Skip to main content

The role of pyroptosis in modulating the tumor immune microenvironment

Abstract

The tumor immune microenvironment (TIME) plays a key role in immunosuppression in cancer, which results in tumorigenesis and tumor progression, and contributes to insensitivity to chemotherapy and immunotherapy. Understanding the mechanism of TIME formation is critical for overcoming cancer. Pyroptosis exerts a dual role in modulating the TIME. In this review, we summarize the regulatory mechanisms of pyroptosis in modulating the TIME and the potential application of targeted pyroptosis therapy in the clinic. Several treatments targeting pyroptosis have been developed; however, the majority of treatments are still in preclinical studies. Only a few agents have been used in clinic, but the outcomes are unsatisfactory. More studies are necessary to determine the role of pyroptosis in cancer, and more research is required to realize the application of treatments targeting pyroptosis in the clinic.

Overview of cancer burden

Cancer is a major global challenge. In 2019, respiratory cancer ranked as the sixth leading cause of death in the world [1]. In China, respiratory cancer was the third leading cause of death after stroke and ischemic heart disease, and liver cancer was the fifth leading cause in 2017 [2]. According to the estimate from Global Cancer Observatory, there will be approximately 4,820,000 and 2,370,000 new cancer cases and 3,210,000 and 640,000 cancer deaths in China and the USA in 2022, respectively [3]. Many cancer treatments have been developed; however, no tumor can be radically cured. For example, pancreatic cancer, the most malignant tumor, has a five-year survival of less than 10% [4]. The low rate of early detection, high incidence of recurrence after resection, and resistance to chemotherapy partly account for the poor prognosis of pancreatic cancer patients. To improve the prognosis of pancreatic cancer, novel therapeutic regimens have been developed, such as immunotherapy and targeted therapy. However, the outcomes of treatments, especially immunotherapy, have not met expectations [5]. The complex mechanisms of tumor development may partly account for the poor prognosis. The tumor immune microenvironment (TIME) plays a vital role in tumorigenesis and tumor progression, as well as to tumor sensitivity to chemotherapy and immunotherapy. Better understanding of the TIME may lead to therapeutic strategies for cancer treatment.

The TIME

The TIME consists of cellular and acellular components, including cancer cells, tumor-associated stomal cells, immune cells, extracellular matrix, and cytokines and other soluble molecules. (Table 1). The processes and mechanisms of TIME formation are extremely complex and vary among tumors depending on the genetic background and heterogeneity. The TIME of pancreatic cancer is representative of its characteristics of fewer tumor cells (accounting for 10% of total cells), more infiltration of immunosuppressive cells and fewer effector T cells, extremely dense matrix, and immunosuppression [6,7,8]. Therefore, in this review, we focus on pancreatic cancer as an example to describe the complex networks of the TIME.

Table 1 The cellular components of the TIME in pancreatic cancer

At the stage of tumorigenesis in pancreatic cancer, mutations of the KRAS gene cause ductal metaplasia of acinar cells, followed by pancreatic intraepithelial neoplasia (PanIN). During the progression from PanIN-I to PanIN-III, mutations in several key genes, including P16, TP53 and SMAD4 genes, ultimately lead to tumor formation. Theoretically, the immune microenvironment stimulated by tumor cells should fight against tumor cells and inhibit tumor formation. However, tumor cells escape immune system surveillance by low expression of major histocompatibility complex class I and overexpression of programmed death-ligand 1 (PD-L1), Fas, and Fas ligand. In addition, immune cells are reprogrammed to promote the formation of a “cold” tumor that is characterized by an absence of T cells within the tumor and at the tumor edges and failure of T cell priming [9].

Tumor-associated macrophages (TAM) are the most common infiltrating immune cells in the TIME and are categorized as either activated M1 or activated M2 macrophages. M1-TAM have antitumor functions, whereas M2-TAM contribute to tumor progression. Macrophages can be induced to M2-TAM by tumor cell–derived C-C motif chemokine ligand 2 (CCL-2), transforming growth factor beta (TGF-β), and intercellular adhesion molecule 1 (ICAM-1) and then enhance the malignant phenotype of tumor cells. Furthermore, M2-TAM secrete arginase 1 (Arg-1) and TGF-β that attenuate the function of CD8+  T cells and natural killer (NK) cells, which also cross talk with regulatory T cells (Treg) and type 1 helper T (Th1) cells by the secretion of interleukin-10 (IL-10) and TGF-β [10]. Cytokines in the TIME, such as granulocyte-macrophage colony stimulating factor (GM-CSF) and granulocyte colony stimulating factors (G-CSF), arrest the maturation of myeloid progenitor cells and induce differentiation to myeloid-derived immunosuppressor cells (MDSC) that exert immunosuppressive functions, including inhibition of T cells and NK cell proliferation and activation, induction of Treg expansion, and promotion of macrophage differentiation to M2-TAM [11,12,13].

The molecular mechanism of the immunosuppression of MDSC has been identified. MDSC are mainly classified into polymorphonuclear MDSC (PMN-MDSC, also called granulocytic-MDSC) and mononuclear-MDSC (M-MDSC) [14]. Both PMN-MDSC and M-MDSC express high amounts of Arg-1, which depletes L-arginine and results in T-cell inhibition [13]. PMN-MDSC and M-MDSC have distinct immunosuppressive mechanisms. PMN-MDSC produce high amounts of reactive oxygen species (ROS) that activate signal transducer and activator of transcription 3 (STAT3) [15]. M-MDSC express high levels of inducible nitric oxide synthase (iNOS), which activate the STAT1 signal and increase the production of nitric oxide (NO) [16]. MDSC exert immunosuppressive effects by interactions with immune cells. However, MDSC also promote tumor progression by directly contacting cancer cells. Panni et al. demonstrated that M-MDSC increased the population of cancer stem cells and promoted epithelial-mesenchymal transition when cocultured with pancreatic cancer cells through a STAT3-dependent mechanism [17].

The tumor stroma also plays a crucial role in the formation of the TIME. In the presence of cancer cells or during injury, pancreatic stellate cells (PSC) are activated, which inhibit the proliferation of T cells and NK cells and attenuate the cytotoxic functions, activate MDSC, and polarize macrophages by secreting cytokines [18, 19]. Furthermore, activated PSC secrete a large amount of extracellular matrix proteins that prevent therapeutic drugs into the tumor [18].

In summary, tumor cells, immune cells, and stromal cells interacting with each other via cytokines constitute a “cold” tumor with abundant extracellular matrix, which is characterized by immunosuppression, and insensitivity to drug therapy (Fig. 1).

Fig. 1
figure 1

The tumor immune microenvironment in pancreatic cancer. Pancreatic cancer is focused as an example to describe the TIME. Tumor cells, immune cells, and stromal cells interacting with each other via cytokines constitute a “cold” tumor with abundant extracellular matrix. aPSC, activated pancreatic stellate cells; DC, dendritic cells; MDSC, myeloid-derived suppressor cells; NK cells, natural killer cells; TAM, M2 tumor-associated macrophages; Th17, helper T cells 17; Treg, regulatory T cells

Pyroptosis

Pyroptosis is a gasdermin (GSDM)-mediated programmed cell death [20] that was first described in 1992 [21]. The GSDM family contains six members in humans (A, B, C, D, E, and DFNB59) and 10 members in mice (A1/2/3, B, C1/2/3/4, D, and DFNB59). All GSDM members in human except for DFNB59 contain two conserved domains (a C-terminal inhibitory domain and a N-terminal effector domain), which are cleaved and mediate pyroptosis [22]. Most research has focused on GSDMD and GSDME. GSDMD-meditated pyroptosis is induced by both the canonical inflammasome pathway and the non-canonical inflammasome pathway (Fig. 2). In the canonical pathway, exogenous pathogens and endogenous damage are recognized by intracellular sensor proteins, including the nucleotide-binding oligomerization domain (NOD)-like receptors (NLRP1, NLRC4 and NLRP3), absent in melanoma 2 (AIM2), and Pyrin. The signal recruits the inflammasome adaptor apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (CARD) through Pyrin domains, bridging inflammasome sensors with pro-caspase-1 and resulting in activation of caspase-1 via self-cleavage [23]. In the non-canonical pathway, intracellular lipopolysaccharide (LPS) binds directly to caspase-4/5 in humans and caspase-11 in mice via the CARD domain, resulting in the activation of these caspases [24]. Activated caspase-1/4/5/11 cleave GSDMD, the N-terminal domain is liberated and forms pores on the cell membrane, resulting in cytoplasmic swelling, membrane rupture, and the release of cytosolic contents. Activated caspases also cleave the pro-inflammatory cytokines IL-1β and IL-18, leading to maturation of IL-1β and IL-18. These pro-inflammatory cytokines are released from the pores on cell membrane and amplify the local or systemic inflammatory effects [22].

Fig. 2
figure 2

The pyroptosis pathways. In the canonical pathway, exogenous pathogens and endogenous damage are recognized by the intracellular sensor proteins, such as NLRP1, NLRC4, NLRP3, AIM2, and Pyrin, and then activate caspase-1. In the non-canonical pathway, intracellular LPS binds directly to caspase-4/5 in humans and caspase-11 in mice and results in activation of these caspases. Activated caspase-1/4/5/11 cleave GSDMD; the N-terminal domain is liberated, which forms pores on cell membrane and results in cytoplasmic swelling, membrane rupture, and the release of cytosolic contents. Activated caspases also cleave pro-inflammatory cytokines IL-1β and IL-18, leading to maturation of IL-1β and IL-18 that are released from the pores on the cell membrane and amplify the local or systemic inflammation

GSDME-meditated pyroptosis is usually induced by drugs. Chemotherapeutic drugs, such as doxorubicin and cisplatin, activate caspase-3 and then cleave GSDME [25]. Other drugs such as metformin enhance B-cell lymphoma-2-associated X expression and cytochrome C release, subsequently mediating caspase-3/GSDME noncanonical pyroptosis [26]. Caspase-7 also cleaves GSDME at the same cleavage site as caspase-3 [27]. There are also reports about the cleavage of GSDMA, GSDMB and GSDMC, which are listed in Table 2.

Table 2 Induction and cleavage of gasdermin family

Involvement of pyroptosis in the TIME

Evidence of the involvement of pyroptosis in TIME has mainly come from bioinformatic analyses, which demonstrates that several genes and non-coding RNAs related to pyroptosis are associated with tumor infiltrating immune cells in pan-cancers [35,36,37]. Here, we review the current understanding of the pyroptosis executor GSDM, inflammasome, and proinflammatory cytokines (IL-1β and IL-18) in modulating the TIME (Fig. 3).

Fig. 3
figure 3

Involvement of pyroptosis in the TIME. The pyroptosis executor GSDM, and proinflammatory cytokines (IL-1β and IL-18) modulate the TIME by regulating immune cells

Fig. 4
figure 4

Remodeling the TIME by targeting pyroptosis. Several treatments targeting pyroptosis have been proposed or developed to overcome the immunosuppression microenvironment, including evocation of GSDMD/GDDME-mediated pyroptosis, prevention of inflammasome activation, antagonizing IL-1β, increase of IL-18. ICB, immune checkpoint blockade

GSDM

Several publications have reported the association between GSDM and the TIME; however, most studies reported results that were obtained from the bioinformatic analyses. The associations of GSDMA and GSDMB with the TIME were just traced by bioinformatic analyses. However, some evidence from basic experimental studies have confirmed the association between GSDMC, GSDMD, GSDME and the TIME. Liu et al. developed an immunotherapy system on the basis of red blood cell membranes encapsulated Listeria monocytogenes (Lmo) with selective deletion of virulence factors (Lmo@RBC). Lmo@RBC-induced GSDMC-mediated cancer cell pyroptosis promoted the secretion of IL-6, TNF-α, and IFN-γ of bone marrow–derived DC and accumulation of DC and CD8+ T cells in tumor tissues [38]. Wang et al. [39] reported that membranous GSDMD expression in colorectal cancer positively correlated with CD68+ macrophages in the tumor center and CD8+ lymphocytes in the tumor invasive front. However, nuclear GSDMD was negatively related to CD68+ macrophages in the tumor invasive front and CD8+ lymphocytes in the tumor center. Cytoplasmic GSDMD was associated with more CD3+ lymphocytes both in the tumor center and tumor invasive front. The role of GSDME in tumor biology is unclear. Croes et al. indicated that GSDME played a role in tumorigenesis in intestinal cancer. However, neither the number of affected mice nor the multiplicity of proliferative lesions in both chemically induced and genetically modified intestinal cancer mouse models between the GSDME knockout and wild-type groups was different. The authors explained the contradictory phenomenon through the effects created by GSDME-mediated necrotic cell death on TIME due to the observation of inflammatory cell infiltration in the tumor [40]. Tan et al. [41] reported that macrophages were polarized to the M1 phenotype when cocultured with D2 dopamine receptor-expressing breast cancer cells, which then activated GSDME-mediated pyroptosis of breast cancer cells.

Inflammasomes (Table 3)

NLRP3

Bioinformatic analyses identified NLRP3-inflammasome-related genes that were differentially expressed between normal and tumor samples in 15 cancer types and were significantly associated with survival of patients with hepatocellular carcinoma or skin cutaneous melanoma. The NLRP3 inflammasome score correlated with tumor-infiltrating lymphocytes and macrophages and was a stronger predictor for immune signatures compared with tumor mutation burden and glycolytic activity [42]. The NLRP3 inflammasome is involved in tumor progression by the recruitment of myeloid cells, such as MDSC and TAM [43]. The inhibition of inflammasomes suppressed TAM infiltration and increased CD4+ and CD8+ T cell infiltration and PD-L1 expression in tumors with high inflammasome signaling activity, amplifying the curative effect of PD-L1 blockade [44].

Table 3 Involvement of inflammasomes in TIME

The NLRP3 inflammasomes in tumor infiltrating immune cells and stromal cells are involved in tumor progression. Inflammasomes in macrophages are activated by the macrophage-colorectal cancer cell crosstalk, then increasing IL-1β secretion and promoting colorectal cell migration [45]. Cancer-derived exosomal tripartite motif-containing 59 activates the NLRP3 inflammasome in macrophages by ubiquitination and degradation of abhydrolase domain containing 5, which facilitated lung cancer growth and metastasis in vivo [46]. Inhibition of the NLRP3 inflammasome in macrophages suppressed the metastatic potential of melanoma cells [47]. Cancer-associated fibroblast–derived NLRP3 inflammasome and IL-1β facilitated tumor growth and metastasis by modulating the tumor microenvironment towards an immune suppressive milieu in breast cancer. The NLRP3 inflammasome increased the recruitment of CD11b+Ly6ChighLy6G myeloid cells into tumors, and IL-1β increased the recruitment of the granulocytic fraction (CD11b+Ly6G+Ly6Clow) in the primary tumor and the monocytic fraction (CD11b+Ly6ChighLy6G−) in metastatic tumors [48].

Several factors activate the NLRP3 inflammasome and alter the TIME. Epstein-Barr virus latent membrane protein 1 promoted the expression of MDSC-related molecules and cytokines by activating the NLRP3 inflammasome, leading to tumor immunosuppression in nasopharyngeal carcinoma [49]. Porphyromonas gingivalis changed the TIME by selectively expanding myeloid-derived immune cells and inducing a proinflammatory microenvironment without the influence of CD4+ T cells, CD8+ T cells and NK cells in a colorectal cancer xenograft mouse model by activating the hematopoietic NLRP3 inflammasome [50]. High mobility group box 1 (HMGB1) increased the sensitivity of glioblastoma cells to temozolomide. Mechanistically, HMGB1 interacted with receptor for advanced glycation end products (RAGE), a major receptor for HMGB1 on TAM, and then activated the NLRP3 inflammasome by nuclear factor-kappaB (NF-κB) signaling, which polarized a M1-like macrophage phenotype by promoting the release of pro-inflammatory cytokines (TNF-α, interferon gamma (IFN-γ), IL-1β, IL-6, IL-8 and CCL2) [51]. The macrophage-specific deletion of glucose transporter 1 in pancreatic cancer mouse models inhibited tumor growth and NK cell infiltration by suppressing the pro-inflammatory NLRP3-IL1β axis [52].

NLRC4

NLRC4 is critical for cytokine production in TAM and necessary for the generation of IFN-γ-producing CD4+ and CD8+ T cells in an inflammasome-independent manner, suppressing tumor growth in a subcutaneous murine model of melanoma [53]. Obesity enhanced NLRC4 expression in tumor-infiltrating CD11b+ cells in breast tumors in mice, resulting in activation of IL-1β, and promotion of disease progression through adipocyte-mediated expression of vascular endothelial growth factor A and angiogenesis [54]. In mouse models for nonalcoholic fatty liver disease, fatty liver enhanced the expression of IL-1β and NLRC4 in metastatic tumors, which promoted TAM infiltration and M2 polarization and increased colorectal cancer liver metastasis [55].

NLRP1

Dipeptidyl peptidase (DPP) inhibitors induced NK and T cell infiltration, enhanced the efficacy of anti-PD1 antibody, and reduced tumor growth. Mechanistically, DPP inhibition activated the NLRP1 inflammasome, resulting in proinflammatory cytokine release and the Th1 cell response, further enhancing the CXCL9/10-CXCR3 axis [56]. However, DPP inhibitors also promoted the recruitment of tumor-infiltrating CD45, MPO, F4/80, CD4, Foxp3-positive cells, and MDSC and decreased CD8-positive lymphocytes in metastatic sites in mouse breast cancer models by ROS-NF-κB-dependent NLRP3 inflammasome activation [57]. The dual-role of DPP inhibitors in the TIME should be considered when the inhibitors are used in cancer treatment.

AIM2

Fukuda et al. reported that AIM2 promoted tumor growth with the increase of Treg proportion and decrease of CD4+ effector T cells and without changes in the numbers of CD8+ or CD4+ T cells and DC in the melanoma microenvironment in mouse models. Additionally, AIM2 inhibited the expression of IFN-β and increased IL-1β and IL-18. Tumor infiltrating DC were the major producers of IFN-β. The authors examined the expression of AIM2 in DC and found that tumor-infiltrating DC with AIM2-expression in human melanoma were increased and correlated with tumor progression. AIM2-deficient DC promoted tumor antigen–specific CD8+ T cell infiltration into the tumor via CXCL10. AIM2 was also required for the production of IL-1β and IL-18, which promoted Treg accumulation and tumor growth in vivo [58].

IL-1β and IL-18

Both IL-1β and IL-18 lead to the infiltration of more immune cells and result in the generation and maintenance of an inflammatory microenvironment surrounding cancer cells [59], however, the biofunctions are diverse.

Il-1β

The function of IL-1β in the TIME is a double-edged sword in tumor progression. IL-1 signaling activates innate immune cells and drives polarization of CD4+ T cells towards Th1 and Th17 cells, which initiates an adaptive anti-tumor response [60]. The protective effects of IL-1β are observed in several tumors [60]. However, IL-1β also creates a tumor immunosuppressive microenvironment predominantly by TAM and MDSC, which promotes tumor development.

IL-1β enhances TAM expansion. Interferon-inducible protein 16 (IFI-16) is increased in pancreatic cancer, and elevated IFI-16 correlates to a poor prognosis. Overexpression of IFI-16 in pancreatic cancer cells induces the maturation, infiltration, and proliferation of TAM in the tumor microenvironment by activating inflammasomes and therefore increasing the release of IL-1β. Neutralization of IL-1β attenuated the effects of IFI-16 overexpression in pancreatic cancer cells on TAM [61]. Tumor cell–derived IL-1β is essential for the formation of the protumorigenic microenvironment in pancreatic cancer, which promotes the activation and secretory phenotype of quiescent PSC, increases stromal accumulation of TAM, MDSC, CD1dhiCD5+ regulatory B cells, and Th17 cells, and decreases intratumoral infiltration and activation of CD8+ cytotoxic T cells [62].

IL-1β also increases cell numbers and infiltration of MDSC in tumor. The levels of MDSC in mice harboring mammary carcinoma were positive association with IL-1β expression levels [63]. IL-1β upregulated cyclooxygenase-2 that encoded prostaglandins then mediate MDSC propagation [64]. IL-1β also activated STAT3 in glioma cells, which decreased the population of DC and increased the population of MDSC [65].

Il-18

Like IL-1β, IL-18 exhibits a dual-role in tumor development by regulating the TIME. The anti-tumor effects of IL-18 are partly ascribed to the production of IFN-γ in Th1 cells and NK cells. IL-18 synergized with IL-12 to promote IFN-γ production in Th1 cells [66], and Th1 cell–derived IFN-γ enhanced the cytolytic activity of CD8+ cells and NK cells. IL-18 also increased IFN-γ production and cytotoxic activity in NK cells without IL-12 [67] or combined with IL-12 or IL-2 [68]. Furthermore, IL-18 combined with IL-12 enhanced the cytotoxic activity of murine spleen-derived CD8+T cells, including inducing IFN-γ and granzyme production [69]. IL-18 in non-small cell lung cancer (NSCLC) tumor district, mostly produced by tumor cells, was associated with classical Th1 cell–associated cytokines (i.e., IFNγ, TNF-α, and IL-12) and contributed to the expansion of IL-18R+ CD8+ T cells expressing T-bet and IFN-γ within the tumor microenvironment [70]. Markowitz et al. reported that IL-18 played an inflammation-dependent tumor-suppressive effect mainly by promoting the differentiation, activity, and survival of tumor-infiltrating T cells in hepatocellular carcinoma [71].

IL-18 also promotes tumor progression by inducing an immunosuppressive microenvironment. Lim et al. demonstrated that IL-18 treatment significantly increased the percentage and the number of M-MDSC through the differentiation of bone marrow progenitor cells, which enhanced the suppression of T cell proliferation and IFN-γ production in vitro. The administration of IL-18 to mice bearing melanoma increased the accumulation of M-MDSC in the tumor microenvironment [72]. The multiple myeloma-niche-derived IL-18 drove the generation of both PMN-MDSC and M-MDSC, which suppressed CD8+T cell proliferation and IFN-γ production and then accelerated disease progression [73]. The tumor-promoting effects of IL-18 were also shown by IL-18 induction of PD-1 expression on NK cells in the TME, leading to a poor prognosis in nasopharyngeal carcinoma [74], and high expression of IL-18 in TAM promoted the migration and invasion of breast cancer cells [75].

Although IL-1β and IL-18 play crucial roles in creating the TIME, the regulatory role is not equivalent to the effect of pyroptosis in consideration of the source of IL-1β and IL-18 [60, 76]. The maturation and release of IL-1β and IL-18 depending on the inflammasome-caspase 1-GSDM signal is just one of the ways. Kiss et al. demonstrated that myeloid cells were the primary source of IL-1β in lung and breast tumors and released IL-1β independent of the inflammasome and GSDMD. Furthermore, the authors showed that IL-1β promoted systemic neutrophil expansion and enhanced the accumulation of T-cell-suppressive neutrophils in tumors. IL-1β deletion attenuated immune suppression in the TME, including inhibition of neutrophil recruitment and increase of cytotoxic CD8+T cells [77]. More evidence is needed to verify the effects of pyroptosis related to IL-1β and IL-18 in the TIME.

Remodeling TIME by targeting pyroptosis: from bench to bedside (Fig. 4)

Activation of GSDM-mediated pyroptosis

Both GSDMD and GSDME have been focused in the field of tumor-targeted therapy, and several small molecule agents or nano-drugs targeting GSDMD and GSDME have been developed. Su et al. [78] designed a carbonic anhydrase IX (CAIX)-anchored rhenium(I) photosensitizer (named CA-Re) that evokes GSDMD-mediated pyroptosis to effectively stimulate tumor immunogenicity. CA-Re enhanced the maturation and antigen-presenting ability of DC and fully activated the T cell–dependent adaptive immune response in vivo. A small molecule agent NBS-1MT with the photosensitizer and indoleamine 2,3-dioxygenase (IDO) inhibitor was synthesized that triggers pyroptosis by activating caspase-1 and then cleaving GSDMD. The activated pyroptosis enhanced the immune-related factor release and promoted the intratumoral infiltration of cytotoxic T lymphocytes and immunogenic cell death of tumor cells [79].

Xiao et al. [80] designed a smart tumor microenvironment ROS/glutathione (GSH) dual-responsive nano-prodrug (denoted as MCPP) with high paclitaxel and photosensitizer purpurin 18 (P18) loading that responded to high ROS/GSH in the tumor microenvironment and was shown to achieve optimal drug release in tumors. The nano-prodrug promoted the release of danger-associated molecular patterns via inducing GSDME-related tumor cell pyroptosis, which then initiated adaptive immunity, boosted the efficiency of the immune checkpoint blockade, achieved tumor regression, generated immunological memory, and prevented tumor recurrence. The engineering of a series of multienzyme-mimicking covalent organic frameworks (COFs), COF-909-Cu, induced GSDME-dependent pyroptosis and remodeled the tumor microenvironment, which activated durable anti-tumor immunity, enhanced the response of PD-1 inhibitor, and restrained tumor metastasis and recurrence [81]. Furthermore, activation of GSDM-mediated pyroptosis increased the effects of targeted therapy for tumors. The combinations of BRAF inhibitors and MEK inhibitors (BRAFi + MEKi) caused durable regression of melanoma by promoting cleavage of GSDME and the release of HMGB1. GSDME-deficient melanoma showed defective HMGB1 release, decreased tumor-associated T cells, and activated DC infiltrates in response to BRAFi + MEKi, and more frequent tumor regrowth was observed after drug removal [82].

Inactivation of inflammasomes

Many agents and molecules have been developed to regulate inflammasome activity. However, few agents have been used in the clinic and several are still in clinical trials [83]. Thalidomide, a drug that inhibits the activation of caspase-1, showed anti-tumor effects in patients with advanced myeloma [83]. A phase II randomized clinical trial indicated that thalidomide combined with docetaxel resulted in the increased median survival in patients with metastatic androgen-independent prostate cancer [84]. However, thalidomide in combination with chemotherapy did not improve survival in patients with NSCLC and increased the risk of thrombotic events [85]. A P2X7R antagonist has been evaluated in a clinical trial. P2X7R promoted tumor growth in several solid tumors by activating the NLRP3 inflammasome. A phase I study was performed to assess the safety and tolerability of the P2X7R antagonist in the treatment of basal cell carcinoma; approximately 65% of patients showed a decrease of the lesion area and the most common adverse event was an allergic reaction at the treatment site [86]. Many agents, such as pralnacasan, Ac-YVAD-CHO, MCC950, glyburide, and terpenoids, are in the preclinical stage and have shown potential anti-tumor effects [83, 87]. Clinical trials are required to determine the potential clinical application of these agents.

Activation of the NLRP3 inflammasome remodeled the TIME and increased infiltration of MDSC, Tregs and TAM and decreased CD4+ and CD8+ T cells [88]. The genetic and pharmacologic inhibition of NLRP3 inhibited PMN-MDSC tumor infiltration and significantly improved the efficacy of anti-PD-1 antibody immunotherapy [89]. However, the combination of NLRP3 blockers with anti-PD-L1 treatment showed antagonistic effects in lymphoma. Lu et al. demonstrated that NLRP3 inflammasome blockade reduced the immunosuppressive cell population and PD-L1 expression and inhibited lymphoma growth. Anti-PD-L1 antibodies partly impaired the effect of MCC950 (an inhibitor of activation of NLRP3 inflammasomes) [90]. The possible reason was that activation of CD8+ T cells in response to PD-1 blockade induced a PD-L1/NLRP3 inflammasome signaling cascade that resulted in the recruitment of PMN-MDSC into the tumor, thus inhibited the resulting anti-tumor immune response [89]. This phenomenon indicates that a complex regulatory net with negative and positive feed-back loops is created in the TME, and inhibition of the NLRP3 inflammasome does not always only produce the desired positive effects. The application of NLRP3 inhibitors should be evaluated by preclinical studies.

IL-1β and IL-18

Il-1β

A pre-clinical study indicated the anti-tumor effects of anti-IL-1β antibody and enhanced response of anti-PD-1 antibody [91]. Blocking IL-1R inhibited tumor growth and metastasis accompanied by inhibition of myeloid cell accumulation in human breast cancer models [43]. Several clinical trials for anti-IL-1β or IL-1R blockage in tumor treatment are ongoing.

The CANOPY program including four phase II or III randomized studies was designed to evaluate the effects of canakinumab (a IgG1k monoclonal anti-human antibody that selectively binds and neutralizes IL-1β) in the treatment of NSCLC [92]. The registered phase 1B study PanCAN-SR1 (NCT04581343, https://clinicaltrials.gov/) is recruiting to confirm the tolerable doses and acceptable safety profile of canakinumab and spartalizumab in combination with nab-paclitaxel and gemcitabine for treatment of patients with metastatic pancreatic cancer. Clinical trials for gevokizumab (an allosteric anti–IL-1β antibody) with standard care of anti-cancer therapies for metastatic colorectal, gastroesophageal and renal cancers, FL-101 (a monoclonal anti-IL-1β antibody) for surgically resectable NSCLC, isunakinra (a fusion protein-based inhibitor of the IL-1R containing domains from both IL-1β and IL-1Ra) for solid tumors, and anakinra (an IL-1 receptor antagonist) for hematological malignancies are ongoing.

Il-18

While preclinical studies and clinical trials have almost only focused on the anti-tumor effects of anti-IL-1β, other research has examined improving the levels of IL-18 in the treatment of tumors. The combination of recombinant IL-18 and immune checkpoint inhibitors led to synergistic inhibition of tumor growth in animal models of peritoneal dissemination of colon cancer [93]. The combination of IL-18 and IL-2 showed a synergistic anti-tumor effect in mice subcutaneously inoculated with a sarcoma [94]. However, recombinant IL-18 had limited activity as a single agent in patients with metastatic melanoma in clinical trials [95]. Several novel methods aiming to improve the anti-tumor effects of IL-18 have been developed.

Saetang et al. reported that engineered E6K + T63A IL-18, which has 16 times higher activity than native IL-18, increased the proportions of Th1 cells and cytotoxic T lymphocytes in mice harboring tumors and inhibited tumor growth [96]. Drakes and colleagues demonstrated that T-cell receptor (TCR)-modified T-cells with IL-18 expression created a superior proinflammatory TME, which improved overall survival of mice in the pmel-1 syngeneic tumor model [97]. Similarly, Kunert et al. engineered T cells with a melanoma-specific TCR and murine IL-18 under the control of a nuclear-factor of activated T-cell (NFAT)-sensitive promoter; the cells produced IL-18 and consequently enhanced levels of IFN-γ after exposure to antigen-positive tumor cells. Adoptive transfer of these cells to melanoma-bearing mice resulted in the accumulation of therapeutic CD8+T cells within tumors, reduction of tumor burden, and prolonged survival without side effects [98].

IL-18 binding protein (a high-affinity IL-18 decoy receptor) limited the anti-tumor activity of IL-18 in mice. Therefore, Zhou et al. engineered a “decoy-resistant” IL-18 (DR-18), which exerted potent anti-tumor effects in mouse tumor models compared with wild-type IL-18 by promoting the development of poly-functional effector CD8+T cells, decreasing the prevalence of exhausted CD8+ T cells, and expanding the pool of stem-like TCF1+ precursor CD8+ T cells. Additionally, DR-18 enhanced the activity and maturation of NK cells to effectively treat anti-PD-1-resistant tumors [99].

Chimeric antigen receptor (CAR) T cell therapy has emerged as an effective cancer treatment [100]. IL-18-secreting CAR T cells presented expansion and persistence and significantly increased long-term survival in mouse models for hematological and solid malignancies. These T cells modulate the TIME and enhance the anti-tumor immune response and might be a promising strategy to improve the clinical outcomes of adoptive T cell therapy [101].

Notably, both IL-1β and IL-18 exert dual effects in tumor progression. A protective effect of IL-1β in mouse models of chemically induced colon carcinoma was observed [102], whereas recombinant IL-18 promoted T-cell acute leukemia growth in vitro [103]. Therefore, more studies are needed to fully determine the role of IL-1β and IL-18 in tumors. The application of anti-IL-1β or recombinant IL-18 in the clinic should consider its dual role in tumors. Furthermore, several anti-tumor agents were shown to induce IL-1β mostly in a NLRP3 inflammasome–dependent manner [60]; therefore, the treatment of tumors with the combination of conventional agents and anti-IL-1β has been adapted. Voloshin et al. [104] demonstrated that paclitaxel chemotherapy induced IL-1β expression in macrophages in the TME, which promoted the invasive properties of cancer cells. Inhibition of IL-1β slightly delayed primary tumor regrowth following paclitaxel treatment but increased spontaneous metastases. The authors suggested that treatments using “add-on” drugs to conventional therapy should be investigated in tumor models consisting of primary tumors and their metastases [104].

Conclusions and future perspectives

The TIME plays a key role in immunosuppression that results in tumorigenesis and tumor progression, as well as resistance to chemotherapy and immunotherapy. Pyroptosis exerts a crucial role in modulating the TIME. Several treatments targeting pyroptosis have been proposed or developed to overcome the immunosuppression microenvironment in tumor; however, most of these treatments are in preclinical studies. Although some agents have been used in the clinic, the outcomes are unsatisfactory.

In the field of drug development, researchers are also focusing on natural products and prescribed drugs. Several natural products associated with pyroptosis show anti-tumor effects, which are promising in developing strategies to improve the effects of chemotherapy and immunotherapy. Terpenoids are bioactive compounds widely distributed in nature and show a wide range of biological activities, such as antimicrobial, antiviral, anti-tumor, antioxidant, and cardio-protection effects [87]. Recently, these compounds were verified to inhibit the activation of the NLRP3 inflammasome in mouse models. Andrographolide, a natural diterpenoid from Andrographis paniculate, protected mice against azoxymethane/dextran sulfate sodium–induced colon carcinogenesis by inhibiting the NLRP3 inflammasome [105]. Triptolide, a compound isolated from the traditional Chinese herb Tripterygium wilfordii Hook f, eliminated head and neck cancer cells by inducing GSDME-mediated pyroptosis [106]. In addition, Spatholobus suberectus Dunn percolation extract exhibited anti-tumor efficacy by activating ROS-induced noncanonical inflammasome pyroptosis in triple-negative breast cancer [107]. Piperlongumine (PL) analogue L50377 suppressed the cell growth of non-small-cell lung cancer cells partly by activation of ROS-mediated pyroptosis [108].

Some prescribed drugs exhibit anti-tumor effects through regulating pyroptosis. Metformin, a widely used anti-diabetic medication, induces both GSDMD-mediated canonical pyroptosis [109] and caspase-3/GSDME-mediated noncanonical pyroptosis [26], which inhibited the proliferation of esophageal squamous cell carcinoma cells [109]. Disulfiram (DSF), an anti-alcoholic drug, was recently reported for the treatment of cancer and inflammatory disorders [110, 111]. However, the mechanism is not well understood. Hu et al. demonstrated that that DSF covalently modified Cys192 of GSDMD and inhibited pore formation of the plasma membrane and the process of pyroptosis [112]. Therefore, we speculated that one mechanism of the anti-tumor activity of DSF might be the inhibition of pyroptosis. Further studies are necessary to identify the potential clinical application of these compounds.

Notably, pyroptosis exhibits both anti-tumor and tumor promoting effects, which limits the application of agents targeting pyroptosis. The effects of agents that target pyroptosis should be well evaluated by rigorous preclinical studies and clinical trials. Furthermore, combined therapy with PD-1 antibody and NLRP3 inhibitor resulted in impaired anti-tumor effects compared with monotherapy by increasing recruitment of PMN-MDSC into the tumor, which highlights the complexity of the TIME. The combination of PD-1 antibody and targeted pyroptosis therapy in tumors might produce unexpected results. Nevertheless, deeper insights into the role of pyroptosis in modulating TIME expands our understanding of cancer and is beneficial to developing novel drugs for overcoming this tricky disease.

Availability of data and materials

Not applicable.

Abbreviations

AIM2:

absent in melanoma 2

Arg-1:

arginase 1

CAR:

chimeric antigen receptor

CARD:

caspase activation and recruitment domain

CCL-2:

C-C motif chemokine ligand 2

COF:

covalent organic framework

DC:

dendritic cell

DPP:

dipeptidyl peptidase

DR-18:

decoy-resistant IL-18

DSF:

disulfiram

G-CSF:

granulocyte colony stimulating factor

GM-CSF:

granulocyte-macrophage colony stimulating factor

GSDM:

gasdermin

GSH:

glutathione

HMGB1:

high mobility group box 1

ICAM-1:

intercellular adhesion molecule 1

IFI-16:

interferon-inducible protein 16

IFN-γ:

interferon gamma

IL:

interleukin

iNOS:

inducible nitric oxide synthase

LPS:

lipopolysaccharide

MDSC:

myeloid-derived immunosuppressor cell

M-MDSC:

mononuclear-MDSC

NF-κB:

nuclear factor-kappaB

NK:

natural killer

NLR:

nucleotide-binding oligomerization domain (NOD)-like receptor

NOD:

nucleotide-binding oligomerization domain

NSCLC:

non-small cell lung cancer

PanIN:

pancreatic intraepithelial neoplasia

PD-L1:

programmed death-ligand 1

PMN-MDSC:

polymorphonuclear MDSC

PSC:

pancreatic stellate cell

ROS:

reactive oxygen species

RAGE:

receptor for advanced glycation end products

STAT3:

signal transducer and activator of transcription 3

TAM:

tumor-associated macrophage

TCR:

T-cell receptor

TGF-β:

transforming growth factor beta

Th1:

type 1 helper T

TIME:

tumor immune microenvironment

References

  1. World Health Orgnization: the top 10 causes of death. 2020. https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death.

  2. Zhou M, Wang H, Zeng X, Yin P, Zhu J, Chen W, et al. Mortality, morbidity, and risk factors in China and its provinces, 1990-2017: a systematic analysis for the global burden of disease study 2017. Lancet. 2019;394(10204):1145–58.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Xia C, Dong X, Li H, Cao M, Sun D, He S, et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chin Med J. 2022;135(5):584–90.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33.

    Article  PubMed  Google Scholar 

  5. Qian Y, Gong Y, Fan Z, Luo G, Huang Q, Deng S, et al. Molecular alterations and targeted therapy in pancreatic ductal adenocarcinoma. J Hematol Oncol. 2020;13(1):130.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Mundry CS, Eberle KC, Singh PK, Hollingsworth MA, Mehla K. Local and systemic immunosuppression in pancreatic cancer: targeting the stalwarts in tumor's arsenal. Biochimica et Biophysica Acta Rev Cancer. 2020;1874(1):188387.

    Article  CAS  Google Scholar 

  7. Kemp SB, Pasca di Magliano M, Crawford HC. Myeloid cell mediated immune suppression in pancreatic Cancer. Cell Mol. Gastroenterol Hepatol. 2021;12(5):1531–42.

    CAS  Google Scholar 

  8. Lim SA, Kim J, Jeon S, Shin MH, Kwon J, Kim TJ, et al. Defective localization with impaired tumor cytotoxicity contributes to the immune escape of NK cells in pancreatic Cancer patients. Front Immunol. 2019;10:496.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019;18(3):197–218.

    Article  CAS  PubMed  Google Scholar 

  10. Du W, Pasca di Magliano M, Zhang Y. Therapeutic potential of targeting stromal crosstalk-mediated immune suppression in pancreatic cancer. Front Oncol. 2021;11:682217.

  11. Szefel J, Danielak A, Kruszewski WJ. Metabolic pathways of L-arginine and therapeutic consequences in tumors. Adv Med Sci. 2019;64(1):104–10.

    Article  PubMed  Google Scholar 

  12. Hu C, Pang B, Lin G, Zhen Y, Yi H. Energy metabolism manipulates the fate and function of tumour myeloid-derived suppressor cells. Br J Cancer. 2020;122(1):23–9.

    Article  CAS  PubMed  Google Scholar 

  13. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Su YL, Banerjee S, White SV, Kortylewski M. STAT3 in tumor-associated myeloid cells: multitasking to disrupt immunity. Int J Mol Sci. 2018;19(6):1803.

  16. Budhwar S, Verma P, Verma R, Rai S, Singh K. The Yin and Yang of myeloid derived suppressor cells. Front Immunol. 2018;9:2776.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Panni RZ, Sanford DE, Belt BA, Mitchem JB, Worley LA, Goetz BD, et al. Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunol Immunother. 2014;63(5):513–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ahmad RS, Eubank TD, Lukomski S, Boone BA. Immune cell modulation of the extracellular matrix contributes to the pathogenesis of pancreatic Cancer. Biomolecules. 2021;11(6):901.

  19. Gorchs L, Kaipe H. Interactions between Cancer-associated fibroblasts and T cells in the pancreatic tumor microenvironment and the role of chemokines. Cancers. 2021;13(12):2995.

  20. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5.

    Article  CAS  PubMed  Google Scholar 

  21. Zychlinsky A, Prevost MC, Sansonetti PJ. Shigella flexneri induces apoptosis in infected macrophages. Nature. 1992;358(6382):167–9.

    Article  CAS  PubMed  Google Scholar 

  22. Kovacs SB, Miao EA. Gasdermins: effectors of Pyroptosis. Trends Cell Biol. 2017;27(9):673–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tan Y, Chen Q, Li X, Zeng Z, Xiong W, Li G, et al. Pyroptosis: a new paradigm of cell death for fighting against cancer. J Experiment Clin Cancer Res. 2021;40(1):153.

    Article  CAS  Google Scholar 

  24. Robinson N, Ganesan R, Hegedus C, Kovacs K, Kufer TA, Virag L. Programmed necrotic cell death of macrophages: focus on pyroptosis, necroptosis, and parthanatos. Redox Biol. 2019;26:101239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang Y, Gao W, Shi X, Ding J, Liu W, He H, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547(7661):99–103.

    Article  CAS  PubMed  Google Scholar 

  26. Zheng Z, Bian Y, Zhang Y, Ren G, Li G. Metformin activates AMPK/SIRT1/NF-κB pathway and induces mitochondrial dysfunction to drive caspase3/GSDME-mediated cancer cell pyroptosis. Cell Cycle (Georgetown, Tex). 2020;19(10):1089–104.

    Article  CAS  Google Scholar 

  27. Chen H, Wu X, Gu Z, Chen S, Zhou X, Zhang Y, et al. Zebrafish gasdermin E cleavage-engaged pyroptosis by inflammatory and apoptotic caspases. Dev Comp Immunol. 2021;124:104203.

    Article  CAS  PubMed  Google Scholar 

  28. Deng W, Bai Y, Deng F, Pan Y, Mei S, Zheng Z, et al. Streptococcal pyrogenic exotoxin B cleaves GSDMA and triggers pyroptosis. Nature. 2022;602(7897):496–502.

    Article  CAS  PubMed  Google Scholar 

  29. Li L, Li Y, Bai Y. Role of GSDMB in Pyroptosis and Cancer. Cancer Manag Res. 2020;12:3033–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhou Z, He H, Wang K, Shi X, Wang Y, Su Y, et al. Granzyme a from cytotoxic lymphocytes cleaves GSDMB to trigger pyroptosis in target cells. Science (New York, NY). 2020;368(6494):eaaz548.

  31. Panganiban RA, Sun M, Dahlin A, Park HR, Kan M, Himes BE, et al. A functional splice variant associated with decreased asthma risk abolishes the ability of gasdermin B to induce epithelial cell pyroptosis. J Allergy Clin Immunol. 2018;142(5):1469–78.e2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hou J, Zhao R, Xia W, Chang CW, You Y, Hsu JM, et al. PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol. 2020;22(10):1264–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang CC, Li CG, Wang YF, Xu LH, He XH, Zeng QZ, et al. Chemotherapeutic paclitaxel and cisplatin differentially induce pyroptosis in A549 lung cancer cells via caspase-3/GSDME activation. Apoptosis. 2019;24(3–4):312–25.

    Article  CAS  PubMed  Google Scholar 

  34. Zhang Z, Zhang Y, Xia S, Kong Q, Li S, Liu X, et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature. 2020;579(7799):415–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Xu L, Hu Y, Liu W. Pyroptosis-mediated molecular subtypes are characterized by distinct tumor microenvironment infiltration characteristics in breast Cancer. J Inflamm Res. 2022;15:345–62.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lu H, Wu J, Liang L, Wang X, Cai H. Identifying a novel defined Pyroptosis-associated long noncoding RNA signature contributes to predicting prognosis and tumor microenvironment of bladder Cancer. Front Immunol. 2022;13:803355.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu A, Shen L, Li N, Shen L, Li Z. Pan-cancer analyses of pyroptosis with functional implications for prognosis and immunotherapy in cancer. J Transl Med. 2022;20(1):109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu Y, Lu Y, Ning B, Su X, Yang B, Dong H, et al. Intravenous delivery of living listeria monocytogenes elicits Gasdmermin-dependent tumor Pyroptosis and motivates anti-tumor immune response. ACS Nano. 2022;16(3):4102–15.

    Article  CAS  PubMed  Google Scholar 

  39. Wang J, Kang Y, Li Y, Sun L, Zhang J, Qian S, et al. Gasdermin D in different subcellular locations predicts diverse progression, immune microenvironment and prognosis in colorectal Cancer. J Inflamm Res. 2021;14:6223–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Croes L, Fransen E, Hylebos M, Buys K, Hermans C, Broeckx G, et al. Determination of the potential tumor-suppressive effects of Gsdme in a chemically induced and in a genetically modified intestinal Cancer mouse model. Cancers. 2019;11(8);1214.

  41. Tan Y, Sun R, Liu L, Yang D, Xiang Q, Li L, et al. Tumor suppressor DRD2 facilitates M1 macrophages and restricts NF-κB signaling to trigger pyroptosis in breast cancer. Theranostics. 2021;11(11):5214–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ju M, Bi J, Wei Q, Jiang L, Guan Q, Zhang M, et al. Pan-cancer analysis of NLRP3 inflammasome with potential implications in prognosis and immunotherapy in human cancer. Brief Bioinform. 2021;22(4):bbaa345.

  43. Guo B, Fu S, Zhang J, Liu B, Li Z. Targeting inflammasome/IL-1 pathways for cancer immunotherapy. Sci Rep. 2016;6:36107.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Liang Q, Wu J, Zhao X, Shen S, Zhu C, Liu T, et al. Establishment of tumor inflammasome clusters with distinct immunogenomic landscape aids immunotherapy. Theranostics. 2021;11(20):9884–903.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Deng Q, Geng Y, Zhao L, Li R, Zhang Z, Li K, et al. NLRP3 inflammasomes in macrophages drive colorectal cancer metastasis to the liver. Cancer Lett. 2019;442:21–30.

    Article  CAS  PubMed  Google Scholar 

  46. Liang M, Chen X, Wang L, Qin L, Wang H, Sun Z, et al. Cancer-derived exosomal TRIM59 regulates macrophage NLRP3 inflammasome activation to promote lung cancer progression. J Experiment Clin Cancer Res. 2020;39(1):176.

    Article  CAS  Google Scholar 

  47. Lee HE, Lee JY, Yang G, Kang HC, Cho YY, Lee HS, et al. Inhibition of NLRP3 inflammasome in tumor microenvironment leads to suppression of metastatic potential of cancer cells. Sci Rep. 2019;9(1):12277.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Ershaid N, Sharon Y, Doron H, Raz Y, Shani O, Cohen N, et al. NLRP3 inflammasome in fibroblasts links tissue damage with inflammation in breast cancer progression and metastasis. Nat Commun. 2019;10(1):4375.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Cai TT, Ye SB, Liu YN, He J, Chen QY, Mai HQ, et al. LMP1-mediated glycolysis induces myeloid-derived suppressor cell expansion in nasopharyngeal carcinoma. PLoS Pathog. 2017;13(7):e1006503.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Wang X, Jia Y, Wen L, Mu W, Wu X, Liu T, et al. Porphyromonas gingivalis promotes colorectal carcinoma by activating the hematopoietic NLRP3 Inflammasome. Cancer Res. 2021;81(10):2745–59.

    Article  CAS  PubMed  Google Scholar 

  51. Li Z, Fu WJ, Chen XQ, Wang S, Deng RS, Tang XP, et al. Autophagy-based unconventional secretion of HMGB1 in glioblastoma promotes chemosensitivity to temozolomide through macrophage M1-like polarization. J Experiment Clin Cancer Res. 2022;41(1):74.

    Article  CAS  Google Scholar 

  52. Penny HL, Sieow JL, Gun SY, Lau MC, Lee B, Tan J, et al. Targeting Glycolysis in Macrophages Confers Protection Against Pancreatic Ductal Adenocarcinoma. Int J Mol Sci. 2021;22(12):6350.

  53. Janowski AM, Colegio OR, Hornick EE, McNiff JM, Martin MD, Badovinac VP, et al. NLRC4 suppresses melanoma tumor progression independently of inflammasome activation. J Clin Invest. 2016;126(10):3917–28.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Kolb R, Phan L, Borcherding N, Liu Y, Yuan F, Janowski AM, et al. Obesity-associated NLRC4 inflammasome activation drives breast cancer progression. Nat Commun. 2016;7:13007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ohashi K, Wang Z, Yang YM, Billet S, Tu W, Pimienta M, et al. NOD-like receptor C4 Inflammasome regulates the growth of Colon Cancer liver metastasis in NAFLD. Hepatol. 2019;70(5):1582–99.

    Article  CAS  Google Scholar 

  56. Fitzgerald AA, Wang S, Agarwal V, Marcisak EF, Zuo A, Jablonski SA, et al. DPP inhibition alters the CXCR3 axis and enhances NK and CD8+ T cell infiltration to improve anti-PD1 efficacy in murine models of pancreatic ductal adenocarcinoma. J Immunother Cancer. 2021;9(11):e002837.

  57. Li R, Zeng X, Yang M, Feng J, Xu X, Bao L, et al. Antidiabetic DPP-4 inhibitors reprogram tumor microenvironment that facilitates murine breast Cancer metastasis through interaction with Cancer cells via a ROS-NF-кB-NLRP3 Axis. Front Oncol. 2021;11:728047.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Fukuda K, Okamura K, Riding RL, Fan X, Afshari K, Haddadi NS, et al. AIM2 regulates anti-tumor immunity and is a viable therapeutic target for melanoma. J Exp Med. 2021;218(9):e2020962.

  59. Ahechu P, Zozaya G, Martí P, Hernández-Lizoáin JL, Baixauli J, Unamuno X, et al. NLRP3 Inflammasome: a possible link between obesity-associated low-grade chronic inflammation and colorectal Cancer development. Front Immunol. 2018;9:2918.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bent R, Moll L, Grabbe S, Bros M. Interleukin-1 Beta-A Friend or Foe in Malignancies? Int J Mol Sci. 2018;19(8):2155.

  61. Chen JX, Cheng CS, Gao HF, Chen ZJ, Lv LL, Xu JY, et al. Overexpression of interferon-inducible protein 16 promotes progression of human pancreatic adenocarcinoma through interleukin-1β-induced tumor-associated macrophage infiltration in the tumor microenvironment. Front Cell Develop Biol. 2021;9:640786.

    Article  Google Scholar 

  62. Das S, Shapiro B, Vucic EA, Vogt S, Bar-Sagi D. Tumor cell-derived IL1β promotes Desmoplasia and immune suppression in pancreatic Cancer. Cancer Res. 2020;80(5):1088–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Bunt SK, Sinha P, Clements VK, Leips J, Ostrand-Rosenberg S. Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression. J Immunol. 2006;176(1):284–90.

  64. Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67(9):4507–13.

    Article  CAS  PubMed  Google Scholar 

  65. Rolim GB, Dantas Lima AJP, Dos Santos Cardoso VI, de Fátima Machado Soares É, Nunes DN, Barros HCS, et al. Can inflammasomes promote the pathophysiology of glioblastoma multiforme? A view about the potential of the anti-inflammasome therapy as pharmacological target. Crit Rev Oncol Hematol 2022;172:103641.

  66. Takeda K, Tsutsui H, Yoshimoto T, Adachi O, Yoshida N, Kishimoto T, et al. Defective NK cell activity and Th1 response in IL-18-deficient mice. Immunity. 1998;8(3):383–90.

    Article  CAS  PubMed  Google Scholar 

  67. Tominaga K, Yoshimoto T, Torigoe K, Kurimoto M, Matsui K, Hada T, et al. IL-12 synergizes with IL-18 or IL-1beta for IFN-gamma production from human T cells. Int Immunol. 2000;12(2):151–60.

    Article  CAS  PubMed  Google Scholar 

  68. Senju H, Kumagai A, Nakamura Y, Yamaguchi H, Nakatomi K, Fukami S, et al. Effect of IL-18 on the expansion and phenotype of human natural killer cells: application to Cancer immunotherapy. Int J Biol Sci. 2018;14(3):331–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Okamoto I, Kohno K, Tanimoto T, Ikegami H, Kurimoto M. Development of CD8+ effector T cells is differentially regulated by IL-18 and IL-12. J Immunol. 1999;162(6):3202–11.

  70. Timperi E, Focaccetti C, Gallerano D, Panetta M, Spada S, Gallo E, et al. IL-18 receptor marks functional CD8(+) T cells in non-small cell lung cancer. Oncoimmunol. 2017;6(7):e1328337.

    Article  Google Scholar 

  71. Markowitz GJ, Yang P, Fu J, Michelotti GA, Chen R, Sui J, et al. Inflammation-dependent IL18 signaling restricts hepatocellular carcinoma growth by enhancing the accumulation and activity of tumor-infiltrating lymphocytes. Cancer Res. 2016;76(8):2394–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lim HX, Hong HJ, Cho D, Kim TS. IL-18 enhances immunosuppressive responses by promoting differentiation into monocytic myeloid-derived suppressor cells. J Immunol (Baltimore, Md : 1950). 2014;193(11):5453–60.

  73. Nakamura K, Kassem S, Cleynen A, Chrétien ML, Guillerey C, Putz EM, 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(4):634–48.e5.

    Article  CAS  PubMed  Google Scholar 

  74. Liou AK, Soon G, Tan L, Peng Y, Cher BM, Goh BC, et al. Elevated IL18 levels in nasopharyngeal carcinoma induced PD-1 expression on NK cells in TILS leading to poor prognosis. Oral Oncol. 2020;104:104616.

    Article  CAS  PubMed  Google Scholar 

  75. Li K, Wei L, Huang Y, Wu Y, Su M, Pang X, et al. Leptin promotes breast cancer cell migration and invasion via IL-18 expression and secretion. Int J Oncol. 2016;48(6):2479–87.

    Article  CAS  PubMed  Google Scholar 

  76. Fung KY, Nguyen PM, Putoczki TL. Emerging roles for Interleukin-18 in the gastrointestinal tumor microenvironment. Adv Exp Med Biol. 2020;1240:59–72.

    Article  CAS  PubMed  Google Scholar 

  77. Kiss M, Vande Walle L, Saavedra PHV, Lebegge E, Van Damme H, Murgaski A, et al. IL1β promotes immune suppression in the tumor microenvironment independent of the Inflammasome and Gasdermin D. Cancer. Immunol Res. 2021;9(3):309–323.

  78. Su X, Wang WJ, Cao Q, Zhang H, Liu B, Ling Y, et al. A carbonic anhydrase IX (CAIX)-anchored rhenium(I) photosensitizer evokes Pyroptosis for enhanced anti-tumor immunity. Angew Chem Int Ed Engl. 2022;61(8):e202115800.

    CAS  PubMed  Google Scholar 

  79. Lu Y, Xu F, Wang Y, Shi C, Sha Y, He G, et al. Cancer immunogenic cell death via photo-pyroptosis with light-sensitive Indoleamine 2,3-dioxygenase inhibitor conjugate. Biomaterials. 2021;278:121167.

    Article  CAS  PubMed  Google Scholar 

  80. Xiao Y, Zhang T, Ma X, Yang QC, Yang LL, Yang SC, et al. Microenvironment-responsive Prodrug-induced Pyroptosis boosts Cancer immunotherapy. Adv Sci (Weinh). 2021;8(24):e2101840.

    Article  CAS  Google Scholar 

  81. Zhang L, Yang QC, Wang S, Xiao Y, Wan SC, Deng H, et al. Engineering multienzyme-mimicking covalent organic frameworks as Pyroptosis inducers for boosting antitumor immunity. Adv Mater. 2021;34(13):e2108174.

  82. Erkes DA, Cai W, Sanchez IM, Purwin TJ, Rogers C, Field CO, et al. Mutant BRAF and MEK inhibitors regulate the tumor immune microenvironment via Pyroptosis. Cancer Discovery. 2020;10(2):254–69.

    Article  CAS  PubMed  Google Scholar 

  83. Xu S, Li X, Liu Y, Xia Y, Chang R, Zhang C. Inflammasome inhibitors: promising therapeutic approaches against cancer. J Hematol Oncol. 2019;12(1):64.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Fourie C, Shridas P, Davis T, de Villiers WJS, Engelbrecht AM. Serum amyloid a and inflammasome activation: a link to breast cancer progression? Cytokine Growth Factor Rev. 2021;59:62–70.

    Article  CAS  PubMed  Google Scholar 

  85. Lee SM, Rudd R, Woll PJ, Ottensmeier C, Gilligan D, Price A, et al. Randomized double-blind placebo-controlled trial of thalidomide in combination with gemcitabine and carboplatin in advanced non-small-cell lung cancer. J Clin Oncol. 2009;27(31):5248–54.

    Article  CAS  PubMed  Google Scholar 

  86. Gilbert SM, Gidley Baird A, Glazer S, Barden JA, Glazer A, Teh LC, et al. A phase I clinical trial demonstrates that nfP2X(7) -targeted antibodies provide a novel, safe and tolerable topical therapy for basal cell carcinoma. Br J Dermatol. 2017;177(1):117–24.

    Article  CAS  PubMed  Google Scholar 

  87. Hortelano S, González-Cofrade L, Cuadrado I, de Las HB. Current status of terpenoids as inflammasome inhibitors. Biochem Pharmacol. 2020;172:113739.

    Article  CAS  PubMed  Google Scholar 

  88. Chen L, Huang CF, Li YC, Deng WW, Mao L, Wu L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma. Cell Mol Life Sci. 2018;75(11):2045–58.

    Article  CAS  PubMed  Google Scholar 

  89. Theivanthiran B, Evans KS, DeVito NC, Plebanek M, Sturdivant M, Wachsmuth LP, et al. A tumor-intrinsic PD-L1/NLRP3 inflammasome signaling pathway drives resistance to anti-PD-1 immunotherapy. J Clin Invest. 2020;130(5):2570–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Lu F, Zhao Y, Pang Y, Ji M, Sun Y, Wang H, et al. NLRP3 inflammasome upregulates PD-L1 expression and contributes to immune suppression in lymphoma. Cancer Lett. 2021;497:178–89.

    Article  CAS  PubMed  Google Scholar 

  91. Kaplanov I, Carmi Y, Kornetsky R, Shemesh A, Shurin GV, Shurin MR, et al. Blocking IL-1β reverses the immunosuppression in mouse breast cancer and synergizes with anti-PD-1 for tumor abrogation. Proc Natl Acad Sci U S A. 2019;116(4):1361–9.

    Article  CAS  PubMed  Google Scholar 

  92. Pretre V, Papadopoulos D, Regard J, Pelletier M, Woo J. Interleukin-1 (IL-1) and the inflammasome in cancer. Cytokine. 2022;153:155850.

    Article  CAS  PubMed  Google Scholar 

  93. Ma Z, Li W, Yoshiya S, Xu Y, Hata M, El-Darawish Y, et al. Augmentation of immune checkpoint Cancer immunotherapy with IL18. Clin Cancer Res. 2016;22(12):2969–80.

    Article  CAS  PubMed  Google Scholar 

  94. Son YI, Dallal RM, Lotze MT. Combined treatment with interleukin-18 and low-dose interleukin-2 induced regression of a murine sarcoma and memory response. J Immunother. 2003;26(3):234–40.

    Article  CAS  PubMed  Google Scholar 

  95. Tarhini AA, Millward M, Mainwaring P, Kefford R, Logan T, Pavlick A, et al. A phase 2, randomized study of SB-485232, rhIL-18, in patients with previously untreated metastatic melanoma. Cancer. 2009;115(4):859–68.

    Article  CAS  PubMed  Google Scholar 

  96. Saetang J, Chonpathompikunlert P, Sretrirutchai S, Roongsawang N, Kayasut K, Voravuthikunchai SP, et al. Anti-cancer effect of engineered recombinant interleukin 18. Adv Clin Exp Med. 2020;29(10):1135–43.

    Article  PubMed  Google Scholar 

  97. Drakes DJ, Rafiq S, Purdon TJ, Lopez AV, Chandran SS, Klebanoff CA, et al. Optimization of T-cell receptor-modified T cells for Cancer therapy. Cancer Immunol Res. 2020;8(6):743–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. 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(1):e1378842.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zhou T, Damsky W, Weizman OE, McGeary MK, Hartmann KP, Rosen CE, et al. IL-18BP is a secreted immune checkpoint and barrier to IL-18 immunotherapy. Nature. 2020;583(7817):609–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Gumber D, Wang LD. Improving CAR-T immunotherapy: overcoming the challenges of T cell exhaustion. EBioMedicine. 2022;77:103941.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Avanzi MP, Yeku O, Li X, Wijewarnasuriya DP, van Leeuwen DG, Cheung K, et al. Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep. 2018;23(7):2130–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. 2010;207(5):1045–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Uzan B, Poglio S, Gerby B, Wu CL, Gross J, Armstrong F, et al. Interleukin-18 produced by bone marrow-derived stromal cells supports T-cell acute leukaemia progression. EMBO Mol Med. 2014;6(6):821–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Voloshin T, Alishekevitz D, Kaneti L, Miller V, Isakov E, Kaplanov I, et al. Blocking IL1β pathway following paclitaxel chemotherapy slightly inhibits primary tumor growth but promotes spontaneous metastasis. Mol Cancer Ther. 2015;14(6):1385–94.

    Article  CAS  PubMed  Google Scholar 

  105. Guo W, Sun Y, Liu W, Wu X, Guo L, Cai P, et al. Small molecule-driven mitophagy-mediated NLRP3 inflammasome inhibition is responsible for the prevention of colitis-associated cancer. Autophagy. 2014;10(6):972–85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Cai J, Yi M, Tan Y, Li X, Li G, Zeng Z, et al. Natural product triptolide induces GSDME-mediated pyroptosis in head and neck cancer through suppressing mitochondrial hexokinase-ΙΙ. J Experiment Clin Cancer Res. 2021;40(1):190.

    Article  CAS  Google Scholar 

  107. Zhang F, Liu Q, Ganesan K, Kewu Z, Shen J, Gang F, et al. The Antitriple negative breast cancer efficacy of Spatholobus suberectus Dunn on ROS-induced noncanonical Inflammasome Pyroptotic pathway. Oxidative Med Cell Longev. 2021;2021:5187569.

    Google Scholar 

  108. Li Q, Chen L, Dong Z, Zhao Y, Deng H, Wu J, et al. Piperlongumine analogue L50377 induces pyroptosis via ROS mediated NF-κB suppression in non-small-cell lung cancer. Chem Biol Interact. 2019;313:108820.

    Article  CAS  PubMed  Google Scholar 

  109. Wang L, Li K, Lin X, Yao Z, Wang S, Xiong X, et al. Metformin induces human esophageal carcinoma cell pyroptosis by targeting the miR-497/PELP1 axis. Cancer Lett. 2019;450:22–31.

    Article  CAS  PubMed  Google Scholar 

  110. Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 2017;552(7684):194–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Guo W, Chen S, Li C, Xu J, Wang L. Application of Disulfiram and its metabolites in treatment of inflammatory disorders. Front Pharmacol. 2021;12:795078.

    Article  CAS  PubMed  Google Scholar 

  112. Hu JJ, Liu X, Xia S, Zhang Z, Zhang Y, Zhao J, et al. FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat Immunol. 2020;21(7):736–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Gabrielle White Wolf, PhD, from Liwen Bianji (Edanz) (www.liwenbianji.cn) for editing the English text of a draft of this manuscript.

Funding

This study was supported by grants from the National Natural Science Foundation of China (81502051, 81800025), the Shandong Provincial Natural Science Foundation, China (ZR2020MH256), Medical Health Science and Technology Project of Shandong Provincial Health Commission (2019WS386), Basic Cancer Research Program Supported by Bethune Charitable Foundation (BCF-NH-ZL-20201119-009).

Author information

Authors and Affiliations

Authors

Contributions

JWX proposed and designed the study. JXW, LW, and JWX wrote the draft, and JXW and LW collected the data. All authors contributed to the design and interpretation of the study and to further drafts. JWX and JXW revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jianwei Xu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, J., Wang, L. & Xu, J. The role of pyroptosis in modulating the tumor immune microenvironment. Biomark Res 10, 45 (2022). https://doi.org/10.1186/s40364-022-00391-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-022-00391-3

Keywords