparted by IL10 genetic variants may be mediated by effects on the tumor micro-environment composition or modulation [47]. The possibility that SNPs participate in the development, establishment, and the prognosis of cancer has been investigated by numerous research groups and includes several group malignances.

Eastern European patients with breast cancer, IL10 −1082A > G, −819 T > C, −592A > C polymorphisms, and phased haplotypes have not revealed a prognostic value [77]. Nevertheless, another study report 592C > A polymorphisms could modify disease-free and overall survival in women with lymph node-positive breast cancer [78]. In a Chinese Han population study, the −1082AA genotype was associated with a significantly increase risk of lymph node involvement and larger tumor size at the time of diagnosis [79]. After meta-analysis, it showed that IL10 rs1800896 and rs1800871 polymorphisms had no association with breast cancer risk, while rs1800872 polymorphism had a decreased risk of breast cancer in Caucasians [3]. However, another meta-analysis suggests that IL10 rs1800871 and rs1800872 polymorphisms might contribute to breast cancer susceptibility in the overall population, but not by ethnicity [6].

Chinese men, diagnostic with prostate cancer, do not show significant differences in (−1082A/G, −819 T/C, and −592A/C) SNPs between patients and control subjects [80]. Meta-analysis performed with a Caucasian and Asiatic population suggests that there is no association between IL10 gene rs1800896, rs1800871, and rs1800872 polymorphisms and prostate cancer [81]. A study looking for the participation of variant alleles at both −819 and −592 polymorphisms was modestly associated with the advanced stages of prostate cancer [82, 83]. Other meta-analysis reports that rs1800896 polymorphism is associated with a decreased risk of prostate cancer [84].

In a meta-analysis including 26 studies designed to search the risk of skin cancer and IL10 polymorphism, the results show that there is not a significant association of −1082G > A or −592C > A and risk to skin cancer. Nevertheless, it suggested a potential association between −819C > T polymorphism and decreased risk of skin cancer [38]. A meta-analysis carried out for possible association between polymorphism and melanoma did not find an association between skin cancer risk and the −592A/C or IL-10-1082G/A. However, there was a correlation between IL10 −819T/C polymorphisms and skin cancer [85].

Association between IL10 −1082A/G, −592C/A, and −819T/C gene polymorphisms and risk of lung cancer was also investigated by meta-analysis, but until now, there are inconsistent results: on Asian populations, IL10 −1082A/G and −819T/C polymorphisms might have a significant association with the risk of lung cancer [86]. Another meta-analysis suggests that the polymorphism associated with a risk factor for lung cancer was IL10 −592A > C polymorphism, especially among Asians and Caucasians. In contrast, the IL10 −819 T > C and −1082A > G polymorphisms are not significantly associated with an increased risk of lung cancer [87].

Cervical cancer associated with a polymorphism at this time is not conclusive: −1082 polymorphism was suggested as a marker of genetic susceptibility to cervical cancer among Japanese women but was not significantly increased in Zimbabwean women or Korean women [88]. In a meta-analysis, including 17 publications of Asian, African, and Caucasian populations, it did not find a significant association between the polymorphism and cervical cancer risk [89]. Other meta-analysis looking for association between −1082 G/A polymorphism and cervical cancer risk, including the same population, report as well that there is no association [90].

Association between rs1800896 polymorphism and head and neck cancer risk and its clinical stages in Caucasian and Asiatic evaluated by meta-analysis gave a significant association with head and neck cancer risk but not with the clinical stages [91]. Other analysis reports significant associations between the IL10 −1082A > G and IL10 −819 T > C polymorphism and increased risks of head and neck cancer similar population [92].

Other studies on the relation between risk or prognosis polymorphisms in cancer have been performed. In there, a possible association with gastric cancer showed a significant association with IL10-819 on the Mexican population. Nevertheless, no significant association was found for IL10 −592C/A (rs1800872) and IL10 −1082A/G (rs1800896) [93]. Looking for a possible predispose to oral squamous cell carcinoma (−592), A/C polymorphism was significantly associated with reduced risk [94]. In colorectal cancer IL10 −819 T > C, polymorphism was associated with significantly increased risk [95].

In a clinical study with child Hokdkin disease, −1082GG genotype was associated with lower IL10 mRNA expression. Nevertheless, genotypes of the −592 SNP showed no association with IL10 mRNA expression. However, −1082AACAG genotypes, ATA haplotype, and the presence of the −592AA genotype were associated with unfavorable prognosis [47, 96].

Genotypic variants of IL10 (−1082G/A) polymorphism in adult Sudanese patients with acute myeloid leukemia were investigated as a possible risk factor. In there, no association between IL10 (−1082G/A) and acute myeloid leukemia was detected [7]. Nevertheless, in the Egyptian population, −819 polymorphism was associated with enhanced risk [97]. In the Chinese population, the −819A allele frequencies in the AML group were higher than in the controls [98]. In pediatric patients, Classical Hodgkin lymphoma −1082AA/AG, −592CC genotypes, and ATA haplotype were associated with unfavorable prognosis; interestingly, −1082 was associated with low IL10 mRNA expression [46].

Unfortunately, to date, the results are controversial; the relationship of polymorphism with the presence of cancer and/or the possible risk of developing cancer is shown in Table 2.

DiseasePopulationPolymorphismAssociationReference
Breast cancerEastern EuropeanIL10 −1082A/G, IL10 −592A, IL10 −819CNo prognostic value[77]
Eastern EuropeanIL10 −592C > AAssociated with survival[78]
Chinese HanIL10 −1082AAAssociated with cancer risk[79]
Chinese Hanrs1800896, rs1800871No associated with cancer risk[3]
Caucasianrs1800872Associated with cancer risk[3]
ProstateChinese men studyIL10 −1082A/G, IL10 −592A/C, IL10 −819 T/CNo associated with cancer risk[80]
Caucasian, African-Americans, and Asiaticrs1800896Associated with reduced cancer risk[84]
Caucasian and Asiaticrs1800896, rs1800871, rs1800872No association with risk cancer[81]
Caucasian, African-Americans, and AsiaticIL10-1082 A4G, IL10-819 C4, IL10-592 C4ANo association with cancer risk[82]
Caucasian and AsiaticIL10--592A > CNo association with cancer risk[83]
Skin cancerCaucasian, African-Americans, and AsiaticIL10 −1082A/G, IL10-592A, IL10-819CNo associated with cancer skin risk[82]
Caucasian, African-Americans, and AsiaticIL10 −592A/C, IL10 −1082G/ANo associated with cancer risk[85]
Lung cancerAsiaticIL10 −1082A/G, IL10-592A, IL10-819CAssociated with cancer risk[86]
CaucasianIL10 −1082A/G, IL10-592A, IL10-819CNo association with cancer risk[86]
CaucasianIL10 −592A > CAssociated with cancer risk[87]
Caucasian and AsiaticIL10 −819 T > C, IL10 −1082A > GNo associated with cancer risk[87]
Cervix cancerJapanese womenIL10 −1081Associated with cancer risk[88]
Zimbabwean women and Korean womenIL10 −1082No associated with risk[88]
Caucasian, African-Americans, and AsiaticIL10 −1082A > GNo associated with risk[89]
Caucasian, African-Americans, and AsiaticIL10 −1082 G/ANo associated with cancer risk[90]
Head and neck cancerCaucasian and Asiaticrs1800896Associated with cancer risk[91]
Caucasian, and AsiaticIL10 −1082A > G, IL10 −819 T > CAssociated with cancer risk[92]
Gastric cancerMexican populationIL10 −819Associated with risk[93]
Mexican populationIL10 −592C/A, IL10 −1082A/GNo associated with risk[93]
Oral mucosa cancerIndian populationIL10-592 A/CAssociated with cancer risk[94]
Colorectal cancerCaucasian and AsiaticIL10819T > CAssociated with cancer risk[95]
Caucasian and Asiaticrs1800871 rs1800872No associated with cancer risk[95]
Caucasian and AsiaticIL10 −1082A > G, IL10 −592C > ANo associated with cancer risk[95]
Pediatric Hodgkin’s diseaseCaucasianIL10-592AAPronostic marker[96]
BrazilianIL10 −1082GGUnfavorable prognosis[47]
Acute myeloid leukemiaEgyptianIL10‑819Associated with risk[97]
Chinese HanIL-10-819, −592Associated with risk[98]
SudaneseIL10 −1082G/AAssociated with risk[7]

Table 2.

Relationship of IL10 polymorphism and cancer risk.

Advertisement

5. Conclusions

High concentrations of IL-10 in the serum of cancer patients seem to correspond not only to the expression of this protein by immune cells. The evidence shows that the cancer cell is capable of synthesizing it, which would cause an imbalance in the homeostasis of the immune system.

Besides, the presence of polymorphisms suggested the possibility that some of them could be involved in the regulation of the activity of IL10; this gave rise to numerous studies seeking opportunities to explore for cancer risks or implications in the development, establishment, and survival of the cancer cell. The relationship between polymorphisms and the risk and prevalence of different types of cancer had evaluated by meta-analysis. Unfortunately, until now, the results are not conclusive.

Nevertheless, the fact is that IL-10 can exert the antitumor effect by mechanisms such as the activation of natural killer cells (NK), lymphocytes T, macrophages, and nitric oxide; its high concentration shows the deregulation of the immune system, where a high level of IL10 allows the tumor to escape. Nevertheless, the fact that IL-10 can exert the antitumor effect by mechanisms such as the activation of natural killer cells, lymphocytes T, macrophages, and nitric oxide results controversial. Its high concentration induces the deregulation of the immune system, where a high level of IL-10 allows the tumor escape. In some cases, it could be propose as a biomarker for patient lifespan or chemotherapy respond.

Because of this, it is essential to study not only the mechanisms that allow this high expression of interleukin IL-10, but its effect on tumor cells and also. It is important to consider its relationship with other cytokines.

Advertisement

Conflict of interest

The authors declare that they have no competing interests.

References

  1. 1. de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nature Reviews Cancer. 2006;6:24-37. DOI: 10.1038/nrc1782
  2. 2. Jordan WJ, Eskdale J, Boniotto M, Lennon GP, Peat J, Campbell JD, et al. Human IL-19 regulates immunity through auto-induction of IL-19 and production of IL-10. European Journal of Immunology. 2005;35:1576-1582. DOI: 10.1002/eji.200425317
  3. 3. Dai ZJ, Wang XJ, Zhao Y, Ma XB, Kang HF, Min WL, et al. Effects of interleukin-10 polymorphisms (rs1800896, rs1800871, and rs1800872) on breast cancer risk: Evidence from an updated meta-analysis. Genetic Testing and Molecular Biomarkers. 2014;18:439-445. DOI: 10.1089/gtmb.2014.0012
  4. 4. Fei C, Yao XM, Sun Y, Gu XZ, Yu LQ , Lai X. Interleukin-10 polymorphisms associated with susceptibility to acute myeloid leukemia. Genetics and Molecular Research. 2015;14:925-930. DOI: 10.4238/2015
  5. 5. Yang Y, Fa X. Role of IL-10 gene polymorphisms on the susceptibility for esophageal cancer and its association with environmental factors. International Journal of Clinical and Experimental Pathology. 2015;8:9580-9585
  6. 6. Moghimi M, Ahrar H, Karimi-Zarchi M, Aghili K, Salari M, Zare-Shehneh M, et al. Association of IL-10 rs1800871 and rs1800872 polymorphisms with breast cancer risk: A systematic review and meta-analysis. Asian Pacific Journal of Cancer Prevention. 2018;19:3353-3359. DOI: 10.31557/APJCP.2018.19.12.3353
  7. 7. Sharif OM, Hassan R, Mohammed Basbaeen AA, Mohmed AH, Ibrahim IK. Interleukin-10 (1082G/A) polymorphism is associated with susceptibility of acute myeloid leukemia patients in Sudanese population. Asian Pacific Journal of Cancer Prevention. 2019;20:1939-1943. DOI: 10.31557/APJCP.2019.20.7.1939
  8. 8. Vieira P, de Waal-Malefyt R, Dang MN, Johnson KE, Kastelein R, Fiorentino DF, et al. Isolation and expression of human cytokine synthesis inhibitory factor cDNA clones: Homology to Epstein-Barr virus open reading frame BCRFI. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:1172-1176. DOI: 10.1073/pnas.88.4.1172
  9. 9. Trinchieri G. Interleukin-10 production by effector T cells: Th1 cells show self control. The Journal of Experimental Medicine. 2007;204:239-243. DOI: 10.1084/jem.20070104
  10. 10. Said EA, Dupuy FP, Trautmann L, Zhang Y, Shi Y, El-Far M, et al. Programmed death-1-induced interleukin-10 production by monocytes impairs CD4+ T cell activation during HIV infection. Nature Medicine. 2010;16:452-459. DOI: 10.1038/nm.2106
  11. 11. Li X, Mai J, Virtue A, Yin Y, Gong R, Sha X, et al. IL-35 is a novel responsive anti-inflammatory cytokine--a new system of categorizing anti-inflammatory cytokines. PLoS One. 2012;7(3):e33628. DOI: 10.1371/journal.pone.0033628
  12. 12. Donnelly RP, Dickensheets H, Finbloom DS. The interleukin-10 signal transduction pathway and regulation of gene expression in mononuclear phagocytes. Journal of Interferon & Cytokine Research. 1999;19:563-573. DOI: 10.1089/107999099313695
  13. 13. Josephson K, Logsdon NJ, Walter MR. Crystal structure of the IL-10/IL-10R1 complex reveals a shared receptor binding site. Immunity. 2001;15:35-46. DOI: 10.1016/s1074-7613(01)00169-8
  14. 14. Saraiva M, O'Garra A. The regulation of IL-10 production by immune cells. Nature Reviews Immunology. 2010;10:170-181. DOI: 10.1038/nri2711
  15. 15. Gabryšová L, Howes A, Saraiva M, O'Garra A. The regulation of IL-10 expression. Current Topics in Microbiology and Immunology. 2014;380:157-190. DOI: 10.1007/978-3-662-43492-5_8
  16. 16. Kubo M, Motomura Y. Transcriptional regulation of the anti-inflammatory cytokine IL-10 in acquired immune cells. Frontiers in Immunology. 2012;3:275. DOI: 10.3389/fimmu.2012.00275
  17. 17. MacKenzie KF, Pattison MJ, Arthur JS. Transcriptional regulation of IL-10 and its cell-specific role in vivo. Critical Reviews in Immunology. 2014;34:315-345. DOI: 10.1615/critrevimmunol.2014010694
  18. 18. Teixeira-Coelho M, Guedes J, Ferreirinha P, Howes A, Pedrosa J, Rodrigues F, et al. Differential post-transcriptional regulation of IL-10 by TLR2 and TLR4-activated macrophages. European Journal of Immunology. 2014;44:856-866. DOI: 10.1002/eji.201343734
  19. 19. Moore KW, Rousset F, Banchereau J. Evolving principles in immunopathology: Interleukin 10 and its relationship to Epstein-Barr virus protein BCRF1. Seminars in Immunopathology. 1991;13:157-166. DOI: 10.1007/bf00201466
  20. 20. de Waal Malefyt R, Abrams J, Bennett B, Figdor CG, de Vries JE. Interleukin 10(IL-10) inhibits cytokine synthesis by human monocytes: An autoregulatory role of IL-10 produced by monocytes. The Journal of Experimental Medicine. 1991;174:1209-1220. DOI: 10.1084/jem.174.5.1209
  21. 21. Akdis CA, Joss A, Akdis M, Blaser K. Mechanism of IL-10-induced T cell inactivation in allergic inflammation and normal response to allergens. International Archives of Allergy and Immunology. 2001;124:180-182. DOI: 10.1159/000053704
  22. 22. Joss A, Akdis M, Faith A, Blaser K, Akdis CA. IL-10 directly acts on T cells by specifically altering the CD28 co-stimulation pathway. European Journal of Immunology. 2000;30:1683-1690. DOI: 10.1002/1521-4141(200006)30:6<1683:AID-IMMU1683>3.0.CO;2-A
  23. 23. Opp MR, Smith EM, Hughes TK Jr. Interleukin-10 (cytokine synthesis inhibitory factor) acts in the central nervous system of rats to reduce sleep. Journal of Neuroimmunology. 1995;60:165-168. DOI: 10.1016/0165-5728(95)00066-b
  24. 24. Aste-Amezaga M, Ma X, Sartori A, Trinchieri G. Molecular mechanisms of the induction of IL-12 and its inhibition by IL-10. Journal of Immunology. 1998;160:5936-5944
  25. 25. Varma TK, Toliver-Kinsky TE, Lin CY, Koutrouvelis AP, Nichols JE, Sherwood ER. Cellular mechanisms that cause suppressed gamma interferon secretion in endotoxin-tolerant mice. Infection and Immunity. 2001;69:5249-5263. DOI: 10.1128/iai.69.9.5249-5263.2001
  26. 26. Maynard CL, Weaver CT. Diversity in the contribution of interleukin-10 to T-cell-mediated immune regulation. Immunological Reviews. 2008;226:219-233. DOI: 10.1111/j.1600-065X.2008.00711.x
  27. 27. Veenbergen S, Li P, Raatgeep HC, Lindenbergh-Kortleve DJ, Simons-Oosterhuis Y, Farrel A, et al. IL-10 signaling in dendritic cells controls IL-1β-mediated IFNγ secretion by human CD4(+) T cells: Relevance to inflammatory bowel disease. Mucosal Immunology. 2019;12:1201-1211. DOI: 10.1038/s41385-019-0194-9
  28. 28. Verma R, Balakrishnan L, Sharma K, Khan AA, Advani J, Gowda H, et al. A network map of interleukin-10 signaling pathway. Journal of Cell Communication and Signaling. 2016;10:61-67. DOI: 10.1007/s12079-015-0302-x
  29. 29. McCarter MD, Baumgartner J, Escobar GA, Richter D, Lewis K, Robinson W, et al. Immunosuppressive dendritic and regulatory T cells are upregulated in melanoma patients. Annals of Surgical Oncology. 2007;14:2854-2860. DOI: 10.1245/s10434-007-9488-3
  30. 30. Rousset F, Garcia E, Defrance T, Péronne C, Vezzio N, Hsu DH, et al. Interleukin 10 is a potent growth and differentiation factor for activated human B lymphocytes. Proceedings of the National Academy of Sciences of the United States of America. 1992;89:1890-1893. DOI: 10.1073/pnas.89.5.1890
  31. 31. Armitage RJ, Macduff BM, Spriggs MK, Fanslow WC. Human B cell proliferation and Ig secretion induced by recombinant CD40 ligand are modulated by soluble cytokines. Journal of Immunology. 1993;150:3671-3680
  32. 32. Briere F, Servet-Delprat C, Bridon JM, Saint-Remy JM, Banchereau J. Human interleukin 10 induces naive surface immunoglobulin D+ (sIgD+) B cells to secrete IgG1 and IgG3. The Journal of Experimental Medicine. 1994;179:757-762. DOI: 10.1084/jem.179.2.757
  33. 33. Itoh K, Hirohata S. The role of IL-10 in human B cell activation, proliferation, and differentiation. Journal of Immunology. 1995;154:4341-4350
  34. 34. Moens L, Tangye SG. Cytokine-mediated regulation of plasma cell generation: IL-21 takes center stage. Frontiers in Immunology. 2014;5:65. DOI: 10.1007/s13277-014-2589-2
  35. 35. Dummer W, Becker JC, Schwaaf A, Leverkus M, Moll T, Bröcker EB. Elevated serum levels of interleukin-10 in patients with metastatic malignant melanoma. Melanoma Research. 1995;5:67-68. DOI: 10.1097/00008390-199502000-00008
  36. 36. Dummer W, Bastian BC, Ernst N, Schänzle C, Schwaaf A, Bröcker EB. Interleukin-10 production in malignant melanoma: Preferential detection of IL-10-secreting tumor cells in metastatic lesions. International Journal of Cancer. 1996;66:607-610. DOI: 10.1002/(SICI)1097-0215(19960529)66:5<607::AID-IJC4>3.0.CO;2-X
  37. 37. Zhao H, Yang J, Yu Z, Shen H, Huang X, Zhang M, et al. Synthetic analysis of associations between IL-10 polymorphisms and skin cancer risk. Oncotarget. 2017;9:6728-6736. DOI: 10.18632/oncotarget.23385
  38. 38. Zhao S, Wu D, Wu P, Wang Z, Huang J. Serum IL-10 predicts worse outcome in cancer patients: A meta-analysis. PLoS One. 2015;10:e0139598. DOI: 10.1371/journal.pone.0139598
  39. 39. Nemunaitis J, Fong T, Shabe P, Martineau D, Ando D. Comparison of serum interleukin-10 (IL-10) levels between normal volunteers and patients with advanced melanoma. Cancer Investigation. 2001;19:239-247. DOI: 10.1081/cnv-100102550
  40. 40. Ikeguchi M, Hatada T, Yamamoto M, Miyake T, Matsunaga T, Fukumoto Y, et al. Serum interleukin-6 and -10 levels in patients with gastric cancer. Gastric Cancer. 2009;12:95-100. DOI: 10.1007/s10120-009-0509-8
  41. 41. Fortis C, Foppoli M, Gianotti L, Galli L, Citterio G, Consogno G, et al. Increased interleukin-10 serum levels in patients with solid tumours. Cancer Letters. 1996;104:1-5. DOI: 10.1016/0304-3835(96)04213-9
  42. 42. Danilova AB, Danilov AO, Fakhrutdinova OL, Baldueva IA, Moiseenko VM. Laboratory evaluation of TGFbeta1, IL-10, VEGF levels in vivo and in vitro in patients with solid tumors. Voprosy Onkologii. 2011;57:759-766
  43. 43. Hsu TI, Wang YC, Hung CY, Yu CH, Su WC, Chang WC, et al. Positive feedback regulation between IL10 and EGFR promotes lung cancer formation. Oncotarget. 2016;7:20840-20854. DOI: 10.18632/oncotarget.7894. 8-870915
  44. 44. Bhattacharjee HK, Bansal VK, Nepal B, Srivastava S, Dinda AK, Misra MC. Is interleukin 10 (IL10) expression in breast cancer a marker of poor prognosis? Indian Journal of Surgical Oncology. 2016;7:320-325. DOI: 10.1007/s13193-016-0512-6
  45. 45. Liu C, Sun B, Xu B, Meng X, Li L, Cong Y, et al. A panel containing PD-1, IL-2Rα, IL-10, and CA15-3 as a biomarker to discriminatebreast cancer from benign breast disease. Cancer Management and Research. 2018;10:1749-1761. DOI: 10.2147/CMAR.S160452
  46. 46. Bohlen H, Kessler M, Sextro M, Diehl V, Tesch H. Poor clinical outcome of patients with Hodgkin's disease and elevated interleukin-10 serum levels. Clinical significance of interleukin-10 serum levels for Hodgkin's disease. Annals of Hematology. 2000;79:110-113. DOI: 10.1007/s002770050564
  47. 47. Vera-Lozada G, Minnicelli C, Segges P, Stefanoff G, Kristcevic F, Ezpeleta J, et al. Interleukin 10 (IL10) proximal promoter polymorphisms beyond clinical response in classical Hodgkin lymphoma: Exploring the basis for the genetic control of the tumor microenvironment. Oncoimmunology. 2018;7:e1389821. DOI: 10.1080/2162402X.2017.1389821
  48. 48. Hsi ED, Li H, Nixon AB, Schöder H, Bartlett NL, LeBlanc M, et al. Serum levels of TARC, MDC, IL-10, and soluble CD163 in Hodgkin lymphoma: A SWOG S0816 correlative study. Blood. 2019;133:1762-1765. DOI: 10.1182/blood-2018-0
  49. 49. Blay JY, Burdin N, Rousset F, Lenoir G, Biron P, Philip T, et al. Serum interleukin-10 in non-Hodgkin's lymphoma: A prognostic factor. Blood. 1993;82:2169-2174
  50. 50. Cortes J, Kurzrock R. Interleukin-10 in non-Hodgkin's lymphoma. Leukemia & Lymphoma. 1997;26:251-259. DOI: 10.3109/10428199709051774
  51. 51. Shokrzadeh M, Mohammadpour A, Hoseini V, Abediankenari S, Ghassemi-Barghi N, Tabari YS. Serum cytokine of il-2, il-10 and il-12 levels in patients with stomach adenocarcinoma. Archives of Gastroenterology. 2018;55:385-389. DOI: 10.1590/S0004-2803.201800000-83
  52. 52. De Vita F, Orditura M, Galizia G, Romano C, Infusino S, Auriemma A, et al. Serum interleukin-10 levels in patients with advanced gastrointestinal malignancies. Cancer. 1999;86:1936-1943
  53. 53. Szaflarska A, Szczepanik A, Siedlar M, Czupryna A, Sierzega M, Popiela T, et al. Preoperative plasma level of IL-10 but not of proinflammatory cytokines is an independent prognostic factor in patients with gastric cancer. Anticancer Research. 2009;29:5005-5012
  54. 54. Feng L, Qi Q , Wang P, Chen H, Chen Z, Meng Z, et al. Serum levels of IL-6, IL-8, and IL-10 are indicators of prognosis in pancreatic cancer. The Journal of International Medical Research. 2018;46:5228-5236. DOI: 10.1177/0300060518800588
  55. 55. Ebrahimi B, Tucker SL, Li D, Abbruzzese JL, Kurzrock R. Cytokines in pancreatic carcinoma: Correlation with phenotypic characteristics and prognosis. Cancer. 2004;101:2727-2736. DOI: 10.1002/cncr.20672
  56. 56. Li B, Wang F, Ma C, Hao T, Geng L, Jiang H. Predictive value of IL-18 and IL-10 in the prognosis of patients with colorectal cancer. Oncology Letters. 2019;18:713-719. DOI: 10.3892/ol.2019.10338
  57. 57. Stanilov N, Miteva L, Deliysky T, Jovchev J, Stanilova S. Advanced colorectal cancer is associated with enhanced IL-23 and IL-10 serum levels. Labmedicine. 2010;41:159-163
  58. 58. Chau GY, Wu CW, Lui WY, Chang TJ, Kao HL, Wu LH, et al. Serum interleukin-10 but not interleukin-6 is related to clinical outcome in patients with resectable hepatocellular carcinoma. Annals of Surgery. 2000;231:552-558
  59. 59. Sarris AH, Kliche KO, Pethambaram P, Preti A, Tucker S, Jackow C, et al. Interleukin-10 levels are often elevated in serum of adults with Hodgkin's disease and are associated withinferior failure-free survival. Annals of Oncology. 1999;10:433-440. DOI: 10.1023/a:1008301602785
  60. 60. Visco C, Vassilakopoulos TP, Kliche KO, Nadali G, Viviani S, Bonfante V, et al. Elevated serum levels of IL-10 are associated with inferior progression-free survival in patients with Hodgkin's disease treated with radiotherapy. Leukemia & Lymphoma. 2004;45:2085-2092. DOI: 10.1080/10428190410001712234
  61. 61. Vassilakopoulos TP, Nadali G, Angelopoulou MK, Siakantaris MP, Dimopoulou MN, Kontopidou FN, et al. Serum interleukin-10 levels are an independent prognostic factor for patients with Hodgkin's lymphoma. Haematologica. 2001;86:274-281
  62. 62. Viviani S, Notti P, Bonfante V, Verderio P, Valagussa P, Bonadonna G. Elevated pretreatment serum levels of Il-10 are associated with a poor prognosis in Hodgkin's disease, the Milan cancer institute experience. Medical Oncology. 2000;17:59-63. DOI: 10.1007/bf02826218
  63. 63. De Vita F, Orditura M, Galizia G, Romano C, Roscigno A, Lieto E, et al. Serum interleukin-10 levels as a prognostic factor in advanced non-small cell lung cancer patients. Chest. 2000;117:365-373. DOI: 10.1378/chest.117.2.365
  64. 64. Wang H, Wang L, Chi PD, Wang WD, Chen XQ , Geng QR, et al. High level of interleukin-10 in serum predicts poor prognosis in multiple myeloma. British Journal of Cancer. 2016;114:463-468. DOI: 10.1038/bjc.2016.11
  65. 65. Shekarriz R, Janbabaei G, Abedian Kenari S. Prognostic value of IL-10 and its relationship with disease stage in Iranian patients with multiple myeloma. Asian Pacific Journal of Cancer Prevention. 2018;19:27-32. DOI: 10.22034/APJCP.2018.19.1.27
  66. 66. Gupta M, Han JJ, Stenson M, Maurer M, Wellik L, Hu G, et al. Elevated serum IL-10 levels in diffuse large B-cell lymphoma: A mechanism of aberrant JAK2 activation. Blood. 2012;119:2844-2853. DOI: 10.1182/blood-2011-10-388538
  67. 67. Fayad L, Keating MJ, Reuben JM, O'Brien S, Lee BN, Lerner S, et al. Interleukin-6 and interleukin-10 levels in chronic lymphocytic leukemia: Correlation with phenotypic characteristics and outcome. Blood. 2001;97:256-263. DOI: 10.1182/blood.v97.1.256
  68. 68. Nakagomi H, Pisa P, Pisa EK, Yamamoto Y, Halapi E, Backlin K, et al. Lack of i international journal of cancer nterleukin-2 (IL-2) expression and selective expression of IL-10 mRNA in human renal cell carcinoma. International Journal of Cancer. 1995;63:366-371. DOI: 10.1002/ijc.2910630311
  69. 69. Welsh TJ, Green RH, Richardson D, Waller DA, O’Byrne KJ, Bradding P. Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer. Journal of Clinical Oncology. 2005;23:8959-8967. DOI: 10.1200/JCO.2005.01.4910
  70. 70. Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Research. 2006;66:605-612. DOI: 10.1158/0008-5472.CAN-05-4005
  71. 71. Redente EF, Orlicky DJ, Bouchard RJ, Malkinson AM. Tumor signaling to the bone marrow changes the phenotype of monocytes and pulmonary macrophages during urethane-induced primary lung tumorigenesis in a/J mice. The American Journal of Pathology. 2007;170:693-708. DOI: 10.2353/ajpath.2007.060566
  72. 72. Zeni E, Mazzetti L, Miotto D, Lo Cascio N, Maestrelli P, Querzoli P, et al. Macrophage expression of interleukin-10 is a prognostic factor in nonsmall cell lung cancer. The European Respiratory Journal. 2007;30:627-632. DOI: 10.1183/09031936.00129306
  73. 73. Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against cancer: A counterpoint. Journal of Leukocyte Biology. 2005;78:1043-1051. DOI: 10.1189/jlb.0705358
  74. 74. Hatanaka H, Abe Y, Kamiya T, Morino F, Nagata J, Tokunaga T, et al. Clinical implications of interleukin (IL)-10 induced by non-small-cell lung cancer. Annals of Oncology. 2000;11:815-819. DOI: 10.1023/a:1008375208574
  75. 75. Vahl JM, Friedrich J, Mittler S, Trump S, Heim L, Kachler K, et al. Interleukin-10-regulated tumour tolerance in non-small cell lung cancer. British Journal of Cancer. 2017;117:1644-1655. DOI: 10.1038/bjc.2017.336
  76. 76. Howell MW, Rose-zerilli MJ. Cytokine gene polymorphisms, cancer susceptibility and prognosis. Journal of Nutrition. 2007;137(Suppl 1):194-199
  77. 77. Korobeinikova E, Myrzaliyeva D, Ugenskiene R, Raulinaityte D, Gedminaite J, Smigelskas K, et al. The prognostic value of IL10 and TNF alpha functional polymorphisms in premenopausal early-stage breast cancer patients. BMC Genetics. 2015;16:70. DOI: 10.1186/s12863-015-0234-8
  78. 78. Knechtel G, Hofmann G, Gerger A, Renner W, Langsenlehner T, Szkandera J, et al. Analysis of common germline polymorphisms as prognostic factors in patients with lymph node-positive breast cancer. Journal of Cancer Research and Clinical Oncology. 2010;136:1813-1819. DOI: 10.1007/s00432-010-0839-2
  79. 79. Kong F, Liu J, Liu Y, Song B, Wang H, Liu W. Association of interleukin-10 gene polymorphisms with breast cancer in a Chinese population. Journal of Experimental & Clinical Cancer Research. 2010;29:72. DOI: 10.1186/1756-9966-29-72
  80. 80. Liu J, Song B, Bai X, Liu W, Li Z, Wang J, et al. Association of genetic polymorphisms in the interleukin-10 promoter with risk of prostate cancer in Chinese. BMC Cancer. 2010;10:456. DOI: 10.1186/1471-2407-10-456
  81. 81. Zou YF, Wang F, Feng XL, Tian YH, Tao JH, Pan FM, et al. Lack of association of IL-10 gene polymorphisms with prostate cancer: Evidence from 11,581 subjects. European Journal of Cancer. 2011;47:1072-1079. DOI: 10.1016/j.ejca.2010.11.034
  82. 82. Shao N, Xu B, Mi YY, Hua LX. IL-10 polymorphisms and prostate cancer risk: A meta-analysis. Prostate Cancer and Prostatic Diseases. 2011;14:129-135. DOI: 10.1038/pcan.2011.6
  83. 83. Men T, Yu C, Wang D, Liu F, Li J, Qi X, et al. The impact of interleukin-10 (IL-10) gene 4 polymorphisms on peripheral blood IL-10 variation and prostate cancer risk based on published studies. Oncotarget. 2017;8:45994-46005. DOI: 10.18632/oncotarget.17522
  84. 84. Chen H, Tang J, Shen N, Ren K. Interleukin 10 gene rs1800896 polymorphism is associated with the risk of prostate cancer. Oncotarget. 2017;8:66204-66214. DOI: 10.18632/oncotarget.19857
  85. 85. Wu N, Sun H, Sun Q , Cui M, Jiang R, Cong X. Associations between IL-10 polymorphisms and susceptibility to melanoma, basal cell carcinoma, and squamous cell carcinoma: A meta-analysis. Genetic Testing and Molecular Biomarkers. 2018;22:693-701. DOI: 10.1089/gtmb.2018.0172
  86. 86. Liu L, Zheng F. IL-10 -1082A/G, -592C/a, and -819T/C polymorphisms in association with lung cancer susceptibility: A meta-analysis. Onco Targets Therapy. 2016;9:6083-6091. DOI: 10.2147/OTT.S118481
  87. 87. Jafari-Nedooshan J, Moghimi M, Zare M, Heiranizadeh N, Morovati-Sharifabad M, Akbarian-Bafghi MJ, et al. Association of promoter region polymorphisms of IL-10 gene with susceptibility to lung cancer: Systematic review and meta-analysis. Asian Pacific Journal of Cancer Prevention. 2019;20:1951-1957. DOI: 10.31557/APJCP.2019.20.7.1951
  88. 88. Wang Y, Liu XH, Li YH, Li O. The paradox of IL-10-mediated modulation in cervical cancer. Biomedical Reports. 2013;1:347-351. DOI: 10.3892/br.2013.69
  89. 89. Guo C, Wen L, Song JK, Zeng WJ, Dan C, Niu YM, et al. Significant association between interleukin-10 gene polymorphisms and cervical cancer risk: A meta-analysis. Oncotarget. 2018;9:12365-12375. DOI: 10.18632/oncotarget.24193
  90. 90. Zhang S, Kong YL, Li YL, Yin YW. Interleukin-10 gene −1082 G/a polymorphism in cervical cancer and cervical intraepithelial neoplasia: Meta-analysis. The Journal of International Medical Research. 2014;42:1193-1201. DOI: 10.1177/0300060514544388
  91. 91. Huang W, Song J, Jia XW, Chen YX, Shi J, Jiang X. Interleukin-10 rs1800896 polymorphism is associated with increased head and neck cancer risk but not associated with its clinical stages. Oncotarget. 2017;8:37217-37224. DOI: 10.18632/oncotarget.16660
  92. 92. Niu YM, Du XY, Cai HX, Zhang C, Yuan RX, Zeng XT, et al. Increased risks between Interleukin-10 gene polymorphisms and haplotype and head and neck cancer: A meta-analysis. Scientific Reports. 2015;5:17149. DOI: 10.1038/srep17149
  93. 93. Martínez-Campos C, Torres-Poveda K, Camorlinga-Ponce M, Flores-Luna L, Maldonado-Bernal C, Madrid-Marina V, et al. Polymorphisms in IL-10 and TGF-β gene promoter are associated with lower risk to gastric cancer in a Mexican population. BMC Cancer. 2019;19:453. DOI: 10.1186/s12885-019-5627-z
  94. 94. Singh PK, Ahmad MK, Kumar V, Gupta R, Kohli M, Jain A, et al. Genetic polymorphism of interleukin-10 (-A592C) among oral cancer with squamous cell carcinoma. Archives of Oral Biology. 2017;77:18-22. DOI: 10.1016/j.archoralbio.2016.12.011
  95. 95. Mirjalili SA, Moghimi M, Aghili K, Jafari M, Abolbaghaei SM, Neamatzadeh H, et al. Association of promoter region polymorphisms of interleukin-10 gene with susceptibility to colorectal cancer: A systematic review and meta-analysis. Arquivos de Gastroenterologia. 2018;55:306-313. DOI: 10.1590/S0004-2803.201800000-66
  96. 96. Hohaus S, Giachelia M, Di Febo A, Martini M, Massini G, Vannata B, et al. Polymorphism in cytokine genes as prognostic markers in Hodgkin’s lymphoma. Annals of Oncology. 2007;18:1376-1381. DOI: 10.1093/annonc/mdm132
  97. 97. Rashed R, Shafik RE, Shafik NF, Shafik HE. Associations of interleukin-10 gene polymorphisms with acute myeloid leukemia in human (Egypt). Journal of Cancer Research and Therapeutics. 2018;14:1083-1086. DOI: 10.4103/0973-1482.187367
  98. 98. Yao CJ, Du W, Chen HB, Xiao S, Wang CH, Fan ZL. Associations of IL-10 gene polymorphisms with acute myeloid leukemia in Hunan, China. Asian Pacific Journal of Cancer Prevention. 2013;14:2439-2442. DOI: 10.7314/apjcp.2013.14.4.2439

Written By

Maria Teresa Gonzalez-Garza, Delia Elva Cruz-Vega and Carmen Maldonado-Bernal

Submitted: 30 September 2019 Reviewed: 09 December 2019 Published: 08 June 2020