Next Article in Journal
Effects of Flavonoids on Cancer, Cardiovascular and Neurodegenerative Diseases: Role of NF-κB Signaling Pathway
Next Article in Special Issue
The Role of P2X7 Purinoceptors in the Pathogenesis and Treatment of Muscular Dystrophies
Previous Article in Journal
Ceramide AD™ Restores Skin Integrity and Function following Exposure to House Dust Mite
Previous Article in Special Issue
Animal Models for the Investigation of P2X7 Receptors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of IL-18 in P2RX7-Mediated Antitumor Immunity

by
Serena Janho dit Hreich
1,2,3,
Paul Hofman
2,4,5 and
Valérie Vouret-Craviari
1,2,3,*
1
Faculty of Medicine, Université Côte d’Azur, CNRS, INSERM, IRCAN, 06108 Nice, France
2
IHU RespirEREA, Université Côte d′Azur, 06108 Nice, France
3
FHU OncoAge, 06108 Nice, France
4
Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur Hospital, 06108 Nice, France
5
Hospital-Related Biobank, Pasteur Hospital, 06108 Nice, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9235; https://doi.org/10.3390/ijms24119235
Submission received: 26 April 2023 / Revised: 22 May 2023 / Accepted: 23 May 2023 / Published: 25 May 2023
(This article belongs to the Special Issue The Role of P2X7 Receptor in Human Health and Diseases)

Abstract

:
Cancer is the leading cause of death worldwide despite the variety of treatments that are currently used. This is due to an innate or acquired resistance to therapy that encourages the discovery of novel therapeutic strategies to overcome the resistance. This review will focus on the role of the purinergic receptor P2RX7 in the control of tumor growth, through its ability to modulate antitumor immunity by releasing IL-18. In particular, we describe how the ATP-induced receptor activities (cationic exchange, large pore opening and NLRP3 inflammasome activation) modulate immune cell functions. Furthermore, we recapitulate our current knowledge of the production of IL-18 downstream of P2RX7 activation and how IL-18 controls the fate of tumor growth. Finally, the potential of targeting the P2RX7/IL-18 pathway in combination with classical immunotherapies to fight cancer is discussed.
Keywords:
P2RX7; IL-18; cancer

1. Introduction

Current treatments in cancer not only target the cancerous cells, but also take advantage of the composition of the tumor microenvironment (TME) that include immune and stromal cells that have been shown to impact tumor growth [1,2]. Indeed, boosting the antitumor activity of the immune system using blocking antibodies targeting immune checkpoints, such as PD-L1 and CTLA-4, has markedly increased patients’ survival [3,4,5]. Despite the efficacy of immunotherapies and the vast diversity of therapies in cancer, the majority of cancer patients remain resistant to treatments due to innate or acquired resistance mechanisms. Therefore, new strategies to fight this disease are still needed.
The TME is characterized by high extracellular ATP (eATP) that are released from dying cancer cells or immune cells, as reviewed in [6,7]. Such levels of eATP activate the purinergic P2RX7 receptor that is expressed on all cells in the TME [6]. Since ATP is normally present inside the cell, eATP constitutes a damage-associated molecular pattern (DAMP), enabling the recruitment and activation of immune cells into the TME, but can also be recognized by P2RX7-expressing non-immune cells, such as tumor cells. Given that the role of P2RX7 on cancer cell death and proliferation has been widely studied [8], this review will mainly focus on the role of P2RX7 in modulating the antitumor immune response and its potential to constitute a novel antitumoral strategy.

2. P2RX7: A Unique P2X Receptor with Several Activities Impacting Tumor Growth

P2RX7 belongs to the family of P2X receptors that are assembled and active when in their trimeric form. Each monomer is composed of two transmembrane domains that are connected by a large extracellular loop, and an N- and C- termini domain located intracellularly. However, unlike other members, P2RX7 has a long intracellular C-terminal domain that structurally distinguishes it from the others and confers its unique biological activities.
Even though all seven members of the P2X receptors recognize eATP, they are activated with various affinities that range from 0.5 µM for P2RX3 to over 100 µM for P2RX7 [9]. Thus, activation of P2RX7 requires high levels of eATP, levels that are found in the TME [10] which controls the three main activities of the receptor: cationic exchange, macropore opening and NLRP3 inflammasome activation.

2.1. Cationic Exchange

P2RX7, as well as the other P2X receptors, are non-selective cationic channels that lead to membrane depolarization when bound to eATP. Indeed, activation of P2RX7 leads to calcium and sodium influx as well as potassium efflux, which ultimately activate various signaling pathways that range from cell proliferation to immune-related events [11].
It has been reported that low levels of eATP can induce cell proliferation in a P2RX7-dependent manner through calcium influx [8]. Indeed, intracellular calcium is a second messenger that controls various cellular functions. It has been shown that the ATP/P2RX7 axis leads to the activation of MAP kinases (ERK1/2, p38, JNK), B kinases (PKB, Akt) and PKC involved in cell proliferation in several types of cancer, in mouse and human models [8,11]. Moreover, increase of intracellular calcium levels following P2RX7 activation has been shown to promote cancer cell survival via mitochondrial ion homeostasis [12], calcineurin activation and NFAT translocation into the nucleus [13].
P2RX7-dependent calcium influx has also been shown to be implicated in the immune response. P2RX7 stimulation in mouse microglial cells induced NFAT and NF-κB translocation into the nucleus in a calcineurin-dependent manner, and both are required for the expression of inflammatory cytokines [14] and chemokines such as CCL3 [15]. On the other hand, the increase of intracellular calcium and extracellular potassium concentrations have been shown to limit B cell activation by decreasing NFAT translocation into the nucleus [16]. P2RX7 has also been shown to have a role in immune cell proliferation [17]. Indeed, the activation of the ATP/P2RX7 axis in human T cells induces NFAT translocation and IL-2 secretion, necessary for T cell survival and proliferation [18]. Accordingly, it has been shown that the expression of P2RX7 on mouse CD8+ T cells enhances mitochondrial functions through calcium influx and AMPK activation, required to support the generation and survival of memory T cells [19] and efficiently eradicate tumor cells [20]. In line with these findings, the same authors have shown, in viral infections mouse models, that P2RX7 induces the expression of the TGBRII receptor through calcineurin activation, thereby sustaining the generation of tissue-resident memory T cells [21] that require TGFβ sensing for their survival. However, a contradictory study had previously shown, in a mouse model of melanoma, that the expression of P2RX7 on tumor-infiltrating lymphocytes has a detrimental role by limiting T cell proliferation and inducing T cell senescence through mitochondrial reactive oxygen species (ROS) and p38 MAPK activation [22]. The discrepancy between the studies could be due to a different activation protocol of T cells, as explained by the authors [20].

2.2. Macropore Opening

One particular characteristic of P2RX7 is the capacity to induce cell permeabilization and death, which paved the way to the discovery of the receptor. The receptor was initially named P2Z since high levels of ATP were shown to induce cell death in human macrophages [23]. Its cloning in 1996 highlighted that its structure is close to the P2X receptors [24], which led to it becoming the seventh member of the P2X family.
The P2RX7-dependent cell permeabilization is linked to its ability to cause the opening of macropores of 8.5 Å on the plasma membrane, leading to cell death [25]. The pores allow the passing of macromolecules up to 900 Da in a non-selective and bidirectional manner, disturbing the intracellular homeostasis [25]. The nature of this pore is still under debate. Either (1) P2RX7 recruits pore-forming proteins following its activation, such as pannexine-1 or connexine-43 [26,27,28], or (2) the pores are shaped due to the dilatation of the canal formed by the three monomers of P2RX7 [29,30,31,32]. Nevertheless, it is certain that the pore-forming activity of P2RX7 requires the presence of 177 aminos acids in the C-terminal domain [24], which explains the selective activity of P2RX7, as compared to the other P2X receptors.
Given its ability to induce cell death, one can speculate that the expression of P2RX7 on tumor cells is beneficial. Indeed, studies using tumor cell lines and preclinical mouse models in melanoma [33,34], non-melanoma skin cancers [35], intestinal carcinomas [36] and breast cancer [37] have shown that activation of P2RX7 reduced cell proliferation by inducing cell death. On the other hand, many reports show an increase of P2RX7 expression in various tumor cell lines and in patients [38,39,40,41,42] and this increase is linked to tumor growth, as discussed in the previous section. When the human P2RX7 gene is subjected to alternative splicing, a C-terminal-deleted variant named P2RX7-B [43] is generated, preventing the receptor from forming macropores. This accords with reports that P2RX7-B is overexpressed in lung adenocarcinomas [44], acute myeloid leukemia [45], osteosarcoma [46], neuroblastoma [47] and glioblastoma [48]. In addition, a non-functional form of P2RX7 that lacks macropore activity has been reported in several types of cancers including lung and glioblastomas [49,50]. Whether the expression of the non-functional form is linked to P2RX7-B is currently unknown. However, the existence of isoforms unable to form macropores can explain how a cell death-inducing receptor can favor tumor growth.
Not only is the macropore activity associated with tumor cell death, but also with immune functions. P2RX7 is implicated in the regulation of T cell homeostasis through induction of cell death, which is linked to the levels of expression of the receptor. Indeed, the expression levels of P2RX7 are regulated according to the T cell subset, its activation status as well as its localization as extensively reviewed in [51,52]. For instance, it has been shown that the expression of P2RX7 is downregulated following the activation of T cells, protecting them from cell death [53]. This is the case for follicular T cells in Peyer’s Patches [54] and tissue-resident memory T cells [55,56]. The purpose of P2RX7’s downregulation is not only to prevent T cell death by protecting antigen-reactive T cells, but also to favor the ionic activity of the receptor and to confer a better fitness of CD8+ T cells [19], which consequently enhances the control of tumor growth [20].
On the other hand, T regulatory cells (Tregs) and natural killer T cells (NKT) express high levels of P2RX7 and are more susceptible to P2RX7-induced cell death [51,57]. We have shown that p2rx7-/- mice with colitis-associated cancer develop bigger and more aggressive lesions than WT mice, which was associated with an accumulation of Tregs within the lesions [58]. Moreover, early depletion of Tregs in vivo by activation of P2RX7-expressing T cells using the NAD+/ART2 axis (known to activate P2RX7 in mouse T cells [53]) in tumor mouse models increased the anti-tumor effector functions of CD8+ T cells [59].

2.3. NLRP3 Inflammasome Activation

Another feature of P2RX7 that differentiates it from other P2X receptors is its ability to activate the NLRP3 inflammasome. The NLRP3 (NOD-like receptor family, pyrin domain containing 3) protein belongs to the NOD-Like receptors (NLR) family which are pattern recognition receptors (PRR), meaning that NLRP3 is implicated in danger signal recognition [60], highlighting its importance in the establishment of the immune response, notably in the context of cancer. It is, therefore, mainly studied in the innate immune cells from the myeloid lineage such as macrophages, monocytes and dendritic cells. This is supported by the high expression levels of P2RX7 on these cells compared to other immune cells [11,61].
Even though NLRP3 is not a receptor per se, it is a cytosolic sensor of stress that interacts with other proteins to form a multimeric complex of proteins called the NLRP3 inflammasome. The complex involves the NLRP3 protein (containing a pyrin domain), the apoptosis-associated speck-like protein containing a CARD (ASC, with a CARD and pyrin domain) and the effector protein pro-caspase-1 (with a CARD domain). The proteins associate by way of ASC, which acts as an anchor through the respective binding of the CARD and pyrin domains of NLRP3 and pro-caspase-1. The NEK7 protein (NIMA-related kinase 7) is necessary for the oligomerization of the NLRP3 subunits to form the NLRP3 inflammasome [62]. The oligomerized NLRP3 inflammasome causes the self-cleavage of the pro-caspase-1 into active caspase-1, which, in turn, cleaves the inactive precursors of the IL-1β and IL-18 inflammatory cytokines intracellularly to shape the immune response.
The ATP/P2RX7 axis is not the only activator of the NLRP3 inflammasome but is described as the most potent [63]. Indeed, the NLRP3 inflammasome can also be activated by a variety of stress signals such as pore-forming toxins, ionophores or uric acid crystals [64]. It has been reported that the main event leading to the assembly of the NLRP3 inflammasome is potassium efflux, which is common for all its activators, including ATP [64,65]. Potassium efflux is a common feature of all the P2X receptors as well as other potassium channels, yet they are unable to independently activate the NLRP3 inflammasome. Indeed, the C-terminal domain of P2RX7 is necessary for the activation of NLRP3 by ATP, since its absence prevents NLRP3 activation and IL-1β release [32]. Moreover, unlike the other P2X receptors, P2RX7 does not desensitize [9], leading to continuous potassium efflux that may facilitate NLRP3 activation. On the other hand, NLRP3 and P2RX7 have been shown to interact at the plasma membrane either directly [66] or indirectly via the Paxillin protein [67] to induce the assembly of NLRP3. It has also been shown that P2RX7-mediated NLRP3 activation requires the potassic channel TWIK2, where both receptors act in synergy to decrease intracellular potassium levels and activate NLRP3 [68]. Besides potassium efflux, P2RX7-mediated calcium influx has also been shown to assemble NLRP3, through ROS production and mitochondrial depolarization [69]. Therefore, the potency of P2RX7 to activate NLRP3 could be explained by the fact that P2RX7 can induce calcium influx, potassium efflux and interact with other potassic proteins as well as with NLRP3 itself, leading to a robust activation of the NLRP3 inflammasome.
The role of NLRP3 in cancer progression is not clear. Indeed, there are several conflicting studies in various types of cancer showing that NLRP3 can either favor tumor growth or act as a tumor suppressor [70,71]. Since the readout activity of NLRP3 is the release of mature inflammatory cytokines (Figure 1), we will focus in the next section on the role of IL-1β and IL-18 in cancer progression.

3. IL-1β and IL-18: The Main Players in the Immunomodulation by P2RX7

Even though the activation P2RX7 induces a plethora of immune-related events such as T cell activation via the shedding of CD62L [51], the control of phagocytosis [72] and the release of cytokines [73,74], the main and most studied function of P2RX7 is its ability to release IL-1β and IL-18.

3.1. Biology and Signaling of IL-1β and IL-18

As mentioned before, the IL-1β and IL-18 cytokines are released from the cell after the cleavage of their precursors (pro-IL-1β and pro-IL-18, respectively) by caspase-1. However, a priming step prior to the activation of P2RX7 is required, involving the activation of TLR4/NF-κB pathway by Pathogen Associated Molecular Patterns (PAMPs). Not only does this priming step allow post translational modifications on NLRP3 for an optimal assembly [75,76], but it is also crucial in inducing the expression of pro-IL-1β and upregulating the expression of the other components of the inflammasome. Unlike pro-IL-1β, the expression of pro-IL-18 is constitutive, as seen in immune cells [77] and in epithelial cells and pulmonary fibroblasts [78,79]. It was reported in the past that the activation of the NLRP3 inflammasome cannot occur without the priming step; however, it has recently been shown that it can be dispensable in the release of IL-18 from human monocytes [80].
Apart from the fact that the differential expression of the cytokines contributes to the regulation of their activity, both cytokines are regulated on another level that involves their receptors and antagonists, as discussed hereafter.

3.1.1. IL-1β

IL-1β is recognized by the IL-1R1 receptor. The binding of IL-1β to its receptor triggers the recruitment of the IL-1RAcP adaptor protein to form an IL-1β/IL-1R1/IL-R1AcP complex required for the activation of the downstream MYD88/NF-κB signaling. Another cell surface receptor called IL-1R2 can also bind IL-1β with higher affinity than IL-1R1, but is a decoy receptor. IL-1R2 prevents the recruitment of the MYD88 protein and the activation the NF-κB pathway, since it lacks the intracellular TIR domain required for MYD88 recruitment. The IL-1β signaling can also be inhibited by another antagonist called IL-1Ra that binds the IL-1R1 receptor and blocks the association of IL-1RAcP to the complex [81,82,83].

3.1.2. IL-18

IL-18 signaling is similar to that of IL-1β. IL-18 binds to the IL-18Rα (IL-18R1) subunit recruiting the high affinity IL-18Rβ (IL-18RAP) subunit for MYD88/NF-κB activation. However, IL-18Rβ is only expressed by T cells, NK cells and dendritic cells [84]. Indeed, IL-18 was initially named IGIF (IFN-γ inducing factor) [85], as this cytokine induces IFN-γ production by T cells and NK cells after its binding to the IL-18Rα/IL-18Rβ complex. IL-18 binds, however, with higher affinity to its natural antagonist called IL-18 Binding Protein (IL-18BP). IL-18BP is constitutively secreted in high amounts to prevent the binding of IL-18 to its receptor and acts as a negative feedback loop to IL-18 activity, since its expression is increased when high levels of IFN-γ are produced [84].
As IL-1β was discovered long before IL-18, its role in inflammation and cancer progression has been vastly studied and well-reviewed [70,86,87,88]. IL-1β has been shown to have both pro- and anti-tumor effects. For instance, it is known that IL-1β can favor tumor growth by inducing an immunosuppressive environment through the recruitment of myeloid-derived suppressors cells (MDSCs) and tumor-associated macrophages (TAMs) [89,90], as well as by favoring angiogenesis [91,92]. On another hand, IL-1β has also been shown to shape the T cell response and favor antitumor immunity through the ATP/P2RX7/NLRP3 axis in dendritic cells [93,94]. As IL-18 has recently attracted attention in tumor biology, and the implication of IL-1β was extensively discussed elsewhere, the rest of this review will focus on the role of IL-18 in cancer progression.

3.2. IL-18: An Antitumoral Cytokine

The role of IL-18 has been studied in several types of cancer. Preclinical studies have shown that IL-18 is required to inhibit tumor growth using either il18-/- or il18r1-/- mice, or even recombinant IL-18 administration.
In AOM/DSS-induced colon cancer, it has been shown that IL-18 levels were decreased in tumor-bearing mice [95], that the NLRP3/ASC/CASP1/IL-18 axis was important to dampen tumor growth [95,96] and this was dependent on IFN-γ production [97]. Indeed, there is an absence of the components of the NLRP3 inflammasome, IL-18 exacerbated cancer progression [97] and inflammatory markers [98]. In line with this, systemic administration of IL-18 in the same model and in the MC38 subcutaneous model reduced disease aggressiveness as well as the cytotoxicity of NK cells, CD4+ and CD8+ T cells [96,99,100,101], thus, demonstrating the ability of IL-18 to enhance antitumor immunity by boosting IFN-γ production. Moreover, IL-18 production and administration was shown to limit the migration of colon MC38 tumor cells into the liver by increasing the activation of NK cells and their cytotoxicity through FASL [102]. Similar results have been shown in pancreatic, metastatic and non-metastatic melanoma mouse models, where in vivo administration of IL-18 slowed tumor growth by increasing the activation and cytotoxicity of CD4+ and CD8+ T cells and NK cells [101,103,104,105], as well as their interaction with tumor cells and the robust generation of memory T cells [106]. It has also been recently shown that IL-18 administration after bone marrow transplantation induced the cytotoxicity of T cells in a mouse model of myeloma and enhanced their lethality in a leukemia model [107].
The role of the ATP/P2RX7/NLRP3 axis in IL-18 release and antitumor immunity was investigated. We have shown that activation of P2RX7 using a positive modulator (HEI3090) inhibits tumor growth in lung and melanoma mouse models. This effect relies on IL-18 production by dendritic cells to boost antitumor immunity, e.g., IFN-γ production by CD4+ T cells and NK cells [85,103]. Moreover, using blocking antibodies against the ectonucleotidases responsible for ATP degradation, namely CD73 (or NT5E) and CD39 (or ENTPD1), antitumor immunity was boosted through the P2RX7/NLRP3 pathway, and tumor growth was inhibited in mouse models of melanoma, fibrosarcoma, colon and prostate tumors [108,109]. It is interesting to note that these studies have also shown that IL-18 can synergize with αPD-1 immunotherapy or chemotherapy for stronger antitumor immunity and tumor growth inhibition. Indeed, activation of the P2RX7/IL-18 axis with HEI3090 sensitized lung and melanoma tumors to immunotherapy, cured 80% of mice and protected them from a tumor rechallenge, highlighting the potency of IL-18 in generating long lasting memory T cells in tumor models [110]. The ability of IL-18 to generate memory T cells is mainly studied in infection models, where IL-18R is shown to be upregulated [111,112]. This is supported by the fact that the release of IL-18 in DC and the IL-18R/MyD88/IFN-γ axis in T cells during T cell activation favors the expansion of memory T cells [113,114,115]. Interestingly, IL-18 can also induce IFN-γ production even in absence of TCR stimulation [111,112]. These studies underly the power of IL-18 in enhancing antitumor immunity through T cell cytotoxicity and T memory cell formation.
IL-18 increases the activation and cytotoxicity of T cells and NK cells, as evidenced in tumor models. In vitro activation of effector NK and T cells before their transfer in vivo has shown powerful antitumor effects and constitute a promising strategy. Transfer of in vitro pre-activated NK cells with IL-18, IL-12 (another inducer of IFN-γ) and IL-15 (activator of NK cells) in a melanoma mouse model under radiation therapy enhanced antitumor immunity and decreased lung metastases, by increasing the accumulation and cytotoxicity of in vivo NK cells that required the presence of CD4+ T cells [116]. The same efficacy in NK cell cytotoxicity was observed in human NK cells derived from patients with metastatic melanoma [117] and healthy subjects, and this enhanced the killing of human colon, melanoma, glioblastoma, prostate, breast and ovarian tumor cell line spheroids [118]. Due to the powerful antitumor potential of IL-18, it has been used in preclinical studies as a tool to increase the proliferation, survival and cytotoxicity of CAR T cells (Chimeric Antigen Receptor T cells) in melanoma [119,120,121,122,123,124] and small cell lung cancer [125].
The antitumor potential of IL-18 has also been evaluated in cancer patients. Levels of IL-18 and IFN-γ were shown to be lower in plasmas of non-small cell lung cancer patients [126] and in tumor tissue of colon cancer, melanoma, thyroid and esophageal cancer patients compared to those of healthy subjects, and low levels of IL-18 correlated with the presence of metastases, a poor outcome [127,128,129,130,131] and increased risk of developing acute myeloid leukemia [132]. Moreover, IL-18 levels were shown to be correlated with higher CD8+ T cells, NK cells infiltration and activity in colorectal cancer [133], melanoma [129] and thyroid carcinoma [130]. Accordingly, functional intratumoral CD8+ T cells were shown to express high levels of IL-18R in non-small cell lung cancer patients and were, furthermore, cytotoxic in the presence of IL-18 [126]. IL-18R was clearly shown to be correlated to immune infiltrate in lung squamous carcinoma and to improved overall survival [134]. However, IL-18BP levels have been shown to be high in sera of patients with ovarian [135], non-small cell lung cancer [101] and prostate cancer [136], as well as in tumor tissue of melanoma, breast, head and neck, gastric, ovarian and prostate cancers [101,135,136]. These findings are in alignment with the observation that high levels of IL-18 in sera and tissue of ovarian cancer patients were shown to be inactive given the lack of IFN-γ production [137]. This is consistent with the fact that IL-18 signaling is linked to better antitumor response and survival. Indeed, it was shown that after the first round of αPD-L1 immunotherapy, IL-18 levels increase in sera of patients with multiple myeloma, non-small cell lung cancer, renal cell carcinoma and melanoma, which is linked to a reactivation and cytotoxicity of antitumor effector cells, namely IFN-γ production [138,139].
Given the potency of IL-18 to trigger the activation and cytotoxicity of immune effector cells and its ability to enhance the efficacy of immunotherapy, and given the lack of biomarkers to predict patients’ response to immunotherapy, recent studies have investigated the potential of IL-18 in predicting patient outcome. High levels of IL-18 were shown to be found in plasmas and tissue of immunotherapy-responding non-small cell lung cancer patients, and were linked to an antitumor immune gene signature [140] and a reduced tumor burden [139,141].

3.3. IL-18: A Pro Tumoral Cytokine

Even though IL-18 has extensively been shown to boost antitumor immunity and to be associated with a better outcome in both murine and human settings, several studies have pointed out a pro-tumoral role of IL-18.
Indeed, IL-18 is associated with increased migration of cancer cells and metastatic activity. In vitro studies have shown that the migration ability and proliferation of human leukemic cells, lymphoma [142,143], human gastric cells and murine melanoma was dependent on their production of IL-18 in a NLRP3/caspase-1-dependent manner through VEGF production [144,145,146], and this was further increased when cells were treated with recombinant IL-18 [147]. Furthermore, in vivo neutralization of IL-18 with IL-18BP reduced melanoma metastases into the liver in a mouse model [148] and il18-/- mice exhibited less tumor growth in a multiple myeloma mouse models [149]. In multiple myeloma, the pro-tumoral effect of IL-18 was shown to be due of its release in a NLRP3-dependent from TAMs [150] and favored the immunosuppressive activity of MDSCs [149]. Not only levels of IL-18 were increased in tumor tissue of mouse models of pancreatic cancers [151,152], it was also shown to have a pro-tumoral activity. Indeed, IL-18 release is dependent on NLRP3 activation, and its signaling through the IL-18R receptor induces the exhaustion of CD8+ T cells [153] and limited migration into the tumor [154]. This is supported by another study in melanoma and colon tumor mouse models showing that IL-18 induces the expression of PD-1 on NK cells [155]. An earlier study had shown that NF-κB-dependent expression of IL-18 by human and murine pancreatic tumor cells increased their proliferation and invasion in vitro and in vivo, and they were blocked in the presence of IL-18BP. However, the authors also showed that IL-18 enhanced the cytotoxicity of NK cells and T cells, making IL-18 a cytokine with both pro- and antitumoral proprieties. The combination of NF-κB inhibition and IL-18 administration to mice significantly reduced tumor growth, taking advantage of both properties of the cytokine [152]. IL-18 would have a different effect depending on the cell type, which demonstrated by the facts that human pancreatic tumor tissue expresses high levels of IL-18 [152,153,156,157] and low levels of IL-18BP [156] and these are linked to shorter survival and increased metastasis, whereas high levels of plasmatic IL-18 are associated with a better outcome [152]. Similarly, NLRP3-associated IL-18 is found in high levels in plasma of lymphoma [142] and multiple myeloma patients where IL-18 is a predictor of poor survival in multiple myeloma patients [149,158], esophageal carcinoma [159], renal cell carcinoma [160] and gastric carcinoma [161]. High expression of IL-18 is also associated with increased metastases in hepatocellular and breast carcinomas [162,163]. As for the role of IL-18 in predicting patients’ response to immunotherapy, it was shown that patients with non-small cell lung cancer who respond to treatment had lower levels of IL-18 compared to non-responders [164], which contrasts with other studies in the same tumor type [140,141].

4. The P2RX7/IL-18 Pathway: A Target in Cancer?

P2RX7 can be considered to be a fine tuner of the tumor microenvironment, given that it is able to induce cell proliferation and death as well as to modulate the immune response through various pathways, notably through the release of IL-18 following the activation of the NLRP3 inflammasome.
Even though P2RX7 has been shown to promote tumor growth when expressed by tumor cells [8], its expression and activity on immune cells has mostly been associated with antitumor effects by enhancing antitumor immunity. As described by us and others, this effect relies on the NLRP3-dependent release of IL-18 by dendritic cells, which enhances the cytotoxicity of antitumor effector cells. This is in agreement with other work showing a strong antitumor activity of IL-18 as well as its potential as a biomarker for tumor progression and patient outcomes. However, several other studies pointed out a pro-tumoral role of IL-18.
The dual roles of IL-18 could have several explanations. IL-18 could have a pro-tumoral activity in certain types of cancers, such as in pancreatic cancer and multiple myeloma, as discussed in this review, and the source and frequency of IL-18 release may contribute to its effect. Indeed, it was shown that daily systemic administration of rIL-18 had antitumor effects, whereas once weekly or bi-weekly administration favored lung metastases of melanoma cells and induced a strong immunosuppressive environment [155], hinting that the levels of IL-18 are critical for its effect. The pro-tumoral role of IL-18 seems also to be linked to its release from tumor cells, whereas its antitumoral role seems mainly be linked to its release from dendritic cells, therefore, shaping the immune response. This supports the predictive value of plasmatic IL-18, since it can be released from tumor and immune cells. In line with this, it has been shown that the processing of IL-18 from ovarian tumor cells can be defective [165,166], which could explain its pro-tumoral activity when released from tumor cells. Only a few studies have addressed the activity of IL-18, i.e., the free, unbound IL-18 from IL-18BP. Indeed, IL-18BP is present physiologically at high levels and has been shown to be further increased in several cancers, inhibiting even more IL-18 signaling. Therefore, determining the levels of free, unbound IL-18 is key to understanding its role. It should be cautioned that the exact role of IL-18 can be misinterpreted when looking at mRNA levels. Since it is constitutively expressed and requires a proteolytic cleavage to be released and active, looking at the proteic levels of IL-18 would be more meaningful. Additionally, the pro-tumoral role of IL-18 seems to be linked to T cell and NK cell exhaustion. This could be beneficial when combined with immune checkpoint inhibitors. This was in fact the case when using HEI3090 or anti-CD39/CD73 or even DR-IL-18 (a decoy resistant IL-18) [101,108,109,110]. The exact role of IL-18 in cancer progression warrants further investigation to determine (i) the cancer type, (ii) the cell source and (iii) its activity (free unbound IL-18).
In any case, the studies have been consistent in showing that IL-18 release in an immune-P2RX7-dependent manner is highly antitumoral, and suggesting that boosting the P2RX7/IL-18 pathway is promising for several types of cancer. However, one cannot exclude the studies that show P2RX7 itself has also pro-tumoral effects. Even though we and others have shown in vivo that boosting the activation of P2RX7 occurs mostly in immune cells [108,110], caution should also be taken with the functionality of the receptor expressed by tumor cells.

5. Conclusions

Preclinical studies point towards strong antitumor efficacy of the P2RX7/IL-18 axis in several types of cancers. Targeting the P2RX7/IL-18 axis is therefore promising in humans and encourages further studies to confirm these results in human settings.

Author Contributions

Writing—original draft preparation: S.J.d.H.; Review and editing: S.J.d.H. and V.V.-C.; Validation: P.H.; Funding acquisition: V.V.-C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Cancéropôle PACA, Bristol-Myers Squibb Foundation for Research in Immuno-Oncology, the French Government (National Research Agency, ANR through the “Investments for the Future”: program reference #ANR-11-LABX-0028-01). S.J.d.H. is funded by the “Ligue Nationale Contre le Cancer”, the “Fondation pour la recherche médicale” grant number #FDT202106013099 and ARC (grant number ARCTHEM2021020003478).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This publication is based upon discussion from PRESTO COST action CA21130 supported by COST (European Cooperation in Science and Technology). Figure was edited with BioRender application (BioRender.com).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ping, Q.; Yan, R.; Cheng, X.; Wang, W.; Zhong, Y.; Hou, Z.; Shi, Y.; Wang, C.; Li, R. Cancer-Associated Fibroblasts: Overview, Progress, Challenges, and Directions. Cancer Gene Ther. 2021, 28, 984–999. [Google Scholar] [CrossRef] [PubMed]
  2. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  3. Antonia, S.J.; Borghaei, H.; Ramalingam, S.S.; Horn, L.; De Castro Carpeño, J.; Pluzanski, A.; Burgio, M.A.; Garassino, M.; Chow, L.Q.M.; Gettinger, S.; et al. Four-Year Survival with Nivolumab in Patients with Previously Treated Advanced Non-Small-Cell Lung Cancer: A Pooled Analysis. Lancet Oncol. 2019, 20, 1395–1408. [Google Scholar] [CrossRef] [PubMed]
  4. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.-G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in Combination with Carboplatin plus Nab-Paclitaxel Chemotherapy Compared with Chemotherapy Alone as First-Line Treatment for Metastatic Non-Squamous Non-Small-Cell Lung Cancer (IMpower130): A Multicentre, Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef] [PubMed]
  5. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef] [PubMed]
  6. Janho dit Hreich, S.; Benzaquen, J.; Hofman, P.; Vouret-Craviari, V. The Purinergic Landscape of Non-Small Cell Lung Cancer. Cancers 2022, 14, 1926. [Google Scholar] [CrossRef]
  7. Kepp, O.; Bezu, L.; Yamazaki, T.; Di Virgilio, F.; Smyth, M.J.; Kroemer, G.; Galluzzi, L. ATP and Cancer Immunosurveillance. EMBO J. 2021, 40, e108130. [Google Scholar] [CrossRef]
  8. Janho Dit Hreich, S.; Benzaquen, J.; Hofman, P.; Vouret-Craviari, V. To Inhibit or to Boost the ATP/P2RX7 Pathway to Fight Cancer-That Is the Question. Purinergic Signal. 2021, 17, 619–631. [Google Scholar] [CrossRef]
  9. Illes, P.; Müller, C.E.; Jacobson, K.A.; Grutter, T.; Nicke, A.; Fountain, S.J.; Kennedy, C.; Schmalzing, G.; Jarvis, M.F.; Stojilkovic, S.S.; et al. Update of P2X Receptor Properties and Their Pharmacology: IUPHAR Review 30. Br. J. Pharmacol. 2021, 178, 489–514. [Google Scholar] [CrossRef]
  10. Pellegatti, P.; Raffaghello, L.; Bianchi, G.; Piccardi, F.; Pistoia, V.; Di Virgilio, F. Increased Level of Extracellular ATP at Tumor Sites: In Vivo Imaging with Plasma Membrane Luciferase. PLoS ONE 2008, 3, e2599. [Google Scholar] [CrossRef]
  11. Kopp, R.; Krautloher, A.; Ramírez-Fernández, A.; Nicke, A. P2X7 Interactions and Signaling—Making Head or Tail of It. Front. Mol. Neurosci. 2019, 12, 183. [Google Scholar] [CrossRef]
  12. Adinolfi, E.; Callegari, M.G.; Ferrari, D.; Bolognesi, C.; Minelli, M.; Wieckowski, M.R.; Pinton, P.; Rizzuto, R.; Di Virgilio, F. Basal Activation of the P2X7 ATP Receptor Elevates Mitochondrial Calcium and Potential, Increases Cellular ATP Levels, and Promotes Serum-Independent Growth. Mol. Biol. Cell 2005, 16, 3260–3272. [Google Scholar] [CrossRef]
  13. Adinolfi, E.; Callegari, M.G.; Cirillo, M.; Pinton, P.; Giorgi, C.; Cavagna, D.; Rizzuto, R.; Di Virgilio, F. Expression of the P2X7 Receptor Increases the Ca2+ Content of the Endoplasmic Reticulum, Activates NFATc1, and Protects from Apoptosis. J. Biol. Chem. 2009, 284, 10120–10128. [Google Scholar] [CrossRef] [PubMed]
  14. Ferrari, D.; Stroh, C.; Schulze-Osthoff, K. P2X7/P2Z Purinoreceptor-Mediated Activation of Transcription Factor NFAT in Microglial Cells. J. Biol. Chem. 1999, 274, 13205–13210. [Google Scholar] [CrossRef]
  15. Kataoka, A.; Tozaki-Saitoh, H.; Koga, Y.; Tsuda, M.; Inoue, K. Activation of P2X7 Receptors Induces CCL3 Production in Microglial Cells through Transcription Factor NFAT. J. Neurochem. 2009, 108, 115–125. [Google Scholar] [CrossRef]
  16. Pippel, A.; Beßler, B.; Klapperstück, M.; Markwardt, F. Inhibition of Antigen Receptor-Dependent Ca(2+) Signals and NF-AT Activation by P2X7 Receptors in Human B Lymphocytes. Cell Calcium 2015, 57, 275–289. [Google Scholar] [CrossRef]
  17. Baricordi, O.R.; Ferrari, D.; Melchiorri, L.; Chiozzi, P.; Hanau, S.; Chiari, E.; Rubini, M.; Di Virgilio, F. An ATP-Activated Channel Is Involved in Mitogenic Stimulation of Human T Lymphocytes. Blood 1996, 87, 682–690. [Google Scholar] [CrossRef]
  18. Yip, L.; Woehrle, T.; Corriden, R.; Hirsh, M.; Chen, Y.; Inoue, Y.; Ferrari, V.; Insel, P.A.; Junger, W.G. Autocrine Regulation of T-cell Activation by ATP Release and P2X7 Receptors. FASEB J. 2009, 23, 1685–1693. [Google Scholar] [CrossRef]
  19. Borges da Silva, H.; Beura, L.K.; Wang, H.; Hanse, E.A.; Gore, R.; Scott, M.C.; Walsh, D.A.; Block, K.E.; Fonseca, R.; Yan, Y.; et al. The Purinergic Receptor P2RX7 Directs Metabolic Fitness of Long-Lived Memory CD8+ T Cells. Nature 2018, 559, 264–268. [Google Scholar] [CrossRef]
  20. Wanhainen, K.M.; Peng, C.; Zhou, M.H.; de Gois Macedo, B.; O’Flanagan, S.; Yang, T.; Kelekar, A.; Burbach, B.J.; Borges da Silva, H.; Jameson, S.C. P2RX7 Enhances Tumor Control by CD8+ T Cells in Adoptive Cell Therapy. Cancer Immunol. Res. 2022, 10, 871–884. [Google Scholar] [CrossRef] [PubMed]
  21. Borges da Silva, H.; Peng, C.; Wang, H.; Wanhainen, K.M.; Ma, C.; Lopez, S.; Khoruts, A.; Zhang, N.; Jameson, S.C. Sensing of ATP via the Purinergic Receptor P2RX7 Promotes CD8+ Trm Cell Generation by Enhancing Their Sensitivity to the Cytokine TGF-β. Immunity 2020, 53, 158–171.e6. [Google Scholar] [CrossRef] [PubMed]
  22. Romagnani, A.; Rottoli, E.; Mazza, E.M.C.; Rezzonico-Jost, T.; De Ponte Conti, B.; Proietti, M.; Perotti, M.; Civanelli, E.; Perruzza, L.; Catapano, A.L.; et al. P2X7 Receptor Activity Limits Accumulation of T Cells within Tumors. Cancer Res. 2020, 80, 3906–3919. [Google Scholar] [CrossRef]
  23. Falzoni, S.; Munerati, M.; Ferrari, D.; Spisani, S.; Moretti, S.; Di Virgilio, F. The Purinergic P2Z Receptor of Human Macrophage Cells. Characterization and Possible Physiological Role. J. Clin. Investig. 1995, 95, 1207–1216. [Google Scholar] [CrossRef]
  24. Surprenant, A.; Rassendren, F.; Kawashima, E.; North, R.A.; Buell, G. The Cytolytic P2Z Receptor for Extracellular ATP Identified as a P2X Receptor (P2X7). Science 1996, 272, 735–738. [Google Scholar] [CrossRef]
  25. Di Virgilio, F.; Schmalzing, G.; Markwardt, F. The Elusive P2X7 Macropore. Trends Cell Biol. 2018, 28, 392–404. [Google Scholar] [CrossRef]
  26. Beyer, E.C.; Steinberg, T.H. Evidence That the Gap Junction Protein Connexin-43 Is the ATP-Induced Pore of Mouse Macrophages. J. Biol. Chem. 1991, 266, 7971–7974. [Google Scholar] [CrossRef] [PubMed]
  27. Locovei, S.; Scemes, E.; Qiu, F.; Spray, D.C.; Dahl, G. Pannexin1 Is Part of the Pore Forming Unit of the P2X7 Receptor Death Complex. FEBS Lett. 2007, 581, 483–488. [Google Scholar] [CrossRef]
  28. Pelegrin, P.; Surprenant, A. Pannexin-1 Mediates Large Pore Formation and Interleukin-1β Release by the ATP-Gated P2X7 Receptor. EMBO J. 2006, 25, 5071–5082. [Google Scholar] [CrossRef]
  29. Alberto, A.V.P.; Faria, R.X.; Couto, C.G.C.; Ferreira, L.G.B.; Souza, C.A.M.; Teixeira, P.C.N.; Fróes, M.M.; Alves, L.A. Is Pannexin the Pore Associated with the P2X7 Receptor? Naunyn Schmiedebergs Arch. Pharmacol. 2013, 386, 775–787. [Google Scholar] [CrossRef]
  30. Karasawa, A.; Michalski, K.; Mikhelzon, P.; Kawate, T. The P2X7 Receptor Forms a Dye-Permeable Pore Independent of Its Intracellular Domain but Dependent on Membrane Lipid Composition. eLife 2017, 6, e31186. [Google Scholar] [CrossRef] [PubMed]
  31. Smart, M.L.; Gu, B.; Panchal, R.G.; Wiley, J.; Cromer, B.; Williams, D.A.; Petrou, S. P2X7 Receptor Cell Surface Expression and Cytolytic Pore Formation Are Regulated by a Distal C-Terminal Region. J. Biol. Chem. 2003, 278, 8853–8860. [Google Scholar] [CrossRef]
  32. Solle, M.; Labasi, J.; Perregaux, D.G.; Stam, E.; Petrushova, N.; Koller, B.H.; Griffiths, R.J.; Gabel, C.A. Altered Cytokine Production in Mice Lacking P2X7Receptors. J. Biol. Chem. 2001, 276, 125–132. [Google Scholar] [CrossRef]
  33. White, N.; Butler, P.E.M.; Burnstock, G. Human Melanomas Express Functional P2X7 Receptors. Cell Tissue Res. 2005, 321, 411–418. [Google Scholar] [CrossRef]
  34. White, N.; Knight, G.E.; Butler, P.E.M.; Burnstock, G. An in Vivo Model of Melanoma: Treatment with ATP. Purinergic Signal. 2009, 5, 327–333. [Google Scholar] [CrossRef]
  35. Greig, A.V.H.; Burnstock, G.; Linge, C.; Healy, V.; Lim, P.; Clayton, E.; Rustin, M.H.A.; Angus McGrouther, D. Expression of Purinergic Receptors in Non-Melanoma Skin Cancers and Their Functional Roles in A431 Cells. J. Investig. Dermatol. 2003, 121, 315–327. [Google Scholar] [CrossRef]
  36. Coutinho-Silva, R.; Stahl, L.; Cheung, K.-K.; de Campos, N.E.; de Oliveira Souza, C.; Ojcius, D.M.; Burnstock, G. P2X and P2Y Purinergic Receptors on Human Intestinal Epithelial Carcinoma Cells: Effects of Extracellular Nucleotides on Apoptosis and Cell Proliferation. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 288, G1024–G1035. [Google Scholar] [CrossRef]
  37. Huang, S.; Chen, Y.; Wu, W.; Ouyang, N.; Chen, J.; Li, H.; Liu, X.; Su, F.; Lin, L.; Yao, Y. MiR-150 Promotes Human Breast Cancer Growth and Malignant Behavior by Targeting the Pro-Apoptotic Purinergic P2X7 Receptor. PLoS ONE 2013, 8, e80707. [Google Scholar] [CrossRef]
  38. Adinolfi, E.; Raffaghello, L.; Giuliani, A.L.; Cavazzini, L.; Capece, M.; Chiozzi, P.; Bianchi, G.; Kroemer, G.; Pistoia, V.; Di Virgilio, F. Expression of P2X7 Receptor Increases In Vivo Tumor Growth. Cancer Res. 2012, 72, 2957–2969. [Google Scholar] [CrossRef]
  39. Zhang, Y.; Cheng, H.; Li, W.; Wu, H.; Yang, Y. Highly-expressed P2X7 Receptor Promotes Growth and Metastasis of Human HOS/MNNG Osteosarcoma Cells via PI3K/Akt/GSK3β/Β-catenin and MTOR/HIF1α/VEGF Signaling. Int. J. Cancer 2019, 145, 1068–1082. [Google Scholar] [CrossRef]
  40. Qiu, Y.; Li, W.; Zhang, H.; Liu, Y.; Tian, X.-X.; Fang, W.-G. P2X7 Mediates ATP-Driven Invasiveness in Prostate Cancer Cells. PLoS ONE 2014, 9, e114371. [Google Scholar] [CrossRef]
  41. Qian, F.; Xiao, J.; Hu, B.; Sun, N.; Yin, W.; Zhu, J. High Expression of P2X7R Is an Independent Postoperative Indicator of Poor Prognosis in Colorectal Cancer. Hum. Pathol. 2017, 64, 61–68. [Google Scholar] [CrossRef] [PubMed]
  42. Lara, R.; Adinolfi, E.; Harwood, C.A.; Philpott, M.; Barden, J.A.; Di Virgilio, F.; McNulty, S. P2X7 in Cancer: From Molecular Mechanisms to Therapeutics. Front. Pharmacol. 2020, 11, 793. [Google Scholar] [CrossRef] [PubMed]
  43. Benzaquen, J.; Heeke, S.; Janho Dit Hreich, S.; Douguet, L.; Marquette, C.H.; Hofman, P.; Vouret-Craviari, V. Alternative Splicing of P2RX7 Pre-Messenger RNA in Health and Diseases: Myth or Reality? Biomed. J. 2019, 42, 141–154. [Google Scholar] [CrossRef] [PubMed]
  44. Benzaquen, J.; Dit Hreich, S.J.; Heeke, S.; Juhel, T.; Lalvee, S.; Bauwens, S.; Saccani, S.; Lenormand, P.; Hofman, V.; Butori, M.; et al. P2RX7B Is a New Theranostic Marker for Lung Adenocarcinoma Patients. Theranostics 2020, 10, 10849–10860. [Google Scholar] [CrossRef]
  45. Pegoraro, A.; Orioli, E.; De Marchi, E.; Salvestrini, V.; Milani, A.; Di Virgilio, F.; Curti, A.; Adinolfi, E. Differential Sensitivity of Acute Myeloid Leukemia Cells to Daunorubicin Depends on P2X7A versus P2X7B Receptor Expression. Cell Death Dis. 2020, 11, 876. [Google Scholar] [CrossRef]
  46. Tattersall, L.; Shah, K.M.; Lath, D.L.; Singh, A.; Down, J.M.; De Marchi, E.; Williamson, A.; Di Virgilio, F.; Heymann, D.; Adinolfi, E.; et al. The P2RX7B Splice Variant Modulates Osteosarcoma Cell Behaviour and Metastatic Properties. J. Bone Oncol. 2021, 31, 100398. [Google Scholar] [CrossRef]
  47. Arnaud-Sampaio, V.F.; Bento, C.A.; Glaser, T.; Adinolfi, E.; Ulrich, H.; Lameu, C. P2X7 Receptor Isoform B Is a Key Drug Resistance Mediator for Neuroblastoma. Front. Oncol. 2022, 12, 1–19. [Google Scholar] [CrossRef]
  48. Zanoni, M.; Sarti, A.C.; Zamagni, A.; Cortesi, M.; Pignatta, S.; Arienti, C.; Tebaldi, M.; Sarnelli, A.; Romeo, A.; Bartolini, D.; et al. Irradiation Causes Senescence, ATP Release, and P2X7 Receptor Isoform Switch in Glioblastoma. Cell Death Dis. 2022, 13, 80. [Google Scholar] [CrossRef]
  49. Barden, J.A. Non-Functional P2X7: A Novel and Ubiquitous Target in Human Cancer. J. Clin. Cell Immunol. 2014, 5, 2155–9899. [Google Scholar] [CrossRef]
  50. Gilbert, S.; Oliphant, C.; Hassan, S.; Peille, A.; Bronsert, P.; Falzoni, S.; Di Virgilio, F.; McNulty, S.; Lara, R. ATP in the Tumour Microenvironment Drives Expression of NfP2X7, a Key Mediator of Cancer Cell Survival. Oncogene 2019, 38, 194–208. [Google Scholar] [CrossRef]
  51. Rissiek, B.; Haag, F.; Boyer, O.; Koch-Nolte, F.; Adriouch, S. P2X7 on Mouse T Cells: One Channel, Many Functions. Front. Immunol. 2015, 6, 204. [Google Scholar] [CrossRef]
  52. Grassi, F. The P2X7 Receptor as Regulator of T Cell Development and Function. Front. Immunol. 2020, 11, 1179. [Google Scholar] [CrossRef]
  53. Adriouch, S.; Hubert, S.; Pechberty, S.; Koch-Nolte, F.; Haag, F.; Seman, M. NAD+ Released during Inflammation Participates in T Cell Homeostasis by Inducing ART2-Mediated Death of Naive T Cells In Vivo. J. Immunol. 2007, 179, 186–194. [Google Scholar] [CrossRef]
  54. Proietti, M.; Cornacchione, V.; Rezzonico Jost, T.; Romagnani, A.; Faliti, C.E.; Perruzza, L.; Rigoni, R.; Radaelli, E.; Caprioli, F.; Preziuso, S.; et al. ATP-Gated Ionotropic P2X7 Receptor Controls Follicular T Helper Cell Numbers in Peyer’s Patches to Promote Host-Microbiota Mutualism. Immunity 2014, 41, 789–801. [Google Scholar] [CrossRef]
  55. Stark, R.; Wesselink, T.H.; Behr, F.M.; Kragten, N.A.M.; Arens, R.; Koch-Nolte, F.; van Gisbergen, K.P.J.M.; van Lier, R.A.W. TRM Maintenance Is Regulated by Tissue Damage via P2RX7. Sci. Immunol. 2018, 3, eaau1022. [Google Scholar] [CrossRef]
  56. Vardam-Kaur, T.; van Dijk, S.; Peng, C.; Wanhainen, K.M.; Jameson, S.C.; Borges da Silva, H. The Extracellular ATP Receptor P2RX7 Imprints a Promemory Transcriptional Signature in Effector CD8+ T Cells. J. Immunol. 2022, 208, 1686–1699. [Google Scholar] [CrossRef]
  57. Borges da Silva, H.; Wang, H.; Qian, L.J.; Hogquist, K.A.; Jameson, S.C. ARTC2.2/P2RX7 Signaling during Cell Isolation Distorts Function and Quantification of Tissue-Resident CD8+ T Cell and Invariant NKT Subsets. J. Immunol. 2019, 202, 2153–2163. [Google Scholar] [CrossRef]
  58. Hofman, P.; Cherfils-Vicini, J.; Bazin, M.; Ilie, M.; Juhel, T.; Hébuterne, X.; Gilson, E.; Schmid-Alliana, A.; Boyer, O.; Adriouch, S.; et al. Genetic and Pharmacological Inactivation of the Purinergic P2RX7 Receptor Dampens Inflammation but Increases Tumor Incidence in a Mouse Model of Colitis-Associated Cancer. Cancer Res. 2015, 75, 835–845. [Google Scholar] [CrossRef]
  59. Hubert, S.; Rissiek, B.; Klages, K.; Huehn, J.; Sparwasser, T.; Haag, F.; Koch-Nolte, F.; Boyer, O.; Seman, M.; Adriouch, S. Extracellular NAD+ Shapes the Foxp3+ Regulatory T Cell Compartment through the ART2-P2X7 Pathway. J. Exp. Med. 2010, 207, 2561–2568. [Google Scholar] [CrossRef]
  60. Ting, J.P.-Y.; Lovering, R.C.; Alnemri, E.S.; Bertin, J.; Boss, J.M.; Davis, B.K.; Flavell, R.A.; Girardin, S.E.; Godzik, A.; Harton, J.A.; et al. The NLR Gene Family: A Standard Nomenclature. Immunity 2008, 28, 285–287. [Google Scholar] [CrossRef]
  61. Di Virgilio, F.; Sarti, A.C.; Grassi, F. Modulation of Innate and Adaptive Immunity by P2X Ion Channels. Curr. Opin. Immunol. 2018, 52, 51–59. [Google Scholar] [CrossRef] [PubMed]
  62. He, Y.; Zeng, M.Y.; Yang, D.; Motro, B.; Núñez, G. NEK7 Is an Essential Mediator of NLRP3 Activation Downstream of Potassium Efflux. Nature 2016, 530, 354–357. [Google Scholar] [CrossRef] [PubMed]
  63. Pelegrin, P. P2X7 Receptor and the NLRP3 Inflammasome: Partners in Crime. Biochem. Pharmacol. 2021, 187, 114385. [Google Scholar] [CrossRef] [PubMed]
  64. Muñoz-Planillo, R.; Kuffa, P.; Martínez-Colón, G.; Smith, B.L.; Rajendiran, T.M.; Núñez, G. K+ Efflux Is the Common Trigger of NLRP3 Inflammasome Activation by Bacterial Toxins and Particulate Matter. Immunity 2013, 38, 1142–1153. [Google Scholar] [CrossRef] [PubMed]
  65. Perregaux, D.G.; Gabel, C.A. Human Monocyte Stimulus-Coupled IL-1β Posttranslational Processing: Modulation via Monovalent Cations. Am. J. Physiol. Cell Physiol. 1998, 275, C1538–C1547. [Google Scholar] [CrossRef]
  66. Franceschini, A.; Capece, M.; Chiozzi, P.; Falzoni, S.; Sanz, J.M.; Sarti, A.C.; Bonora, M.; Pinton, P.; Di Virgilio, F. The P2X7 Receptor Directly Interacts with the NLRP3 Inflammasome Scaffold Protein. FASEB J. 2015, 29, 2450–2461. [Google Scholar] [CrossRef]
  67. Wang, W.; Hu, D.; Feng, Y.; Wu, C.; Song, Y.; Liu, W.; Li, A.; Wang, Y.; Chen, K.; Tian, M.; et al. Paxillin Mediates ATP-Induced Activation of P2X7 Receptor and NLRP3 Inflammasome. BMC Biol. 2020, 18, 182. [Google Scholar] [CrossRef]
  68. Di, A.; Xiong, S.; Ye, Z.; Malireddi, R.K.S.; Kometani, S.; Zhong, M.; Mittal, M.; Hong, Z.; Kanneganti, T.-D.; Rehman, J.; et al. The TWIK2 Potassium Efflux Channel in Macrophages Mediates NLRP3 Inflammasome-Induced Inflammation. Immunity 2018, 49, 56–65.e4. [Google Scholar] [CrossRef]
  69. Lee, G.-S.; Subramanian, N.; Kim, A.I.; Aksentijevich, I.; Goldbach-Mansky, R.; Sacks, D.B.; Germain, R.N.; Kastner, D.L.; Chae, J.J. The Calcium-Sensing Receptor Regulates the NLRP3 Inflammasome through Ca2+ and CAMP. Nature 2012, 492, 123–127. [Google Scholar] [CrossRef]
  70. Hamarsheh, S.; Zeiser, R. NLRP3 Inflammasome Activation in Cancer: A Double-Edged Sword. Front. Immunol. 2020, 11, 1444. [Google Scholar] [CrossRef]
  71. Sharma, B.R.; Kanneganti, T.-D. NLRP3 Inflammasome in Cancer and Metabolic Diseases. Nat. Immunol. 2021, 22, 550–559. [Google Scholar] [CrossRef]
  72. Campagno, K.E.; Mitchell, C.H. The P2X7 Receptor in Microglial Cells Modulates the Endolysosomal Axis, Autophagy, and Phagocytosis. Front. Cell Neurosci. 2021, 15, 645244. [Google Scholar] [CrossRef]
  73. Di Virgilio, F.; Dal Ben, D.; Sarti, A.C.; Giuliani, A.L.; Falzoni, S. The P2X7 Receptor in Infection and Inflammation. Immunity 2017, 47, 15–31. [Google Scholar] [CrossRef]
  74. Adinolfi, E.; Giuliani, A.L.; De Marchi, E.; Pegoraro, A.; Orioli, E.; Di Virgilio, F. The P2X7 Receptor: A Main Player in Inflammation. Biochem. Pharmacol. 2018, 151, 234–244. [Google Scholar] [CrossRef]
  75. Lopez-Castejon, G. Control of the Inflammasome by the Ubiquitin System. FEBS J. 2020, 287, 11–26. [Google Scholar] [CrossRef]
  76. Xu, J.; Núñez, G. The NLRP3 Inflammasome: Activation and Regulation. Trends Biochem. Sci. 2023, 48, 331–344. [Google Scholar] [CrossRef]
  77. Puren, A.J.; Fantuzzi, G.; Dinarello, C.A. Gene Expression, Synthesis, and Secretion of Interleukin 18 and Interleukin 1beta Are Differentially Regulated in Human Blood Mononuclear Cells and Mouse Spleen Cells. Proc. Natl. Acad. Sci. USA 1999, 96, 2256–2261. [Google Scholar] [CrossRef]
  78. Cameron, L.A.; Taha, R.A.; Tsicopoulos, A.; Kurimoto, M.; Olivenstein, R.; Wallaert, B.; Minshall, E.M.; Hamid, Q.A. Airway Epithelium Expresses Interleukin-18. Eur. Respir. J. 1999, 14, 553–559. [Google Scholar] [CrossRef]
  79. Artlett, C.M.; Sassi-Gaha, S.; Rieger, J.L.; Boesteanu, A.C.; Feghali-Bostwick, C.A.; Katsikis, P.D. The Inflammasome Activating Caspase 1 Mediates Fibrosis and Myofibroblast Differentiation in Systemic Sclerosis. Arthritis Rheum. 2011, 63, 3563–3574. [Google Scholar] [CrossRef]
  80. Gritsenko, A.; Yu, S.; Martin-Sanchez, F.; Diaz-del-Olmo, I.; Nichols, E.-M.; Davis, D.M.; Brough, D.; Lopez-Castejon, G. Priming Is Dispensable for NLRP3 Inflammasome Activation in Human Monocytes In Vitro. Front. Immunol. 2020, 11, 565924. [Google Scholar] [CrossRef]
  81. Wang, D.; Zhang, S.; Li, L.; Liu, X.; Mei, K.; Wang, X. Structural Insights into the Assembly and Activation of IL-1β with Its Receptors. Nat. Immunol. 2010, 11, 905–911. [Google Scholar] [CrossRef] [PubMed]
  82. Dripps, D.J.; Brandhuber, B.J.; Thompson, R.C.; Eisenberg, S.P. Interleukin-1 (IL-1) Receptor Antagonist Binds to the 80-KDa IL-1 Receptor but Does Not Initiate IL-1 Signal Transduction. J. Biol. Chem. 1991, 266, 10331–10336. [Google Scholar] [CrossRef] [PubMed]
  83. Fields, J.K.; Günther, S.; Sundberg, E.J. Structural Basis of IL-1 Family Cytokine Signaling. Front. Immunol. 2019, 10, 1412. [Google Scholar] [CrossRef] [PubMed]
  84. Dinarello, C.A.; Novick, D.; Kim, S.; Kaplanski, G. Interleukin-18 and IL-18 Binding Protein. Front. Immunol. 2013, 4, 289. [Google Scholar] [CrossRef] [PubMed]
  85. Okamura, H.; Tsutsi, H.; Komatsu, T.; Yutsudo, M.; Hakura, A.; Tanimoto, T.; Torigoe, K.; Okura, T.; Nukada, Y.; Hattori, K. Cloning of a New Cytokine That Induces IFN-Gamma Production by T Cells. Nature 1995, 378, 88–91. [Google Scholar] [CrossRef]
  86. Rébé, C.; Ghiringhelli, F. Interleukin-1β and Cancer. Cancers 2020, 12, 1791. [Google Scholar] [CrossRef]
  87. Pretre, V.; Papadopoulos, D.; Regard, J.; Pelletier, M.; Woo, J. Interleukin-1 (IL-1) and the Inflammasome in Cancer. Cytokine 2022, 153, 155850. [Google Scholar] [CrossRef]
  88. Missiroli, S.; Perrone, M.; Boncompagni, C.; Borghi, C.; Campagnaro, A.; Marchetti, F.; Anania, G.; Greco, P.; Fiorica, F.; Pinton, P.; et al. Targeting the NLRP3 Inflammasome as a New Therapeutic Option for Overcoming Cancer. Cancers 2021, 13, 2297. [Google Scholar] [CrossRef]
  89. Tengesdal, I.W.; Dinarello, A.; Powers, N.E.; Burchill, M.A.; Joosten, L.A.B.; Marchetti, C.; Dinarello, C.A. Tumor NLRP3-Derived IL-1β Drives the IL-6/STAT3 Axis Resulting in Sustained MDSC-Mediated Immunosuppression. Front. Immunol. 2021, 12, 3439. [Google Scholar] [CrossRef]
  90. Tengesdal, I.W.; Menon, D.R.; Osborne, D.G.; Neff, C.P.; Powers, N.E.; Gamboni, F.; Mauro, A.G.; D’Alessandro, A.; Stefanoni, D.; Henen, M.A.; et al. Targeting Tumor-Derived NLRP3 Reduces Melanoma Progression by Limiting MDSCs Expansion. Proc. Natl. Acad. Sci. USA 2021, 118, e2000915118. [Google Scholar] [CrossRef]
  91. Voronov, E.; Shouval, D.S.; Krelin, Y.; Cagnano, E.; Benharroch, D.; Iwakura, Y.; Dinarello, C.A.; Apte, R.N. IL-1 Is Required for Tumor Invasiveness and Angiogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 2645–2650. [Google Scholar] [CrossRef]
  92. Mantsounga, C.S.; Lee, C.; Neverson, J.; Sharma, S.; Healy, A.; Berus, J.M.; Parry, C.; Ceneri, N.M.; López-Giráldez, F.; Chun, H.J.; et al. Macrophage IL-1β Promotes Arteriogenesis by Autocrine STAT3- and NF-ΚB-Mediated Transcription of pro-Angiogenic VEGF-A. Cell Rep. 2022, 38, 110309. [Google Scholar] [CrossRef]
  93. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 Inflammasome in Dendritic Cells Induces IL-1β–Dependent Adaptive Immunity against Tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef]
  94. Zhivaki, D.; Borriello, F.; Chow, O.A.; Doran, B.; Fleming, I.; Theisen, D.J.; Pallis, P.; Shalek, A.K.; Sokol, C.L.; Zanoni, I.; et al. Inflammasomes within Hyperactive Murine Dendritic Cells Stimulate Long-Lived T Cell-Mediated Anti-Tumor Immunity. Cell Rep. 2020, 33, 108381. [Google Scholar] [CrossRef]
  95. Allen, I.C.; TeKippe, E.M.; Woodford, R.-M.T.; Uronis, J.M.; Holl, E.K.; Rogers, A.B.; Herfarth, H.H.; Jobin, C.; Ting, J.P.-Y. The NLRP3 Inflammasome Functions as a Negative Regulator of Tumorigenesis during Colitis-Associated Cancer. J. Exp. Med. 2010, 207, 1045–1056. [Google Scholar] [CrossRef]
  96. Zaki, M.H.; Boyd, K.L.; Vogel, P.; Kastan, M.B.; Lamkanfi, M.; Kanneganti, T.-D. The NLRP3 Inflammasome Protects against Loss of Epithelial Integrity and Mortality during Experimental Colitis. Immunity 2010, 32, 379–391. [Google Scholar] [CrossRef]
  97. Zaki, M.H.; Vogel, P.; Body-Malapel, M.; Lamkanfi, M.; Kanneganti, T.-D. IL-18 Production Downstream of the Nlrp3 Inflammasome Confers Protection against Colorectal Tumor Formation. J. Immunol. 2010, 185, 4912–4920. [Google Scholar] [CrossRef]
  98. Salcedo, R.; Worschech, A.; Cardone, M.; Jones, Y.; Gyulai, Z.; Dai, R.-M.; Wang, E.; Ma, W.; Haines, D.; O’hUigin, C.; et al. MyD88-Mediated Signaling Prevents Development of Adenocarcinomas of the Colon: Role of Interleukin 18. J. Exp. Med. 2010, 207, 1625–1636. [Google Scholar] [CrossRef]
  99. Malik, A.; Sharma, D.; Malireddi, R.K.S.; Guy, C.S.; Chang, T.-C.; Olsen, S.R.; Neale, G.; Vogel, P.; Kanneganti, T.-D. SYK-CARD9 Signaling Axis Promotes Gut Fungi-Mediated Inflammasome Activation to Restrict Colitis and Colon Cancer. Immunity 2018, 49, 515–530.e5. [Google Scholar] [CrossRef]
  100. Sharma, D.; Malik, A.; Guy, C.S.; Karki, R.; Vogel, P.; Kanneganti, T.-D. Pyrin Inflammasome Regulates Tight Junction Integrity to Restrict Colitis and Tumorigenesis. Gastroenterology 2018, 154, 948–964.e8. [Google Scholar] [CrossRef]
  101. Zhou, T.; Damsky, W.; Weizman, O.-E.; McGeary, M.K.; Hartmann, K.P.; Rosen, C.E.; Fischer, S.; Jackson, R.; Flavell, R.A.; Wang, J.; et al. IL-18BP Is a Secreted Immune Checkpoint and Barrier to IL-18 Immunotherapy. Nature 2020, 583, 609–614. [Google Scholar] [CrossRef] [PubMed]
  102. Dupaul-Chicoine, J.; Arabzadeh, A.; Dagenais, M.; Douglas, T.; Champagne, C.; Morizot, A.; Rodrigue-Gervais, I.G.; Breton, V.; Colpitts, S.L.; Beauchemin, N.; et al. The Nlrp3 Inflammasome Suppresses Colorectal Cancer Metastatic Growth in the Liver by Promoting Natural Killer Cell Tumoricidal Activity. Immunity 2015, 43, 751–763. [Google Scholar] [CrossRef]
  103. Osaki, T.; Péron, J.M.; Cai, Q.; Okamura, H.; Robbins, P.D.; Kurimoto, M.; Lotze, M.T.; Tahara, H. IFN-Gamma-Inducing Factor/IL-18 Administration Mediates IFN-Gamma-and IL-12-Independent Antitumor Effects. J. Immunol. 1998, 160, 1742–1749. [Google Scholar] [CrossRef] [PubMed]
  104. Nishio, S.; Yamada, N.; Ohyama, H.; Yamanegi, K.; Nakasho, K.; Hata, M.; Nakamura, Y.; Fukunaga, S.; Futani, H.; Yoshiya, S.; et al. Enhanced Suppression of Pulmonary Metastasis of Malignant Melanoma Cells by Combined Administration of α-Galactosylceramide and Interleukin-18. Cancer Sci. 2008, 99, 113–120. [Google Scholar] [CrossRef] [PubMed]
  105. Choi, I.-K.; Lee, J.-S.; Zhang, S.-N.; Park, J.; Sonn, C.H.; Lee, K.-M.; Yun, C.-O. Oncolytic Adenovirus Co-Expressing IL-12 and IL-18 Improves Tumor-Specific Immunity via Differentiation of T Cells Expressing IL-12Rβ2 or IL-18Rα. Gene Ther. 2011, 18, 898–909. [Google Scholar] [CrossRef]
  106. Savid-Frontera, C.; Viano, M.E.; Baez, N.S.; Lidon, N.L.; Fontaine, Q.; Young, H.A.; Vimeux, L.; Donnadieu, E.; Rodriguez-Galan, M.C. Exploring the Immunomodulatory Role of Virtual Memory CD8+ T Cells: Role of IFN Gamma in Tumor Growth Control. Front. Immunol. 2022, 13, 971001. [Google Scholar] [CrossRef]
  107. Minnie, S.A.; Waltner, O.G.; Ensbey, K.S.; Nemychenkov, N.S.; Schmidt, C.R.; Bhise, S.S.; Legg, S.R.W.; Campoy, G.; Samson, L.D.; Kuns, R.D.; et al. Depletion of Exhausted Alloreactive T Cells Enables Targeting of Stem-like Memory T Cells to Generate Tumor-Specific Immunity. Sci. Immunol. 2022, 7, eabo3420. [Google Scholar] [CrossRef]
  108. Li, X.-Y.; Moesta, A.K.; Xiao, C.; Nakamura, K.; Casey, M.; Zhang, H.; Madore, J.; Lepletier, A.; Aguilera, A.R.; Sundarrajan, A.; et al. Targeting CD39 in Cancer Reveals an Extracellular ATP- and Inflammasome-Driven Tumor Immunity. Cancer Discov. 2019, 9, 1754–1773. [Google Scholar] [CrossRef]
  109. Perrot, I.; Michaud, H.-A.; Giraudon-Paoli, M.; Augier, S.; Docquier, A.; Gros, L.; Courtois, R.; Déjou, C.; Jecko, D.; Becquart, O.; et al. Blocking Antibodies Targeting the CD39/CD73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies. Cell Rep. 2019, 27, 2411–2425.e9. [Google Scholar] [CrossRef]
  110. Douguet, L.; Janho Dit Hreich, S.; Benzaquen, J.; Seguin, L.; Juhel, T.; Dezitter, X.; Duranton, C.; Ryffel, B.; Kanellopoulos, J.; Delarasse, C.; et al. A Small-Molecule P2RX7 Activator Promotes Anti-Tumor Immune Responses and Sensitizes Lung Tumor to Immunotherapy. Nat. Commun. 2021, 12, 653. [Google Scholar] [CrossRef]
  111. Berg, R.E.; Crossley, E.; Murray, S.; Forman, J. Memory CD8+ T Cells Provide Innate Immune Protection against Listeria Monocytogenes in the Absence of Cognate Antigen. J. Exp. Med. 2003, 198, 1583–1593. [Google Scholar] [CrossRef]
  112. Ge, C.; Monk, I.R.; Pizzolla, A.; Wang, N.; Bedford, J.G.; Stinear, T.P.; Westall, G.P.; Wakim, L.M. Bystander Activation of Pulmonary Trm Cells Attenuates the Severity of Bacterial Pneumonia by Enhancing Neutrophil Recruitment. Cell Rep. 2019, 29, 4236–4244.e3. [Google Scholar] [CrossRef]
  113. Iwai, Y.; Hemmi, H.; Mizenina, O.; Kuroda, S.; Suda, K.; Steinman, R.M. An IFN-Gamma-IL-18 Signaling Loop Accelerates Memory CD8+ T Cell Proliferation. PLoS ONE 2008, 3, e2404. [Google Scholar] [CrossRef]
  114. Oliveira, A.-C.; Gomes-Neto, J.F.; Barbosa, C.-H.D.; Granato, A.; Reis, B.S.; Santos, B.M.; Fucs, R.; Canto, F.B.; Nakaya, H.I.; Nóbrega, A.; et al. Crucial Role for T Cell-Intrinsic IL-18R-MyD88 Signaling in Cognate Immune Response to Intracellular Parasite Infection. eLife 2017, 6, 30883. [Google Scholar] [CrossRef]
  115. Jain, A.; Song, R.; Wakeland, E.K.; Pasare, C. T Cell-Intrinsic IL-1R Signaling Licenses Effector Cytokine Production by Memory CD4 T Cells. Nat. Commun. 2018, 9, 3185. [Google Scholar] [CrossRef]
  116. Ni, J.; Miller, M.; Stojanovic, A.; Garbi, N.; Cerwenka, A. Sustained Effector Function of IL-12/15/18-Preactivated NK Cells against Established Tumors. J. Exp. Med. 2012, 209, 2351–2365. [Google Scholar] [CrossRef]
  117. Mirjačić Martinović, K.; Babović, N.; Džodić, R.; Jurišić, V.; Matković, S.; Konjević, G. Favorable in Vitro Effects of Combined IL-12 and IL-18 Treatment on NK Cell Cytotoxicity and CD25 Receptor Expression in Metastatic Melanoma Patients. J. Transl. Med. 2015, 13, 120. [Google Scholar] [CrossRef]
  118. Aarsund, M.; Segers, F.M.; Wu, Y.; Inngjerdingen, M. Comparison of Characteristics and Tumor Targeting Properties of Extracellular Vesicles Derived from Primary NK Cells or NK-Cell Lines Stimulated with IL-15 or IL-12/15/18. Cancer Immunol. Immunother. 2022, 71, 2227–2238. [Google Scholar] [CrossRef]
  119. Blokon-Kogan, D.; Levi-Mann, M.; Malka-Levy, L.; Itzhaki, O.; Besser, M.J.; Shiftan, Y.; Szöőr, Á.; Vereb, G.; Gross, G.; Abken, H.; et al. Membrane Anchored IL-18 Linked to Constitutively Active TLR4 and CD40 Improves Human T Cell Antitumor Capacities for Adoptive Cell Therapy. J. Immunother. Cancer 2022, 10, e001544. [Google Scholar] [CrossRef]
  120. Drakes, D.J.; Rafiq, S.; Purdon, T.J.; Lopez, A.V.; Chandran, S.S.; Klebanoff, C.A.; Brentjens, R.J. Optimization of T-Cell Receptor-Modified T Cells for Cancer Therapy. Cancer Immunol. Res. 2020, 8, 743–755. [Google Scholar] [CrossRef]
  121. Kunert, A.; Chmielewski, M.; Wijers, R.; Berrevoets, C.; Abken, H.; Debets, R. Intra-Tumoral Production of IL18, but Not IL12, by TCR-Engineered T Cells Is Non-Toxic and Counteracts Immune Evasion of Solid Tumors. Oncoimmunology 2017, 7, e1378842. [Google Scholar] [CrossRef] [PubMed]
  122. Hu, B.; Ren, J.; Luo, Y.; Keith, B.; Young, R.M.; Scholler, J.; Zhao, Y.; June, C.H. Augmentation of Antitumor Immunity by Human and Mouse CAR T Cells Secreting IL-18. Cell Rep. 2017, 20, 3025–3033. [Google Scholar] [CrossRef] [PubMed]
  123. Cirella, A.; Bolaños, E.; Di Trani, C.A.; de Andrea, C.E.; Sánchez-Gregorio, S.; Etxeberria, I.; Gonzalez-Gomariz, J.; Olivera, I.; Brocco, D.; Glez-Vaz, J.; et al. Intratumoral Gene Transfer of MRNAs Encoding IL12 in Combination with Decoy-Resistant IL18 Improves Local and Systemic Antitumor Immunity. Cancer Immunol. Res. 2023, 11, 184–198. [Google Scholar] [CrossRef] [PubMed]
  124. Olivera, I.; Bolaños, E.; Gonzalez-Gomariz, J.; Hervas-Stubbs, S.; Mariño, K.V.; Luri-Rey, C.; Etxeberria, I.; Cirella, A.; Egea, J.; Glez-Vaz, J.; et al. MRNAs Encoding IL-12 and a Decoy-Resistant Variant of IL-18 Synergize to Engineer T Cells for Efficacious Intratumoral Adoptive Immunotherapy. Cell Rep. Med. 2023, 4, 100978. [Google Scholar] [CrossRef] [PubMed]
  125. Jaspers, J.E.; Khan, J.F.; Godfrey, W.D.; Lopez, A.V.; Ciampricotti, M.; Rudin, C.M.; Brentjens, R.J. IL-18-Secreting CAR T Cells Targeting DLL3 Are Highly Effective in Small Cell Lung Cancer Models. J. Clin. Investig. 2023, 133, 1–11. [Google Scholar] [CrossRef]
  126. Timperi, E.; Focaccetti, C.; Gallerano, D.; Panetta, M.; Spada, S.; Gallo, E.; Visca, P.; Venuta, F.; Diso, D.; Prelaj, A.; et al. IL-18 Receptor Marks Functional CD8+ T Cells in Non-Small Cell Lung Cancer. Oncoimmunology 2017, 6, e1328337. [Google Scholar] [CrossRef]
  127. Pagès, F.; Berger, A.; Henglein, B.; Piqueras, B.; Danel, C.; Zinzindohoue, F.; Thiounn, N.; Cugnenc, P.H.; Fridman, W.H. Modulation of Interleukin-18 Expression in Human Colon Carcinoma: Consequences for Tumor Immune Surveillance. Int. J. Cancer 1999, 84, 326–330. [Google Scholar] [CrossRef]
  128. Feng, X.; Zhang, Z.; Sun, P.; Song, G.; Wang, L.; Sun, Z.; Yuan, N.; Wang, Q.; Lun, L. Interleukin-18 Is a Prognostic Marker and Plays a Tumor Suppressive Role in Colon Cancer. Dis. Markers 2020, 2020, 6439614. [Google Scholar] [CrossRef]
  129. Gil, M.; Kim, K.E. Interleukin-18 Is a Prognostic Biomarker Correlated with CD8+ T Cell and Natural Killer Cell Infiltration in Skin Cutaneous Melanoma. J. Clin. Med. 2019, 8, 1993. [Google Scholar] [CrossRef]
  130. Jiang, K.; Lin, B.; Zhang, Y.; Lu, K.; Wu, F.; Luo, D. A Novel Pyroptosis-Related Gene Signature for Prediction of Disease-Free Survival in Papillary Thyroid Carcinoma. J. Pers. Med. 2022, 13, 85. [Google Scholar] [CrossRef]
  131. Xiao, W.; Tang, P.; Sui, Z.; Han, Y.; Zhao, G.; Wu, X.; Yang, Y.; Zhu, N.; Gong, L.; Yu, Z.; et al. Establishment of a Risk Model by Integrating Hypoxia Genes in Predicting Prognosis of Esophageal Squamous Cell Carcinoma. Cancer Med. 2023, 12, 2117–2133. [Google Scholar] [CrossRef]
  132. Song, J.; Li, A.; Qian, Y.; Liu, B.; Lv, L.; Ye, D.; Sun, X.; Mao, Y. Genetically Predicted Circulating Levels of Cytokines and the Risk of Cancer. Front. Immunol. 2022, 13, 886144. [Google Scholar] [CrossRef]
  133. Mutala, L.B.; Deleine, C.; Karakachoff, M.; Dansette, D.; Ducoin, K.; Oger, R.; Rousseau, O.; Podevin, J.; Duchalais, E.; Fourquier, P.; et al. The Caspase-1/IL-18 Axis of the Inflammasome in Tumor Cells: A Modulator of the Th1/Tc1 Response of Tumor-Infiltrating T Lymphocytes in Colorectal Cancer. Cancers 2021, 13, 189. [Google Scholar] [CrossRef]
  134. Guo, Q.; Wu, C.-Y.; Jiang, N.; Tong, S.; Wan, J.-H.; Xiao, X.-Y.; Mei, P.-Y.; Liu, H.-S.; Wang, S.-H. Downregulation of T-Cell Cytotoxic Marker IL18R1 Promotes Cancer Proliferation and Migration and Is Associated with Dismal Prognosis and Immunity in Lung Squamous Cell Carcinoma. Front. Immunol. 2022, 13, 986447. [Google Scholar] [CrossRef]
  135. Carbotti, G.; Barisione, G.; Orengo, A.M.; Brizzolara, A.; Airoldi, I.; Bagnoli, M.; Pinciroli, P.; Mezzanzanica, D.; Centurioni, M.G.; Fabbi, M.; et al. The IL-18 Antagonist IL-18-Binding Protein Is Produced in the Human Ovarian Cancer Microenvironment. Clin. Cancer Res. 2013, 19, 4611–4620. [Google Scholar] [CrossRef]
  136. Fujita, K.; Ewing, C.M.; Isaacs, W.B.; Pavlovich, C.P. Immunomodulatory IL-18 Binding Protein Is Produced by Prostate Cancer Cells and Its Levels in Urine and Serum Correlate with Tumor Status. Int. J. Cancer 2011, 129, 424–432. [Google Scholar] [CrossRef]
  137. Orengo, A.M.; Fabbi, M.; Miglietta, L.; Andreani, C.; Bruzzone, M.; Puppo, A.; Cristoforoni, P.; Centurioni, M.G.; Gualco, M.; Salvi, S.; et al. Interleukin (IL)-18, a Biomarker of Human Ovarian Carcinoma, Is Predominantly Released as Biologically Inactive Precursor. Int. J. Cancer 2011, 129, 1116–1125. [Google Scholar] [CrossRef]
  138. Herbst, R.S.; Soria, J.-C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive Correlates of Response to the Anti-PD-L1 Antibody MPDL3280A in Cancer Patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef]
  139. Netterberg, I.; Li, C.-C.; Molinero, L.; Budha, N.; Sukumaran, S.; Stroh, M.; Jonsson, E.N.; Friberg, L.E. A PK/PD Analysis of Circulating Biomarkers and Their Relationship to Tumor Response in Atezolizumab-Treated Non-Small Cell Lung Cancer Patients. Clin. Pharmacol. Ther. 2019, 105, 486–495. [Google Scholar] [CrossRef]
  140. Chen, B.; Yang, M.; Li, K.; Li, J.; Xu, L.; Xu, F.; Xu, Y.; Ren, D.; Zhang, J.; Liu, L. Immune-Related Genes and Gene Sets for Predicting the Response to Anti-Programmed Death 1 Therapy in Patients with Primary or Metastatic Non-Small Cell Lung Cancer. Oncol. Lett. 2021, 22, 540. [Google Scholar] [CrossRef]
  141. Wang, Y.; Chen, H.; Zhang, T.; Yang, X.; Zhong, J.; Wang, Y.; Chi, Y.; Wu, M.; An, T.; Li, J.; et al. Plasma Cytokines Interleukin-18 and C-X-C Motif Chemokine Ligand 10 Are Indicative of the Anti-Programmed Cell Death Protein-1 Treatment Response in Lung Cancer Patients. Ann. Transl. Med. 2021, 9, 33. [Google Scholar] [CrossRef] [PubMed]
  142. Zhao, X.; Zhang, C.; Hua, M.; Wang, R.; Zhong, C.; Yu, J.; Han, F.; He, N.; Zhao, Y.; Liu, G.; et al. NLRP3 Inflammasome Activation Plays a Carcinogenic Role through Effector Cytokine IL-18 in Lymphoma. Oncotarget 2017, 8, 108571–108583. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, B.; Wu, K.-F.; Cao, Z.-Y.; Rao, Q.; Ma, X.-T.; Zheng, G.-G.; Li, G. IL-18 Increases Invasiveness of HL-60 Myeloid Leukemia Cells: Up-Regulation of Matrix Metalloproteinases-9 (MMP-9) Expression. Leuk. Res. 2004, 28, 91–95. [Google Scholar] [CrossRef] [PubMed]
  144. Vidal-Vanaclocha, F.; Fantuzzi, G.; Mendoza, L.; Fuentes, A.M.; Anasagasti, M.J.; Martín, J.; Carrascal, T.; Walsh, P.; Reznikov, L.L.; Kim, S.H.; et al. IL-18 Regulates IL-1beta-Dependent Hepatic Melanoma Metastasis via Vascular Cell Adhesion Molecule-1. Proc. Natl. Acad. Sci. USA 2000, 97, 734–739. [Google Scholar] [CrossRef] [PubMed]
  145. Ahmad, I.; Muneer, K.M.; Tamimi, I.A.; Chang, M.E.; Ata, M.O.; Yusuf, N. Thymoquinone Suppresses Metastasis of Melanoma Cells by Inhibition of NLRP3 Inflammasome. Toxicol. Appl. Pharmacol. 2013, 270, 70–76. [Google Scholar] [CrossRef] [PubMed]
  146. Kim, K.-E.; Song, H.; Kim, T.S.; Yoon, D.; Kim, C.; Bang, S.I.; Hur, D.Y.; Park, H.; Cho, D.-H. Interleukin-18 Is a Critical Factor for Vascular Endothelial Growth Factor-Enhanced Migration in Human Gastric Cancer Cell Lines. Oncogene 2007, 26, 1468–1476. [Google Scholar] [CrossRef]
  147. Jung, M.K.; Song, H.K.; Kim, K.-E.; Hur, D.Y.; Kim, T.; Bang, S.; Park, H.; Cho, D.H. IL-18 Enhances the Migration Ability of Murine Melanoma Cells through the Generation of ROI and the MAPK Pathway. Immunol. Lett. 2006, 107, 125–130. [Google Scholar] [CrossRef]
  148. Carrascal, M.T.; Mendoza, L.; Valcárcel, M.; Salado, C.; Egilegor, E.; Tellería, N.; Vidal-Vanaclocha, F.; Dinarello, C.A. Interleukin-18 Binding Protein Reduces B16 Melanoma Hepatic Metastasis by Neutralizing Adhesiveness and Growth Factors of Sinusoidal Endothelium. Cancer Res. 2003, 63, 491–497. [Google Scholar]
  149. Nakamura, K.; Kassem, S.; Cleynen, A.; Chrétien, M.-L.; Guillerey, C.; Putz, E.M.; Bald, T.; Förster, I.; Vuckovic, S.; Hill, G.R.; et al. Dysregulated IL-18 Is a Key Driver of Immunosuppression and a Possible Therapeutic Target in the Multiple Myeloma Microenvironment. Cancer Cell 2018, 33, 634–648.e5. [Google Scholar] [CrossRef]
  150. Hofbauer, D.; Mougiakakos, D.; Broggini, L.; Zaiss, M.; Büttner-Herold, M.; Bach, C.; Spriewald, B.; Neumann, F.; Bisht, S.; Nolting, J.; et al. Β2-Microglobulin Triggers NLRP3 Inflammasome Activation in Tumor-Associated Macrophages to Promote Multiple Myeloma Progression. Immunity 2021, 54, 1772–1787.e9. [Google Scholar] [CrossRef]
  151. Daley, D.; Mani, V.R.; Mohan, N.; Akkad, N.; Pandian, G.S.D.B.; Savadkar, S.; Lee, K.B.; Torres-Hernandez, A.; Aykut, B.; Diskin, B.; et al. NLRP3 Signaling Drives Macrophage-Induced Adaptive Immune Suppression in Pancreatic Carcinoma. J. Exp. Med. 2017, 214, 1711–1724. [Google Scholar] [CrossRef]
  152. Guo, X.; Zheng, L.; Jiang, J.; Zhao, Y.; Wang, X.; Shen, M.; Zhu, F.; Tian, R.; Shi, C.; Xu, M.; et al. Blocking NF-ΚB Is Essential for the Immunotherapeutic Effect of Recombinant IL18 in Pancreatic Cancer. Clin. Cancer Res. 2016, 22, 5939–5950. [Google Scholar] [CrossRef]
  153. Lutz, V.; Hellmund, V.M.; Picard, F.S.R.; Raifer, H.; Ruckenbrod, T.; Klein, M.; Bopp, T.; Savai, R.; Duewell, P.; Keber, C.U.; et al. IL18 Receptor Signaling Regulates Tumor-Reactive CD8+ T-Cell Exhaustion via Activation of the IL2/STAT5/MTOR Pathway in a Pancreatic Cancer Model. Cancer Immunol. Res. 2023, 11, 421–434. [Google Scholar] [CrossRef]
  154. Nasiri, E.; Student, M.; Roth, K.; Siti Utami, N.; Huber, M.; Buchholz, M.; Gress, T.M.; Bauer, C. IL18 Receptor Signaling Inhibits Intratumoral CD8+ T-Cell Migration in a Murine Pancreatic Cancer Model. Cells 2023, 12, 456. [Google Scholar] [CrossRef]
  155. Terme, M.; Ullrich, E.; Aymeric, L.; Meinhardt, K.; Desbois, M.; Delahaye, N.; Viaud, S.; Ryffel, B.; Yagita, H.; Kaplanski, G.; et al. IL-18 Induces PD-1–Dependent Immunosuppression in Cancer. Cancer Res. 2011, 71, 5393–5399. [Google Scholar] [CrossRef]
  156. Carbone, A.; Vizio, B.; Novarino, A.; Mauri, F.A.; Geuna, M.; Robino, C.; Brondino, G.; Prati, A.; Giacobino, A.; Campra, D.; et al. IL-18 Paradox in Pancreatic Carcinoma: Elevated Serum Levels of Free IL-18 Are Correlated with Poor Survival. J. Immunother. 2009, 32, 920–931. [Google Scholar] [CrossRef]
  157. Ahmed, A.; Klotz, R.; Köhler, S.; Giese, N.; Hackert, T.; Springfeld, C.; Jäger, D.; Halama, N. Immune Features of the Peritumoral Stroma in Pancreatic Ductal Adenocarcinoma. Front. Immunol. 2022, 13, 947407. [Google Scholar] [CrossRef]
  158. Alexandrakis, M.G.; Passam, F.H.; Sfiridaki, K.; Moschandrea, J.; Pappa, C.; Liapi, D.; Petreli, E.; Roussou, P.; Kyriakou, D.S. Interleukin-18 in Multiple Myeloma Patients: Serum Levels in Relation to Response to Treatment and Survival. Leuk. Res. 2004, 28, 259–266. [Google Scholar] [CrossRef]
  159. Tsuboi, K.; Miyazaki, T.; Nakajima, M.; Fukai, Y.; Masuda, N.; Manda, R.; Fukuchi, M.; Kato, H.; Kuwano, H. Serum Interleukin-12 and Interleukin-18 Levels as a Tumor Marker in Patients with Esophageal Carcinoma. Cancer Lett. 2004, 205, 207–214. [Google Scholar] [CrossRef]
  160. Wang, X.; Zhu, W.; Long, Q.; Chen, E.; Sun, H.; Li, X.; Xu, H.; Li, W.; Dong, P.; He, L.; et al. The Prognostic Value and Immune Correlation of IL18 Expression and Promoter Methylation in Renal Cell Carcinoma. Clin. Epigenet. 2023, 15, 14. [Google Scholar] [CrossRef]
  161. Kawabata, T.; Ichikura, T.; Majima, T.; Seki, S.; Chochi, K.; Takayama, E.; Hiraide, H.; Mochizuki, H. Preoperative Serum Interleukin-18 Level as a Postoperative Prognostic Marker in Patients with Gastric Carcinoma. Cancer 2001, 92, 2050–2055. [Google Scholar] [CrossRef] [PubMed]
  162. Merendino, R.A.; Gangemi, S.; Ruello, A.; Bene, A.; Losi, E.; Lonbardo, G.; Purello-Dambrosio, F. Serum Levels of Interleukin-18 and SICAM-1 in Patients Affected by Breast Cancer: Preliminary Considerations. Int. J. Biol. Markers 2001, 16, 126–129. [Google Scholar] [CrossRef] [PubMed]
  163. Tangkijvanich, P.; Thong-Ngam, D.; Mahachai, V.; Theamboonlers, A.; Poovorawan, Y. Role of Serum Interleukin-18 as a Prognostic Factor in Patients with Hepatocellular Carcinoma. World J. Gastroenterol. 2007, 13, 4345–4349. [Google Scholar] [CrossRef] [PubMed]
  164. Musaelyan, A.A.; Lapin, S.V.; Urtenova, M.A.; Odintsova, S.V.; Chistyakov, I.V.; Ulitin, A.M.; Akopov, A.L.; Orlov, S.V. Inflammatory and Autoimmune Predictive Markers of Response to Anti-PD-1/PD-L1 Therapy in NSCLC and Melanoma. Exp. Ther. Med. 2022, 24, 557. [Google Scholar] [CrossRef]
  165. Wang, Z.Y.; Gaggero, A.; Rubartelli, A.; Rosso, O.; Miotti, S.; Mezzanzanica, D.; Canevari, S.; Ferrini, S. Expression of Interleukin-18 in Human Ovarian Carcinoma and Normal Ovarian Epithelium: Evidence for Defective Processing in Tumor Cells. Int. J. Cancer 2002, 98, 873–878. [Google Scholar] [CrossRef]
  166. Gaggero, A.; De Ambrosis, A.; Mezzanzanica, D.; Piazza, T.; Rubartelli, A.; Figini, M.; Canevari, S.; Ferrini, S. A Novel Isoform of Pro-Interleukin-18 Expressed in Ovarian Tumors Is Resistant to Caspase-1 and-4 Processing. Oncogene 2004, 23, 7552–7560. [Google Scholar] [CrossRef]
Figure 1. IL-18 production by the NLRP3 inflammasome. (A) Extracellular ATP (eATP), present at high concentration within the TME, activates P2RX7, which in turn allows the assembly and activation of the NLRP3 inflammasome leading to caspase1 activation, cleavage of the constitutive pro IL-18 cytokine and release of mature IL-18. (B) Released mature IL-18 binds to IL-18 receptor, composed of IL-18R1 and IL-18RAP, to activate NF-kB, which in turn controls the production of INF-γ.
Figure 1. IL-18 production by the NLRP3 inflammasome. (A) Extracellular ATP (eATP), present at high concentration within the TME, activates P2RX7, which in turn allows the assembly and activation of the NLRP3 inflammasome leading to caspase1 activation, cleavage of the constitutive pro IL-18 cytokine and release of mature IL-18. (B) Released mature IL-18 binds to IL-18 receptor, composed of IL-18R1 and IL-18RAP, to activate NF-kB, which in turn controls the production of INF-γ.
Ijms 24 09235 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Janho dit Hreich, S.; Hofman, P.; Vouret-Craviari, V. The Role of IL-18 in P2RX7-Mediated Antitumor Immunity. Int. J. Mol. Sci. 2023, 24, 9235. https://doi.org/10.3390/ijms24119235

AMA Style

Janho dit Hreich S, Hofman P, Vouret-Craviari V. The Role of IL-18 in P2RX7-Mediated Antitumor Immunity. International Journal of Molecular Sciences. 2023; 24(11):9235. https://doi.org/10.3390/ijms24119235

Chicago/Turabian Style

Janho dit Hreich, Serena, Paul Hofman, and Valérie Vouret-Craviari. 2023. "The Role of IL-18 in P2RX7-Mediated Antitumor Immunity" International Journal of Molecular Sciences 24, no. 11: 9235. https://doi.org/10.3390/ijms24119235

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop