Open Access

Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review)

  • Authors:
    • Wanli Liu
    • Xianze Wang
    • Wenming Wu
  • View Affiliations

  • Published online on: August 26, 2024     https://doi.org/10.3892/or.2024.8803
  • Article Number: 144
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Interleukin‑17 (IL‑17), an inflammatory cytokine primarily secreted by T helper 17 cells, serves a crucial role in numerous inflammatory diseases and malignancies via its receptor, IL‑17R. In addition to stimulating inflammatory responses, IL‑17 exhibits dual functions in tumors, exerting both pro‑ and antitumor effects. Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy and accounts for >90% of pancreatic cancer cases. PDAC is characterized by a prominent stromal microenvironment with significant heterogeneity, which contributes to treatment resistance. IL‑17/IL‑17R signaling has a notable effect on tumorigenesis, the tumor microenvironment and treatment efficacy in various cancer types, including PDAC. However, the specific mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer remain uncertain. This review presents a brief overview of the current knowledge and recent advances in the role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer. Furthermore, the potential of IL‑17‑targeted therapeutic strategies for PDAC treatment is also discussed.

Introduction

Pancreatic cancer, predominantly consisting of pancreatic ductal adenocarcinoma (PDAC) cases, ranks among the most lethal neoplasms with a 5-year survival rate of only 12% (1). Despite accounting for only 3% of all tumors, PDAC is the fourth leading cause of cancer-related death (2). PDAC exhibits extreme resistance to traditional chemotherapy and radiotherapy. Furthermore, while immunotherapy has achieved considerable milestones in treating various tumors, standalone immunotherapy has not exhibited an equal efficacy against pancreatic cancer (3). Therefore, exploring innovative targets and mechanisms to enhance the therapeutic effects of chemotherapy and immunotherapy is critical for improving the clinical prognosis of patients with PDAC.

The inflammatory cytokine interleukin-17 (IL-17), primarily released by T helper 17 (Th17) cells, is also produced by other cell populations such as neutrophils, γδT cells, CD8+ cytotoxic T cells and natural killer T cells (NKTs) (4). IL-17 and its receptor, IL-17R, play pivotal roles not only in allergic and autoimmune diseases, inflammation (5) and in response to COVID-19 (6), but also in the microenvironment of various tumors, including PDAC (79). The activation of IL-17/IL-17R and downstream signaling pathways, including the nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, regulates tumor progression and resistance to chemotherapy in several tumor types (1012). Due to the functions and properties of IL-17 in diverse malignancies, comprehensive investigations into the role and mechanisms of IL-17/IL-17R signaling in the pathogenesis of PDAC could reveal a potentially beneficial strategy for improving therapeutic outcomes.

This review presents a comprehensive exploration of the biology of IL-17/IL-17R signaling and its associated pathogenic mechanisms in pancreatic cancer. It provides an in-depth review of the substantial effects of IL-17/IL-17R in PDAC from the perspective of the tumor microenvironment (TME) and treatment responsiveness at the cellular and molecular levels. The present review aims to offer novel insight into the therapeutic management of pancreatic cancer.

IL-17/IL-17R family: Structure, function and signaling pathways

IL-17 family

The cytokine family IL-17 comprises six distinct members: IL-17A, IL-17B, IL-17C, IL-17D, IL-17E (also known as IL-25) and IL-17F. Among this family, IL-17A [or IL-17, alternatively known as cytotoxic T-lymphocyte-associated protein (CTLA)-8], is the prototypical and most extensively studied member (5,13,14). Besides Th17 cells, IL-17 is produced by other cell types including Type 17 CD8+ cytotoxic T cells (Tc17), NKTs, innate lymphoid cells and neutrophils (15,16). Functioning as an immune effector cytokine, IL-17 is derived from both adaptive and innate immune cells, facilitating an integral nexus between adaptive and innate immunity.

Serving as the hallmark of Th17 cells, IL-17A plays a pivotal role in the development of autoimmune diseases, inflammation and tumorigenesis (5). Increased levels of IL-17A have been implicated as a key driver in a series of autoimmune diseases (17) including psoriasis (18), rheumatoid arthritis (19) and multiple sclerosis (20), which are primarily characterized by chronic inflammation. In previous years, an increasing number of studies have focused on the role of IL-17A in tumors. For instance, in lung cancer cell lines, IL-17A stimulation triggers the phosphorylation and activation of extracellular signal-regulated kinase (ERK)1/2, which subsequently phosphorylates a myriad of cytoplasmic proteins, thereby regulating various crucial cellular processes such as proliferation, differentiation, invasion and apoptosis (21). A recent study indicated that the transcriptional activation of IL-17A by Oct4 regulates the p38 signaling pathway and encourages the polarization of M2 macrophages, thereby promoting the metastasis of cervical cancer (22). IL-17B is widely expressed across diverse tissues, including the stomach, intestine and pancreas, and is intimately correlated with tumorigenesis and the progression of tumors, as observed in gastric and pancreatic cancer (23). This underscores an emerging field of research into the IL-17 family. Additionally, IL-17B expression is positively correlated with an unfavorable prognosis in breast (24), lung (25) and pancreatic (26) cancer. IL-17C is predominantly localized within the mucosal epithelium of the oral cavity, skin and respiratory tract (27), and responds to various cytokines and pathogenic stimuli at the mucosal surfaces, thereby orchestrating inflammation, autoimmune diseases and bacterial infections (28,29). Furthermore, in lung cancer, IL-17C contributes to the growth and proliferation of tumor cells by recruiting neutrophils into the TME (30). Conversely, few studies have investigated IL-17D; although its receptor remains obscure, evidence suggests that it may play an immunoregulatory role in tumors and infectious diseases (31). IL-17E, also known as IL-25, is extensively distributed in a number of tissues (32), exerts potent proinflammatory effects, and stimulates Th2 responses. Additionally, it induces IL-4 and IL-13 production in various tissues, and fosters eosinophil expansion by inducing IL-5 release (33). IL-17F, the last member of the IL-17 family, shares >50% amino acid sequence homology with IL-17A. Typically produced by the same cells, IL-17F and IL-17A unite to create homo or heterodimers and execute similar functions (5). IL-17F serves as a less potent inducer of inflammation than IL-17A, displaying notably reduced signaling intensity and downstream gene activation (34).

IL-17R

The cytokine, IL-17, interacts with IL-17R to transmit upstream signals. The IL-17R family consists of five members, ranging from IL-17RA to IL-17RE. The IL-17R transmembrane protein comprises a fibronectin type III domain situated extracellularly and a conserved Sef/IL-17R (SEFIR) domain at the cytoplasmic terminus (5,34). During this process, the NF-κB activator 1, Act1, is recruited downstream of the IL-17R complex upon recognizing the SEFIR domain, thereby relaying IL-17-induced inflammatory response signals (35,36).

Mechanistically, the interaction between IL-17 and IL-17R is not straightforward; it involves the formation of homodimers or heterodimers between IL-17Rs. For instance, IL-17RA functions as a common receptor of the IL-17 family, forming heterodimers with IL-17RC to transmit upstream IL-17A and IL-17F signals in most cases, which is a characteristic feature of IL-17/IL-17R signaling (34,35). Similarly, IL-17RA can form heterodimers with IL-17RB and IL-17RE, which recognize homodimeric IL-17B/IL-17E and IL-17C proteins, respectively (37).

IL-17RB, often referred to as IL-25R, serves as a receptor for IL-17B and IL-17E and exhibits widespread expression across various organs and tissues, including the liver, kidney and mucosal epithelium (25). IL-17B signaling via IL-17RB can promote tumor growth, migration and invasion. Furthermore, activating IL-17B/IL-17RB directly stimulates the proliferation and migration of malignant cells and induces resistance to conventional chemotherapy (38). The downstream signaling pathway of IL-17RB is unclear. However, a recent study revealed the proximal signal transduction pathway of IL-17RB when stimulated by IL-17B in pancreatic cancer. This involves IL-17RB recruiting mixed-lineage kinase 4 to phosphorylate the Y447 residue of IL-17RB. Subsequently, phosphorylated IL-17RB recruits a ubiquitin ligase, tripartite motif containing 56, which adds K63-linked ubiquitin chains onto the K470 residue of IL-17RB. Mutation of either the Y447 or K470 of IL-17RB subsequently propagates downstream oncogenic signaling through assembling Act1 and other factors (39). IL-17RC, also known as IL-17RL, serves a crucial functional role in modulating IL-17-mediated responses, particularly through interactions with IL-17A and IL-17F (40). A recent study suggested that IL-17RC could be a key factor in IL-17-induced responses in tumor cells by regulating the activation of classical IL-17A target genes and proteins such as C-X-C motif chemokine ligand 1 (CXCL1), colony stimulating factor (CSF)1 and programmed death-ligand 1 (PD-L1) at a molecular level. Thus, IL-17RC may serve as a possible indicator of the impact of IL-17/IL-17R signaling on tumor advancement (41).

New evidence shows that IL-17RD forms a heterodimer with IL-17RA, exclusively binding to the IL-17A homodimer instead of the IL-17A/F or IL-17F/F heterodimers (42). This updates the previous understanding that IL-17RD functions as an orphan receptor. The downregulation and loss of IL-17RD have been documented in a range of malignancies, with mounting evidence indicating its involvement in tumorigenesis (43). Mechanistically, the expression of IL-17RD may regulate the aggressiveness of tumor cells by modulating the biological responses to EGF, FGF and Wnt signaling pathways (43). IL-17RE has been less investigated compared with the other IL-17 receptors. However, previous studies revealed interactions between IL-17RE and IL-17C in inflammatory and immune responses (44,45). Additionally, an increased IL-17RE expression level corresponds to a poor prognosis in patients with hepatocellular carcinoma (46).

IL-17/IL-17R signaling

As a pivotal proinflammatory cytokine, the physiological functions of IL-17 are predominantly mediated via the initiation of downstream signaling cascades upon its interaction with IL-17R. In the process of IL-17/IL-17R signal transduction, the intracellular domain of IL-17R includes a conserved SEFIR domain that orchestrates an interaction with the corresponding SEFIR motif on Act1, thereby activating a wide range of downstream signaling pathways (47). Act1 harbors a TNF receptor-associated factor (TRAF) binding site, facilitating interaction with TRAF family proteins (35,36). The binding of TRAF6 to Act1 effectively activates the downstream NF-κB and MAPK pathways, thereby activating a cascade of transcriptional regulatory alterations (48). Additionally, NF-κB participates in a series of physiological and pathological processes, including inflammation, oxidative stress, cell metabolism, proliferation and apoptosis, and exerts pervasive regulatory effects (4951). Furthermore, the binding of TRAF2 and TRAF5 to Act1 enhances posttranscriptional mRNA stability through the modulation of diverse RNA-binding proteins, such as human antigen R (HuR) and AT-rich interactive domain-containing protein 5a (Arid5a) (5).

The binding of IL-17R to Act1 via the SEFIR domain is the initial step in IL-17/IL-17R signal transduction (5,52). By serving as an E3 ubiquitin ligase, Act1 recruits and ubiquitinates TRAF6, subsequently recruiting and activating transforming growth factor β-activated kinase 1 and inhibitor of NF-κB kinase (IKK). The activated IKK complex promotes the phosphorylation and degradation of the IκB subunit on the NF-κB/IκB complex, resulting in the exposure of the nuclear localization signal on NF-κB. This process enables the rapid translocation of NF-κB to the nucleus and the induction of inflammatory gene transcription (52). Furthermore, activated TRAF6 triggers the phosphorylation and activation of the MAPK pathway (5,52). Notably, CCAAT/enhancer-binding protein β (C/EBPβ) is another significant transcription regulatory factor activated directly by IL-17R through the C/EBPβ activation domain. C/EBPβ activation can also occur as a secondary response to the activation of the aforementioned MAPK pathway (5,52).

Compared with the transcriptional regulation mediated by transcription factors, IL-17 triggers posttranscriptional regulation by stabilizing specific mRNAs and proteins, which can be more potent but less well-defined. The complex formed by TRAF2 and TRAF5 binding with Act1 acts as an RNA-binding protein, interacting with target mRNAs, such as CSF2 and CXCL2, and facilitating their fates (53). Consequently, the complex directly influences mRNA metabolism, stability and translation. Additionally, Act1 promotes the binding of HuR to mRNA, enabling mRNA to translocate to polysomes for translation (52). Similarly, the expression of Arid5a, an RNA-binding protein, is upregulated by IL-17, which competes with Regnase-1 to stabilize IL-17-induced transcripts by binding to TRAF2 (54). In addition, IL-17 promotes the interaction between Act1, IKK and TANK-binding kinase 1, resulting in the translocation of these proteins to the nucleus and the phosphorylation of splicing factor 2 (SF2), which in turn inhibits mRNA degradation mediated by SF2 (53). Notably, the IL-17-induced posttranscriptional regulation of mRNA is a component of its self-enhancement mechanisms, amplifying the activity of IL-17 (55).

Overall, the IL-17/IL-17R signaling cascade is centered on Act1. On the one hand, it activates downstream signaling primarily through the NF-κB and MAPK pathways, thereby regulating the expression of corresponding target genes transcriptionally. On the other hand, it exerts a robust proinflammatory effect through posttranscriptional regulation, principally by enhancing the stability of target mRNAs and protein translation. The classification and structure of IL-17 cytokine family members and the corresponding downstream IL-17R, as well as the intracellular IL-17/IL-17R signal transduction pathways are summarized in Fig. 1.

Figure 1.

IL-17 family members and intracellular signal transduction pathways. The IL-17 family comprises six cytokine members (IL-17A to IL-17F) and five receptor members (IL-17RA to IL-17RE). In addition to Th17 cells, IL-17-producing cells include NKT cells, Tc17 cells, ILCs and neutrophils. A homodimer or heterodimer formed by IL-17A and IL-17F binds to the IL-17RA/RC complex, functioning as a hallmark of IL-17/IL-17R signaling. The downstream signaling pathways of IL-17B/IL-17RB have not been thoroughly investigated. After recognizing the IL-17RA/RE complex, the IL-17C homodimer triggers downstream signaling pathways involved in host defense and inflammation. Although the receptor for the IL-17D homodimer remains unidentified, the IL-17RA/RD complex exclusively binds to the IL-17A homodimer. The IL-17E homodimer binds to the IL-17RA/RB complex, activating downstream Th2 responses. The intracellular signaling pathways and critical signaling molecules downstream of the activation of the IL-17RA/RC complex are summarized. Act1, NF-κB activator 1; AP-1, activator protein-1; Arid5a, AT-rich interactive domain-containing protein 5a; C/EBP, CCAAT enhancer-binding protein; CBAD, C/EBPβ activation domain; HuR, human antigen R; IL, interleukin; IL-R, IL-receptor; ILC, innate lymphoid cell; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κB; NKT, natural killer T cell; SEFIR, SEF/IL-17 receptor; SEFEX, SEFIR-extension sequences; Tc17, Type 17 CD8+ cytotoxic cell; Th17, T helper-17 cell; TRAF, TNF-receptor associated factor; TAK1, transforming growth factor-β-activated kinase-1.

IL-17/IL-17R signaling in tumors

IL-17/IL-17R signaling serves a crucial role in the tumorigenesis and progression of diverse neoplasms by stimulating distinct pathways and downstream effectors. For instance, IL-17 upregulates zinc finger E-box binding homeobox 1, a pivotal regulator of epithelial-mesenchymal transition (EMT) through NF-κB, thus fostering tumor cell invasion and metastasis by precipitating EMT (11). Additionally, it has been demonstrated that IL-17 expression is positively correlated with the activation of STAT3 (56). Facilitated by the intermediary IL-6, IL-17 can accelerate the activation of the STAT3 pathway within tumor cells. This leads to resistance to apoptosis and an increase in angiogenic factors, consequently promoting tumor cell growth and angiogenesis, thereby advancing tumor progression (57,58).

IL-17/IL-17R signaling not only has protumor effects but can also achieve antitumor effects by regulating adaptive immune responses. These include the recruitment of T lymphocytes, the enhancement of NKT activity and the stimulation of cytotoxic T lymphocyte (CTL) production (5961). For instance, mice lacking IL-17 displayed reduced levels of IFN-γ+ T cells infiltrating colon cancer cells (62). This resulted in an increased tumor volume and number of metastases. Similarly, there is an inverse relationship between the expression of IL-17 and tumor invasion in esophageal squamous cell carcinoma. By contrast, a positive correlation was observed between IL-17 expression and the abundance of CTLs, NKTs and dendritic cells (DCs) (59). The different roles and corresponding mechanisms of IL-17/IL-17R signaling in various tumors are summarized in Table I.

Table I.

Functions and mechanisms of IL-17/IL-17R signaling in tumors.

Table I.

Functions and mechanisms of IL-17/IL-17R signaling in tumors.

EffectTumorMediatorMechanism(Refs.)
Pro-tumorLung cancerIL-17Induces EMT via the IL-17/NF-κB/ZEB1 pathway and promotes invasion and metastasis(11)
IL-17AElicits the phosphorylation activation of ERK1/2 and modulates cell proliferation and invasion(21)
Liver cancerIL-17Facilitates angiogenesis and tumor progression via the IL-6/STAT3 pathway(57)
IL-17Induces the transformation of LPCs into CSCs through the downregulation of miR-122 activity(102)
Pancreatic cancerIL-17AAccelerates the development of ADM and PanIN(65,103)
IL-17B/IL-17RBFacilitates the recruitment of neutrophils, lymphocytes and endothelial cells(26,89)
Tc17Mediates Tc17-iCAFs interaction, alters the transcriptional profile of tumor cells and promotes proliferation and metabolism(58)
Colorectal cancerIL-17Potentiates tumor progression and resistance to chemotherapy via the IL-17/CXCL17/GPR35 axis(104)
IL-17AInduces mitochondrial dysfunction and cell pyroptosis via the ROS/NLRP3/capsase4/GSDMD pathway(10)
Cervical cancerIL-17AEncourages polarization of M2 macrophages and promotes metastasis through the OCT4/IL-17A/p38 pathway(22)
Breast cancerIL-17RBPromotes tumorigenesis through the inhibition of cell apoptosis mediated by NF-κB(24)
Gastric cancerIL-17B/IL-17RBDrives tumor cell proliferation and migration via AKT-β-catenin activation(105)
Prostate cancerIL-17Attracts M2 macrophages and promotes tumor growth(106)
Oral squamous cell carcinomaIL-17ACo-mediates the protumor phenotype of neutrophils and EMT with TGF-β1(107)
AntitumorEsophageal squamous cell carcinomaIL-17Enhances the cytotoxic activity of NKTs and the migration of T cells and DCs(59)
MelanomaTh17Stimulates CD8+ cytotoxic T lymphocyte responses(60)
ThymomaTh17Facilitates diverse inflammatory leukocyte recruitment (CD4+, CD8+ T cells and DCs)(61)

[i] ADM, acinar duct metaplasia; AKT, AKT serine/threonine kinase; CSC, cancer stem cell; CXCL17, C-X-C motif chemokine ligand 17; DC, dendritic cell; EMT, epithelial-mesenchymal transition; ERK, extracellular signal-regulated kinase; GPR35, G protein-coupled receptor 35; GSDMD, Gasdermin D; iCAF, inflammatory cancer-associated fibroblast; IL, interleukin; IL-R, interleukin receptor; LPC, liver progenitor cell; NF-κB, nuclear factor κB; NKT, natural killer T cell; NLRP3, NLR family pyrin domain containing 3; OCT4, octamer-binding protein 4; p38, mitogen-activated protein kinase 14; PanIN, pancreatic intraepithelial neoplasia; ROS, reactive oxygen species; STAT3, signal transducer and activator of transcription 3; Tc17, Type 17 CD8+ cytotoxic cell; TGF-β1, transforming growth factor β1; Th17, T helper-17 cell; ZEB1, zinc finger E-Box binding homeobox 1.

IL-17/IL-17R signaling in the pathogenesis of PDAC

PDAC, which is derived from pancreatic epithelial cells, is the most common exocrine tumor of the pancreas and accounts for >90% of all pancreatic cancer cases (63). In the presence of oncogenic Kras mutations and acute or chronic inflammatory stimuli, mature pancreatic acinar cells display marked plasticity and undergo differentiation into duct-like cells with ductal features. This transformation, known as acinar-to-ductal metaplasia (ADM), subsequently progresses to pancreatic intraepithelial neoplasia (PanIN) and ultimately gives rise to PDAC (63). A notable increase in the levels of Th17 and IL-17 in the peripheral blood of patients with PDAC is positively correlated with the tumor stage. Moreover, the abundance of Th17 and IL-17A within the TME of patients with PDAC significantly exceeds their level in peripheral blood and adjacent normal tissue of the same patients (64). IL-17/IL17R signaling involves a multitude of pathogenetic processes in PDAC, including ADM, PanIN and advanced tumor progression (6567).

During the precancerous stages of PDAC (ADM and PanIN), IL-17A can accelerate the process of pancreatic ADM while also assisting in the maintenance of stem cell features in tumor cells and the recruitment of immune-suppressive granulocytes (65). IL-17 released from Th17 and γδT17 cells interacts with the IL-17RA located in PanIN epithelial cells, resulting in the expansion of the tumor stroma and the development of PanIN, which can be effectively suppressed pharmacologically by inhibiting the signal transduction of IL-17 (65). In pancreatic epithelial cells with the KrasG12D mutation, IL-17A can directly stimulate the pancreatitis mediator, regenerating islet-derived 3-β, leading to the activation of STAT3, thereby furthering the development of ADM and PanIN (68). Moreover, IL-17/IL-17R signaling acts synergistically with the Notch pathway and aids the differentiation of Th17 cells, to also promote PanIN and PDAC development (68). Furthermore, IL-17 can modulate the stem cell features of PanIN, leading to an enhanced embryonic stem cell signature represented by doublecortin like kinase 1, POU class 2 homeobox 3 and aldehyde dehydrogenase 1 family member A1 and an increased expression level of IL-17RC through the canonical NF-κB and MAPK pathways. This ultimately promotes the initiation and progression of PanIN (69). Similarly, the impact of IL-17 on promoting the tumorigenic potential of cancer stem-like cells has also been observed in ovarian cancer (70).

As PDAC progresses, IL-17/IL-17R signaling also plays a pivotal role. For instance, IL-17A collaborates with IL-4 to activate NF-κB within PDAC tumor cells, thereby inducing the expression of dual oxidase 2 (DUOX2), triggering the accumulation of extracellular hydrogen peroxide and reactive oxygen species (ROS)-induced oxidative stress-related DNA damage and consequently fostering tumor progression (71). Additionally, activating IL-17B/RB on tumor cells can subsequently phosphorylate and activate ERK1/2, enhancing the production of a series of chemokines, such as C-C motif chemokine ligand (CCL)20, CXCL1, IL-8 and trefoil factor 1, ultimately facilitating the recruitment of neutrophils, lymphocytes and endothelial cells, which assists the invasion and metastasis of PDAC (26). Moreover, activating the IL-17B/RB pathway in activated pancreatic stellate cells can also enhance the metabolism and proliferation of PDAC tumor cells (72). Furthermore, in a KPC-OG murine model designed to study antigen-specific immune responses in the context of PDAC, enhanced inflammation, fibrosis and neovascularization were observed in KPC-OG lesions after IL-17 stimulation through the phosphorylation and activation of ERK, STAT3, and EGFR (73).

In general, these findings underscore the significant role of IL-17/IL-17R signaling and its associated pathways in the tumorigenesis and progression of PDAC, suggesting that IL-17/IL-17R is a promising target for therapeutic interventions against PDAC. In particular, studies on the effect of IL-17 on PDAC tumor cells have focused primarily on the initial phases of the tumor, wherein IL-17 modulates the stem cell features of tumor cells, and on the inflammation-cancer transformation of epithelial cells. Therefore, it is imperative to delve further into the involvement of IL-17/IL-17R signaling in the advanced stages of PDAC, as this will provide more compelling evidence for the targeting of IL-17 in PDAC. IL-17/IL-17R signaling in the pathogenesis of PDAC is summarized in Fig. 2.

Figure 2.

IL-17/IL-17R signaling in the pathogenesis of PDAC. In the presence of the oncogenic KrasG12D mutation, mature pancreatic acinar cells can differentiate into duct-like cells. These duct-like cells subsequently progress through stages of PanIN leading to PDAC. During this process, IL-17A released from Th17 and γδT cells stimulates REG3-β in pancreatic epithelial cells to promote the progression of ADM and PanIN. IL-17A helps to maintain the stem cell features of PanIN cells, leading to an enhanced embryonic stem cell signature and expression of IL-17RC. IL-17A collaborates with IL-4 to activate NF-κB in PDAC, thus inducing the expression of DUOX2 and consequently promoting tumor progression by triggering the ROS signature. The activation of NF-κB leads to overexpression of the Notch pathway in PanIN and PDACs. This promotes the progression of PanIN and PDAC and facilitates the differentiation of Th17 cells. Activating IL-17B/RB in PDAC tumor cells can subsequently phosphorylate and activate ERK1/2, enhancing the production of a series of chemokines and facilitating the recruitment of neutrophils, lymphocytes, and endothelial cells, which ultimately contributes to the invasion and metastasis of PDAC. ADM, acinar duct metaplasia; DUOX2, dual oxidase 2; ERK, extracellular signal-regulated kinase; IL, interleukin; IL-R, IL-receptor; NF-κB, nuclear factor κB; PanIN, pancreatic intraepithelial neoplasia; PDAC, pancreatic ductal adenocarcinoma; ROS, reactive oxygen species; Th17, T helper-17 cell; REG3-β, regenerating islet-derived 3β; DCLK1, doublecortin like kinase 1; POU2F3, POU class 2 homeobox 3; ALDH1A1, aldehyde dehydrogenase 1 family member A1; CCL20, C-C motif chemokine ligand 20; CXCL1, C-X-C motif chemokine ligand 1; TFF1, trefoil factor 1.

IL-17/IL-17R signaling in the PDAC TME

The TME of PDAC, characterized by markedly high stromal density and spatial heterogeneity, encompasses a plethora of cell components, such as fibroblasts, endothelial cells and immune cells, that are surrounded by an extracellular matrix and different types of signaling molecules (74). Compared with those of the normal pancreas, the distinct milieu of the PDAC TME exhibits a series of changes in immune cells and immunomodulatory molecules. A study has shown a reduction in regulatory T (Treg) cells and increased IL-17A expression levels in the TME of patients with PDAC in the stable or remission phases. Conversely, patients with unresectable advanced PDAC exhibit elevated Treg infiltration and reduced expression levels of IL-17A in the TME (8). Growing evidence indicates that tumor cells within the PDAC microenvironment promote cell proliferation and immune escape by engaging neighboring immune cells and the tumor stroma (74,75). Activation of IL-17/IL-17R signaling and downstream pathways occurs to varying degrees in different cells within the TME and contributes to shaping an immunosuppressive microenvironment together, thus promoting the progression of PDAC (9,76).

Th17 differentiation from naïve CD4+ T cells can be induced by DCs located in the TME of PDAC. This is achieved through the secretion of cytokines, including IL-23, IL-6 and TGF-β (76). Consequently, Th17 differentiation stimulates the synthesis of IL-17 and the progression of PDAC. Notably, IL-17-stimulated tumor cells can recruit neutrophils through the release of several cytokines and chemokines (CCXL5, CXCL3, CSF3, CCL20 and CCXL1), triggering neutrophil extracellular traps (NETs) and cytotoxic CD8+ T-cell exclusion and maintaining the immunosuppressive microenvironment (12). Nonetheless, research has indicated that neutrophils can suppress IL-17+ γδT cells through ROS signaling, thereby exerting antitumor effects (77). A recent study indicated that eliminating IL-17RA in PDAC tumor epithelial cells could lead to the enhanced infiltration of CD8+ T cells into the TME. Mechanistically, IL-17/IL-17R signaling regulates immune responses via the expression of the co-stimulatory molecule, B7-H4, in tumor cells, hence promoting an immunosuppressive microenvironment (78).

A prominent feature of the PDAC TME is its high stromal density, where cancer-associated fibroblasts (CAFs) contribute greatly (76). A previous study has demonstrated that IL-17 can activate fibroblasts through metabolic reprogramming and proliferation enhancement via the induction of hypoxia inducible factor 1 subunit α (HIF1α) expression (7). Accordingly, depletion of IL-17A in the TME of PDAC can reshape the features of CAFs and the tumor stroma, resulting in increased IL-17F levels in the serum and elevated IL-17R expression levels in CAFs, ultimately fostering an antitumor microenvironment. Specifically, the presence of IL-17A leads to collagen accumulation and a progressive increase in stiffness in the TME, thus hindering immune cell infiltration and promoting EMT (9). Conversely, IL-17A depletion in PDAC causes enhanced infiltration of α smooth muscle actin (αSMA)+ fibroblasts, macrophages (especially CD80+ macrophages), CD3+ T cells and CD8+ T cells and downregulated infiltration of Treg cells (9). The influence of IL-17/IL-17R signaling on CAFs is also evident in the interaction between Tc17 cells and inflammatory CAFs (iCAFs). IL-17A secreted by Tc17 cells may synergistically promote the differentiation of protumor iCAFs upon TNF stimulation, and activated iCAFs can in turn promote the differentiation of Tc17 cells through an IL-6 positive feedback loop. iCAFs activated by Tc17 cells can transform PDAC tumor cells toward a ‘basal-like’ transcriptional subtype signature, with enhanced proliferation and metabolism (58).

Macrophages also play an essential role in modulating the immunosuppressive microenvironment of PDAC. Depletion of IL-17A in the TME can reshape macrophage characteristics through metabolic reprogramming, with reduced tumor-associated macrophage (TAM) and Treg infiltration and increased CD8+ T-cell infiltration (79). Notably, receptor-interacting serine/threonine-protein kinase 1 suppression in TAMs can trigger a distinctive ‘mixed’ Th1/Th17 cell phenotype in PDAC, which is associated with the superior efficacy of immunotherapy (80).

The influence of the gut microbiome on the TME has been extensively documented (81). Recent research has indicated that gut dysbiosis triggered by enteric IL-17RA deficiency results in the expansion and infiltration of Th17 and CD20+ B cells in PDAC. Specifically, there exists a compensatory IL-17 loop between extra-tumoral sites, such as the gut, and the PDAC TME. In this context, the deficiency of IL-17RA in enteric epithelial cells leads to gut microbial changes, which in turn drive systemic IL-17 production and promote the growth of pancreatic tumors through IL-17RA signaling pathways within tumor cells. In tumor cells, IL-17/IL-17RA signaling stimulates tumor growth through the activation of the oxidative stress-related gene, DUOX2 (82). Another study demonstrated that the inhibitory effect of gut microbiome depletion by antibiotics on pancreatic tumor growth was mitigated in mice following treatment with an IL-17 neutralizing antibody (83). This underscores the significant role of IL-17 signaling in the interaction between the gut microbiome and pancreatic cancer progression. A potential mechanism of IL-17 signaling in the progression of pancreatic cancer mediated by alterations in gut microbiota involves the elevation of the Bacteroides phylum, as observed in mice with enteric epithelial IL-17RA deficiency [Il17ra(−/-) and Il17ra(fl/fl); Villin-Cre mice] (82). Similarly, it has been reported that a reduction in the levels of Bacteroides phylum in the gut microbiome can impede Th17 cell differentiation and IL-17 production, resulting in the suppression of pancreatic tumor growth (84).

These findings provide significant evidence for the role of IL-17 in shaping the immunosuppressive environment of PDAC. Although the specific underlying mechanisms remain intricate and require further exploration, previous studies on neutrophils (12,65) and CAFs (58) have established a strong basis for understanding the potential immunosuppressive effects of IL-17/IL-17R signaling in the TME. Future research should place more emphasis on specific cell subtypes in the microenvironment to uncover the precise mechanisms by which IL-17/IL-17R signaling influences the modulation of the PDAC TME. This detailed exploration will enhance the understanding of the PDAC TME and pave the way for novel immunotherapeutic strategies for PDAC. IL-17/IL-17R signaling involved in the PDAC TME is summarized in Fig. 3.

Figure 3.

IL-17/IL-17R signaling in the PDAC tumor microenvironment. A schematic diagram depicting the role of IL-17 in maintaining the immunosuppressive microenvironment of PDAC. IL-23, IL-6 and TGF-β released by dendritic cells stimulate the differentiation of Th17 cells. IL-17-stimulated tumor cells recruit neutrophils through the release of several cytokines and chemokines, triggering NETs and cytotoxic CD8+ T-cell exclusion. Neutrophils in the TME can suppress IL-17+ γδ T cells via ROS signaling. IL-17/IL-17R regulates the expression of the co-stimulatory molecule B7-H4 in PDAC tumor epithelial cells, thereby downregulating the infiltration of CD8+ T cells. IL-17A in the TME can reshape the PDAC stromal microenvironment and CAF characteristics, with a progressive increase in stiffness in the presence of IL-17A. Conversely, IL-17A depletion causes the enhanced infiltration of αSMA+ fibroblasts, macrophages, CD3+ T cells and CD8+ T cells and downregulated infiltration of Treg cells. Tc17 cells interact with iCAFs through a positive feedback loop featuring IL-17A, TNF and IL-6. Depletion of IL-17A in the TME can also reshape macrophage characteristics through metabolic reprogramming and the recruitment of effector T cells. RIP1 suppression in PDAC tumor-associated macrophages can trigger a distinctive ‘mixed’ Th1/Th17 cell phenotype. Additionally, gut dysbiosis triggered by enteric IL-17RA deficiency induces tumor-promoting Th17 and B cells, with systemic increases in IL-17 levels and the subsequent growth of PDAC. CAFs, cancer-associated fibroblasts; iCAFs, inflammatory CAFs; IL, interleukin; IL-R, IL-receptor; EMT, epithelial-mesenchymal transition; NETs, neutrophil extracellular traps; TME, tumor microenvironment; CCL20, C-C motif chemokine ligand 20; CXCL, C-X-C motif chemokine ligand; PDAC, pancreatic ductal adenocarcinoma; ROS, reactive oxygen species; Th17, T helper-17 cell; αSMA, α smooth muscle actin; RIP1, receptor-interacting serine/threonine-protein kinase 1; Treg, regulatory T cell.

IL-17/IL-17R signaling in PDAC cancer therapy

Chemotherapy

Chemotherapy remains the cornerstone of non-surgical treatment for patients with PDAC, and gemcitabine is one of the first-line chemotherapeutic agents (85). Nevertheless, the clinical efficacy of gemcitabine is often limited by the rapid emergence of resistance that arises within a brief timeframe for a number of patients (86). Studies have validated the contribution of cytokines in the TME, such as interleukins and TNF, to the emergence of chemotherapy resistance (87). Notably, IL-17 has been identified as a promoter of resistance to chemotherapy across various tumor types (88). Within PDAC, elevated IL-17RB levels are closely associated with the enhanced expression of mucin (MUC)1 and MUC4. The upregulated production of these MUCs by IL-17RB contributes to the enhancement of tumor stem cell-like features and resistance to gemcitabine (38). Moreover, another study revealed that IL-17RB may serve as a predictive marker for the prognosis of patients with surgically resectable PDAC and the efficacy of gemcitabine therapy (89). Therefore, targeting IL-17RB and MUC1/4 may represent a viable strategy for overcoming resistance to gemcitabine (38). Another noteworthy discovery is that the combination of the IL-17A antibody with gemcitabine can induce a distinctive ‘M1-like’ phenotype in macrophages, with an increased phagocytosis rate and enhanced antitumor response (79). Notably, IL-17F may be linked to improved outcomes for patients with PDAC treated with gemcitabine (90). Consequently, improving the efficacy of chemotherapy in PDAC may involve the administration of gemcitabine in conjunction with IL-17A blockade therapies, including either neutralizing antibodies or small molecule inhibitors.

Indeed, these discoveries are currently insufficient to overcome the major challenge of overcoming chemotherapy resistance in patients with PDAC. Further investigations are required to explore the potential mechanisms and contributors to IL-17-mediated chemoresistance and the development of novel PDAC chemotherapeutic strategies.

Immune checkpoint inhibitors (ICIs)

The combination of immunotherapy approaches, chiefly based on ICIs, has yielded noteworthy therapeutic advantages for patients with PDAC in recent years (91). According to recent research, members of the IL-17 family are indicators of the potential therapeutic effect of ICIs and are related to the infiltration of immune cells in tumors (92). The impact of IL-17 and Th17 cells on the therapeutic efficacy of ICIs has been reported in different types of tumor (7,93,94). For instance, in cutaneous squamous cell carcinoma, the IL-17-Act1-HIFα pathway facilitates the proliferation and collagen deposition of CAFs, promoting resistance to αPD-L1 (7). In colorectal cancer, a similar increase in resistance to αPD-L1 is attributed to the activation of the p65/nuclear respiratory factor 1/miRNA-15b-5p axis induced by IL-17A (94). PD-L1/programmed death-1 (PD-1) expression levels are reduced after IL-17 depletion, which modulates the function and frequency of myeloid-derived suppressor cells and enhances antitumor responses in breast cancer (93). In essence, anti-IL-17 sensitization to ICI immunotherapy is effective across different types of cancer.

In PDAC, the function of CD8+ T cells can be inhibited by CD25+ Th17 cells through CTLA-4-dependent mechanisms, thereby decreasing the concentration of IL-17. This finding indicates the potential role of Th17 and IL-17 in PDAC immunotherapy (95). Coincidentally, the combined use of IL-17 blockade with αPD-1 or αCTLA-4 effectively reduces pancreatic tumor volume by activating CD8+ T cells and mitigating the resistance caused by the single use of the aforementioned drugs (12). This observation underscores the promising clinical application of such combination strategies in PDAC. The relevance of Th17 cells to therapeutic efficacy was demonstrated in a preclinical study that investigated combination immunotherapy for PDAC. This study revealed that Th17 levels increased in a pancreatic cancer mouse model following anti-PD-L1 immunotherapy. Notably, Th17 cells increased both at the onset of PDAC and post-immunotherapy, indicating the context dependency of this signaling pathway, as Th17 cells play distinct roles across different stages of PDAC (96). Mechanistically, research has indicated that although IL-17 regulates the expression of PD-L1 in myeloid cells, it is not directly responsible for the expression of PD-L1 in tumor cells (97). This discovery suggests that IL-17 mediates the sensitivity of PDAC to anti-PD-L1 immunotherapy through complex interactions with components of the TME. Further comprehensive studies are required to elucidate these interactions.

In a recent study, the application of virus-like vesicles for the delivery of three immunomodulators (IL-12, PD-L1 short hairpin RNA and dnIL17-RA), in combination with IL-17 blockade and αPD-L1, decreased tumor growth and prolonged the overall survival of mice (98). The future implementation of innovative immunotherapies targeting IL-17 in combination with immune checkpoint molecules holds significant potential and prospects for PDAC treatment.

Innovative immunotherapy strategies

Tumor vaccines have represented a burgeoning field in tumor immunotherapy in recent years. A previous study has indicated that IL-17 may inhibit the therapeutic efficacy of tumor vaccines by restricting the development of efficient antitumor CD8+ T-cell responses (98). A previous study on the application of the GM-CSF-modified tumor cell vaccine (GVAX) in patients with PDAC revealed that following GVAX vaccine immunotherapy, Th17 signaling was enhanced, while Treg signaling was weakened in tertiary lymphoid structures (TLSs), which was correlated with prognosis (99). Moreover, a study has confirmed that Th17 signaling can stimulate the production and development of TLSs in PDAC (100). Thus, the IL-17/IL-17R signaling-based Th17 signature could serve as a prognostic indicator for patients with PDAC after GVAX vaccine immunotherapy.

The gut microbiome has garnered increasing attention in the field of immunotherapy in recent years. Researchers have shown that the gut microbiome and its products can promote the progression of gastrointestinal tumors through interactions with the host immune system (101). A study has shown that the oral administration of broad-spectrum antibiotics can reduce the number of IL-17-producing cells and increase the number of IFN-γ+ T cells, thereby inhibiting the growth of PDAC (83). Hence, regulating the gut microbiome could become a new strategy for PDAC immunotherapy. The IL-17/IL-17R signaling involved in different PDAC treatments is summarized in Table II.

Table II.

IL-17/IL-17R signaling in PDAC treatment response.

Table II.

IL-17/IL-17R signaling in PDAC treatment response.

Therapeutic strategyMediatorBiological effect and mechanism(Refs.)
GemcitabineIL-17RBBenefits gemcitabine resistance by upregulating MUC1 and MUC4 expression(38)
GemcitabineIL-17ARemodels the phenotype of macrophages, improving gemcitabine resistance with the IL-17A antibody(79)
GemcitabineIL-17FAssociated with favorable outcomes in patients with PDAC undergoing gemcitabine treatment(90)
Anti-PD-1/CTLA-4IL-17A/IL-17RAEnhances the responsiveness of ICIs via CD8+ T cell activation after IL-17 blocking(12)
Anti-PD-1/CTLA-4Th17CD25+ Th17 impedes the function of Tc cells by overexpressing CTLA-4; increases Th17 levels in mice following anti-PD-L1 immunotherapy(95,96)
GVAXTh17Stimulates the production and development of tertiary lymph nodes in PDAC and serves as a prognostic indicator for patients with PDAC post-GVAX immunotherapy(99)
Anti-intestinal microbiomeIL-17Reduces the level of IL-17-producing cells with oral broad-spectrum antibiotics, thus inhibiting PDAC growth(83)

[i] CTLA-4, cytotoxic T-lymphocyte-associated protein 4; GVAX, GM-CSF-modified tumor cell vaccine; ICI, immune checkpoint inhibitor; MUC, mucin; PD-1, programmed cell death-1; PD-L1, programmed cell death-ligand 1.

Conclusion and perspectives

In summary, the present review provides a detailed discussion on the functional mechanisms involving the IL-17 family and its receptors in the tumorigenesis and progression of pancreatic cancer. These findings highlight the crucial roles played by IL-17/IL-17R signaling and the associated pathways in this exceptionally malignant tumor, when considering both the TME and treatment responsiveness. Treating pancreatic cancer presents a considerable challenge when focusing on a single target, such as ICIs or IL-17, owing to the unique characteristics of the TME. An enhanced understanding of the interplay among immune cells, tumor cells and diverse regulatory factors of the immune system is imperative for developing more efficacious therapeutic approaches. At present, monoclonal antibodies targeting IL-17, such as brodalumab and secukinumab, have received approval for treating autoimmune diseases (5). We consider that IL-17 blockade could serve as a propellant to overcome chemotherapy resistance in various malignancies, including PDAC, and will serve as a crucial component of future immunotherapeutic innovations.

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant no. 82102940).

Availability of data and materials

Not applicable.

Authors' contributions

WL and XW contributed equally to this manuscript. WL was responsible for the initial conceptualization of the review, developed the research questions and coordinated the overall writing process. XW also conducted the literature search, selected relevant studies and drafted the initial section on ‘IL-17/IL-17R signaling in the pathogenesis of PDAC’. XW assisted with the writing of the background section and the implications of the findings in the context of ‘IL-17/IL-17R signaling in PDAC cancer therapy’. Additionally, XW assisted in the revision of the manuscript and responded to reviewer comments. WW was responsible for the overall coordination and supervision of the project, ensured the integrity and coherence of the manuscript, facilitated communication with the co-authors and managed the submission process. WW also contributed to the writing of the introduction and conclusion sections, providing a comprehensive overview of the objectives and implications of the review. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

Act1

NF-κB activator 1

ADM

acinar-to-ductal metaplasia

CAF

cancer-associated fibroblast

iCAF

inflammatory CAF

CTLA-4/8

cytotoxic T-lymphocyte-associated protein 4/8

EMT

epithelial-mesenchymal transition

ERK

extracellular signal-regulated kinase

GVAX

GM-CSF-modified tumor cell vaccine

ICI

immune checkpoint inhibitor

IL-17

interleukin-17

IL-17R

interleukin-17 receptor

MAPK

mitogen-activated protein kinase

NF-κB

nuclear factor κB

NKT

natural killer T cell

PanIN

pancreatic intraepithelial neoplasia

PDAC

pancreatic ductal adenocarcinoma

PD-1

programmed death-1

PD-L1

programmed death-ligand 1

ROS

reactive oxygen species

SEFIR

SEF/IL-17 receptor

Tc17

Type 17 CD8+ cytotoxic T cell

Th17

T helper 17 cell

TLS

tertiary lymphoid structure

TME

tumor microenvironment

TRAF

TNF-receptor associated factor

Treg

regulatory T cell

References

1 

Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM and Matrisian LM: Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74:2913–2921. 2014. View Article : Google Scholar : PubMed/NCBI

3 

DeLeo AB and Appella E: The p53 saga: Early steps in the development of tumor immunotherapy. J Immunol. 204:2321–2328. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Iwanaga N and Kolls JK: Updates on T helper type 17 immunity in respiratory disease. Immunology. 156:3–8. 2019. View Article : Google Scholar : PubMed/NCBI

5 

McGeachy MJ, Cua DJ and Gaffen SL: The IL-17 family of cytokines in health and disease. Immunity. 50:892–906. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Pacha O, Sallman MA and Evans SE: COVID-19: A case for inhibiting IL-17? Nat Rev Immunol. 20:345–346. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Chen X, Zhao J, Herjan T, Hong L, Liao Y, Liu C, Vasu K, Wang H, Thompson A, Fox PL, et al: IL-17-induced HIF1α drives resistance to anti-PD-L1 via fibroblast-mediated immune exclusion. J Exp Med. 219:e202106932022. View Article : Google Scholar : PubMed/NCBI

8 

Liu C, Cheng H, Luo G, Lu Y, Jin K, Guo M, Ni Q and Yu X: Circulating regulatory T cell subsets predict overall survival of patients with unresectable pancreatic cancer. Int J Oncol. 51:686–694. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Mucciolo G, Curcio C, Roux C, Li WY, Capello M, Curto R, Chiarle R, Giordano D, Satolli MA, Lawlor R, et al: IL17A critically shapes the transcriptional program of fibroblasts in pancreatic cancer and switches on their protumorigenic functions. Proc Natl Acad Sci USA. 118:e20203951182021. View Article : Google Scholar : PubMed/NCBI

10 

Feng WQ, Zhang YC, Xu ZQ, Yu SY, Huo JT, Tuersun A, Zheng MH, Zhao JK, Zong YP and Lu AG: IL-17A-mediated mitochondrial dysfunction induces pyroptosis in colorectal cancer cells and promotes CD8 + T-cell tumour infiltration. J Transl Med. 21:3352023. View Article : Google Scholar : PubMed/NCBI

11 

Gu K, Li MM, Shen J, Liu F, Cao JY, Jin S and Yu Y: Interleukin-17-induced EMT promotes lung cancer cell migration and invasion via NF-κB/ZEB1 signal pathway. Am J Cancer Res. 5:1169–1179. 2015.PubMed/NCBI

12 

Zhang Y, Chandra V, Riquelme Sanchez E, Dutta P, Quesada PR, Rakoski A, Zoltan M, Arora N, Baydogan S, Horne W, et al: Interleukin-17-induced neutrophil extracellular traps mediate resistance to checkpoint blockade in pancreatic cancer. J Exp Med. 217:e201903542020. View Article : Google Scholar : PubMed/NCBI

13 

Liu S, Song X, Chrunyk BA, Shanker S, Hoth LR, Marr ES and Griffor MC: Crystal structures of interleukin 17A and its complex with IL-17 receptor A. Nat Commun. 4:18882013. View Article : Google Scholar : PubMed/NCBI

14 

Rouvier E, Luciani MF, Mattéi MG, Denizot F and Golstein P: CTLA-8, cloned from an activated T cell, bearing AU-rich messenger RNA instability sequences, and homologous to a herpesvirus saimiri gene. J Immunol. 150:5445–5456. 1993. View Article : Google Scholar : PubMed/NCBI

15 

Marques HS, de Brito BB, da Silva FAF, Santos MLC, de Souza JCB, Correia TML, Lopes LW, Neres NSM, Dórea RSDM, Dantas ACS, et al: Relationship between Th17 immune response and cancer. World J Clin Oncol. 12:845–867. 2021. View Article : Google Scholar : PubMed/NCBI

16 

Lückel C, Picard FSR and Huber M: Tc17 biology and function: Novel concepts. Eur J Immunol. 50:1257–1267. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Kuwabara T, Ishikawa F, Kondo M and Kakiuchi T: The role of IL-17 and related cytokines in inflammatory autoimmune diseases. Mediators Inflamm. 2017:39080612017. View Article : Google Scholar : PubMed/NCBI

18 

Yu Z, Yu Q, Xu H, Dai X, Yu Y, Cui L, Chen Y, Gu J, Zhang X, Guo C and Shi Y: IL-17A promotes psoriasis-associated keratinocyte proliferation through ACT1-dependent activation of YAP-AREG axis. J Invest Dermatol. 142:2343–2352. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Hot A and Miossec P: Effects of interleukin (IL)-17A and IL-17F in human rheumatoid arthritis synoviocytes. Ann Rheum Dis. 70:727–732. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Kolbinger F, Huppertz C, Mir A and Padova FD: IL-17A and multiple sclerosis: Signaling pathways, producing cells and target cells in the central nervous system. Curr Drug Targets. 17:1882–1893. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Chen X, Wan J, Liu J, Xie W, Diao X, Xu J, Zhu B and Chen Z: Increased IL-17-producing cells correlate with poor survival and lymphangiogenesis in NSCLC patients. Lung Cancer. 69:348–354. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Bian Z, Wu X, Chen Q, Gao Q, Xue X and Wang Y: Oct4 activates IL-17A to orchestrate M2 macrophage polarization and cervical cancer metastasis. Cancer Immunol Immunother. 73:732024. View Article : Google Scholar : PubMed/NCBI

23 

Bastid J, Dejou C, Docquier A and Bonnefoy N: The emerging role of the IL-17B/IL-17RB pathway in cancer. Front Immunol. 11:7182020. View Article : Google Scholar : PubMed/NCBI

24 

Huang CK, Yang CY, Jeng YM, Chen CL, Wu HH, Chang YC, Ma C, Kuo WH, Chang KJ, Shew JY and Lee WH: Autocrine/paracrine mechanism of interleukin-17B receptor promotes breast tumorigenesis through NF-κB-mediated antiapoptotic pathway. Oncogene. 33:2968–2977. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Yang YF, Lee YC, Lo S, Chung YN, Hsieh YC, Chiu WC and Yuan SF: A positive feedback loop of IL-17B-IL-17RB activates ERK/β-catenin to promote lung cancer metastasis. Cancer Lett. 422:44–55. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Wu HH, Hwang-Verslues WW, Lee WH, Huang CK, Wei PC, Chen CL, Shew JY, Lee EY, Jeng YM, Tien YW, et al: Targeting IL-17B-IL-17RB signaling with an anti-IL-17RB antibody blocks pancreatic cancer metastasis by silencing multiple chemokines. J Exp Med. 212:333–349. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Al-Samadi A, Kouri VP, Salem A, Ainola M, Kaivosoja E, Barreto G, Konttinen YT, Hietanen J and Häyrinen-Immonen R: IL-17C and its receptor IL-17RA/IL-17RE identify human oral epithelial cell as an inflammatory cell in recurrent aphthous ulcer. J Oral Pathol Med. 43:117–124. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Nies JF and Panzer U: IL-17C/IL-17RE: Emergence of a unique axis in TH17 biology. Front Immunol. 11:3412020. View Article : Google Scholar : PubMed/NCBI

29 

Ramirez-Carrozzi V, Sambandam A, Luis E, Lin Z, Jeet S, Lesch J, Hackney J, Kim J, Zhou M, Lai J, et al: IL-17C regulates the innate immune function of epithelial cells in an autocrine manner. Nat Immunol. 12:1159–1166. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Jungnickel C, Schmidt LH, Bittigkoffer L, Wolf L, Wolf A, Ritzmann F, Kamyschnikow A, Herr C, Menger MD, Spieker T, et al: IL-17C mediates the recruitment of tumor-associated neutrophils and lung tumor growth. Oncogene. 36:4182–4190. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Liu X, Sun S and Liu D: IL-17D: A less studied cytokine of IL-17 family. Int Arch Allergy Immunol. 181:618–623. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Borowczyk J, Shutova M, Brembilla NC and Boehncke WH: IL-25 (IL-17E) in epithelial immunology and pathophysiology. J Allergy Clin Immunol. 148:40–52. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Xu M and Dong C: IL-25 in allergic inflammation. Immunol Rev. 278:185–191. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Gaffen SL: Structure and signalling in the IL-17 receptor family. Nat Rev Immunol. 9:556–567. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Qian Y, Liu C, Hartupee J, Altuntas CZ, Gulen MF, Jane-Wit D, Xiao J, Lu Y, Giltiay N, Liu J, et al: The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat Immunol. 8:247–256. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Herjan T, Hong L, Bubenik J, Bulek K, Qian W, Liu C, Li X, Chen X, Yang H, Ouyang S, et al: IL-17-receptor-associated adaptor Act1 directly stabilizes mRNAs to mediate IL-17 inflammatory signaling. Nat Immunol. 19:354–365. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Song X, Zhu S, Shi P, Liu Y, Shi Y, Levin SD and Qian Y: IL-17RE is the functional receptor for IL-17C and mediates mucosal immunity to infection with intestinal pathogens. Nat Immunol. 12:1151–1158. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Tsai LH, Hsu KW, Chiang CM, Yang HJ, Liu YH, Yang SF, Peng PH, Cheng WC and Wu HH: Targeting interleukin-17 receptor B enhances gemcitabine sensitivity through downregulation of mucins in pancreatic cancer. Sci Rep. 10:178172020. View Article : Google Scholar : PubMed/NCBI

39 

Wu HH, Tsai LH, Huang CK, Hsu PH, Chen MY, Chen YI, Hu CM, Shen CN, Lee CC, Chang MC, et al: Characterization of initial key steps of IL-17 receptor B oncogenic signaling for targeted therapy of pancreatic cancer. Sci Transl Med. 13:eabc28232021. View Article : Google Scholar : PubMed/NCBI

40 

Ho AW and Gaffen SL: IL-17RC: A partner in IL-17 signaling and beyond. Semin Immunopathol. 32:33–42. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Rodriguez C, Araujo Furlan CL, Tosello Boari J, Bossio SN, Boccardo S, Fozzatti L, Canale FP, Beccaria CG, Nuñez NG, Ceschin DG, et al: Interleukin-17 signaling influences CD8+ T cell immunity and tumor progression according to the IL-17 receptor subunit expression pattern in cancer cells. Oncoimmunology. 12:22613262023. View Article : Google Scholar : PubMed/NCBI

42 

Su Y, Huang J, Zhao X, Lu H, Wang W, Yang XO, Shi Y, Wang X, Lai Y and Dong C: Interleukin-17 receptor D constitutes an alternative receptor for interleukin-17A important in psoriasis-like skin inflammation. Sci Immunol. 4:eaau96572019. View Article : Google Scholar : PubMed/NCBI

43 

Pande S, Yang X and Friesel R: Interleukin-17 receptor D (Sef) is a multi-functional regulator of cell signaling. Cell Commun Signal. 19:62021. View Article : Google Scholar : PubMed/NCBI

44 

Vella G, Lunding L, Ritzmann F, Honecker A, Herr C, Wegmann M, Bals R and Beisswenger C: The IL-17 receptor IL-17RE mediates polyIC-induced exacerbation of experimental allergic asthma. Respir Res. 21:1762020. View Article : Google Scholar : PubMed/NCBI

45 

Pätzold L, Stark A, Ritzmann F, Meier C, Tschernig T, Reichrath J, Bals R, Bischoff M and Beisswenger C: IL-17C and IL-17RE promote wound closure in a Staphylococcus aureus-Based murine wound infection model. Microorganisms. 9:18212021. View Article : Google Scholar : PubMed/NCBI

46 

Liao R, Sun J, Wu H, Yi Y, Wang JX, He HW, Cai XY, Zhou J, Cheng YF, Fan J and Qiu SJ: High expression of IL-17 and IL-17RE associate with poor prognosis of hepatocellular carcinoma. J Exp Clin Cancer Res. 32:32013. View Article : Google Scholar : PubMed/NCBI

47 

Gu C, Wu L and Li X: IL-17 family: Cytokines, receptors and signaling. Cytokine. 64:477–485. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Schwandner R, Yamaguchi K and Cao Z: Requirement of tumor necrosis factor receptor-associated factor (TRAF)6 in interleukin 17 signal transduction. J Exp Med. 191:1233–1240. 2000. View Article : Google Scholar : PubMed/NCBI

49 

Napetschnig J and Wu H: Molecular basis of NF-κB signaling. Annu Rev Biophys. 42:443–468. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Mitchell S, Vargas J and Hoffmann A: Signaling via the NFκB system. Wiley Interdiscip Rev Syst Biol Med. 8:227–241. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Oeckinghaus A, Hayden MS and Ghosh S: Crosstalk in NF-κB signaling pathways. Nat Immunol. 12:695–708. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Wu NL, Huang DY, Tsou HN, Lin YC and Lin WW: Syk mediates IL-17-induced CCL20 expression by targeting Act1-dependent K63-linked ubiquitination of TRAF6. J Invest Dermatol. 135:490–498. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Tanaka H, Arima Y, Kamimura D, Tanaka Y, Takahashi N, Uehata T, Maeda K, Satoh T, Murakami M and Akira S: Phosphorylation-dependent Regnase-1 release from endoplasmic reticulum is critical in IL-17 response. J Exp Med. 216:1431–1449. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Amatya N, Childs EE, Cruz JA, Aggor FEY, Garg AV, Berman AJ, Gudjonsson JE, Atasoy U and Gaffen SL: IL-17 integrates multiple self-reinforcing, feed-forward mechanisms through the RNA binding protein Arid5a. Sci Signal. 11:eaat46172018. View Article : Google Scholar : PubMed/NCBI

55 

Gaffen SL, Jain R, Garg AV and Cua DJ: The IL-23-IL-17 immune axis: From mechanisms to therapeutic testing. Nat Rev Immunol. 14:585–600. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Wang L, Yi T, Kortylewski M, Pardoll DM, Zeng D and Yu H: IL-17 can promote tumor growth through an IL-6-Stat3 signaling pathway. J Exp Med. 206:1457–1464. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Gu FM, Li QL, Gao Q, Jiang JH, Zhu K, Huang XY, Pan JF, Yan J, Hu JH, Wang Z, et al: IL-17 induces AKT-dependent IL-6/JAK2/STAT3 activation and tumor progression in hepatocellular carcinoma. Mol Cancer. 10:1502011. View Article : Google Scholar : PubMed/NCBI

58 

Picard FSR, Lutz V, Brichkina A, Neuhaus F, Ruckenbrod T, Hupfer A, Raifer H, Klein M, Bopp T, Pfefferle PI, et al: IL-17A-producing CD8+ T cells promote PDAC via induction of inflammatory cancer-associated fibroblasts. Gut. 72:1510–1522. 2023. View Article : Google Scholar : PubMed/NCBI

59 

Lu L, Pan K, Zheng HX, Li JJ, Qiu HJ, Zhao JJ, Weng DS, Pan QZ, Wang DD, Jiang SS, et al: IL-17A promotes immune cell recruitment in human esophageal cancers and the infiltrating dendritic cells represent a positive prognostic marker for patient survival. J Immunother. 36:451–458. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Ankathatti Munegowda M, Deng Y, Mulligan SJ and Xiang J: Th17 and Th17-stimulated CD8+ T cells play a distinct role in Th17-induced preventive and therapeutic antitumor immunity. Cancer Immunol Immunother. 60:1473–1484. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Guéry L, Dubrot J, Lippens C, Brighouse D, Malinge P, Irla M, Pot C, Reith W, Waldburger JM and Hugues S: Ag-presenting CpG-activated pDCs prime Th17 cells that induce tumor regression. Cancer Res. 74:6430–6440. 2014. View Article : Google Scholar : PubMed/NCBI

62 

Kryczek I, Wei S, Szeliga W, Vatan L and Zou W: Endogenous IL-17 contributes to reduced tumor growth and metastasis. Blood. 114:357–359. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Storz P: Acinar cell plasticity and development of pancreatic ductal adenocarcinoma. Nat Rev Gastroenterol Hepatol. 14:296–304. 2017. View Article : Google Scholar : PubMed/NCBI

64 

He S, Fei M, Wu Y, Zheng D, Wan D, Wang L and Li D: Distribution and clinical significance of Th17 cells in the tumor microenvironment and peripheral blood of pancreatic cancer patients. Int J Mol Sci. 12:7424–7437. 2011. View Article : Google Scholar : PubMed/NCBI

65 

McAllister F, Bailey JM, Alsina J, Nirschl CJ, Sharma R, Fan H, Rattigan Y, Roeser JC, Lankapalli RH, Zhang H, et al: Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell. 25:621–637. 2014. View Article : Google Scholar : PubMed/NCBI

66 

McAllister F and Leach SD: Targeting IL-17 for pancreatic cancer prevention. Oncotarget. 5:9530–9531. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Wang X, Wang L, Mo Q, Dong Y, Wang G and Ji A: Changes of Th17/Treg cell and related cytokines in pancreatic cancer patients. Int J Clin Exp Pathol. 8:5702–5708. 2015.PubMed/NCBI

68 

Loncle C, Bonjoch L, Folch-Puy E, Lopez-Millan MB, Lac S, Molejon MI, Chuluyan E, Cordelier P, Dubus P, Lomberk G, et al: IL17 Functions through the Novel REG3β-JAK2-STAT3 inflammatory pathway to promote the transition from chronic pancreatitis to pancreatic cancer. Cancer Res. 75:4852–4862. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Zhang Y, Zoltan M, Riquelme E, Xu H, Sahin I, Castro-Pando S, Montiel MF, Chang K, Jiang Z, Ling J, et al: Immune cell production of interleukin 17 induces stem cell features of pancreatic intraepithelial neoplasia cells. Gastroenterology. 155:210–223.e3. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Xiang T, Long H, He L, Han X, Lin K, Liang Z, Zhuo W, Xie R and Zhu B: Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer. Oncogene. 34:165–176. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Wu Y, Konaté MM, Lu J, Makhlouf H, Chuaqui R, Antony S, Meitzler JL, Difilippantonio MJ, Liu H, Juhasz A, et al: IL-4 and IL-17A cooperatively promote hydrogen peroxide production, oxidative DNA damage, and upregulation of dual oxidase 2 in human colon and pancreatic cancer cells. J Immunol. 203:2532–2544. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Li J, Wu X, Schiffmann L, MacVicar T, Zhou C, Wang Z, Li D, Camacho OV, Heuchel R, Odenthal M, et al: IL-17B/RB activation in pancreatic stellate cells promotes pancreatic cancer metabolism and growth. Cancers (Basel). 13:53382021. View Article : Google Scholar : PubMed/NCBI

73 

Hegde S, Krisnawan VE, Herzog BH, Zuo C, Breden MA, Knolhoff BL, Hogg GD, Tang JP, Baer JM, Mpoy C, et al: Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell. 37:289–307.e9. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Sherman MH and Beatty GL: Tumor microenvironment in pancreatic cancer pathogenesis and therapeutic resistance. Annu Rev Pathol. 18:123–148. 2023. View Article : Google Scholar : PubMed/NCBI

75 

Hosein AN, Brekken RA and Maitra A: Pancreatic cancer stroma: An update on therapeutic targeting strategies. Nat Rev Gastroenterol Hepatol. 17:487–505. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Barilla RM, Diskin B, Caso RC, Lee KB, Mohan N, Buttar C, Adam S, Sekendiz Z, Wang J, Salas RD, et al: Specialized dendritic cells induce tumor-promoting IL-10+IL-17+ FoxP3neg regulatory CD4+ T cells in pancreatic carcinoma. Nat Commun. 10:14242019. View Article : Google Scholar : PubMed/NCBI

77 

Mensurado S, Rei M, Lança T, Ioannou M, Gonçalves-Sousa N, Kubo H, Malissen M, Papayannopoulos V, Serre K and Silva-Santos B: Tumor-associated neutrophils suppress pro-tumoral IL-17+γδ T cells through induction of oxidative stress. PLoS Biol. 16:e20049902018. View Article : Google Scholar : PubMed/NCBI

78 

Castro-Pando S, Howell RM, Li L, Mascaro M, Faraoni EY, Le Roux O, Romanin D, Tahan V, Riquelme E, Zhang Y, Kolls JK, Allison JP, Lozano G, Moghaddam SJ and McAllister F: Pancreatic epithelial IL-17/IL-17RA signaling drives B7-H4 expression to promote tumorigenesis. Cancer Immunol Res. doi: 10.1158/2326-6066.CIR-23-0527, Published online June 6. 2024. View Article : Google Scholar : PubMed/NCBI

79 

Roux C, Mucciolo G, Kopecka J, Novelli F, Riganti C and Cappello P: IL17A depletion affects the metabolism of macrophages treated with gemcitabine. Antioxidants (Basel). 10:4222021. View Article : Google Scholar : PubMed/NCBI

80 

Wang W, Marinis JM, Beal AM, Savadkar S, Wu Y, Khan M, Taunk PS, Wu N, Su W, Wu J, et al: RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer. Cancer Cell. 34:757–774.e7. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Wong-Rolle A, Wei HK, Zhao C and Jin C: Unexpected guests in the tumor microenvironment: Microbiome in cancer. Protein Cell. 12:426–435. 2021. View Article : Google Scholar : PubMed/NCBI

82 

Chandra V, Li L, Le Roux O, Zhang Y, Howell RM, Rupani DN, Baydogan S, Miller HD, Riquelme E, Petrosino J, et al: Gut epithelial Interleukin-17 receptor a signaling can modulate distant tumors growth through microbial regulation. Cancer Cell. 42:85–100.e6. 2024. View Article : Google Scholar : PubMed/NCBI

83 

Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B, et al: Gut Microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 155:33–37.e6. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Ivanov II, Frutos R de L, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB and Littman DR: Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 4:337–349. 2008. View Article : Google Scholar : PubMed/NCBI

85 

Grossberg AJ, Chu LC, Deig CR, Fishman EK, Hwang WL, Maitra A, Marks DL, Mehta A, Nabavizadeh N, Simeone DM, et al: Multidisciplinary standards of care and recent progress in pancreatic ductal adenocarcinoma. CA Cancer J Clin. 70:375–403. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Amrutkar M and Gladhaug IP: Pancreatic cancer chemoresistance to gemcitabine. Cancers (Basel). 9:1572017. View Article : Google Scholar : PubMed/NCBI

87 

Senthebane DA, Rowe A, Thomford NE, Shipanga H, Munro D, Mazeedi MAMA, Almazyadi HAM, Kallmeyer K, Dandara C, Pepper MS, et al: The role of tumor microenvironment in chemoresistance: To survive, keep your enemies closer. Int J Mol Sci. 18:15862017. View Article : Google Scholar : PubMed/NCBI

88 

Cochaud S, Giustiniani J, Thomas C, Laprevotte E, Garbar C, Savoye AM, Curé H, Mascaux C, Alberici G, Bonnefoy N, et al: IL-17A is produced by breast cancer TILs and promotes chemoresistance and proliferation through ERK1/2. Sci Rep. 3:34562013. View Article : Google Scholar : PubMed/NCBI

89 

Song Y, Ji B, Jiang CX, Chen ZM, Yao NH, Mukaida N and Huang H: IL17RB expression might predict prognosis and benefit from gemcitabine in patients with resectable pancreatic cancer. Pathol Res Pract. 215:1526502019. View Article : Google Scholar : PubMed/NCBI

90 

Innocenti F, Owzar K, Cox NL, Evans P, Kubo M, Zembutsu H, Jiang C, Hollis D, Mushiroda T, Li L, et al: A genome-wide association study of overall survival in pancreatic cancer patients treated with gemcitabine in CALGB 80303. Clin Cancer Res. 18:577–584. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Morrison AH, Byrne KT and Vonderheide RH: Immunotherapy and prevention of pancreatic cancer. Trends Cancer. 4:418–428. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Han X, Ye J, Huang R, Li Y, Liu J, Meng T and Song D: Pan-cancer analysis reveals interleukin-17 family members as biomarkers in the prediction for immune checkpoint inhibitor curative effect. Front Immunol. 13:9002732022. View Article : Google Scholar : PubMed/NCBI

93 

Moaaz M, Lotfy H, Motawea MA and Fadali G: The interplay of interleukin-17A and breast cancer tumor microenvironment as a novel immunotherapeutic approach to increase tumor immunogenicity. Immunobiology. 226:1520682021. View Article : Google Scholar : PubMed/NCBI

94 

Liu C, Liu R, Wang B, Lian J, Yao Y, Sun H, Zhang C, Fang L, Guan X, Shi J, et al: Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer. J Immunother Cancer. 9:e0018952021. View Article : Google Scholar : PubMed/NCBI

95 

Lang C, Wang J and Chen L: CD25-expressing Th17 cells mediate CD8+ T cell suppression in CTLA-4 dependent mechanisms in pancreatic ductal adenocarcinoma. Exp Cell Res. 360:384–389. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Li Y, Hong YK, Wang X, Pandit H, Zheng Q, Yu Y, Shi X, Chen Y, Tan M, Pulliam Z, et al: Epigenetic modulation enhances immunotherapy for pancreatic ductal adenocarcinoma. Clin Transl Immunol. 11:e14302022. View Article : Google Scholar : PubMed/NCBI

97 

Du W, Menjivar RE, Donahue KL, Kadiyala P, Velez-Delgado A, Brown KL, Watkoske HR, He X, Carpenter ES, Angeles CV, et al: WNT signaling in the tumor microenvironment promotes immunosuppression in murine pancreatic cancer. J Exp Med. 220:e202205032023. View Article : Google Scholar : PubMed/NCBI

98 

Chen J, Madina BR, Ahmadi E, Yarovinsky TO, Krady MM, Meehan EV, Wang IC, Ye X, Pitmon E, Ma XY, et al: Cancer immunotherapy with enveloped self-amplifying mRNA CARG-2020 that modulates IL-12, IL-17 and PD-L1 pathways to prevent tumor recurrence. Acta Pharm Sin B. 14:335–349. 2024. View Article : Google Scholar : PubMed/NCBI

99 

Lutz ER, Wu AA, Bigelow E, Sharma R, Mo G, Soares K, Solt S, Dorman A, Wamwea A, Yager A, et al: Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation. Cancer Immunol Res. 2:616–631. 2014. View Article : Google Scholar : PubMed/NCBI

100 

Grogan JL and Ouyang W: A role for Th17 cells in the regulation of tertiary lymphoid follicles. Eur J Immunol. 42:2255–2262. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Shalapour S and Karin M: Cruel to be kind: Epithelial, microbial, and immune cell interactions in gastrointestinal cancers. Annu Rev Immunol. 38:649–671. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Gasmi I, Machou C, Rodrigues A, Brouillet A, Nguyen TC, Rousseau B, Guillot A, Rodriguez C, Demontant V, Ait-Ahmed Y, et al: Interleukin-17 programs liver progenitor cell transformation into cancer stem cells through miR-122 downregulation with increased risk of primary liver cancer initiation. Int J Biol Sci. 18:1944–1960. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Wang X, Chen H, Jiang R, Hong X, Peng J, Chen W, Jiang J, Li J, Huang D, Dai H, et al: Interleukin-17 activates and synergizes with the notch signaling pathway in the progression of pancreatic ductal adenocarcinoma. Cancer Lett. 508:1–12. 2021. View Article : Google Scholar : PubMed/NCBI

104 

Bu J, Yan W, Huang Y and Lin K: Activation of the IL-17 signalling pathway by the CXCL17-GPR35 axis affects drug resistance and colorectal cancer tumorigenesis. Am J Cancer Res. 13:2172–2187. 2023.PubMed/NCBI

105 

Bie Q, Sun C, Gong A, Li C, Su Z, Zheng D, Ji X, Wu Y, Guo Q, Wang S and Xu H: Non-tumor tissue derived interleukin-17B activates IL-17RB/AKT/β-catenin pathway to enhance the stemness of gastric cancer. Sci Rep. 6:254472016. View Article : Google Scholar : PubMed/NCBI

106 

Duan Z, Miller HD, Fu X, Ge D, Jin B, Moustafa AA, Lan R, Zhang K, Chen Z and You Z: Th17 cells promote tumor growth in an immunocompetent orthotopic mouse model of prostate cancer. Am J Clin Exp Urol. 7:249–261. 2019.PubMed/NCBI

107 

Yu T, Tang Q, Chen X, Fan W, Zhou Z, Huang W and Liang F: TGF-β1 and IL-17A comediate the protumor phenotype of neutrophils to regulate the epithelial-mesenchymal transition in oral squamous cell carcinoma. J Oral Pathol Med. 50:353–361. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 52 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu W, Wang X and Wu W: Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review). Oncol Rep 52: 144, 2024.
APA
Liu, W., Wang, X., & Wu, W. (2024). Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review). Oncology Reports, 52, 144. https://doi.org/10.3892/or.2024.8803
MLA
Liu, W., Wang, X., Wu, W."Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review)". Oncology Reports 52.5 (2024): 144.
Chicago
Liu, W., Wang, X., Wu, W."Role and functional mechanisms of IL‑17/IL‑17R signaling in pancreatic cancer (Review)". Oncology Reports 52, no. 5 (2024): 144. https://doi.org/10.3892/or.2024.8803