Hostname: page-component-848d4c4894-x24gv Total loading time: 0 Render date: 2024-04-30T18:43:25.741Z Has data issue: false hasContentIssue false

Pharmacogenomic and epigenomic approaches to untangle the enigma of IL-10 blockade in oncology

Published online by Cambridge University Press:  08 January 2024

Noha M. Elemam
Affiliation:
Research Instiute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
Radwa Y. Mekky
Affiliation:
Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
Gowhar Rashid
Affiliation:
Amity Medical School, Amity University, Gurugram (Manesar) 122413, Haryana, India
Maria Braoudaki
Affiliation:
Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
Rana A. Youness*
Affiliation:
Biology and Biochemistry Department, Faculty of Biotechnology, German International University, Cairo 11835, Egypt
*
Corresponding author: Rana A. Youness; Email: rana.youness21@gmail.com
Rights & Permissions [Opens in a new window]

Abstract

The host immune system status remains an unresolved mystery among several malignancies. An immune-compromised state or smart immune-surveillance tactics orchestrated by cancer cells are the primary cause of cancer invasion and metastasis. Taking a closer look at the tumour-immune microenvironment, a complex network and crosstalk between infiltrating immune cells and cancer cells mediated by cytokines, chemokines, exosomal mediators and shed ligands are present. Cytokines such as interleukins can influence all components of the tumour microenvironment (TME), consequently promoting or suppressing tumour invasion based on their secreting source. Interleukin-10 (IL-10) is an interlocked cytokine that has been associated with several types of malignancies and proved to have paradoxical effects. IL-10 has multiple functions on cellular and non-cellular components within the TME. In this review, the authors shed the light on the regulatory role of IL-10 in the TME of several malignant contexts. Moreover, detailed epigenomic and pharmacogenomic approaches for the regulation of IL-10 were presented and discussed.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Introduction

The use of immunotherapy as a novel therapeutic approach in preventing cancer has become widespread (Ref. Reference Elemam1). Immune checkpoint blockade modalities targeting PD-1 and CTLA-4 provide long-lasting immune responses with established therapeutic benefits for some cancer patients (Refs Reference Sponghini2Reference Selem6). Although, targeting cytokines is considered a crucial approach in immunotherapy as evidenced in the treatment of solid tumours, such as renal cell carcinoma (RCC) and melanoma, only interferons (IFNs) and IL-2 have been approved by Food and Drug Administration (FDA) for use as cancer therapies (Ref. Reference Conlon, Miljkovic and Waldmann7).

IL-10 is considered one of the very promising targets for immunotherapy; however, its controversial role in carcinogenesis hinders the applicability of benefiting from its blockade in cancer treatment (Ref. Reference Soliman8). IL-10 has been shown to possess both anti- and pro-inflammatory roles in cancer (Ref. Reference Mocellin9). The intensity of the immunological response to both self and foreign antigens is reduced by IL-10. In light of this, IL-10 signalling blockage improves vaccine-induced T-cell responses and tumour growth inhibition (Ref. Reference Shen10). On the other hand, tumour regression is also induced by exogenous IL-10, particularly PEGylated (PEG)-IL-10 (Ref. Reference Mumm and Oft11). This paradoxical data urges the need to investigate the role of pharmacogenomics, epigenetics and genetic variants in IL-10 and its receptor to identify those patients that might benefit from IL-10 targeted therapies. In this review, the authors will address the role of IL-10 in cancer, the currently available IL-10-based immunotherapy, the epigenetic regulation of IL-10 and the single nucleotide polymorphisms (SNPs) present in IL-10 that might influence patient responses to therapy.

The tumour microenvironment

Cancer definition has been revolutionized over the past few decades from the concept of being abnormal cells to a plethora of complex network that is made up of both neoplastic cells with their surrounding stroma (Refs Reference Elemam1, Reference Ramzy4, Reference Selem6, Reference Hanahan and Weinberg12). The multifaceted dynamic milieu of cellular components along with non-cellular compartments portrays what is now known as the tumour microenvironment (TME) (Refs Reference Selem6, Reference Ocana13, Reference Youness14). Such a microenvironment could control the aggressiveness, rate of growth and metastatic potential of the tumour (Refs Reference Nyberg, Salo and Kalluri15Reference Youness18). These cellular components include immune cells such as T lymphocytes (Refs Reference Fridman19Reference Tan24), regulatory T cells (Tregs) (Ref. Reference Tan25), B lymphocytes, natural killer (NK) cells (Refs Reference Mekky16, Reference Baek26Reference Awad29), mesenchymal stem cells (Refs Reference Hall, Andreeff and Marini30, Reference Jimenez31), tumour-associated-macrophages (Refs Reference Guerriero32, Reference Van Overmeire33), tumour-associated neutrophils (Refs Reference Hurt34, Reference Mensurado35), dendritic cells (DCs) (Ref. Reference Hansen and Andersen36) and non-immune cells such as pericytes (Ref. Reference Guerra37), adipocytes (Refs Reference Reina-Campos38, Reference Akutagawa39), myeloid-derived suppressor cells (MDSCs) (Refs Reference Kumar40Reference Shen42) and cancer-associated fibroblastic cells (Refs Reference Ahirwar43, Reference Liotta and Kohn44). Interestingly, these immune cells drive the production of soluble components that include cytokines, chemokines, growth factors and extra-cellular remodelling enzymes (Refs Reference Youness27, Reference Rahmoon28). Such mediators, particularly cytokines, assist in the communication between the cellular TME components and cancer cells as shown in Figure 1 (Refs Reference Burkholder45, Reference Zigrino, Loffek and Mauch46).

Figure 1. Snapshot of cellular and non-cellular components of the tumour microenvironment

Interleukin-10 (IL-10)

One of these cytokines is the paradoxical interleukin ‘IL-10’, which remains an integral part of several malignancies, and regulates the secretion of other cytokines. This pleiotropic cytokine was characterized early in the late 1980s and was named cytokine synthesis inhibitory factor (Refs Reference Fiorentino, Bond and Mosmann47, Reference Moore48). Later on, six immune mediators (IL-10, IL-19, IL-20, IL-22, IL-24 and IL-26) were grouped into the IL-10 family of cytokines based on their similarities with respect to the structure and location of their encoding genes, their primary and secondary protein structures and the receptor complexes (Refs Reference Kotenko49Reference Fickenscher51). Out of these six members, IL-10 has been recognized as a major member mediating different functions within the immune system and cancer cells (Ref. Reference Mannino52).

Paradoxical role of IL-10 in oncology

IL-10 produced by immune cells

IL-10 has also been causally linked to immunity in both the innate and adaptive immune arms. Different triggers have been shown to induce IL-10 production in various immune cells (Ref. Reference Nagpal53). The main source of IL-10 appears to be monocytes, and different T-cell subsets (Ref. Reference Sabat54). Moreover, DCs, B cells, NK cells, mast cells, as well as neutrophils, and eosinophils can also synthesize IL-10 (Ref. Reference Sabat54). During infection, macrophages are considered a major source of IL-10. Several toll-like receptors (TLRs), including TLR2, TLR4, TLR5, TLR7 and TLR9 have been shown to induce IL-10 production in macrophages and DCs (Refs Reference Agrawal55Reference Rogers63). Also, IL-10 production in DCs is enhanced by the co-activation of TLR2 and Dectin-1 (Ref. Reference Dennehy64). Following exposure to IL-10, DCs can initiate the development of regulatory T cells (Tregs) that limit these effector responses (Refs Reference Kapsenberg65, Reference Mills66). B cells also express several TLRs which have been shown to promote IL-10 production including TLR2, TLR4 or TLR9 (Refs Reference Agrawal and Gupta67Reference Sayi69). Nonetheless, it is also worth mentioning that IFN-α augments IL-10 production if combined with TLR agonists from B cells (Refs Reference Shaw70, Reference Zhang71). Additionally, neutrophils produce IL-10 in response to TLR and C-type lectin co-activation through myeloid differentiation primary response 88 (MyD88) and spleen tyrosine kinase (SYK), respectively (Ref. Reference Zhang72).

The key producer of IL-10 is Treg cells that produce other immunoregulatory cytokines, such as TGF-β (Ref. Reference Peppa73). The production and action of both cytokines IL-10 and TGF-β are involved in a positive feedback loop (Ref. Reference Cottrez and Groux74). Concerning the mechanism of IL-10 production from Tregs, it has been shown that IL-2 and IL-4 induce IL-10 production from Tregs (Refs Reference Barthlott75Reference de la Rosa77). Additionally, a study concluded that TGF-β is required for the differentiation and production of IL-10 from Tregs (Ref. Reference Maynard78). IL-2 and IL-27 are responsible for inducing IL-10 expression in cytotoxic CD8+ T cells (Ref. Reference Sun79). However, IL-12 and IL-23 prime CD8+ and CD4+ T cells for IL-10 production (Refs Reference Gerosa80Reference Meyaard82).

Some studies reported IL-10 immunosuppressive effects such as inhibiting IFN-γ and TNF-α production by NK cells in-vitro (Ref. Reference Moore83). However, other studies reported IL-10 immunostimulatory effects via the promotion of NK cell cytotoxicity in preclinical models (Refs Reference Mocellin9, Reference Mocellin84). Adding to the complexity of this master cytokine, one of the studies has shown that the exposure of malignant cells to IL-10 resulted in a reduction in their sensitivity to cytotoxic T cells but an increase in NK cell cytotoxicity (Ref. Reference Abd Razak85). This might suggest that IL-10 contributes to fighting malignant cells by stimulating the immune innate arm (Ref. Reference Kelly86).

As mentioned earlier, one of the main drivers of IL-10 expression in many immune cells is TLR signalling (Ref. Reference Boonstra56). TLR ligation leads to the activation of several downstream pathways, including the mitogen-activated protein kinase (MAPK) pathway and the phosphoinositide 3-kinases (PI3K)/AKT pathways (Ref. Reference Kawai and Akira87). Activation of the MAPK and downstream extracellular-signal-regulated kinase (ERK1 and ERK2) are critical for IL-10 production in macrophages and DCs in response to several TLR activators (Refs Reference Dillon58, Reference Kaiser62, Reference Banerjee88, Reference Yi89). The MAPK pathway eventually results in the activation of several transcription family members such as the activator protein-1 (AP-1) which activates IL-10 transcription (Refs Reference Agrawal55, Reference Dillon58, Reference Kaiser62, Reference Hu90). Moreover, ERK and p38 also contribute to IL-10 production in TLR-stimulated macrophages, monocytes, and DCs (Refs Reference Chi57, Reference Yi89Reference Kim92). Both ERK and p38 may function cooperatively in their regulation of IL-10 production, through their joined activation of mitogen and stress-activated protein kinases (MSK1 and MSK2) which promote IL-10 production in TLR-stimulated macrophages. Downstream of MSK1 and MSK2 are the transcription factors, cAMP-response element binding protein (CREB), and AP-1, which also bind and transactivate the IL-10 promoter (Refs Reference Saraiva and O'Garra93Reference Kallies95). Moreover, it is worth mentioning that both ERK and p38 were shown also to directly phosphorylate Sp1, one of the IL-10 transcription factors (Refs Reference D'Addario, Arora and McCulloch96, Reference Tan and Khachigian97).

The phosphatidylinositol-3-kinase (PI3K/AKT) pathway also contributes to IL-10 expression in myeloid cells either by antagonizing glycogen synthase kinase 3 beta (GSK3-β), a constitutively active kinase that inhibits the production of IL-10 or through ERK and mammalian target of rapamycin (mTOR) and STAT-3 activation (Refs Reference Asnagli, Afkarian and Murphy98Reference Oliver100).

IL-10 produced by cancer cells

IL-10 has been linked to many types of cancers such as gastric cancer (Ref. Reference Sanchez-Zauco101), cervical cancer (Ref. Reference Berti102), lung cancer (Ref. Reference Vahl103), breast cancer (Ref. Reference Alotaibi104), colon adenocarcinoma (Ref. Reference Townsend105), head and neck cancer (Ref. Reference Bornstein106), oesophageal cancer, nasopharyngeal cancer, oral cancer (Ref. Reference Li, Yang and Li107) and colorectal cancer (Ref. Reference Abtahi108). Its role in tumourigenesis is reported to be controversial where it could be a tumour suppressor or promoter. However, due to the complex nature of IL-10, its role in shaping the TME remains a gap that needs further research. Most of the literature is directed towards presenting the pro-tumoural activity of IL-10 in different oncological settings. This could be through the positive feedback loop with STAT-3, as IL-10 has been shown to activate STAT-3 resulting in the upregulation of B-cell lymphoma 2 (BCL-2) or B-cell lymphoma-extra-large (BCL-xL), and stimulation of cell proliferation by cyclins D1, D2, B, and proto-oncogene c-Myc, thus contributing to cancer progression (Ref. Reference Saraiva and O'Garra93). On the other hand, IL-10 immunosuppressive activity has been reported on macrophages and DCs, where it was found to dampen antigen presentation, cell maturation, and differentiation resulting in tumour immune evasion as shown in Figure 2 (Ref. Reference Shrihari109). Several studies have examined the role of IL-10 in different types of malignancies as listed in Table 1 below.

Figure 2. Paradoxical pro- and anti-tumour roles of IL-10 in oncology

Table 1. Role of IL-10 in different solid malignancies

Previous studies highlighted a significant correlation between IL-10 and the percentage of plasma cells in multiple myeloma patients as it induces the proliferation of plasma cells (Refs Reference Shekarriz, Janbabaei and Abedian Kenari117Reference Kovacs119). Other studies indicated an elevation of IL-10 in different haematological malignancies such as Hodgkin lymphoma and non-Hodgkin lymphoma (Refs Reference Gupta120, Reference Visco121). High IL-10 levels were reported to be associated with a shorter survival rate among patients with diffuse large-cell lymphoma (Ref. Reference Gupta120). Similarly, high IL-10 levels was found to be a prognostic factor in peripheral T cell lymphoma, which can lead to worsening of overall survival, low complete response rate, and higher early relapse rate (Ref. Reference Zhang122). Moreover, elevated IL-10 at diagnosis was found to be an independent prognostic marker in adult hemophagocytic lymphohistiocytosis patients in order to find the right treatment strategy (Ref. Reference Zhou123).

The riddle of IL-10 at the tumour-immune cell synapse

The balance between pro-inflammatory and anti-inflammatory signals is generally crucial for the maintenance of normal physiology and the prevention of cancer and a wide variety of diseases (Refs Reference Youness14, Reference Abdallah124Reference Youness126). In the context of IL-10, it plays a dual function acting either as a pro-inflammatory or an anti-inflammatory mediator (Ref. Reference Hatanaka127). Regarding its role in cancer, studies have reported that IL-10, secreted by tumours or tumour-infiltrating immune cells, has allowed malignant cells to escape from the immune surveillance (Refs Reference Marincola128Reference Mapara and Sykes130). In a study by Neven et al., IL-10 knockout in mice promoted the development of colon cancer. Moreover, the same study showed that humans deficient in IL-10 signalling molecules were more prone to develop lymphomas at a younger age (Ref. Reference Neven131). As an anti-inflammatory cytokine, IL-10 is considered crucial for the homoeostasis of the anti-inflammatory Tregs and the suppression of proinflammatory IL-17-expressing T cells. However, IL-10 action depends on multiple factors such as targeted cells, other stimuli, and the time and duration of its effect (Ref. Reference Couper, Blount and Riley132). Though, with many rationales presented, a question mark continues to rise to explore the nature of this complex cytokine.

Is IL-10 blockade a possible option as a novel immunotherapeutic approach for cancer patients?

Controversial data exists regarding the effectiveness of IL-10 immunotherapy in cancer (Ref. Reference Nafea133). Cancer vaccines that utilized monoclonal antibody (mAb) against IL-10 receptors succeeded to increase CD8+ T cell responses and to inhibit tumour growth whether injected intraperitoneally or subcutaneously (Refs Reference Ni134, Reference Chen135). The beneficial effect of IL-10 blockade is best explained through the inhibition of IL-10-induced suppression of DCs and prevention of their antigen presentation capacity by decreasing the expression of MHC class II and co-stimulator molecules (Ref. Reference Llopiz136). Thus, DC-based vaccinations that disrupt IL-10 signalling provide more potent anti-tumour responses (Ref. Reference Llopiz136). On the contrary, others claimed that antibodies targeting IL-10R had no protective effect against tumour growth when used with vaccines containing adjuvants that do not induce IL-10, such as the TLR3 ligand poly (I: C) or anti-CD40 agonistic antibodies (Ref. Reference Llopiz137). Such a controversy regarding the effectiveness of therapeutic immunization could be explained and summarized by vaccine-induced IL-10 rather than IL-10 produced by tumours (Ref. Reference Llopiz137).

It was previously reported that the prognosis of cancer patients is inversely correlated with elevated serum and tumour IL-10 levels (Ref. Reference Soliman138). Despite that, exogenous administration of IL-10 was tested in clinical studies, and resulted in immunological activation, as evidenced by higher granzymes and IFN in the serum of those patients receiving treatment. Pegylated recombinant (PEG) murine IL-10 promoted rejection of tumours and metastases by enhancing CD8 + T cell-mediated immune responses (Ref. Reference Mumm and Oft139). In addition, PEG-IL-10 exhibited immunologic and clinical advantages in solid tumours in clinical trials, particularly in RCC and uveal melanoma (Ref. Reference Naing140). CD8 + tumour-infiltrating lymphocytes (TILs) in metastatic melanoma co-upregulate IL-10R and PD-1. While PD-1 blockade or IL-10 neutralization as monotherapies were insufficient to produce anti-tumour activity, combination therapies of PD-L1 blockers with IL-10R blockers were shown to exert anti-tumour effects by enhancing T cell responses, thereby suppressing the tumour growth (Ref. Reference Sun141). Similarly, mice with ovarian tumours treated with PD-1 blocking antibodies have higher levels of IL-10 in their serum and ascites. Moreover, infiltration of immunosuppressive MDSCs was reduced, and the immunological activity was increased when IL-10 and PD-1 blockers were used together (Ref. Reference Lamichhane142). On the other hand, a multi-centred trial involving 111 patients with advanced malignant solid tumours unresponsive to previous therapies revealed that anti-PD-1 treatment (pembrolizumab or nivolumab) in combination with PEG-IL-10 offered a new therapeutic option (Ref. Reference Naing143).

Most of the immune cells express IL-10 receptors and can activate subsequent downstream signalling pathways. Therefore, the paradox underlying the IL-10 blockade and whether it carries a beneficial or detrimental role in cancer treatment might be deciphered if we understood how exactly these cells react to IL-10 signalling through comprehensive genomic, epigenomic, and proteomic analysis.

Epigenomic approach

Epigenetic regulations include DNA methylation, histone modifications, histone acetylation, and the action of non-coding RNAs (ncRNAs) (Refs Reference Li144Reference El-Daly146). Epigenetics arising from an alteration in the chromatin usually leads to alterations in gene expression. Moreover, epigenetic changes could either activate or suppress an oncogene or a tumour suppressor gene (Refs Reference Reis, Vargas and Lemos147Reference Kilany150). It has been recently revealed that IL-10 is highly epigenetically regulated (Refs Reference Saraiva and O'Garra93, Reference Fahmy151). It is worth noting that such a level of post-transcriptional regulation of IL-10 expression might be a relevant explanation for the differential expression and effects of IL-10 in different cells at the TME despite the existence of common pathways for IL-10 induction as previously mentioned in this review, via the action of non-coding RNAs including microRNAs (miRNAs) (Refs Reference ElKhouly, Youness and Gad152, Reference ZeinElAbdeen, AbdAlSeed and Youness153), long non-coding RNAs (lncRNAs) (Refs Reference Youness and Gad154Reference El-Aziz156), and circular RNAs (circRNAs) (Refs Reference Li144, Reference El-Aziz156, Reference Dawoud157).

Epigenetic modulation of IL-10 on the post-transcriptional has been highly evident in several reports via DNA methylation, histone modifications and histone acetylation, which have been extensively studied before in several studies (Refs Reference Zhang and Kuchroo158, Reference Larsson159) and recently reviewed in (Ref. Reference Zhang and Kuchroo158). However, the epigenetic regulation of IL-10 via ncRNAs, miRNAs, lncRNAs and circRNAs is recently being explored. Therefore, a closer approach to exploring the epigenetic regulation of IL-10 via ncRNAs could aid in understanding the complex nature of this cytokine.

microRNAs (miRNAs) regulating IL-10

miRNAs are short ncRNAs around 18–25 nucleotides long that widely exist in plants, viruses and animals (Refs Reference Awad29, Reference Kilany150, Reference Bartel160, Reference Youssef161). These miRNAs can regulate gene expression by either degrading the mRNA target or by suppressing mRNA translation and reducing mRNA stability by binding to the 3′UTR (untranslated region) of a target gene (Refs Reference Youness125, Reference ZeinElAbdeen, AbdAlSeed and Youness153). Thus, a miRNA could therefore inhibit or activate the expression of tumour suppressors or oncogenes. Generally, oncogenic miRNAs (oncomiRs) are found to be over-expressed in cancers, whereas miRNAs with tumour-suppressive function are found to be under-expressed (Refs Reference Abdallah124, Reference El-Daly146, Reference Kilany150). When these oncomiRs or tumour suppressor miRNAs are inhibited or stimulated, respectively, cancer cell metastasis, proliferation and survival may be reduced, depending on the specific miRNA being affected and the type of cancer (Refs Reference Rahmoon28, Reference Awad29, Reference Nafea133). Moreover, some cancers are dependent on specific oncomiRs, and suppressing such oncomiRs could completely regress cancer growth (Refs Reference Shaalan149, Reference Fahmy151, Reference Medina, Nolde and Slack162).

Few studies have presented miRNAs that could modulate IL-10 expression. In a study, testing for the possible post-transcriptional modulation of IL-10Rα and IL-10Rβ expression by miRNAs, three miRNAs were shown to have seed regions that target the 3′UTR of IL-10Rα; miR-15a, miR-185 and miR-211. These miRNAs were shown to inhibit the proliferation of IL-10-treated melanoma cells, while their inhibitors caused an increase in cell proliferation in melanoma (Ref. Reference Venza163). IL-10 was also shown to be targeted by several other miRNAs (Ref. Reference Fillatreau, O'Garra and Springer-Verlag Gmb164). Another study showed that miR-106a could bind to the 3′UTR of IL-10 and significantly downregulate its expression in-vitro (Ref. Reference Sharma165). Two transcription factors; early growth response 1 (Egr1) and Sp1 were implicated in the induction of miR-106a, which consequently reduced IL-10 levels (Ref. Reference Fillatreau, O'Garra and Springer-Verlag Gmb164). Furthermore, an inverse relation was reported between Egr1-stimulated miR-106a and IL-10 levels. It is also worth mentioning that miR-106a is part of a cluster that is known to be dysregulated in 46% of human T-cell leukaemias. Thus, it was deduced that the promotion of leukaemic cell survival by IL-10 might be through its modulation via miR-106a (Ref. Reference Fillatreau, O'Garra and Springer-Verlag Gmb164).

Another miRNA reported to positively regulate IL-10 was miRNA-4661. The miR-4661 binding to the 3′UTR of IL-10 resulted in a net increase in the half-life of IL-10. This action was favoured by preventing tristetraprolin (TTP) from binding to the IL-10 mRNA (Ref. Reference Ma166). TTP is an RNA binding protein that plays a critical role in regulating proinflammatory immune responses by destabilizing target mRNAs via binding to their AU-rich elements (AREs) in the 3′-UTRs of mRNAs (Ref. Reference Patial and Blackshear167). Moreover, miRNA/IL-10 interactions were reported in a study by Liu et al. revealing that miR-98-mediated post-transcriptional control could potentially be involved in fine-tuning IL-10 production in endotoxin tolerance (Refs Reference Liu168, Reference Swaminathan169). On the other hand, IL-10 was reported to upregulate miRNAs that contribute towards an anti-inflammatory response such as miR-187 or downregulate those that are highly pro-inflammatory, such as miR-155 (Ref. Reference Fillatreau, O'Garra and Springer-Verlag Gmb164). IL-10 was able to downregulate the induction of miR-155 induced by LPS (Ref. Reference McCoy170). Moreover, in-vivo studies on mice deficient in miR-155, could not generate a protective immune response (Ref. Reference Rodriguez171). Whereas in IL-10 mice-deficient cells, miR-155 levels were shown to highly increase. It was previously known that miR-155 could target a number of genes involved in the immune response, such as suppressor of cytokine signalling (SOCS), inhibitor of NK-κB kinase subunit epsilon (IKBKE) and Fas-associated death domain (FADD). Thus, targeting this miRNA by IL-10 is likely to elucidate key mechanisms through which IL-10 exerts control in the cell. Another study uncovered details of the IL-10 pathway by examining the effect of IL-10 on miRNAs, using IL-10 deficient mice for expression. Ten miRNAs were found to be upregulated in IL-10 deficient mice (miR-19a, miR-21, miR-31, miR-101, miR-223, miR-326, miR-142-3p, miR-142-5p, miR-146a and miR-155) (Ref. Reference Schaefer172). miR-223 could hinder Roquin ubiquitin ligase by binding to its 3′UTR, eventually regulating IL-17 production and its inhibitor IL-10. Thus, this suggested a mechanism by which IL-10 could modulate the expression of IL-17 through miR-223. As previously mentioned, IL-10 can also induce the expression of anti-inflammatory miRNAs, such as miR-198 which is known to suppress TNF-α and IL-6. Consequently, this resulted in the promotion of an anti-inflammatory environment (Ref. Reference Rossato173). Collectively, such interesting findings of the mutual interaction between IL-10 and miRNAs discussed in the previous section highlighted an important role in the miRNA-mediated regulation of IL-10 expression and provided new insights into the intertwined mechanistic details of such immunomodulatory cytokine.

LncRNAs regulating IL-10

Long transcripts of RNA having more than 200 nucleotides, and not involved in protein translation are regarded as lncRNAs (Refs Reference Mekky16, Reference Youness18, Reference Youness and Gad154). LncRNAs play a significant role in the occurrence and development of cancer and thus, regulate the expression of cytokines such as IL-10 and IFN-γ as reported in a study by Tang et al. on non-small cell lung cancer (NSCLC) (Ref. Reference Tang174). A large number of lncRNAs has been associated with cancer as recognized by genome-wide association studies on numerous tumours (Ref. Reference Youness126). They are believed to exhibit functions such as tumour suppression and promotion, hence depicting to have a promising novel approach as biomarkers and therapeutic targets for cancers (Ref. Reference Bhan, Soleimani and Mandal175). An increased expression of lncRNA SNHGI in cancerous breast cells of CD4 + TILs was also reported, whereas the expression of FOX and IL-10 was seen to be greatly reduced by siRNA SNHGI (Ref. Reference Pei, Wang and Li176). Moreover, silencing the lncRNA cox-2 was believed to increase the expression of IL-10, Arg-1 and Fizz-1 in M2 macrophages (Ref. Reference Ye177). A study conducted by Zhou et al. reported reduced expression of IL-10 via suppression of lnc-LINC00473 (Ref. Reference Zhou178). Additionally, increased expression of IL-10 has been associated with the knockdown of lncRNA growth arrest-specific transcript 5 (GAS5) and reduced CRC cell proliferation while knockout of GAS5 promoted CRC colony formation and proliferation (Ref. Reference Li179). LncRNAs are known to regulate various signalling pathways such as TGF-β, STAT3, Hippo, EGF, Wnt, PI3 K/AKT and p53, whilst IL-10 is mostly involved in T-cell immune surveillance and suppression of cancer-associated inflammation. The expression of interleukins is regulated by lncRNAs that are known to be involved in various types of cancer. For instance, previous work by our group highlighted the potential of miRNA and lncRNA in the regulation of IL-10 in breast cancer, where miR-17-5p was identified as a dual regulator of TNF-α and IL-10. Additionally, knocking down the lncRNAs MALAT1 and/or H19 induced miR-17-5p and decreased TNF-α and IL-10 expression levels (Ref. Reference Soliman8). Such reports ed the immune-activator potential of miRNAs and the oncogenic potential of lncRNAs in cancers by regulating immunological targets in the TME. Hence, the extensive research on the relationship between the lncRNAs regulating IL-10 in various cancer needs to be validated further to establish a valid therapeutic link (Ref. Reference Elton180).

CircRNAs regulating IL-10

CircRNAs are recognized as special ncRNA molecules with a distinctive ring structure and play significant roles as gene regulators and are considered one of the recently discovered epigenetic factors (Refs Reference ZeinElAbdeen, AbdAlSeed and Youness153, Reference Dawoud157). Abnormal production of circRNAs was found to influence the onset, progression and metastasis of cancer by acting as either tumour-suppressive or oncogenic factors (Refs Reference ElKhouly, Youness and Gad152, Reference Sun181Reference Wang183). This happens via interactions with proteins, miRNA sponge function and posttranscriptional regulation (Refs Reference Selem, Youness and Gad155, Reference Dawoud157, Reference Memczak184). Moreover, a line of evidence showed that circRNAs play pivotal roles in the chemoresistance (Refs Reference Dawoud157, Reference Kun-Peng185). Recently, specific circRNAs were found to possess an immunomodulatory function and alter the response of the TME by regulating the functions of tumour-infiltrating immune cells. For instance, CD4 + T cells activity is enhanced by circ0005519 through promoting the expression of IL-13 and IL-6 via affecting the expression of hsa-let-7a-5p (Ref. Reference Huang186). On the other hand, circNT5C2 could attenuate the immune response by targeting miR-448 and serve as an oncogene via promoting tumour proliferation and metastasis (Ref. Reference Liu187).

Since IL-10 function represents an unresolved enigma in cancer therapy, and since circRNAs also have dual roles in cancer therapy, the comprehensive understanding of circRNAs regulating IL-10 expression and function might be the key to answering numerous questions. Therefore, several studies that shed the light on novel circRNAs regulating IL-10 in different oncological and non-oncological contexts are highlighted. Some circRNAs can either enhance or inhibit IL-10 production and consequently could either promote or inhibit carcinogenesis. For example, circMERTK was reported to inhibit IL-10 production in colorectal cancer. The same study came to the conclusion that circMERTK knockdown reduced the activity of CD8 + T cells, suggesting that circMERTK may affect immunosuppressive activity through the circMERTK/miR-125a-3p/IL-10 axis (Ref. Reference Zhu188). According to another in vitro study, the downregulation of secreted PD-L1 by non-small cell lung cancer cells upon knockdown of circCPA4 resulted in the activation of CD8 + T cells in the TME (Ref. Reference Zhu188). In addition, the study found that PD-L1 abrogation reduced the expression of IL-10 in CD8 + T cells (Ref. Reference Hong189). Circ103516 expression was found to be inversely correlated with IL-10 in inflammatory bowel diseases and thus it was postulated to play a proinflammatory role by sponging miR-19b. Additionally, it was discovered that circRNA HECTD1 contributed to the development of acute ischaemic stroke and that it was inversely linked with IL-10 production, suggesting that IL-10 played a protective function in acute ischaemic stroke (Ref. Reference Peng190). In another cardiac context, the synthesis of IL-10 was decreased as a result of the overexpression of circFoxo3, a circRNA that is crucial in avoiding cardiac dysfunction brought on by myocardial infarction (Ref. Reference Sun191). Downregulation of circ00074854 was reported to prevent polarization of M2 macrophages, which consequently alleviated the invasion and migration of hepatocellular carcinoma cells. According to the same study, macrophages exposed to exosomes produced by HepG2 cells that contained lower amounts of circ00074854 had significantly lower levels of IL-10 than those exposed to exosomes produced by HepG2 cells, demonstrating the direct relationship between Circ00074854 and IL-10 in different cancer settings (Ref. Reference Wang192). Furthermore, a recent study emphasized the potential of CircSnx5 as a therapeutic target for immunological disorders since it has the ability to regulate the immunity and tolerance induced by DCs. It is interesting to note that knockdown of CircSnx5 led to a significant drop in IL-10, whilst overexpression of CircSnx5 was found to block DC maturation and boost IL-10 expression (Ref. Reference Chen193). Another study focused on Circ0001598 as a potential target for treating breast cancer. It was discovered that circ0001598 regulates miR-1184 and PD-L1 via significantly increasing breast cancer proliferation, chemo-resistance and escape from immune surveillance. According to the same study mentioned above, depletion of circ0001598 increased breast cancer cells' susceptibility to Tratuzumab-induced CD8 + T cell cytotoxicity while decreasing the production of IL-10 (Ref. Reference Huang, Ma and Cui194). Another study showed that the knockdown of circRNA PLCE1 ablated IL-10 production from macrophages while PLCE1 encouraged the transformation of epithelial cells into mesenchymal tissue, thus aiding glycolysis in colorectal cancer (Ref. Reference Yi195). Another recently identified circRNA; circZNF609 has been linked to the pathogenesis of coronary artery disease, and forced overexpression of circZNF609 resulted in augmenting IL-10 expression (Ref. Reference Liang196). It is also worth mentioning that a recent study discovered that circRNA NF1-419 attenuated inflammatory factors such as IL-10 and aging markers to postpone the onset of senile dementia (Ref. Reference Ghafouri-Fard, Gholipour and Taheri197). Also, circGFRA1 has been indicated as a potential therapeutic target in prostate cancer; where Meng et al. reported that through a reduction in IL-10, knocking down circGFRA1 lessens the tumourigenic and immune-evading characteristics of prostate cancer cells (Ref. Reference Meng and Wu198). Zhang et al. also discovered the role of circ0005075 in mediating neuroinflammation where silencing of circ0005075 in rat models resulted in a decrease in IL-10 production and protected against neuro-inflammation (Ref. Reference Zhang199). Another in vitro study revealed that circCdr1 overexpression enhanced the transcription of IL-10 both in naïve and pro-inflammatory macrophages (Ref. Reference Gonzalez200). CircCHST15 was recently reported to possess an oncogenic role by promoting immune escape through upregulating the expression of IL-10 and a sponging effect on miR-155 and miR-194 in lung cancer (Ref. Reference Yang201). Additionally, circ_0046523 was found to promote carcinogenesis, mediate immunosuppression and abrogate CD8 + T cells function in pancreatic cancer via enhancing the secretion of IL-10 and TGF-β (Ref. Reference Fu202). Furthermore, silencing circDNMT3B was discovered to decrease cell survival, promote apoptosis and increase IL-10 production in rat intestinal tissue (Ref. Reference Liu203).

Collectively, it is quite clear that the circRNAs that inhibit IL-10 production from tumour cells act as tumour suppressors, while those that increase the production of IL-10 from tumour cells promote oncogenesis, cell survival, drug resistance and mediate immunosuppression. This highlights the promising role of such circRNAs as novel immunotherapeutic molecules that could ablate IL-10 production and act as a powerful immunomodulatory anti-cancer treatment for several cancer patients.

Pharmacogenomic approach: single nucleotide polymorphisms in IL-10 and its receptor

IL-10 gene

A very important basis for studies and research in IL-10 regulation is the examination of its genomic location and promoter structure. IL-10 gene encodes a protein, 178 amino acids long, which is secreted after cleavage to be comprised of 18 amino acids (Ref. Reference Sabat54). At the proximal promoter sequence of IL-10 in the human genome, there is a TATA box located upstream of the translation start site, for several transcription family members, including nuclear factor-κB (NF-κB), STAT, specificity protein (Sp), CREB, CCATT enhancer/binding protein (C/EBP), c-musculoaponeurotic fibrosarcoma factor (c-MAF), which have been characterized as ‘critical’ factors in regulating IL-10 expression (Ref. Reference Iyer and Cheng204).

IL-10 signalling

Next, it is necessary to understand how IL-10 can signal through its receptor. IL-10R is a heterodimeric receptor complex composed of two chains (IL-10Rα ‘R1’ and IL-10Rβ ‘R2’). The α-chain binds directly to IL-10, while the β-chain is subsequently recruited into the IL-10/IL-10Rα complex (Ref. Reference Asadullah, Sterry and Volk205). The binding of IL-10 to IL-10Rα induces a conformational change in the receptor, allowing it to dimerize with IL-10Rβ. This dimerization leads to signal transduction in target cells (Ref. Reference Josephson, Logsdon and Walter206). When the IL-10 complex is formed, tyrosine kinases Tyk2 and Jak1 become activated and phosphorylate specific tyrosine residues. This phosphorylation further activates the cytoplasmic inactive transcription factor; STAT-3 resulting in the translocation and transcriptional activation (Ref. Reference Finbloom and Winestock207). IL-10 rapidly activates STAT-3 and remains phosphorylated over a sustained period, unlike the transient phosphorylation of IL-6 (Ref. Reference Williams208). The STAT-3 docking sites in IL-10R1 appear to be sufficient to induce IL-10-mediated proliferative responses (Ref. Reference Riley209). While IL-10R2 intracellular domain seems to provide the docking site for Tyk2. Thus, most IL-10-specific cellular functions appear to reside in the IL-10R1 chain, whereas IL-10R2 recruits the downstream signalling kinases (Ref. Reference Walter210).

SNPs affecting IL-10

The IL-10 gene promoter and IL-10R have been found to include a significant number of SNPs (Refs Reference El Din145, Reference Youssef211). There is strong evidence that several of these polymorphisms are linked to the differential expression of IL-10 in vitro and in some situations, in vivo (Refs Reference Youssef161, Reference Li and Li212, Reference Chagas213). Some of these IL-10 variants have been associated with either low or high expression in several cancer types. For example, some genotypes have been evidenced to be correlated with a decreased expression of IL-10 and a higher risk to develop prostate cancer or non-Hodgkin's lymphoma (Refs Reference Faupel-Badger214, Reference Cunningham215). On the other hand, other evidence concluded that some IL-10 variants are associated with higher expression of IL-10 and consequently, an elevated risk for cancer development of multiple myeloma, cervical cancer and gastric cancer in patients harbouring a particular IL-10 variant (Refs Reference Shahzad216Reference El-Omar218). Also, it has been demonstrated that the IL-10 gene transcription and translation were impacted by the SNPs in the IL-10 promoter region, leading to aberrant cell division and emergence of breast cancer (Ref. Reference Moghimi219). Table 2 summarizes most of the IL-10 polymorphisms documented in the literature and their association with cancer development and risk.

Table 2. IL-10 polymorphisms and their association with cancer development and risk

Since IL-10 has a role in malignancy, it is regarded to be the subject of numerous disputes in the literature, whether it has a positive or negative effect. As a result, whether IL-10 blockage is effective as an immunotherapeutic strategy is another unsolved puzzle. This opens the door to a crucial query that might provide the answer. However, it has not yet been addressed in the literature. It remains unclear whether SNPs in the IL-10 or its receptor account for the varying effects of IL-10 inhibition on cancer treatment. A clinical investigation addressing the existence of SNPs in IL-10 or its receptors and their impact on the response to IL-10 therapy is necessary. These pharmacogenomic investigations will aid in the development of immunotherapeutic modalities by identifying the most qualified individuals to provide these cutting-edge drugs.

Conclusions

This review highlighted the controversial functions of IL-10 in oncology. Such contradictory information prevented researchers from determining whether exogenous IL-10 administration or blockage will boost the immune system and combat changes at the TME. This could be explained by the fact that IL-10 has two distinct functions depending on which immune cell and which receptor would be activated. Also, epigenetic regulation of IL-10 in cancer via ncRNAs is quite complex (Fig. 3). Also, the relationship between IL-10 SNPs will help us better understand the precise function of IL-10 in the TME and will help us develop more individualized immunotherapeutic approaches by classifying patients into responders and non-responders.

Figure 3. Epigenomic and pharmacogenomic regulation of IL-10 in oncology

Authors's contributions

Conceptualization, R. Y. M., N. M. E. and R. A. Y.; writing – original draft preparation, R. Y. M., R. A. S., G. R. and R. A. Y.; writing – review and editing, R. Y. M., N. M. E., M. B. and R. A. Y. All authors have read and agreed to the published version of the manuscript.

Funding statement

This work is not supported by any funds or grants.

Competing interests

None.

Footnotes

a

Current address: Biology and Biochemistry Department, Head of Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), Cairo 11835, Egypt

References

Elemam, NM et al. (2023) Editorial: understanding the crosstalk between immune cells and the tumor microenvironment in cancer and its implications for immunotherapy. Frontiers in Medicine (Lausanne) 10, 1202581.CrossRefGoogle ScholarPubMed
Sponghini, A et al. (2017) Complete response to anti-PD-1 nivolumab in massive skin metastasis from melanoma: efficacy and tolerability in an elderly patient. Anti-Cancer Drugs 28, 808810.CrossRefGoogle Scholar
Abdel-Latif, M and Youness, RA (2020) Why natural killer cells in triple negative breast cancer? World Journal of Clinical Oncology 11, 464476.CrossRefGoogle ScholarPubMed
Ramzy, A et al. (2022) Drugless nanoparticles tune-up an array of intertwined pathways contributing to immune checkpoint signaling and metabolic reprogramming in triple-negative breast cancer. Biomedical Materials 18. doi: 10.1088/1748-605X/aca85d.Google ScholarPubMed
Soliman, AH et al. (2023) Phytochemical-derived tumor-associated macrophage remodeling strategy using Phoenix dactylifera L. Boosted photodynamic therapy in melanoma via H19/iNOS/PD-L1 axis. Photodiagnosis and Photodynamic Therapy 44, 103792.CrossRefGoogle ScholarPubMed
Selem, NA et al. (2023) Let-7a/cMyc/CCAT1/miR-17-5p circuit re-sensitizes atezolizumab resistance in triple negative breast cancer through modulating PD-L1. Pathology Research and Practice 248, 154579.CrossRefGoogle ScholarPubMed
Conlon, KC, Miljkovic, MD and Waldmann, TA (2019) Cytokines in the treatment of cancer. Journal of Interferon and Cytokine Research 39, 621.CrossRefGoogle ScholarPubMed
Soliman, R-A et al. (2022) Uncoupling tumor necrosis factor-α and interleukin-10 at tumor immune microenvironment of breast cancer through miR-17-5p/MALAT-1/H19 circuit. BIOCELL 46, 769783.CrossRefGoogle Scholar
Mocellin, S et al. (2003) The dual role of IL-10. Trends in Immunology 24, 3643.CrossRefGoogle ScholarPubMed
Shen, L et al. (2018) Local blockade of interleukin 10 and C-X-C motif chemokine ligand 12 with nano-delivery promotes antitumor response in murine cancers. ACS Nano 12, 98309841.CrossRefGoogle ScholarPubMed
Mumm, JB and Oft, M (2013) Pegylated IL-10 induces cancer immunity. BioEssays 35, 623631.CrossRefGoogle ScholarPubMed
Hanahan, D and Weinberg, RA (2011) Hallmarks of cancer: the next generation. Cell 144, 646674.CrossRefGoogle ScholarPubMed
Ocana, MC et al. (2019) Metabolism within the tumor microenvironment and its implication on cancer progression: an ongoing therapeutic target. Medicinal Research Reviews 39, 70113.CrossRefGoogle ScholarPubMed
Youness, RA et al. (2023) Heat shock proteins: central players in oncological and immuno-oncological tracks. Advances in Experimental Medicine and Biology 1409, 193203.CrossRefGoogle ScholarPubMed
Nyberg, P, Salo, T and Kalluri, R (2008) Tumor microenvironment and angiogenesis. Frontiers in Bioscience 13, 65376553.CrossRefGoogle ScholarPubMed
Mekky, RY et al. (2023) MALAT-1: immunomodulatory lncRNA hampering the innate and the adaptive immune arms in triple negative breast cancer. Translational Oncology 31, 101653.CrossRefGoogle ScholarPubMed
Abdel-Latif, M et al. (2022) MALAT-1/p53/miR-155/miR-146a ceRNA circuit tuned by methoxylated quercitin glycoside alters immunogenic and oncogenic profiles of breast cancer. Molecular and Cellular Biochemistry 477, 12811293.CrossRefGoogle ScholarPubMed
Youness, RA et al. (2018) A novel role of sONE/NOS3/NO signaling cascade in mediating hydrogen sulphide bilateral effects on triple negative breast cancer progression. Nitric Oxide 80, 1223.CrossRefGoogle ScholarPubMed
Fridman, WH et al. (2012) The immune contexture in human tumours: impact on clinical outcome. Nature Reviews Cancer 12, 298306.CrossRefGoogle ScholarPubMed
Strauss, L et al. (2007) The frequency and suppressor function of CD4 + CD25highFoxp3 + T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clinical Cancer Research 13, 63016311.CrossRefGoogle ScholarPubMed
Chen, L et al. (2018) IL-6 influences the polarization of macrophages and the formation and growth of colorectal tumor. Oncotarget 9, 1744317454.CrossRefGoogle ScholarPubMed
Kaimala, S et al. (2018) Attenuated bacteria as immunotherapeutic tools for cancer treatment. Frontiers in Oncology 8, 136.CrossRefGoogle ScholarPubMed
Wang, JC et al. (2018) Metformin's antitumour and anti-angiogenic activities are mediated by skewing macrophage polarization. Journal of Cellular and Molecular Medicine 22, 38253836.CrossRefGoogle ScholarPubMed
Tan, B et al. (2018) Inhibition of rspo-Lgr4 facilitates checkpoint blockade therapy by switching macrophage polarization. Cancer Research 78, 49294942.CrossRefGoogle ScholarPubMed
Tan, YS et al. (2018) Mitigating SOX2-potentiated immune escape of head and neck squamous cell carcinoma with a STING-inducing nanosatellite vaccine. Clinical Cancer Research 24, 42424255.CrossRefGoogle ScholarPubMed
Baek, JH et al. (2018) Knockdown of end-binding protein 1 induces apoptosis in radioresistant A549 lung cancer cells via p38 kinase-dependent COX-2 upregulation. Oncology Reports 39, 15651572.Google ScholarPubMed
Youness, RA et al. (2016) Contradicting interplay between insulin-like growth factor-1 and miR-486-5p in primary NK cells and hepatoma cell lines with a contemporary inhibitory impact on HCC tumor progression. Growth Factors 34, 128140.CrossRefGoogle ScholarPubMed
Rahmoon, MA et al. (2017) MiR-615-5p depresses natural killer cells cytotoxicity through repressing IGF-1R in hepatocellular carcinoma patients. Growth Factors 35, 7687.CrossRefGoogle ScholarPubMed
Awad, AR et al. (2021) An acetylated derivative of vitexin halts MDA-MB-231 cellular progression and improves its immunogenic profile through tuning miR- 20a-MICA/B axis. Natural Product Research 35, 31263130.CrossRefGoogle ScholarPubMed
Hall, B, Andreeff, M and Marini, F (2007) The participation of mesenchymal stem cells in tumor stroma formation and their application as targeted-gene delivery vehicles. Handbook of Experimental Pharmacology 180, 263283.CrossRefGoogle Scholar
Jimenez, G et al. (2018) Mesenchymal stem cell's secretome promotes selective enrichment of cancer stem-like cells with specific cytogenetic profile. Cancer Letters 429, 7888.CrossRefGoogle ScholarPubMed
Guerriero, JL (2018) Macrophages: the road less traveled, changing anticancer therapy. Trends in Molecular Medicine 24, 472489.CrossRefGoogle ScholarPubMed
Van Overmeire, E et al. (2014) Mechanisms driving macrophage diversity and specialization in distinct tumor microenvironments and parallelisms with other tissues. Frontiers in Immunology 5, 127.CrossRefGoogle ScholarPubMed
Hurt, B et al. (2017) Cancer-promoting mechanisms of tumor-associated neutrophils. American Journal of Surgery 214, 938944.CrossRefGoogle ScholarPubMed
Mensurado, S et al. (2018) Tumor-associated neutrophils suppress pro-tumoral IL-17 + gammadelta T cells through induction of oxidative stress. PLoS Biology 16, e2004990.CrossRefGoogle ScholarPubMed
Hansen, M and Andersen, MH (2017) The role of dendritic cells in cancer. Seminars in Immunopathology 39, 307316.CrossRefGoogle ScholarPubMed
Guerra, DAP et al. (2018) Targeting glioblastoma-derived pericytes improves chemotherapeutic outcome. Angiogenesis 21, 667675.CrossRefGoogle ScholarPubMed
Reina-Campos, M et al. (2018) Metabolic reprogramming of the tumor microenvironment by p62 and its partners. Biochimica et Biophysica Acta, Reviews on Cancer 1870, 8895.CrossRefGoogle ScholarPubMed
Akutagawa, T et al. (2018) Cancer-adipose tissue interaction and fluid flow synergistically modulate cell kinetics, HER2 expression, and trastuzumab efficacy in gastric cancer. Gastric Cancer: Official Journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association 21, 946955.CrossRefGoogle ScholarPubMed
Kumar, V et al. (2016) The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends in Immunology 37, 208220.CrossRefGoogle ScholarPubMed
Yin, Z et al. (2019) Myeloid-derived suppressor cells: roles in the tumor microenvironment and tumor radiotherapy. International Journal of Cancer 144, 933946.CrossRefGoogle ScholarPubMed
Shen, M et al. (2018) A novel MDSC-induced PD-1(-)PD-L1(+) B-cell subset in breast tumor microenvironment possesses immuno-suppressive properties. Oncoimmunology 7, e1413520.CrossRefGoogle ScholarPubMed
Ahirwar, DK et al. (2018) Fibroblast-derived CXCL12 promotes breast cancer metastasis by facilitating tumor cell intravasation. Oncogene 37, 44284442.CrossRefGoogle ScholarPubMed
Liotta, LA and Kohn, EC (2001) The microenvironment of the tumour-host interface. Nature 411, 375379.CrossRefGoogle ScholarPubMed
Burkholder, B et al. (2014) Tumor-induced perturbations of cytokines and immune cell networks. Biochimica et Biophysica Acta 1845, 182201.Google ScholarPubMed
Zigrino, P, Loffek, S and Mauch, C (2005) Tumor-stroma interactions: their role in the control of tumor cell invasion. Biochimie 87, 321328.CrossRefGoogle ScholarPubMed
Fiorentino, DF, Bond, MW and Mosmann, TR (1989) Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. Journal of Experimental Medicine 170, 20812095.CrossRefGoogle Scholar
Moore, KW et al. (1990) Homology of cytokine synthesis inhibitory factor (IL-10) to the Epstein-Barr virus gene BCRFI. Science (New York, N.Y.) 248, 12301234.CrossRefGoogle Scholar
Kotenko, SV (2002) The family of IL-10-related cytokines and their receptors: related, but to what extent? Cytokine & Growth Factor Reviews 13, 223240.CrossRefGoogle ScholarPubMed
Volk, H et al. (2001) IL-10 and its homologs: important immune mediators and emerging immunotherapeutic targets. Trends in Immunology 22, 414417.CrossRefGoogle ScholarPubMed
Fickenscher, H et al. (2002) The interleukin-10 family of cytokines. Trends in Immunology 23, 8996.CrossRefGoogle ScholarPubMed
Mannino, MH et al. (2015) The paradoxical role of IL-10 in immunity and cancer. Cancer Letters 367, 103107.CrossRefGoogle ScholarPubMed
Nagpal, G et al. (2017) Computer-aided designing of immunosuppressive peptides based on IL-10 inducing potential. Scientific Reports 7, 42851.CrossRefGoogle ScholarPubMed
Sabat, R et al. (2010) Biology of interleukin-10. Cytokine & Growth Factor Reviews 21, 331344.CrossRefGoogle ScholarPubMed
Agrawal, S et al. (2003) Cutting edge: different Toll-like receptor agonists instruct dendritic cells to induce distinct Th responses via differential modulation of extracellular signal-regulated kinase-mitogen-activated protein kinase and c-Fos. Journal of Immunology 171, 49844989.CrossRefGoogle ScholarPubMed
Boonstra, A et al. (2006) Macrophages and myeloid dendritic cells, but not plasmacytoid dendritic cells, produce IL-10 in response to MyD88- and TRIF-dependent TLR signals, and TLR-independent signals. Journal of Immunology 177, 75517558.CrossRefGoogle Scholar
Chi, H et al. (2006) Dynamic regulation of pro- and anti-inflammatory cytokines by MAPK phosphatase 1 (MKP-1) in innate immune responses. Proceedings of the National Academy of Sciences of the USA 103, 22742279.CrossRefGoogle ScholarPubMed
Dillon, S et al. (2004) A Toll-like receptor 2 ligand stimulates Th2 responses in vivo, via induction of extracellular signal-regulated kinase mitogen-activated protein kinase and c-Fos in dendritic cells. Journal of Immunology 172, 47334743.CrossRefGoogle ScholarPubMed
Geijtenbeek, TB et al. (2003) Mycobacteria target DC-SIGN to suppress dendritic cell function. Journal of Experimental Medicine 197, 717.CrossRefGoogle ScholarPubMed
Hacker, H et al. (2006) Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6. Nature 439, 204207.CrossRefGoogle ScholarPubMed
Higgins, SC et al. (2003) Toll-like receptor 4-mediated innate IL-10 activates antigen-specific regulatory T cells and confers resistance to Bordetella pertussis by inhibiting inflammatory pathology. Journal of Immunology 171, 31193127.CrossRefGoogle ScholarPubMed
Kaiser, F et al. (2009) TPL-2 negatively regulates interferon-beta production in macrophages and myeloid dendritic cells. Journal of Experimental Medicine 206, 18631871.CrossRefGoogle ScholarPubMed
Rogers, NC et al. (2005) Syk-dependent cytokine induction by Dectin-1 reveals a novel pattern recognition pathway for C type lectins. Immunity 22, 507517.CrossRefGoogle ScholarPubMed
Dennehy, KM et al. (2009) Reciprocal regulation of IL-23 and IL-12 following co-activation of Dectin-1 and TLR signaling pathways. European Journal of Immunology 39, 13791386.CrossRefGoogle ScholarPubMed
Kapsenberg, ML (2003) Dendritic-cell control of pathogen-driven T-cell polarization. Nature Reviews Immunology 3, 984993.CrossRefGoogle ScholarPubMed
Mills, KH (2004) Regulatory T cells: friend or foe in immunity to infection? Nature Reviews Immunology 4, 841855.CrossRefGoogle ScholarPubMed
Agrawal, S and Gupta, S (2011) TLR1/2, TLR7, and TLR9 signals directly activate human peripheral blood naive and memory B cell subsets to produce cytokines, chemokines, and hematopoietic growth factors. Journal of Clinical Immunology 31, 8998.CrossRefGoogle ScholarPubMed
Lampropoulou, V et al. (2008) TLR-activated B cells suppress T cell-mediated autoimmunity. Journal of Immunology 180, 47634773.CrossRefGoogle ScholarPubMed
Sayi, A et al. (2011) TLR-2-activated B cells suppress Helicobacter-induced preneoplastic gastric immunopathology by inducing T regulatory-1 cells. Journal of Immunology 186, 878890.CrossRefGoogle ScholarPubMed
Shaw, MH et al. (2006) Tyk2 negatively regulates adaptive Th1 immunity by mediating IL-10 signaling and promoting IFN-gamma-dependent IL-10 reactivation. Journal of Immunology 176, 72637271.CrossRefGoogle ScholarPubMed
Zhang, X et al. (2007) Type I interferons protect neonates from acute inflammation through interleukin 10-producing B cells. Journal of Experimental Medicine 204, 11071118.CrossRefGoogle ScholarPubMed
Zhang, X et al. (2009) Coactivation of Syk kinase and MyD88 adaptor protein pathways by bacteria promotes regulatory properties of neutrophils. Immunity 31, 761771.CrossRefGoogle ScholarPubMed
Peppa, D et al. (2010) Blockade of immunosuppressive cytokines restores NK cell antiviral function in chronic hepatitis B virus infection. PLoS Pathogens 6, e1001227.CrossRefGoogle ScholarPubMed
Cottrez, F and Groux, H (2001) Regulation of TGF-beta response during T cell activation is modulated by IL-10. Journal of Immunology 167, 773778.CrossRefGoogle Scholar
Barthlott, T et al. (2005) CD25 + CD4 + T Cells compete with naive CD4 + T cells for IL-2 and exploit it for the induction of IL-10 production. International Immunology 17, 279288.CrossRefGoogle ScholarPubMed
Cretney, E et al. (2011) The transcription factors Blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells. Nature Immunology 12, 304311.CrossRefGoogle ScholarPubMed
de la Rosa, M et al. (2004) Interleukin-2 is essential for CD4 + CD25 + regulatory T cell function. European Journal of Immunology 34, 24802488.CrossRefGoogle ScholarPubMed
Maynard, CL et al. (2007) Regulatory T cells expressing interleukin 10 develop from Foxp3 + and Foxp3- precursor cells in the absence of interleukin 10. Nature Immunology 8, 931941.CrossRefGoogle ScholarPubMed
Sun, J et al. (2011) CD4 + T Cell help and innate-derived IL-27 induce blimp-1-dependent IL-10 production by antiviral CTLs. Nature Immunology 12, 327334.CrossRefGoogle ScholarPubMed
Gerosa, F et al. (1996) Interleukin-12 primes human CD4 and CD8 T cell clones for high production of both interferon-gamma and interleukin-10. Journal of Experimental Medicine 183, 25592569.CrossRefGoogle ScholarPubMed
Vanden Eijnden, S et al. (2005) IL-23 up-regulates IL-10 and induces IL-17 synthesis by polyclonally activated naive T cells in human. European Journal of Immunology 35, 469475.CrossRefGoogle ScholarPubMed
Meyaard, L et al. (1996) IL-12-induced IL-10 production by human T cells as a negative feedback for IL-12-induced immune responses. Journal of Immunology 156, 27762782.CrossRefGoogle Scholar
Moore, KW et al. (2001) Interleukin-10 and the interleukin-10 receptor. Annual Review of Immunology 19, 683765.CrossRefGoogle ScholarPubMed
Mocellin, S et al. (2004) IL-10 stimulatory effects on human NK cells explored by gene profile analysis. Genes and Immunity 5, 621630.CrossRefGoogle ScholarPubMed
Abd Razak, NA et al. (2016) Patient-specific interface pressure case study at transradial prosthetic socket: comparison trials between ICRC polypropylene socket and air splint socket. European Journal of Physical and Rehabilitation Medicine. PMID: 26771916.Google ScholarPubMed
Kelly, JM et al. (2002) Induction of tumor-specific T cell memory by NK cell-mediated tumor rejection. Nature Immunology 3, 8390.CrossRefGoogle ScholarPubMed
Kawai, T and Akira, S (2010) The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nature Immunology 11, 373384.CrossRefGoogle ScholarPubMed
Banerjee, A et al. (2006) Diverse Toll-like receptors utilize Tpl2 to activate extracellular signal-regulated kinase (ERK) in hemopoietic cells. Proceedings of the National Academy of Sciences of the USA 103, 32743279.CrossRefGoogle ScholarPubMed
Yi, AK et al. (2002) Role of mitogen-activated protein kinases in CpG DNA-mediated IL-10 and IL-12 production: central role of extracellular signal-regulated kinase in the negative feedback loop of the CpG DNA-mediated Th1 response. Journal of Immunology 168, 47114720.CrossRefGoogle ScholarPubMed
Hu, X et al. (2006) IFN-gamma suppresses IL-10 production and synergizes with TLR2 by regulating GSK3 and CREB/AP-1 proteins. Immunity 24, 563574.CrossRefGoogle ScholarPubMed
Foey, AD et al. (1998) Regulation of monocyte IL-10 synthesis by endogenous IL-1 and TNF-alpha: role of the p38 and p42/44 mitogen-activated protein kinases. Journal of Immunology 160, 920928.CrossRefGoogle ScholarPubMed
Kim, L et al. (2005) p38 MAPK autophosphorylation drives macrophage IL-12 production during intracellular infection. Journal of Immunology 174, 41784184.CrossRefGoogle ScholarPubMed
Saraiva, M and O'Garra, A (2010) The regulation of IL-10 production by immune cells. Nature Reviews Immunology 10, 170181.CrossRefGoogle ScholarPubMed
Ananieva, O et al. (2008) The kinases MSK1 and MSK2 act as negative regulators of toll-like receptor signaling. Nature Immunology 9, 10281036.CrossRefGoogle ScholarPubMed
Kallies, A et al. (2009) Blimp-1 transcription factor is required for the differentiation of effector CD8(+) T cells and memory responses. Immunity 31, 283295.CrossRefGoogle ScholarPubMed
D'Addario, M, Arora, PD and McCulloch, CA (2006) Role of p38 in stress activation of Sp1. Gene 379, 5161.CrossRefGoogle ScholarPubMed
Tan, NY and Khachigian, LM (2009) Sp1 phosphorylation and its regulation of gene transcription. Molecular and Cellular Biology 29, 24832488.CrossRefGoogle ScholarPubMed
Asnagli, H, Afkarian, M and Murphy, KM (2002) Cutting edge: identification of an alternative GATA-3 promoter directing tissue-specific gene expression in mouse and human. Journal of Immunology 168, 42684271.CrossRefGoogle ScholarPubMed
Martins, GA et al. (2006) Transcriptional repressor Blimp-1 regulates T cell homeostasis and function. Nature Immunology 7, 457465.CrossRefGoogle ScholarPubMed
Oliver, PM et al. (2006) Ndfip1 protein promotes the function of itch ubiquitin ligase to prevent T cell activation and T helper 2 cell-mediated inflammation. Immunity 25, 929940.CrossRefGoogle ScholarPubMed
Sanchez-Zauco, N et al. (2017) Circulating blood levels of IL-6, IFN-gamma, and IL-10 as potential diagnostic biomarkers in gastric cancer: a controlled study. BMC Cancer 17, 384.CrossRefGoogle ScholarPubMed
Berti, FCB et al. (2017) The role of interleukin 10 in human papilloma virus infection and progression to cervical carcinoma. Cytokine & Growth Factor Reviews 34, 113.CrossRefGoogle ScholarPubMed
Vahl, JM et al. (2017) Interleukin-10-regulated tumour tolerance in non-small cell lung cancer. British Journal of Cancer 117, 16441655.CrossRefGoogle ScholarPubMed
Alotaibi, MR et al. (2018) Characterization of apoptosis in a breast cancer cell line after IL-10 silencing. Asian Pacific Journal of Cancer Prevention: APJCP 19, 777783.Google Scholar
Townsend, MH et al. (2018) Metastatic colon adenocarcinoma has a significantly elevated expression of IL-10 compared with primary colon adenocarcinoma tumors. Cancer biology & therapy 19, 913920.CrossRefGoogle Scholar
Bornstein, S et al. (2016) IL-10 and integrin signaling pathways are associated with head and neck cancer progression. BMC Genomics 17, 38.CrossRefGoogle ScholarPubMed
Li, YF, Yang, PZ and Li, HF (2016) Functional polymorphisms in the IL-10 gene with susceptibility to esophageal, nasopharyngeal, and oral cancers. Cancer Biomarkers: Section A of Disease Markers 16, 641651.CrossRefGoogle ScholarPubMed
Abtahi, S et al. (2017) Dual association of serum interleukin-10 levels with colorectal cancer. Journal of Cancer Research and Therapeutics 13, 252256.Google ScholarPubMed
Shrihari, TG (2017) Dual role of inflammatory mediators in cancer. Ecancermedicalscience 11, 721.CrossRefGoogle ScholarPubMed
Tanikawa, T et al. (2012) Interleukin-10 ablation promotes tumor development, growth, and metastasis. Cancer Research 72, 420429.CrossRefGoogle ScholarPubMed
Rizzuti, D et al. (2015) Helicobacter pylori inhibits dendritic cell maturation via interleukin-10-mediated activation of the signal transducer and activator of transcription 3 pathway. Journal of Innate Immunity 7, 199211.CrossRefGoogle ScholarPubMed
Li, Y et al. (2014) [prognostic value of IL-10 expression in tumor tissues of breast cancer patients]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 30, 517520.Google ScholarPubMed
Yang, C et al. (2015) Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway. Medical Oncology 32, 352.CrossRefGoogle ScholarPubMed
Lane, D et al. (2018) Ascites IL-10 promotes ovarian cancer cell migration. Cancer Microenvironment 11, 115124.CrossRefGoogle ScholarPubMed
Wang, X et al. (2017) Bladder cancer cells induce immunosuppression of T cells by supporting PD-L1 expression in tumour macrophages partially through interleukin 10. Cell Biology International 41, 177186.CrossRefGoogle ScholarPubMed
Bermudez-Morales, VH et al. (2008) Correlation between IL-10 gene expression and HPV infection in cervical cancer: a mechanism for immune response escape. Cancer Investigation 26, 10371043.CrossRefGoogle ScholarPubMed
Shekarriz, R, Janbabaei, G and Abedian Kenari, S (2018) Prognostic value of IL-10 and Its relationship with disease stage in Iranian patients with multiple myeloma. Asian Pacific Journal of Cancer Prevention: APJCP 19, 2732.Google ScholarPubMed
Alexandrakis, MG et al. (2013) Relationship between circulating BAFF serum levels with proliferating markers in patients with multiple myeloma. Biomed Research International 2013, 389579.CrossRefGoogle ScholarPubMed
Kovacs, E (2010) Interleukin-6 leads to interleukin-10 production in several human multiple myeloma cell lines. Does interleukin-10 enhance the proliferation of these cells? Leukemia Research 34, 912916.CrossRefGoogle ScholarPubMed
Gupta, M et al. (2012) Elevated serum IL-10 levels in diffuse large B-cell lymphoma: a mechanism of aberrant JAK2 activation. Blood 119, 28442853.CrossRefGoogle ScholarPubMed
Visco, C et al. (2004) Elevated serum levels of IL-10 are associated with inferior progression-free survival in patients with Hodgkin's disease treated with radiotherapy. Leukemia & Lymphoma 45, 20852092.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2020) Increased serum level of interleukin-10 predicts poor survival and early recurrence in patients with peripheral T-cell lymphomas. Frontiers in Oncology 10, 584261.Google ScholarPubMed
Zhou, Y et al. (2021) Increased levels of serum interleukin-10 are associated with poor outcome in adult hemophagocytic lymphohistiocytosis patients. Orphanet Journal of Rare Diseases 16, 347.CrossRefGoogle ScholarPubMed
Abdallah, RM et al. (2022) Hindering the synchronization between miR-486-5p and H19 lncRNA by hesperetin halts breast cancer aggressiveness through tuning ICAM-1. Anti-Cancer Agents in Medicinal Chemistry 22, 586595.CrossRefGoogle ScholarPubMed
Youness, RA et al. (2021) Targeting hydrogen sulphide signaling in breast cancer. Journal of Advanced Research 27, 177190.CrossRefGoogle ScholarPubMed
Youness, RA et al. (2019) The long noncoding RNA sONE represses triple-negative breast cancer aggressiveness through inducing the expression of miR-34a, miR-15a, miR-16, and let-7a. Journal of Cellular Physiology 234, 2028620297.CrossRefGoogle Scholar
Hatanaka, H et al. (2001) Significant correlation between interleukin 10 expression and vascularization through angiopoietin/TIE2 networks in non-small cell lung cancer. Clinical Cancer Research 7, 12871292.Google ScholarPubMed
Marincola, FM et al. (2000) Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Advances in Immunology 74, 181273.CrossRefGoogle ScholarPubMed
Yang, L and Carbone, DP (2004) Tumor-host immune interactions and dendritic cell dysfunction. Advances in Cancer Research 92, 1327.CrossRefGoogle ScholarPubMed
Mapara, MY and Sykes, M (2004) Tolerance and cancer: mechanisms of tumor evasion and strategies for breaking tolerance. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 22, 11361151.CrossRefGoogle ScholarPubMed
Neven, B et al. (2013) A Mendelian predisposition to B-cell lymphoma caused by IL-10R deficiency. Blood 122, 37133722.CrossRefGoogle ScholarPubMed
Couper, KN, Blount, DG and Riley, EM (2008) IL-10: the master regulator of immunity to infection. Journal of Immunology 180, 57715777.CrossRefGoogle ScholarPubMed
Nafea, H et al. (2021) LncRNA HEIH/miR-939-5p interplay modulates triple-negative breast cancer progression through NOS2-induced nitric oxide production. Journal of Cellular Physiology 236, 53625372.CrossRefGoogle ScholarPubMed
Ni, G et al. (2020) Targeting interleukin-10 signalling for cancer immunotherapy, a promising and complicated task. Human Vaccines & Immunotherapeutics 16, 23282332.CrossRefGoogle ScholarPubMed
Chen, S et al. (2014) IL-10 signalling blockade at the time of immunization inhibits human papillomavirus 16 E7 transformed TC-1 tumour cells growth in mice. Cellular Immunology 290, 145151.CrossRefGoogle ScholarPubMed
Llopiz, D et al. (2018) Enhancement of antitumor vaccination by targeting dendritic cell-related IL-10. Frontiers in Immunology 9, doi:10.3389/fimmu.2018.01923CrossRefGoogle ScholarPubMed
Llopiz, D et al. (2016) Vaccine-induced but not tumor-derived interleukin-10 dictates the efficacy of interleukin-10 blockade in therapeutic vaccination. Oncoimmunology 5, e1075113.CrossRefGoogle Scholar
Soliman, R et al. (2021) Uncoupling tumor necrosis factor-α and interleukin-10 at tumor immune microenvironment of breast cancer through miR-17-5p/MALAT-1/H19 circuit.CrossRefGoogle Scholar
Mumm, JB and Oft, M (2013) Pegylated IL-10 induces cancer immunity: the surprising role of IL-10 as a potent inducer of IFN-γ-mediated CD8(+) T cell cytotoxicity. Bioessays 35, 623631.CrossRefGoogle ScholarPubMed
Naing, A et al. (2016) Safety, antitumor activity, and immune activation of pegylated recombinant human interleukin-10 (AM0010) in patients with advanced solid tumors. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 34, 35623569.CrossRefGoogle ScholarPubMed
Sun, Z et al. (2015) IL10 And PD-1 cooperate to limit the activity of tumor-specific CD8 + T cells. Cancer Research 75, 16351644.CrossRefGoogle ScholarPubMed
Lamichhane, P et al. (2017) IL10 Release upon PD-1 blockade sustains immunosuppression in ovarian cancer. Cancer Research 77, 66676678.CrossRefGoogle ScholarPubMed
Naing, A et al. (2019) Pegilodecakin combined with pembrolizumab or nivolumab for patients with advanced solid tumours (IVY): a multicentre, multicohort, open-label, phase 1b trial. The Lancet. Oncology 20, 15441555.CrossRefGoogle ScholarPubMed
Li, L et al. (2016) Epigenetic inactivation of the CpG demethylase TET1 as a DNA methylation feedback loop in human cancers. Scientific Reports 6, 26591.CrossRefGoogle ScholarPubMed
El Din, GS et al. (2020) miRNA-506-3p directly regulates rs10754339 (A/G) in the immune checkpoint protein B7-H4 in breast cancer. MicroRNA 9, 346353.CrossRefGoogle ScholarPubMed
El-Daly, SM et al. (2023) Editorial: recent breakthroughs in the decoding of circulating nucleic acids and their applications to human diseases. Frontiers in Molecular Biosciences 10, 1203495.CrossRefGoogle ScholarPubMed
Reis, AH, Vargas, FR and Lemos, B (2016) Biomarkers of genome instability and cancer epigenetics. Tumour Biology: The Journal of the International Society for Oncodevelopmental Biology and Medicine 37, 1302913038.CrossRefGoogle ScholarPubMed
Ahmed Youness, R et al. (2020) A methoxylated quercetin glycoside harnesses HCC tumor progression in a TP53/miR-15/miR-16 dependent manner. Natural Product Research 34, 14751480.CrossRefGoogle Scholar
Shaalan, YM et al. (2018) Destabilizing the interplay between miR-1275 and IGF2BPs by Tamarix articulata and quercetin in hepatocellular carcinoma. Natural Product Research 32, 22172220.CrossRefGoogle ScholarPubMed
Kilany, E, et al. FH (2021) miR-744/eNOS/NO axis: a novel target to halt triple negative breast cancer progression. Breast Disease 40, 161169.CrossRefGoogle Scholar
Fahmy, SA et al. (2022) Molecular engines, therapeutic targets, and challenges in pediatric brain tumors: a special emphasis on hydrogen sulfide and RNA-based nano-delivery. Cancers (Basel) 14, 5244.CrossRefGoogle ScholarPubMed
ElKhouly, AM, Youness, RA and Gad, MZ (2020) MicroRNA-486-5p and microRNA-486-3p: multifaceted pleiotropic mediators in oncological and non-oncological conditions. Non-Coding RNA Research 5, 1121.CrossRefGoogle ScholarPubMed
ZeinElAbdeen, YA, AbdAlSeed, A and Youness, RA (2022) Decoding insulin-like growth factor signaling pathway from a non-coding RNAs perspective: a step towards precision oncology in breast cancer. Journal of Mammary Gland Biology and Neoplasia 27, 7999.CrossRefGoogle ScholarPubMed
Youness, RA and Gad, MZ (2019) Long non-coding RNAs: functional regulatory players in breast cancer. Non-coding RNA Research 4, 3644.CrossRefGoogle ScholarPubMed
Selem, NA, Youness, RA and Gad, MZ (2021) What is beyond LncRNAs in breast cancer: a special focus on colon cancer-associated transcript-1 (CCAT-1). Non-Coding RNA Research 6, 174186.CrossRefGoogle ScholarPubMed
El-Aziz, MKA et al. (2023) Decoding hepatocarcinogenesis from a noncoding RNAs perspective. Journal of Cellular Physiology 238, 19822009.CrossRefGoogle ScholarPubMed
Dawoud, A et al. (2023) Circular RNAs: new layer of complexity evading breast cancer heterogeneity. Non-Coding RNA Research 8, 6074.CrossRefGoogle ScholarPubMed
Zhang, H and Kuchroo, V (2019) Epigenetic and transcriptional mechanisms for the regulation of IL-10. Seminars in Immunology 44, 101324.CrossRefGoogle ScholarPubMed
Larsson, L et al. (2012) Influence of epigenetic modifications of the interleukin-10 promoter on IL10 gene expression. European Journal of Oral Sciences 120, 1420.CrossRefGoogle ScholarPubMed
Bartel, DP (2004) MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281297.CrossRefGoogle ScholarPubMed
Youssef, SS et al. (2022) miR-516a-3P, a potential circulating biomarker in hepatocellular carcinoma, correlated with rs738409 polymorphism in PNPLA3. Personalized Medicine 19, 483493.CrossRefGoogle ScholarPubMed
Medina, PP, Nolde, M and Slack, FJ (2010) OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature 467, 8690.CrossRefGoogle Scholar
Venza, I et al. (2015) IL-10Ralpha expression is post-transcriptionally regulated by miR-15a, miR-185, and miR-211 in melanoma. BMC Medical Genomics 8, 81.CrossRefGoogle ScholarPubMed
Fillatreau, S and O'Garra, A and Springer-Verlag Gmb, H (2016) Interleukin-10 in Health and Disease.Google Scholar
Sharma, A et al. (2009) Posttranscriptional regulation of interleukin-10 expression by hsa-miR-106a. Proceedings of the National Academy of Sciences of the USA 106, 57615766.CrossRefGoogle ScholarPubMed
Ma, F et al. (2010) MicroRNA-466l upregulates IL-10 expression in TLR-triggered macrophages by antagonizing RNA-binding protein tristetraprolin-mediated IL-10 mRNA degradation. Journal of Immunology 184, 60536059.CrossRefGoogle ScholarPubMed
Patial, S and Blackshear, PJ (2016) Tristetraprolin as a therapeutic target in inflammatory disease. Trends in Pharmacological Sciences 37, 811821.CrossRefGoogle ScholarPubMed
Liu, Y et al. (2011) MicroRNA-98 negatively regulates IL-10 production and endotoxin tolerance in macrophages after LPS stimulation. FEBS Letters 585, 19631968.CrossRefGoogle ScholarPubMed
Swaminathan, S et al. (2012) Differential regulation of the Let-7 family of microRNAs in CD4 + T cells alters IL-10 expression. Journal of Immunology 188, 62386246.CrossRefGoogle ScholarPubMed
McCoy, CE et al. (2010) IL-10 inhibits miR-155 induction by toll-like receptors. Journal of Biological Chemistry 285, 20492–8.CrossRefGoogle ScholarPubMed
Rodriguez, A et al. (2007) Requirement of bic/microRNA-155 for normal immune function. Science 316, 608611.CrossRefGoogle ScholarPubMed
Schaefer, JS et al. (2011) Selective upregulation of microRNA expression in peripheral blood leukocytes in IL-10-/- mice precedes expression in the colon. Journal of Immunology 187, 58345841.CrossRefGoogle ScholarPubMed
Rossato, M et al. (2012) IL-10-induced microRNA-187 negatively regulates TNF-alpha, IL-6, and IL-12p40 production in TLR4-stimulated monocytes. Proceedings of the National Academy of Sciences of the USA 109, E3101E3110.CrossRefGoogle ScholarPubMed
Tang, XD et al. (2018) lncRNA AFAP1-AS1 promotes migration and invasion of non-small cell lung cancer via up-regulating IRF7 and the RIG-I-like receptor signaling pathway. Cellular Physiology and Biochemistry 50, 179195.CrossRefGoogle ScholarPubMed
Bhan, A, Soleimani, M and Mandal, SS (2017) Long noncoding RNA and cancer: a new paradigm. Cancer Research 77, 39653981.CrossRefGoogle Scholar
Pei, X, Wang, X and Li, H (2018) LncRNA SNHG1 regulates the differentiation of Treg cells and affects the immune escape of breast cancer via regulating miR-448/IDO. International Journal of Biological Macromolecules 118, 2430.CrossRefGoogle ScholarPubMed
Ye, Y et al. (2018) Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. Journal of Cellular Biochemistry 119, 29512963.CrossRefGoogle ScholarPubMed
Zhou, WY et al. (2019) Long noncoding RNA LINC00473 drives the progression of pancreatic cancer via upregulating programmed death-ligand 1 by sponging microRNA-195-5p. Journal of Cellular Physiology 234, 2317623189.CrossRefGoogle ScholarPubMed
Li, Y et al. (2017) Long non-coding RNA growth arrest specific transcript 5 acts as a tumour suppressor in colorectal cancer by inhibiting interleukin-10 and vascular endothelial growth factor expression. Oncotarget 8, 1369013702.CrossRefGoogle ScholarPubMed
Elton, TS et al. (2013) Regulation of the MIR155 host gene in physiological and pathological processes. Gene 532, 112.CrossRefGoogle ScholarPubMed
Sun, YM et al. (2019) circMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood 134, 15331546.CrossRefGoogle Scholar
Yu, J et al. (2018) Circular RNA cSMARCA5 inhibits growth and metastasis in hepatocellular carcinoma. Journal of Hepatology 68, 12141227.CrossRefGoogle ScholarPubMed
Wang, S et al. (2019) Circular RNA FOXP1 promotes tumor progression and Warburg effect in gallbladder cancer by regulating PKLR expression. Molecular Cancer 18, 145.CrossRefGoogle ScholarPubMed
Memczak, S et al. (2013) Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 495, 333338.CrossRefGoogle ScholarPubMed
Kun-Peng, Z et al. (2018) Screening circular RNA related to chemotherapeutic resistance in osteosarcoma by RNA sequencing. Epigenomics 10, 13271346.CrossRefGoogle ScholarPubMed
Huang, Z et al. (2019) Hsa_circ_0005519 increases IL-13/IL-6 by regulating hsa-let-7a-5p in CD4 + T cells to affect asthma. Clinical & Experimental Allergy 49, 11161127.CrossRefGoogle ScholarPubMed
Liu, X et al. (2017) Circular RNA circ-NT5C2 acts as an oncogene in osteosarcoma proliferation and metastasis through targeting miR-448. Oncotarget 8, 114829114838.CrossRefGoogle ScholarPubMed
Zhu, M et al. (2023) CircMERTK modulates the suppressive capacity of tumor-associated macrophage via targeting IL-10 in colorectal cancer. Human Cell 36, 276285.CrossRefGoogle ScholarPubMed
Hong, W et al. (2020) Circular RNA circ-CPA4/ let-7 miRNA/PD-L1 axis regulates cell growth, stemness, drug resistance and immune evasion in non-small cell lung cancer (NSCLC). Journal of Experimental & Clinical Cancer Research 39, 149.CrossRefGoogle ScholarPubMed
Peng, X et al. (2019) The role of circular RNA HECTD1 expression in disease risk, disease severity, inflammation, and recurrence of acute ischemic stroke. Journal of Clinical Laboratory Analysis 33, e22954.CrossRefGoogle ScholarPubMed
Sun, G et al. (2021) Circular RNA Foxo3 relieves myocardial ischemia/reperfusion injury by suppressing autophagy via inhibiting HMGB1 by repressing KAT7 in myocardial infarction. Journal Of inflammation Research 14, 63976407.CrossRefGoogle ScholarPubMed
Wang, Y et al. (2021) Downregulation of hsa_circ_0074854 suppresses the migration and invasion in hepatocellular carcinoma via interacting with HuR and via suppressing exosomes-mediated macrophage M2 polarization. International Journal of Nanomedicine 16, 28032818.CrossRefGoogle ScholarPubMed
Chen, Q et al. (2020) Circular RNA circSnx5 controls immunogenicity of dendritic cells through the miR-544/SOCS1 axis and PU.1 activity regulation. Molecular Therapy 28, 25032518.CrossRefGoogle ScholarPubMed
Huang, L, Ma, J and Cui, M (2021) Circular RNA hsa_circ_0001598 promotes programmed death-ligand-1-mediated immune escape and trastuzumab resistance via sponging miR-1184 in breast cancer cells. Immunologic Research 69, 558567.CrossRefGoogle ScholarPubMed
Yi, B et al. (2022) Circular RNA PLCE1 promotes epithelial mesenchymal transformation, glycolysis in colorectal cancer and M2 polarization of tumor-associated macrophages. Bioengineered 13, 62436256.CrossRefGoogle ScholarPubMed
Liang, B et al. (2020) CircRNA ZNF609 in peripheral blood leukocytes acts as a protective factor and a potential biomarker for coronary artery disease. Annals of Translational Medicine 8, 741.CrossRefGoogle Scholar
Ghafouri-Fard, S, Gholipour, M and Taheri, M (2021) The emerging role of long Non-coding RNAs and circular RNAs in coronary artery disease. Frontiers in Cardiovascular Medicine 8, 632393.CrossRefGoogle ScholarPubMed
Meng, M and Wu, YC (2022) LMX1B Activated circular RNA GFRA1 modulates the tumorigenic properties and immune Escape of prostate cancer. Journal of Immunology Research 2022, 7375879.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2021) Circ_0005075 targeting miR-151a-3p promotes neuropathic pain in CCI rats via inducing NOTCH2 expression. Gene 767, 145079.CrossRefGoogle ScholarPubMed
Gonzalez, C et al. (2022) Role of circular RNA cdr1as in modulation of macrophage phenotype. Life Sciences 309, 121003.CrossRefGoogle ScholarPubMed
Yang, J et al. (2021) Circular RNA CHST15 sponges miR-155-5p and miR-194-5p to promote the immune Escape of lung cancer cells mediated by PD-L1. Frontiers in Oncology 11, doi:10.3389/fonc.2021.595609Google ScholarPubMed
Fu, X et al. (2022) Hsa_circ_0046523 mediates an immunosuppressive tumor microenvironment by regulating MiR-148a-3p/PD-L1 axis in pancreatic cancer. Frontiers in Oncology 12, 877376.CrossRefGoogle ScholarPubMed
Liu, J et al. (2020) Down-regulation of circDMNT3B is conducive to intestinal mucosal permeability dysfunction of rats with sepsis via sponging miR-20b-5p. Journal of Cellular and Molecular Medicine 24, 67316740.CrossRefGoogle ScholarPubMed
Iyer, SS and Cheng, G (2012) Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Critical Reviews in Immunology 32, 2363.CrossRefGoogle ScholarPubMed
Asadullah, K, Sterry, W and Volk, HD (2003) Interleukin-10 therapy--review of a new approach. Pharmacological Reviews 55, 241269.CrossRefGoogle ScholarPubMed
Josephson, K, Logsdon, NJ and Walter, MR (2001) Crystal structure of the IL-10/IL-10R1 complex reveals a shared receptor binding site. Immunity 15, 3546.CrossRefGoogle ScholarPubMed
Finbloom, DS and Winestock, KD (1995) IL-10 induces the tyrosine phosphorylation of tyk2 and Jak1 and the differential assembly of STAT1 alpha and STAT3 complexes in human T cells and monocytes. Journal of Immunology 155, 10791090.CrossRefGoogle Scholar
Williams, LM et al. (2004) Interleukin-10 suppression of myeloid cell activation--a continuing puzzle. Immunology 113, 281292.CrossRefGoogle ScholarPubMed
Riley, JK et al. (1999) Interleukin-10 receptor signaling through the JAK-STAT pathway. Requirement for two distinct receptor-derived signals for anti-inflammatory action. Journal of Biological Chemistry 274, 1651316521.CrossRefGoogle ScholarPubMed
Walter, MR (2014) The molecular basis of IL-10 function: from receptor structure to the onset of signaling. Current Topics in Microbiology and Immunology 380, 191212.Google Scholar
Youssef, SS et al. (2022) PNPLA3 and IL 28B signature for predicting susceptibility to chronic hepatitis C infection and fibrosis progression. Archives of Physiology and Biochemistry 128, 483489.CrossRefGoogle ScholarPubMed
Li, G and Li, D (2016) Relationship between IL-10 gene polymorphisms and the risk of non-Hodgkin lymphoma: a meta-analysis. Human Immunology 77, 418425.CrossRefGoogle ScholarPubMed
Chagas, BS et al. (2013) An interleukin-10 gene polymorphism associated with the development of cervical lesions in women infected with human papillomavirus and using oral contraceptives. Infection Genetics and Evolution 19, 3237.CrossRefGoogle ScholarPubMed
Faupel-Badger, JM et al. (2008) Association of IL-10 polymorphisms with prostate cancer risk and grade of disease. Cancer Causes & Control 19, 119124.CrossRefGoogle ScholarPubMed
Cunningham, LM et al. (2003) Polymorphisms in the interleukin 10 gene promoter are associated with susceptibility to aggressive non-Hodgkin's Lymphoma. Leukemia & Lymphoma 44, 251255.CrossRefGoogle ScholarPubMed
Shahzad, MN et al. (2020) Association between interleukin gene polymorphisms and multiple myeloma susceptibility. Molecular and Clinical Oncology 12, 212224.Google ScholarPubMed
Stanczuk, GA et al. (2001) Cancer of the uterine cervix may be significantly associated with a gene polymorphism coding for increased IL-10 production. International Journal of Cancer 94, 792794.CrossRefGoogle ScholarPubMed
El-Omar, EM et al. (2003) Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology 124, 11931201.CrossRefGoogle ScholarPubMed
Moghimi, M et al. (2018) Association of IL-10 rs1800871 and rs1800872 polymorphisms with breast cancer risk: a systematic review and meta-analysis. Asian Pacific Journal of Cancer Prevention: APJCP 19, 33533359.CrossRefGoogle Scholar
Gamaleldin, M, Moussa, M and Eldin Imbaby, S (2022) Role of interleukin-10 (1082G/A) and splicing factor 3B subunit 1 (2098A/G) gene polymorphisms in chronic lymphocytic leukemia. Journal of Applied Hematology 13, 7683.CrossRefGoogle Scholar
McCarron, SL et al. (2002) Influence of cytokine gene polymorphisms on the development of prostate cancer. Cancer Research 62, 33693372.Google ScholarPubMed
Khorrami, S et al. (2022) Association of a genetic variant in interleukin-10 gene with increased risk and inflammation associated with cervical cancer. Gene 807, 145933.CrossRefGoogle ScholarPubMed
Li, L et al. (2022) Association of interleukin-10 polymorphism (rs1800896, rs1800871, and rs1800872) with breast cancer risk: an updated meta-analysis based on different ethnic groups. Frontiers in Genetics 13, https://doi.org/10.3389/fgene.2022.829283Google ScholarPubMed
Namazi, A et al. (2018) Association of interleukin-10 −1082 A/G (rs1800896) polymorphism with susceptibility to gastric cancer: meta-analysis of 6,101 cases and 8,557 controls. Arquivos de Gastroenterologia 55, 3340.CrossRefGoogle Scholar
Li, F et al. (2020) Association between interleukin-10 gene polymorphisms and risk of oral carcinoma: a meta-analysis. Histology and Histopathology 35, 13291336.Google ScholarPubMed
Zheng, C et al. (2001) Interleukin-10 gene promoter polymorphisms in multiple myeloma. International Journal of Cancer 95, 184188.3.0.CO;2-V>CrossRefGoogle ScholarPubMed
Jafari-Nedooshan, J et al. (2019) 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: APJCP 20, 19511957.CrossRefGoogle ScholarPubMed
Cunningham, LM et al. (2003) Polymorphisms in the interleukin 10 gene promoter are associated with susceptibility to aggressive non-Hodgkin's lymphoma. Leukemia & Lymphoma 44, 251255.CrossRefGoogle ScholarPubMed
Lauten, M et al. (2002) Association of initial response to prednisone treatment in childhood acute lymphoblastic leukaemia and polymorphisms within the tumour necrosis factor and the interleukin-10 genes. Leukemia 16, 14371442.CrossRefGoogle ScholarPubMed
Rashed, R et al. (2018) Associations of interleukin-10 gene polymorphisms with acute myeloid leukemia in human (Egypt). Journal of Cancer Research and Therapeutics 14, 10831086.CrossRefGoogle ScholarPubMed
Mirjalili, SA et al. (2018) Association of promoter region polymorphisms of interleukin-10 gene with susceptibility to colorectal cancer: a systematic review and meta-analysis. Arquivos de Gastroenterologia 55, 306313.CrossRefGoogle ScholarPubMed
Martínez-Escribano, JA et al. (2002) Interleukin-10, interleukin-6 and interferon-gamma gene polymorphisms in melanoma patients. Melanoma Research 12, 465469.CrossRefGoogle ScholarPubMed
Howell, WM et al. (2001) IL-10 promoter polymorphisms influence tumour development in cutaneous malignant melanoma. Genes and Immunity 2, 2531.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Snapshot of cellular and non-cellular components of the tumour microenvironment

Figure 1

Figure 2. Paradoxical pro- and anti-tumour roles of IL-10 in oncology

Figure 2

Table 1. Role of IL-10 in different solid malignancies

Figure 3

Table 2. IL-10 polymorphisms and their association with cancer development and risk

Figure 4

Figure 3. Epigenomic and pharmacogenomic regulation of IL-10 in oncology