- Review
- Open access
- Published:
Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication
Molecular Cancer volume 23, Article number: 268 (2024)
Abstract
In the realm of cancer research, the tumor microenvironment (TME) plays a crucial role in tumor initiation and progression, shaped by complex interactions between cancer cells and surrounding non-cancerous cells. Cytokines, as essential immunomodulatory agents, are secreted by various cellular constituents within the TME, including immune cells, cancer-associated fibroblasts, and cancer cells themselves. These cytokines facilitate intricate communication networks that significantly influence tumor initiation, progression, metastasis, and immune suppression. Pyroptosis contributes to TME remodeling by promoting the release of pro-inflammatory cytokines and sustaining chronic inflammation, impacting processes such as immune escape and angiogenesis. However, challenges remain due to the complex interplay among cytokines, pyroptosis, and the TME, along with the dual effects of pyroptosis on cancer progression and therapy-related complications like cytokine release syndrome. Unraveling these complexities could facilitate strategies that balance inflammatory responses while minimizing tissue damage during therapy. This review delves into the complex crosstalk between cytokines, pyroptosis, and the TME, elucidating their contribution to tumor progression and metastasis. By synthesizing emerging therapeutic targets and innovative technologies concerning TME, this review aims to provide novel insights that could enhance treatment outcomes for cancer patients.
Background
Cancer represents a spectrum of diseases characterized by uncontrolled cell proliferation and is influenced by a multifaceted interplay of genetic, environmental, and microenvironmental factors [1, 2]. In tumor research, the primary focus has traditionally been on the intrinsic properties of cancer cells, such as their proliferation, apoptosis, and mechanisms of drug resistance [3, 4]. However, an increasing body of evidence underscores the necessity of viewing cancer as an intricate evolutionary and ecological process that encompasses extensive interactions between cancer cells and the tumor microenvironment (TME) [5, 6]. The TME constitutes a dynamic and supportive milieu comprising various cellular and non-cellular components, including stromal cells like cancer-associated fibroblasts (CAFs), immune cells (e.g., macrophages and neutrophils), endothelial cells, and the extracellular matrix (ECM) [7, 8]. This complex landscape facilitates sophisticated communications between cancer and non-cancerous cells through the release of soluble factors, such as cytokines and chemokines, as well as various signaling molecules and ECM components [9, 10].
Cytokines, produced by diverse cellular constituents within the TME, play a pivotal role in regulating tumor initiation, progression, and metastasis [10, 11]. For instance, CAFs release various cytokines (e.g. IL6), chemokines (e.g. CXCL12), and growth factors (e.g. LIF) that contribute to the sustenance of pro-tumor microenvironment [12,13,14]. Tumor-associated macrophages (TAMs) serve as a significant source of cytokines and chemokines and play a critical role in the initiation and perpetuation of chronic inflammation, which is closely associated with tumorigenesis and tumor progression [15]. TAMs secrete pro-inflammatory cytokines such as IL-1β, IL-6, and IL-23, thereby promoting tumor growth and progression in colorectal cancer and other malignancies [16,17,18,19]. Additionally, TAMs facilitate the angiogenic switch by releasing key pro-angiogenic factors, including vascular endothelial growth factors (VEGF) and IL-8, which enhance the recruitment and activation of endothelial cells and other cells that support the development of vascular networks [20,21,22]. Furthermore, TAMs contribute to adaptive resistance to targeted cancer therapies. For example, TAM-derived TNF-α, acting as a crucial melanoma growth factor, induces resistance to MAPK pathway inhibitors [23]. Thus, understanding the intricate crosstalk between cytokines and other signaling molecules is crucial for uncovering immune-related mechanisms of cancer development. This knowledge may lead to novel therapeutic strategies targeting the TME to disrupt tumor-promoting signaling pathways and bolster anti-tumor immunity.
In multicellular organisms, maintaining a delicate balance between cell proliferation and cell death is crucial. Regulation imbalance may result in cellular replicative immortality, hence subsequently leading to tumor formation, progression, and even therapeutic interventions [24]. In these intricate processes, programmed cell death (PCD) assumes a critical role in maintaining the internal balance [25]. Apoptosis, the most extensively studied modality of PCD, serves as a natural barrier against tumors; however, the emergence of chemotherapy resistance limits the efficacy of traditional therapies that rely solely on the induction of apoptosis [26]. Thus, in addressing apoptosis resistance, exploring novel therapeutic strategies that target non-apoptotic forms of PCD may offer effective alternatives for cancer treatment. Pyroptosis is a form of inflammatory PCD that is activated by inflammasomes. It is characterized by the cleavage of gasdermin family proteins, leading to the release of cytokines such as IL-1β and IL-18 [27, 28]. Recent years have witnessed promising descriptions of novel modes of cell death, such as cuproptosis [29], disulfidptosis [30], and ammonia-induced cell death [31]. Nevertheless, their roles within the TME and their potential clinical applications require further research and substantiation. Over the past few decades, various forms of PCD have been extensively investigated, including necroptosis, ferroptosis, and pyroptosis [32,33,34]. Compared with necroptosis and ferroptosis, pyroptosis is a more prevalent mechanism of immune defense that is intricately linked to immune cell infiltration into the TME across various cancers [25]. Additionally, the diversity of the gasdermin family and the complexity of inflammasomes contribute to a multifaceted regulatory network governing the pyroptosis pathway and its associated cytokines. Notably, in spite of the positive role of pyroptosis in TME, several studies have reported cytokine release syndrome, a severe side effect resulting from an exaggerated inflammatory response mediated by pyroptosis [35, 36]. Therefore, summarizing the precise mechanisms and elucidating the intricate interactions between pyroptosis and cytokines in the tumor microenvironment are significant for the development of more effective anticancer therapies.
The interplay between pyroptosis and cytokine signaling within the TME forms a complex network characterized by numerous pathways and feedback loops. However, the underlying molecular mechanism remains unclear. Pyroptosis is known to amplify inflammation and alter the TME, enhancing cytokine expression and consequently promoting cancer cell invasion and metastasis [37, 38]. Elevated levels of inflammatory cytokines have been associated with poor prognosis in cancer patients, underscoring their potential as valuable prognostic markers [10, 39]. However, the relationship between pyroptosis, cytokines, and tumor growth is multifaceted; while pyroptosis can promote tumor-promoting inflammation and enhance cancer cell invasiveness, it can also stimulate anti-tumor immune responses that inhibit tumor progression [40, 41]. Recent research has highlighted the potential of therapeutic strategies aimed at inducing pyroptosis within tumor cells. By triggering this form of cell death, it is possible to activate the immune system and bolster anti-tumor responses, thus providing a promising avenue for cancer therapy [42]. The intersection of pyroptosis and cytokine dynamics presents opportunities for innovative treatments that could modulate inflammation and immune response in the TME, potentially overcoming limitations associated with current therapies focused on apoptosis.
In this review, we provide a succinct overview of the fundamental principles and characteristics of cytokines and pyroptosis, as well as highlight their respective functions and recent advancements in the TME. Furthermore, we extensively discuss and elucidate the intricate crosstalk and connections between cytokines and pyroptosis within the TME, delving into the complex molecular mechanisms that regulate networks critical to tumor occurrence, metastasis, immune evasion, and angiogenesis. By synthesizing and summarizing emerging therapeutic targets and novel technologies (e.g. single-cell sequencing and spatial transcriptomics) in anti-tumor therapy, we hope to present novel insights that could drive future research and clinical applications aimed at improving cancer treatment outcomes in the context of tumor inflammation and immunobiology.
Definition and mechanism of pyroptosis
Pyroptosis is a notably inflammatory form of lytic PCD which has a key role in innate immunity and tumor development [34, 43, 44]. The term “pyroptosis” was initially proposed by Cookson and Brennan in 2001 to characterize this distinct process [45]. Originating from Greek roots, “pyroptosis” combines “Pyro,” meaning fire, which signifies the inflammatory nature of this process, and “Ptosis,” meaning to “fall off,” commonly used as a suffix in cell death terminology to illustrate the falling off or dying of cells [46, 47]. Triggered by various inflammatory signals, pyroptosis exhibits both shared traits with other types of PCDs and unique characteristics like pro-inflammatory cytokines and the formation of gasdermin protein pores in the cell membrane that set it apart [48,49,50]. The role of diverse cytokines and related signal pathways in different PCDs were summarized (Table 1).
Various inflammasomes, triggered by stimuli originating from the extracellular or intracellular environment, serve as crucial platforms for the subsequent activation of various caspases, thereby initiating or executing cellular processes [63]. The pyroptosis pathways are distinguished by the involvement of different caspases, notably including caspase-1-dependent pathway, caspase-4/5/11-dependent pathway, and other pathways like caspase-3-dependent pathway, caspase-8-dependent pathway, caspase-free pathways, etc. (Fig. 1).
Summary of the mechanism of pyroptosis. In caspase-1-dependent pathway, the inflammasome sensors triggered by DAMPs and/or PAMPs could activate caspase-1. Subsequently, a cascade of events like the cleavage of GSDMD, maturation of pro-IL-1β and pro-IL-18, release of pro-inflammation cytokines, formation of GSDMD pores and ultimately cell membrane rupture. In caspase-4/5/11-dependent pathway, inflammasome sensors can be directly activated by LPS, which is also a GSDMD-dependent pyroptotic pathway. In other pathways, caspase-3 triggers pyroptosis via GSDME while caspase-8 initiates pyroptosis via GSDMC. Additionally, pyroptosis can be activated without caspase family. CD8 + T cells and NK cells can secrete granzyme A and cause pyroptosis mediated by GSDMB, while release of granzyme B could induce pyroptosis via GSDME
Caspase-1-dependent pathway
Caspase-1-dependent pathway is also named as canonical pyroptotic death, which is orchestrated by the assembly of inflammasomes and characterized by caspase 1 [64]. Upon recognition of PAMPs or DAMPs by classical inflammasome sensors (NLRs, AIM2, P2 × 7R, and pyrin), the inflammasomes undergo an automatic assembly process [65, 66]. This assembly event triggers the activation of pro-caspase-1, leading to its self-cleavage and the subsequent formation of the active p10/p20 heterotetramer [67, 68]. Upon activation, pro-caspase-1 initiates a cascade of events that include the cleavage of gasdermin D (GSDMD), resulting in the release of the functional gasdermin N-terminal fragment. Concurrently, pro-IL-1β and pro-IL-18, which are dormant precursors of the pro-inflammatory cytokine IL-1β and IL-18, are processed into their mature, secretory forms. The gasdermin N-terminal fragment translocates to the cell membrane where it interacts with acidic lipids, leading to the formation of gasdermin pores with a specific inner diameter of 10–14 nm [69]. These pores, characterized by a negative charge, play a crucial role in the selective release of IL-1β and IL-18 through electrostatic filtering mechanisms [70, 71]. The culmination of these processes ultimately results in cell membrane rupture and the induction of pyroptosis.
Caspase-4/5/11-dependent pathway
As opposed to the canonical pyroptosis featured by caspase 1, the pathway initiated by caspase-4/5 in human and caspase-11 in mice is known as a noncanonical pyroptotic pathway [72, 73]. In this alternative pathway, the inflammasome sensors can be directly activated by intracellular bacteria and lipopolysaccharide (LPS), subsequently activating caspase-4/5/11 [74, 75]. Specifically, caspase-4/5/11 have the ability to directly interact with lipid A present in the outer membrane of Gram-negative bacteria, resulting in their oligomerization and subsequent activation [76,77,78]. Following activation, these caspases cleave GSDMD to release the gasdermin N-terminal p30 fragment, which has the capability to form pores leading to pyroptotic cell death [79, 80]. It is important to note that unlike the canonical pathway, caspase-4/5/11 pathways are involved in the maturation and secretion of pro-inflammatory cytokines such as IL-1β and IL-18 without directly cleaving them [81]. This distinction sets the noncanonical pathway apart from the canonical pathway in terms of its mechanism of action and downstream effects.
Other pathways
Apart from caspase-1-dependent and caspase-4/5/11-dependent pathways, pyroptosis can be triggered by caspase-3, caspase-8, and even caspase-independent pathways. Caspase-3 typically known as the executioner caspase in apoptosis [82], can cleave GSDME to release the gasdermin N-terminal that triggers pyroptosis when GSDME is highly expressed or caspase-3 is stimulated by chemotherapy drugs via the BAK/BAX-caspase-3-GSDME pathway [83]. Notably, tissue cells usually have higher expression of GSDME compared to most cancer cells, which might explain a range of chemotherapy-induced adverse effects such as inflammation and tissue damage [84, 85]. As a cell-permeable analog of α-KG, DM-αKG can activate caspase-8 in HeLa and other cancer cell lines [86]. Afterwards, the activated caspase-8 initiates the cleavage of gasdermin C (GSDMC), which subsequently ensembles and forms pores in the cell membrane [86]. This phenomenon is also observed under hypoxia conditions, PD-L1 in the nucleus together with p-Stat3 co-upregulate the expression of GSDMC, thus eventually causing pyroptosis [87]. Intriguingly, pyroptosis can be triggered without caspase family. In a study of cytokine release syndrome (CRS), CAR T cells release perforin to form pores and rapidly activate caspase 3 in target cells through the entry of granzyme B. This in turn cleaves GSDME, leading to extensive pyroptosis [35, 88]. In contrast, CD8 + T cells and NK cells can secrete granzyme A and cause pyroptosis mediated by gasdermin B (GSDMB) [89, 90].
Overview of pyroptosis and cytokine in TME
Pyroptosis is characterized by the release of pro-inflammatory cytokines such as IL-1β and IL-18 [91, 92], alongside the liberation of damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) from expiring cells [93,94,95]. Within the TME, the pro-inflammatory cytokines emanating from pyroptotic cells play a pivotal role in fomenting inflammation and fostering the infiltration of immune cells, thereby potentially advancing tumor progression and metastasis [96,97,98]. Concurrently, the release of DAMPs from pyroptotic cells serves to activate immune cells and bolster the recruitment of immune cells to the TME, culminating in the initiation of anti-tumor immune responses and the suppression of tumor growth and metastasis [99, 100]. Consequently, the involvement of pyroptosis in TME restructuring and metastasis is intricate and multifaceted. The ensuing discourse will expound upon three focal points: an overview of the TME, the impact of pyroptosis on TME remodeling and metastasis, and the influence of cytokines on TME remodeling and metastasis.
Definition and composition of TME
The TME encompasses non-cancerous cells and their associated components within tumors, including the molecules they generate and release. Cancer cells directly engage with the TME and interact with non-cancerous elements, including infiltrating immune cells like TANs and TAMs, CAFs, and the extracellular matrix [101, 102]. These interactions are facilitated by signaling molecules such as cytokines and chemokines. The persistent interaction between tumor cells and the TME significantly influences the initiation, progression, metastasis, and response to treatment of tumors [103]. As a result, the TME has garnered significant research and clinical interest as a potential therapeutic target for tumors [104].
Among the diverse stromal cell populations within the TME, CAFs have been identified as the most abundant cell type [105]. The pro-tumor activities of CAFs have been extensively documented in various cancer types, including prostate cancer, breast cancer, pancreatic cancer, and colorectal cancer [13, 106,107,108,109]. TAMs, which are prevalent in the TME across different cancer types, have consistently been associated with unfavorable clinical outcomes in cancer patients [110, 111]. TANs represent a critical component of the TME and actively contribute to tumor progression and metastasis. In addition to secreting proinflammatory cytokines and chemokines, TANs also produce immunosuppressive factors such as arginase 1 and TGFβ, which effectively suppress adaptive immunity [112].
Researchers are dedicated to gaining a deeper understanding of the critical role played by the TME in tumor development and treatment resistance. By focusing on TME components, therapeutic benefits for cancer patients can be achieved. However, the successful implementation of this strategy necessitates a comprehensive understanding of the molecular and cellular distinctions between tumor-promoting host cells and normal host cells within the TME. By discerning these differences, it becomes possible to identify specific targets within the TME that can be effectively manipulated to impede tumor progression and enhance treatment outcomes. This knowledge is crucial for the development of tailored therapeutic approaches that selectively target the TME, ultimately benefiting cancer patients.
The TME exerts a significant influence on immune cell responses, activation, differentiation, and cytokine secretion. It can either enhance the pro- or anti-tumor response, mediate inflammation, or contribute to oncogenesis depending on the interplay of cytokines. In the context of solid tumors, including breast cancer, the presence of diverse cell populations within the TME leads to intricate networks of interactions mediated by a variety of cytokines. Pyroptosis, a form of programmed cell death, can trigger the release of cytokines, facilitate the activation of macrophages and T lymphocytes, elicit a robust inflammatory response within the body, and induce immune phagocytosis [48, 113]. Furthermore, specific cytokines have been identified to regulate the expression and activation of crucial pyroptosis-related proteins, such as gasdermins [38]. This reciprocal relationship between pyroptosis and cytokine signaling transduction highlights the interconnectedness and complexity of these processes within the TME. Understanding this crosstalk is crucial for unraveling the mechanisms underlying cancer progression and developing targeted therapeutic approaches that exploit the interplay between pyroptosis and cytokine signaling.
Pyroptosis in TME remodeling and metastasis
Inducing factors of pyroptosis in TME
The TME is a complex circumstance composed of surrounding blood vessels, extracellular matrix (ECM), an array of signaling molecules and a variety of cells like immune cells, fibroblasts and so on [114,115,116]. Within this intricate landscape, inducing factors of pyroptosis in the TME mainly encompass inflammasome activation, pro-inflammatory cytokines, hypoxia, and therapy-related inducing factors (Fig. 2).
Inflammasome activation
Inflammasomes, acting as receptors or sensors within the innate immune system, manage the activation of caspase-1 and stimulate inflammation in response to infectious microbes and molecules derived from host proteins [117]. Pattern Recognition Receptors (PRRs) are a type of immune receptor that identify PAMPs or DAMPs [118]. The stimulation of various PRRs can lead to the formation of inflammasomes within the TME. Activated PRRs promote the downstream signaling pathway, and cause type I interferons generation and pro-inflammatory cytokines release [119, 120].
So far, five PRRs (NLRP1, NLRP3, NLRC4, Pyrin, and AIM2) have been identified to form inflammasomes [65, 121]. In TME, NLRP1 has been implicated in promoting melanoma tumorigenesis by activating the inflammasome through caspase-1, while simultaneously inhibiting mitochondrial apoptosis associated with caspase-2 and caspase-9 [122]. Likewise, without NLRP3, the quantity of activated NK cells increased, leading to more IFN-γ secretion and enhanced tumor cell destruction, thereby reducing B16F10 lung metastasis [123, 124]. The activation of the NLRP3 inflammasome, induced by E2, can also initiate pyroptosis and impede autophagy in HCC cells [125]. The pro-tumor effect of microbes can be partially ascribed to the activation of inflammasomes and the subsequent stimulation of the IL-1β/NF-κB/IL-6/signal transducer and activator of transcription 3 (STAT3) pathways [126]. LPS can trigger non-standard inflammasome caspase-11-mediated pyroptosis in lung cancer cells [127]. Also, simvastatin provokes pyroptosis in A549 and H1299 cells by stimulating the NLRP3 pathway [128].
Pro-inflammatory cytokines
The presence of pro-inflammatory cytokines within the TME can trigger pyroptosis in tumor cells. Tumor necrosis factor-α (TNF-α) is predominantly secreted by immune cells like macrophages, T cells, and natural killer cells upon encountering tumor cells or other activating signals [129, 130]. Studies have demonstrated that TNF-α can initiate the caspase-8-dependent pyroptotic pathway, leading to the cleavage of GSDMD and subsequent cellular swelling, lysis, and rupture [131,132,133]. Furthermore, IL-1β and IL-18, crucial components of pro-inflammatory cytokines, are downstream effectors of caspase-1 in the canonical pyroptotic pathway [134,135,136]. They significantly contribute to the peripheral inflammatory response associated with dying cells.
Hypoxia
TME usually presents as hypoxic due to rapid tumor growth [137]. In hypoxic conditions, phosphorylated Stat3 physically associates with PD-L1, aiding its movement into the nucleus and boosting the transcription of GSDMC gene [87]. Upon treatment with TNFα, GSDMC is specifically cleaved by caspase-8, producing a GSDMC N-terminal domain [86, 87]. This domain forms pores on the cell membrane, leading to the induction of pyrop.
Therapy-related inducing factors
Novel drug therapies can variously induce pyroptosis. ZIF-8 nanoparticles (NPs) can intrinsically induce pyroptosis by a caspase-1/GSDMD-dependent pathway [138]. Chemotherapeutic paclitaxel and cisplatin differentially induce pyroptosis in A549 lung cancer cells via caspase-3/GSDME activation [139].
The inducing factors of pyroptosis in tumor microenvironment. Inflammasome activation, pro-inflammatory cytokines, hypoxia, and drug therapies can initiate pyroptosis in the TME. PRRs identify PAMPs or DAMPs and further trigger the formation of inflammasomes within the TME. Eventually, pro-inflammatory cytokines release and pyroptosis occurs. Hypoxia in TME promotes the GSDMC-dependent pyroptosis with PD-L1 and phosphorylated Stat3. Diverse drug therapies can induce pyroptosis in multi-forms. Pro-inflammatory cytokines secreted by immune cells and released by pyroptotic tumor cells may cause peripheral inflammation, which may further recruit and activate more immune cells. Additionally, immune cells may directly kill tumor cells, thus enhancing the peripheral inflammation
Role of pyroptosis in TME remodeling and metastasis
Pyroptosis serves as a pivotal mechanism in TME remodeling and metastasis through the facilitation of inflammation, activation of immune cells, and regulation of tumor cells [32, 41, 51]. The release of cytokines such as IL-1β and IL-18 via the pyroptosis pathway not only triggers inflammation and recruits immune cells but also potentially fosters tumor progression and metastasis [97, 140]. Furthermore, the discharge of DAMPs from pyroptotic cells can activate immune cells, thereby intensifying the inflammatory response and establishing a positive feedback loop [141]. This part will delve into the impact of pyroptosis in direct killing effect on tumor cells, regulation of immune cells, and the impact on tumor angiogenesis, metabolism, and metastasis (Fig. 3).
Dual role of pyroptosis in tumor microenvironment remodeling and metastasis. Pyroptosis plays dual roles in TME. The anti-tumor effects could be concluded in two ways, the direct killing effect on tumor cells and immune cell activation. Membrane lysis by gasdermins and disruption of cellular homeostasis are two primary mechanisms through which pyroptosis mediates the killing of tumor cells. While dying cells may release pro-inflammation cytokines and DAMPs which may further attract and recruit immune cells. The pro-tumor effects of pyroptosis can be multifactorial. Peripheral inflammation caused by pyroptosis facilitates the tumor progression. Additionally, NLRP3 inflammasome-mediated pyroptosis can promote angiogenesis in tumors. The impact of pyroptosis on tumor metastasis is complex and context-dependent, and may enhance tumor infiltration and metastasis via IL-1 and IL-18 cytokine release
Direct killing effect on tumor cells
There are two primary mechanisms through which pyroptosis mediates the killing of tumor cells: membrane lysis by gasdermins and disruption of cellular homeostasis [142, 143]. In the former, tumor cells are destroyed through cell lysis, which is mediated by the cleavage of GSDMD initiated by caspases, reducing the overall tumor burden [27]. In the latter, caspase activation can also cleave other cellular components, leading to the dysfunction of organelles, protein degradation, and metabolic shutdown, which may even ultimately contribute to cell death [144, 145].
Influence and regulation of immune cells
Overall, the enrichment of pyroptosis showed a stronger positive correlation with the immune score than with the stromal score [146]. Analyzing the immune composition of the TME revealed a significant positive correlation between pyroptosis levels and the infiltration of major T cell subtypes, consistent across various tumors [146, 147]. Meanwhile, pyroptosis leads to the release of DAMPs and pro-inflammatory cytokines like IL-1β and IL-18. These may further attract and activate immune cells, particularly dendritic cells, which can then prime T cells to attack cancer cells, activating adaptive immunity [148, 149].
Impact on tumor angiogenesis, metabolism, and metastasis
Recent research shows that in some conditions, there is a positive correlation between angiogenic capacity and pyroptosis level which have something to do with NLRP3 inflammasome. The activation of NLRP3 inflammasome-mediated pyroptosis influences angiogenesis in endometriosis in a manner that is dependent on Notch1 [150]. NLRP3/IL-1β signaling pathway activation can also cause pathological micro-angiogenesis [151].
Theoretically, Caspase activation can disrupt essential metabolic pathways within tumor cells. However, Caspase-11 plays a significant role in maintaining dual-fuel bioenergetics glycolysis and oxidative phosphorylation (OXPHOS) to promote pyroptosis in macrophages [152]. A study also found pyroptosis leads to mitochondrial damage. GSDME promoted mitochondrial depolarization, trafficking defects, and neurite retraction. Frontotemporal dementia (FTD)/amyotrophic lateral sclerosis (ALS)-associated proteins TDP-43 and PR-50 induced GSDME-mediated damage to mitochondria and neurite loss [153].
The influence of pyroptosis on tumor metastasis is intricate and contingent on various factors. On one hand, as previously discussed, pyroptosis can exert a direct killing effect on tumor cells, thereby impeding metastasis. Moreover, the release of pro-inflammatory cytokines during pyroptosis can incite an immune response against the tumor, further hindering its spread. For instance, one study found CBD could induce an integrative stress response and mitochondrial stress in HCC tumor cells, leading to increased ATF4 activation and CHOP expression. This, in turn, promoted the expression of Bax protein from the BCL-2 family causing caspase-3/9-GSDME-dependent pyroptosis [154]. Additionally, an immunotherapy system called Lmo@RBC-triggered GSDMC-dependent pyroptosis in tumors, can reverse the immunosuppressive TME and inhibit tumor metastasis [155]. On the other hand, The cytokine release triggered by pyroptosis, including IL-1 and IL-18, can enhance tumor infiltration, potentially increasing the likelihood of tumorigenesis and metastasis [156]. Overall, the impact of pyroptosis on tumor metastasis is complex and context-dependent. Further research is needed to fully understand the mechanisms underlying this process and to develop targeted therapies that can harness the potential anti-metastatic effects of pyroptosis.
Cytokines in TME remodeling and metastasis
Definition and types of cytokines in TME
Cytokines are small signaling proteins that play critical roles in the TME by mediating communication between cells. These signaling molecules can be broadly classified based on their functions and origins, influencing both the immune response and tumor behaviors. Cytokines serve as crucial mediators for cell communication within the TME [157]. Although cytokines like IL-2, IFNα and IFNγ play a role in anti-tumor responses within the TME [158], irregular cytokine production by malignant cells, immune cells, and stromal cells contributes to all stages of carcinogenesis and therapy responses [159]. Thus, there’s therapeutic promise in utilizing cytokines’ immune-stimulating effects and in mitigating their dysregulated actions [160]. Specific cytokines play a significant role in tumor development, advancement, and spread (Table 2).
Role of cytokines in TME remodeling
Cytokines play a vital role in shaping the TME by modulating immune responses and inflammation [162]. Pyroptosis, a form of inflammatory cell death, is closely linked with the action of these cytokines such as IL-1β and IL-18 [162]. These cytokines amplify local immune responses and actively recruit and activate immune cells, including macrophages and T cells, thereby further enhancing inflammation within the TME [163]. For instance, IL-1β can promote immune cell infiltration and angiogenesis, thereby contributing to tumor progression [164]. This reciprocal interaction between cytokines and pyroptosis not only remodels the TME but also influences tumor growth and metastasis [165]. Consequently, a thorough understanding of the complex crosstalk between pyroptosis and cytokines is essential for elucidating how TME remodeling affects tumor dynamics and responses to immunotherapies [166]. In this context, we summarize the involvement of cytokines in the TME concisely to enhance understanding of their intricate interplay from two key perspectives: immune response and tumor progression (Fig. 4).
Immune cell-mediated pyroptosis activation and pro-inflammatory cytokines
Cytokines play a pivotal role in remodeling TME, significantly influencing the behavior of tumor and immune cells [167]. In the context of pyroptosis, pro-inflammatory cytokines are not only key drivers of inflammation but also crucial mediators in pyroptotic signaling, which shapes immune cell behavior [168].
Pro-inflammatory cytokine IL-1β has been reported to accumulate in MDSCs via the IL-6-STAT3 axis in melanoma [169], suggesting its role in suppressing immune cell activity. Moreover, research indicates that M1 macrophages can induce PD-L1 expression in HCC through IL-1β [170], enabling tumor cells to evade immune responses. Additionally, IL-1β collaborates with VEGF to enhance endothelial cell permeability, promoting angiogenesis in retinal endothelial cells [171]. The induction of PD-L1 and colony-stimulating factor 1 (CSF1) through IL-1β-triggered pathways, including the αKG/HIF1α axis, promotes tumor-associated macrophage (TAM) and MDSC infiltration, underscoring the complex role of pyroptosis in immune regulation within the TME [172]. Targeting IL-1β-related pyroptotic pathways, therefore, presents a promising strategy to mitigate immune suppression and tumor progression. Tumor-infiltrating dendritic cells (DCs), crucial in orchestrating both innate and adaptive immunity, are heavily influenced by cytokines released during pyroptosis [173]. They achieve this through the high expression of class I and class II MHC complexes, adhesion molecules, and costimulatory molecules [174]. Interestingly, cytokines like IL-10 and TGF-β, also prevalent in the TME, can skew DCs toward a tolerogenic phenotype, dampening immune responses. The dual role of pyroptosis in either promoting immune activation or contributing to immunosuppressive environments hinges on the balance of these cytokines, reflecting its complex regulatory role in immune surveillance within the TME [168].
Pro-inflammatory cytokines also participate in natural killer (NK) cells mediated-pyroptosis activation. The effectiveness of NK cells relies on the expression and activation of receptors, either activating or inhibitory, on their cell surface [175]. In the context of NK cell activity, the induction of pyroptosis in tumor cells can potentially reverse immune evasion by releasing intracellular contents that activate NK cells [176]. Researchers found that Interferon-α (IFN-α), Interferon-β (IFN-β), IFN-γ, and to a lesser extent, TNF-α, enhanced GSDMB expression and promoted pyroptosis driven by GzmA [90]. This indicates that interferons can potentially boost NK cell-mediated pyroptosis. However, in solid tumors, soluble inhibitory factors and cellular components, such as CAFs, constitute the immunosuppressive TME, thereby contributing to the compromised functionality of infiltrating NK cells [177]. Research indicates that TGF-β significantly inhibits NK cell activation and cytotoxicity, potentially by reducing IFN-γ production and suppressing activating receptors like NKG2D [178, 179]. Additionally, TGF-β downregulates the transcription of DAP12 and decreases NKp30 and NKG2D expression, further attenuating NK cell activity [180, 181].
Cytokine-mediated enhancement of antitumor immunity
Cytokines also play a key role in enhancing tumor immunity. Met supplementation might restore anti-tumor immunity by stimulating the secretion of IL-2, TNF-a, and IFN-r from TILs [182]. A study shows that NLRP3-dependent pyroptosis, induced by ChS-Ce6 nanovesicles in combination with laser treatment, significantly remodels the TME by enhancing the immunogenicity of the tumor [183]. This process involves the upregulation of NLRP3 and subsequent pyroptosis markers such as N-GSDMD, which leads to the release of immunogenic cell death (ICD) markers like CRT. The increased presence of CRT suggests a heightened anti-tumor immune response. Furthermore, the maturation of dendritic cells (DCs), essential for antigen presentation and the initiation of adaptive immune responses, is markedly enhanced in the ChS-Ce6 + laser group. This maturation promotes the activation and proliferation of cytotoxic T lymphocytes (CTLs) within the tumor, thereby strengthening the overall antitumor immune response. Consequently, ChS-Ce6 + laser-induced pyroptosis effectively boosts antitumor immunity by transforming the TME into a more immunogenic state and enhancing the immune system’s ability to target and eliminate tumor cells [184]. Additionally, studies demonstrated that exogenous IL-24 significantly enhances IFN-γ production in CD4 + and CD8 + T cells [185]. IL-24 upregulation may result from HMGB1 downregulation induced by pyroptosis, as HMGB1 knockdown has been reported to elevate IL-24 levels [186].
ECM modulation of pyroptosis and cytokine signaling in TME
The extracellular matrix (ECM) is a complex network of various macromolecules, such as collagens, fibrin, glycoproteins, and proteoglycans, which maintain the architecture, integrity, development, and homeostasis of normal tissue [187]. Alterations in the ECM within the tumor microenvironment (TME) are common in cancer tissues and are often associated with cancer progression [188]. In vitro studies demonstrated that MA blocked ECM degradation and reduced inflammation by suppressing the PI3K/AKT/NF-κB pathway and NLRP3 inflammasome-mediated pyroptosis. This led to higher anabolic protein expression, lower catabolic protein expression, and decreased secretion of inflammatory mediators like IL-18 and IL-1β [189]. Moreover, the ECM promotes the infiltration of other immunoinhibitory subpopulations. Increased collagen density or stiffness in the ECM triggers extensive FAK activation within cells, which subsequently leads to the direct exhaustion of CD8 + T cells and enhances the recruitment of Tregs, MDSCs, and TAMs, thereby contributing to the formation of an immunosuppressive TME [190].
Role of cytokines and related immune cells in TME metastasis
Cytokines play a critical role in tumor progression and metastasis within the TME by modulating immune responses, inflammation, and cellular communication [191]. Produced by immune cells and other cells, cytokines can either promote or inhibit tumor growth, depending on their types and context. Understanding their roles in TME metastasis is essential for developing targeted therapies that disrupt pro-tumorigenic interactions and enhance anti-tumor immunity (Table 3). Notably, pyroptosis is closely linked to cytokine release and the activity of immune cells, including myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), Treg cells, tumor-associated macrophages (TAMs), and others. Certain cytokines can worsen the pro-inflammatory TME, thus influencing pyroptosis, while immune cells can modulate pyroptosis through cytokine signaling. Therefore, exploring the interplay between cytokines and pyroptosis in TME remodeling is crucial for identifying therapeutic targets that can disrupt tumor-promoting interactions and strengthen anti-tumor immunity.
Cytokine-pyroptosis interplay in MDSC-mediated immunosuppression
Cytotoxic CD8 + T cells, CD4 + T cells, and NK cells collaborate to uphold immune surveillance. In contrast, various immune cells within tumors, such as MDSCs, Treg cells, and TAMs, aid in immune evasion and promote tumor progression. Normally, these cell types play essential roles in regulating the immune response, contributing to homeostasis and self-tolerance [213]. MDSCs are a diverse group of immature myeloid cells that inhibit the effector functions of CTLs and NK cells, exhibiting significant immunosuppressive activity in tumor-bearing hosts [214]. Numerous cytokines originating from tumors, including IL-6, IL-1β, GM-CSF, G-CSF, VEGF, and MCP-1, have been reported to induce MDSC accumulation in preclinical tumor cell models. These cytokines also closely connect with pyroptosis, for instance, IL-1β is a key inflammatory mediator released during this process [215]. One study found that in hepatocellular carcinoma (HCC), HCC-specific cell cycle-related kinase (CCRK) can upregulate IL-6 production through EZH2/NF-KB signaling, leading to extensive polymorphonuclear MDSC infiltration [216]. Similarly, hypoxia in tumors enhances pyroptosis-related inflammation through HIF-1α, further recruiting MDSCs via the CCL26/CX3CR1 pathway [217]. This interplay between cytokines and pyroptosis together regulates MDSCs, which helps shape the immune-suppressive environment.
CAF-mediated immune modulation and pyroptosis in TME
Tissue-resident fibroblasts, also known as quiescent fibroblasts, represent a significant origin of CAFs [218]. In certain types of tumors, stellate cells may serve as an additional origin of CAFs [219, 220]. Numerous studies have suggested that mesenchymal stem cells (MSCs) serve as precursors for CAFs [167, 221]. Drawing from mounting evidence, CAFs within the TME exert significant influence over the anti-tumor functions of immune cells infiltrating the tumor, spanning both innate and adaptive immune responses in the TME [221]. Through the secretion of cytokines, chemokines, and other effector molecules, such as TGF-β, CXCL2, collagens, MMPs, and laminin, CAFs can stimulate immune cell involvement in cancer onset and progression, while also aiding in the degradation and remodeling of the ECM [222]. Certainly, certain notable impacts of various immune cells on CAFs have also been recognized. To this day, numerous investigations have demonstrated that the interplays among CAFs, immune cells, and other immune elements can regulate the TME, consequently impeding the anti-tumor immune response [223]. Tumor-associated fibroblasts (CAFs) can also promote pyroptosis [224]. One study found that human Fibroblast Activation Protein-Chimeric Antigen Receptor Natural killer-92 cells (hFAP-CAR-NK-92 cells) were successfully constructed by using CAFs and other cells. It was confirmed that hFAP-CAR-NK-92 cells can target hFAP-positive NSCLC and inhibit the progression of NSCLC by activating the Caspase-3/GSDME cell pyroptosis pathway [225].
Cytokine can influence Treg cells activity and tumor progression via pyroptosis
The physiological role of Treg cells is to suppress excessive immune responses, maintaining homeostasis and autoimmune tolerance. However, the hyperactivity of Treg cells in tumor cell promotes tumor invasiveness and leads to a compromised T-cell immune response through Cytokines [226]. A greater number of CD4 + CD25 + Treg cells are enriched in the TME compared to healthy individuals. Treg cells are recruited through the CCR6 and CCL20 axis and activated by the interaction of TCR with IL-10 and TGF-b signaling [227]. Pyroptosis, as a form of inflammatory programmed cell death, can significantly influence this immune suppression by releasing cytokines such as ILs or activating pathways, which disrupt the Treg-mediated immunosuppressive environment. For example, Sorafenib, a multi-kinase inhibitor for HCC, has been shown to reduce hepatic Treg infiltration by suppressing TGF-b signaling. Moreover, IL-35, often associated with pyroptosis-driven immune evasion, correlates with the infiltration of CD39 + FoxP3 + Treg cells, underscoring its role in immune suppression and poor treatment outcomes [228].
Impact of cytokines and pyroptosis on TAMs in the TME
Macrophages infiltrating tumors, referred to as TAMs, are categorized into two distinct subsets activated by diverse polarizing cytokines: M1 (stimulated by lipopolysaccharide (LPS) alone or with Th1 cytokines) and M2 (induced by Th2 cytokines) [229]. M1-type macrophages primarily exhibit an anti-tumor function within the TME by facilitating antibody-dependent cellular cytotoxicity and generating reactive oxygen species (ROS) along with tumor necrosis factor (TNF) [230]. M2-type macrophages promote tumor progression through their involvement in tumor angiogenesis, immune suppression, cancer cell invasion and metastasis, as well as extracellular matrix (ECM) remodeling [231].
As a major component of the TME, TAM often indicates a poorer prognosis in tumor cells [232]. TAMs originate from marrow-derived monocytes and acquire diverse immunosuppressive functions throughout differentiation. Numerous studies indicate that M1-polarized macrophages generate pro-inflammatory cytokines and inhibit malignancy progression, while M2-polarized cells produce tumor growth factors (IL-6), angiogenic molecules (VEGF), and immunosuppressive agents (Arg1, IL-10, TGF-b, and IDO) [233]. The interplay between Ca²⁺-mediated pyroptosis and tumor-associated macrophage (TAM) remodeling demonstrates a synergistic enhancement of antitumor efficacy in colorectal cancer models [166]. This induced immunogenic cell death (ICD) promotes M1-type TAM polarization, mitigating immunosuppression, fostering dendritic cell maturation, and activating CD8⁺ T cell-dependent systemic antitumor immunity [166]. Such crosstalk highlights the potential of targeting pyroptosis and TAM dynamics to reshape the TME for improved cancer therapies.
Various cytokines originating from tumor cells, such as IL-4, IL-13, CSF-1, CCL2, CXCL12, and CTG, stimulate the differentiation of CCR2 + inflammatory monocytes into TAMs within the TME [234]. Furthermore, TGF-b derived from the TME enhances the expression of TIM-3 on TAMs, promoting both HCC progression and immune tolerance [235]. TAMs additionally generate cytokines and chemokines to promote immune suppression in tumor cell. For instance, CCL17, CCL18, and CCL22 derived from TAMs could facilitate the infiltration of Treg cells into the TME [235]. The interaction between MDSCs and TAMs decreases the synthesis of IL-6, IL-12, and MHC-II while enhancing IL-10 secretion. IL-10 from TAMs impairs the cytotoxicity of downstream CD8 + T cells and NK cells but elevates the frequency of CD4 + CD25 + FOXP3 + Treg cells [236]. Activated TAMs in the peritumoral stroma of tumor cells produce a range of pro-inflammatory cytokines, including IL-6, IL-23, IL-b, and TNF-a. These cytokines induce the proliferation of Th17 cells, which overexpress PD-1, CTLA-4, and GITR, thereby exerting an immunosuppressive effect [237]. In summary, TAMs represent a potential target for future cancer therapies.
Others
Less frequent immunosuppressive cell types observed in human tumor cells including B cell subset expressing PD-1, Th17 cells, CD4 + T cells expressing CCR4 and CCR6, CD14 + DCs expressing CTLA-4 and PD-1, tumor-associated neutrophils, tumor-associated fibroblasts, and type-II T helper cells (Th2) [238,239,240,241]. The collaboration among these cells contributes to the establishment of an immunosuppressive environment, typically associated with a dismal prognosis in few cancers.
Other impact of cytokines in TME
In the above sections, we focused on the role of cytokines in TME remodeling and their interactions with related immune cells in TME metastasis in the context of pyroptosis. In this part, we concisely discussed other involvement of cytokines and its role in TME.
The dynamic interplay between cytokines participates in immune modulation by dictating the recruitment and polarization of various immune cell subsets [242, 243]. For example, the presence of GM-CSF and G-CSF can drive the accumulation of MDSCs, which suppress T cell activity and promote tumor growth [167]. Cytokines like IL-6, TNF-α, and IL-1β promote tumor growth by activating signaling pathways that prevent apoptosis and enhance cell cycle progression, while IL-10 fosters an immunosuppressive environment [244, 245]. Additionally, TGF-β and IL-8 facilitate tumor cell migration and invasion by inducing epithelial-mesenchymal transition and promoting matrix metalloproteinase expression, respectively [246,247,248]. Cytokines like VEGF, IL-8, and FGF (fibroblast growth factor) promote angiogenesis by stimulating endothelial cell proliferation and migration, which is essential for tumor growth and metastasis [249, 250]. Cytokines such as IL-6 and TNF-α influence metabolic reprogramming in tumor cells, enhancing aerobic glycolysis (the Warburg effect) in cancer cells [251, 252]. What’s more, cytokines can modulate the metabolic activity of stromal cells within the TME, such as fibroblasts and immune cells, further supporting tumor growth and survival [253, 254].
In summary, cytokines in the TME significantly impact other aspects like tumor formation, proliferation, metabolism and so on. Understanding these processes is crucial for developing targeted therapies that can disrupt these cytokine-mediated interactions and inhibit tumor progression [250, 252,253,254].
Crosstalk between pyroptosis and cytokine in TME
The interplay between pyroptosis and cytokine
The interplay between pyroptosis and cytokines is integral to the dynamics of the TME, influencing both immune responses and tumor progression. This section will explore how pyroptosis regulates the generation and release of cytokines, the reciprocal role of cytokines in modulating pyroptotic pathways, and the intricate regulatory networks that govern these processes within the TME. By understanding these relationships, we can gain insight into the complex mechanisms that underlie tumor behavior and potential therapeutic avenues.
The regulation of pyroptosis in cytokine generation and release
Inflammasomes activation mechanisms
Inflammasomes are large molecular complexes that assemble in response to DAMPs and PAMPs. Their activation leads to the maturation of interleukin-1 (IL-1) family members and GSDMD, resulting in the secretion of IL-1 and the induction of pyroptosis, respectively. Various types of inflammasomes, each capable of detecting different types of threats, have been identified [191]. Inflammasome sensors briefly recruit caspase-1 family members, with or without the help of apoptosis-associated speck-like protein containing CARD (ASC), to initiate caspase-1 auto-cleavage. Activated caspase-1 then cleaves precursors of GSDMD and IL-1 family members, releasing these cytokines and inducing pyroptosis [191]. Canonical inflammasomes, composed of sensors, ASC, and caspase-1, play a crucial role in regulating pyroptosis and the subsequent generation and release of cytokines [121]. Upon activation, inflammasome sensors oligomerize and recruit ASC to form an "ASC speck," which then recruits caspase-1 [255]. Caspase-1 activation leads to the cleavage of pro-IL-1β and pro-IL-18 into their active forms, IL-1β and IL-18 [256]. Additionally, caspase-1 cleaves GSDMD, whose amino-terminal domain forms pores in the plasma membrane, resulting in pyroptosis [79, 257, 258]. These GSDMD pores facilitate the release of mature IL-1β and IL-18 into the extracellular environment, thereby contributing to the inflammatory response. This process highlights the intricate connection between inflammasome activation, pyroptosis, and cytokine release [70, 191]. Additionally, caspase-5 and caspase-11 are responsible for cleaving pro-IL-1α at D103 in senescent humans and mice [259]. Another noncanonical inflammasome component, caspase-8, is known to trigger various cell death pathways, including apoptosis, anoikis, necroptosis, autophagy, and pyroptosis [132]. Additionally, dendritic cell-associated C-type lectin-1 (dectin-1) triggers caspase-8 activation and subsequent IL-1β maturation in dendritic cells (DCs) stimulated by fungi and mycobacteria [260]. A similar effect has been observed in macrophages [261]. Coordination and cross-talk appear to exist between non-canonical caspase-8 inflammasomes and canonical inflammasomes via ASC and NLRP3[262, 263]. Canonical inflammasomes with ASC recruit caspase-8, which aids in IL-1β maturation independently of caspase-1 [263].
Pyroptosis in TME remodeling
Pyroptosis, which involves the breakdown and recycling of cellular materials, has been reported to both inhibit and promote tumor progression. In the context of malignancy, autophagy activation can generate reactive oxygen species (ROS), leading to compensatory cell proliferation via protein kinase C (PKC) λ/ι in hepatocellular carcinoma (HCC) [264]. Some researchers have indicated that E2, which activates the NLRP3 inflammasome, can kill cancer cells. Autophagy inhibits caspase-1, which is strongly associated with inducing apoptosis and pyroptosis pathways via the E2/Erβ/AMPK/mTOR pathway in HepG2 cells, rather than inducing pyroptotic death in cancer cells [125]. Additionally, studies on alpinum isoflavone have shown that inhibiting autophagy can enhance the effectiveness of inducing inflammasome-mediated pyroptosis in HCC [265].
A study highlights a novel approach to modulate the TME by simultaneously activating pyroptosis and the cGAS-STING pathway through a light-controlled, tumor-specific nanotheranostic platform [266]. This strategy enhances the expression of STING and GSDME, promotes the release of DNA fragments to potentiate the cGAS-STING pathway, and activates caspase-3 to cleave GSDME, leading to pyroptosis [266]. The subsequent release of inflammatory cytokines matures antigen-presenting cells, triggering T cell-mediated antitumor immunity [266]. This approach shows significant promise in overcoming the limitations of current pyroptosis inducers and STING agonists, offering a potent method for reshaping the TME and enhancing systemic antitumor immunity [267].
Another study found that cisplatin-induced the activation of GSDME and the release of cytokines including IL-12, which enhance the expression of IFN-γ in T cells in the TME and subsequently improve anti-PD-L1 response [268]. Altogether, their work demonstrates that cisplatin could induce GSDME-dependent cell pyroptosis to improve the response of anti-PD-L1 therapy though switching the TME from "cold" to "hot" in small-cell lung cancer, indicating GSDME as a response biomarker and pyroptosis as a pathway for combination therapy of anti-PD-L1 and chemotherapy, as well as a potential target to sensitize the response to PD-L1 inhibitor therapy in future [268].
Role of cytokine in pyroptosis
As mentioned above, cytokines play a critical role in the initiation and amplification of pyroptosis. They prominently participate in the activation of inflammasomes, regulation of intracellular signaling pathways, and the release of inflammatory cytokines, etc. Understanding the role of cytokines in pyroptosis is crucial for comprehending the crosstalk between pyroptosis and cytokines and their roles in TME. Herein, we take IL family and TGF-β as examples to provide a detailed introduction to the role of cytokines in pyroptosis.
IL family
As mentioned above, IL-1β and IL-18 participated in pyroptosis process and matured from pro-IL-1β and pro-IL-18, exerting a critical role in the pro-inflammation cytokines release [135]. One study demonstrated that N-GSDMD trafficking to neutrophil organelles facilitated IL-1β release independently of plasma membrane pores and pyroptosis [269]. In inflammasome-activated macrophages, GSDMD cleavage leads to pyroptosis and IL-1β release, whereas in neutrophils, N-GSDMD is essential for IL-1β secretion via an autophagy-dependent mechanism without causing pyroptosis, demonstrating distinct GSDMD trafficking between these cell types [269]. Similar to IL-1β, IL-18 is also released during pyroptosis. Cells expressing IL-18 receptors, such as NK cells and Th1 cells, can be activated and produce interferon-γ (IFN-γ) [270]. Notably, IFN-γ can stimulate the production of granzyme B in CD8+ T lymphocytes, which can induce cancer cell apoptosis by triggering pyroptosis through GSDME [35] or modulate the expression of apoptosis-related genes [271]. Inhibition of pyroptosis via utilizing rapamycin could reduce the release of IL-1β and IL-18 in vitro in the septic response [272]. Interestingly, one research found that IL-1β can interact with IL-6 and IL-23 to activate naïve CD4+ T cells, leading to the formation of Th17 cells, which in turn recruit neutrophils and release pro-inflammatory factors [273,274,275]. Moreover, IL-1β can drive the differentiation of Th17 cells and diminish the effects of TGF-β, which promotes the differentiation of T cells into Tregs [276, 277]. In summary, IL-1β and IL-18 not only dive themselves into pyroptosis but can also affect antigen-presenting cells (APCs) and other immune cells to indirectly influence the process.
Several other IL family members are also discovered to participate in pyroptosis. For instance, IL-33 is produced through GSDMD-mediated pyroptosis and functions as a pro-inflammatory chemokine. It has been shown to mediate type 2 immunity by activating various immune cells, including macrophages, with the IL-33/ST2 signaling pathway playing a significant role. Consequently, some researchers are targeting this cytokine to inhibit inflammation [278]. IL-17A, primarily secreted by γδT cells in the colorectal TME, regulates the TME in various ways [279]. W. Q. Feng et al. [280] found that it induces mitochondrial dysfunction and pyroptosis through the ROS/NLRP3/caspase-4/GSDMD pathway, leading to intracellular ROS accumulation. Additionally, IL-17A promotes the secretion of inflammatory factors like IL-1β and IL-18, as well as immune antigens, and recruits CD8+ T cells to infiltrate tumors [280].
TGF-β
Transforming growth factor-β (TGF-β) is a multifunctional cytokine expressed in almost all cell types [281]. Several studies discovered the involvement of TGF-β in pyroptosis. NLRP3-mediated pyroptosis in hepatic stellate cells (HSCs) can lead to the production of TGF-β through the IL-1β/IL-1R pathway, thereby regulating TGF-β expression [282]. Meanwhile, TGF-β can activate the NLRP3 inflammasome in HSCs through TGF-β receptor-mediated TAK1-NF-κB signaling or pathways generating ROS in the intracellular space, thus elevating NLRP3 inflammasome levels [282]. This creates a positive feedback loop. However, TGF-β recruitment of Tregs and inhibition of APCs make this type of pyroptosis detrimental to human body [282]. Similarly, another study found that lactate in the tumor microenvironment activates the NLRP3 inflammasome in macrophages, while TGF-β inhibits inflammasome activation and induces autophagy to clear reactive oxygen species (ROS), aiding tumor cells in evading immune surveillance. This indicates that tumor cells modulate TGF-β to counteract immune responses [283]. These findings suggest that targeting TGF-β may represent a viable strategy to inhibit the progression of tumor cells.
There are other cytokines involved in the process of pyroptosis. Inhibiting Nrf2 enhances the sensitivity of colorectal cancer (CRC) cells to oxaliplatin by promoting ferroptosis and pyroptosis, indicated by increased TNF-α release and the modulation of GPX4 expression, thereby offering a novel target to mitigate chemoresistance in CRC treatment [284]. Similarly, elevated levels of inflammatory cytokines like TNF-α and pyroptosis-related proteins were observed in bovine endometritis through neutrophil extracellular traps (NETs) released by neutrophils, resulting in inflammation and tissue damage [285]. Additionally, the combination of TNF-α and IFN-γ induced PANoptosis, a form of inflammatory cell death that exacerbated lung damage in COVID-19 by activating the JAK/STAT1/IRF1 signaling axis [93]. In summary, these studies indicate that inhibiting the cytokine-mediated inflammatory cell death signaling pathway identified could be advantageous for patients with cancer or other infectious and autoinflammatory diseases by reducing tissue damage and inflammation.
Regulation network of pyroptosis and cytokine in TME
Pyroptosis and cytokine interplay
As pyroptotic cells release inflammatory factors, the resulting damaged plasma membranes stimulate chemokine production and attract a variety of immune cells [286, 287]. Studies have demonstrated that pyroptosis amplifies cellular immunity, as cytotoxic lymphocytes, including natural killer (NK) and CD8+T cells, release granzymes like GZMA and GZMB, which cleave GSDMB and GSDME, respectively [88, 90]. Furthermore, the key molecule in pyroptosis, NLRP3, is essential for the TH2 cell transcriptome program in CD4(+) T cells, and its deficiency modulates the TME and promotes tumor cells growth [288, 289]. Paradoxically, NLRP3 exerts an immunosuppressive effect in some tumor cells such as melanoma tumor cells [288, 290] by recruiting MDSCs [291]. Additionally, in vivo experiments demonstrate that inhibiting GSDMC transcription, thereby suppressing pyroptosis, alleviates tumor necrosis symptoms and prolongs the survival of tumor-bearing mice [87]. These results indicate that pyroptosis functions as a double-edged sword in tumors, highlighting the importance of the specific executor involved and the cell type in which the process takes place.
Wenqiong Chen et al. [288]. conducted an association analysis between PScore and the enrichment score of hallmark gene sets from MSigDB across seven melanoma datasets. They observed significant positive correlations between PScore and immune-related hallmark pathways in all datasets. In contrast, multiple carcinogenic signaling pathways were found to correlate significantly negatively with PScore in multiple datasets [288]. This well explains the relationship between cytokines and the formation mechanism of pyroptosis, and the interaction forms a regulatory network.
Extracellular signaling in pyroptosis
After cell rupture during pyroptosis, danger signals are released into the extracellular space. For instance, ATP can bind to and activate the P2X7 channel, a type of potassium ion channel [292]. In the physiological environment, the concentration of K+ inside the cell is higher than in the extracellular space. When the intracellular concentration decreases, this can activate the NLRP3 inflammasome through NEK7, an intracellular potassium sensor [293]. This process indicates that pyroptosis can propagate among cells. Regardless of the trigger, pyroptosis ultimately leads to the production of IL-1β and IL-18 via this pathway. Additionally, ATP can recruit macrophages and dendritic cells to the extracellular matrix (ECM), presenting an “eat-me” signal to pyroptotic cells [294]. The initiation of phagocytosis in pyroptotic cells by macrophages through these signaling pathways enhances CD8+ T-cell activation and promotes IFN-γ production [44].
Comprehensive effects of regulatory networks on TME
As a form of inflammatory cell death (ICD), pyroptosis has the potential to transform the immune "cold" tumors into "hot" tumors by releasing proinflammatory factors and reshaping immune cells within the TME [88]. One of the distinctive features of pyroptosis is the release of inflammatory cytokines, including IL-1β, IL-18, and HMGB1 [295,296,297]. IL-1β and IL-18 are secreted through the GSDMD-forming pores, whereas HMGB1 is released after pyroptosis-induced cell lysis [297]. These inflammatory cytokines, particularly IL-1β and IL-18, play crucial roles in both innate and adaptive immunity [297]. Therefore, the regulation network of pyroptosis and cytokines in the TME shaping and modulation represents a crucial avenue of investigation. Pyroptosis, as a form of inflammatory cell death, not only transforms "cold" tumors into "hot" tumors by releasing proinflammatory factors but also plays a pivotal role in bridging the connection between innate and adaptive immunity. Understanding the intricate mechanisms underlying pyroptosis and its influence on TME reprogramming holds significant promise for devising innovative therapeutic strategies. By elucidating these pathways, novel targeting approaches may be developed to effectively modulate the TME and enhance antitumor immune responses, ultimately improving clinical outcomes for cancer patients.
Signaling pathway in the crosstalk of pyroptosis and cytokine
The interaction between pyroptosis and cytokine signaling pathways forms an intricate network involving numerous participants and potential feedback loops. Multiple pathways are involved in this regulation, including the cGAS-STING pathway (Fig. 5), NF-κB pathway, JAK/STAT pathway, MAPK pathway, and so on. By influencing these pathways, we may have the potential to adjust inflammation, bolster host defense against pathogens, and potentially manage diseases linked to dysregulated pyroptosis or cytokine signaling.
cGAS-STING pathway
Definition and role
The cGAS-STING pathway is the primary sensor for cellular cytosolic double-stranded DNA (dsDNA), enabling the innate immune system to respond to infections, inflammation, and cancer [298,299,300]. Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor/enzyme that catalyzes the formation of 2'-5'-cGAMP, an unusual cyclic di-nucleotide second messenger [301, 302]. This messenger binds to and activates the Stimulator of Interferon Genes (STING), leading to the recruitment of Tank Binding Kinase 1 (TBK1) [302], which results in the activation of the transcription factor Interferon Regulatory Factor 3 (IRF3), and the trans-activation of innate immune response genes [301], including type I Interferon cytokines (IFN-I). The activation of the pro-inflammatory cGAS-STING-IRF3 response is triggered by direct recognition of the DNA genomes of bacteria and viruses [302]. However, it can also occur during RNA virus infection, neoplastic transformation, tumor immunotherapy, and systemic auto-inflammatory diseases [299]. After activation, it then subsequently induces inflammasome activation and the onset of pyroptosis (Fig. 5).
The crosstalk between pyroptosis and cytokines in cGAS-STING pathway. The cGAS-STING pathway (in purple arrow) is the primary sensor for cellular cytosolic dsDNA, enabling the innate immune system to respond to diverse pathogens and dying cells. Cytosolic DNA sensor cGAS binds dsDNA to form cGAS-dsDNA complex, and subsequently activates cGAMP. This initiates a series of downstream effects including STING activation, recruitment of TBK1, and phosphorylation of IRF3. Ultimately, innate immune response and diverse cytokines release. Pyroptosis can regulate cGAS-STING pathway via different components and cytokines (in blue arrow). Additionally, the cGAS-STING pathway can also promote pyroptosis mainly through NLPR3 and AIM2 inflammasomes (in red arrow). The crosstalk between pyroptosis and cytokines in cGAS-STING pathway together forms a complex network
The interplay of pyroptosis and cytokines in cGAS-STING pathway
Caspase family could cleave cGAS to directly regulate cGAS-STING pathway. In activation of canonical pyroptosis, caspase-1 interacts with cGAS, cleaves cGAS, and inhibits STING-mediated interferon production [303]. Upon inflammasome activation, caspase-1 directly binds to cGAS via its p20 domain, cleaving human cGAS at the D140/157 site [303]. This reduces cGAMP production and cytokine expression. Additionally, in non-canonical pyroptotic pathways, caspase-4 and caspase-5 in humans, and caspase-11 in mice, cleave cGAS during lipopolysaccharide (LPS)-induced non-canonical inflammasome activation [303, 304]. In line with this, during Zika virus infection, caspase-1-induced cGAS cleavage inhibits TBK1 and IRF3 phosphorylation, reducing type I interferon production, and thus, bypassing the antiviral response [305].
The GSDMD-K+ efflux axis targets cGAS, decreasing cGAMP synthesis, which in turn inhibits STING signaling and reduces IFN-β production [304]. Researchers find that GSDMD-deficient mice showed an increased IFN-β response to Francisella novicida infection. GSDMD negatively regulates the IFN-β response independently of pyroptosis and IL-1β [306]. The AIM2 inflammasome-activated GSDMD depletes intracellular K+ through membrane pores, which is a process that is both essential and sufficient for inhibiting the cGAS-dependent IFN-β response. This, in turn, suppresses the cGAS-driven type I IFN response to DNA in macrophages [306].
The CARD domain of ASC participates in the regulation of cGAS-STING pathway. ASC protein, acting as a ligand, is composed of two domains: a PYD domain at the N-terminal and a CARD domain at the C-terminal. Through CARD-CARD interactions, ASC assembles the inflammasome by recruiting caspase-1, which also contains a CARD domain. During DNA virus infection, a deficiency in ASC results in elevated IFN production [303]. The CARD domain of ASC in the AIM2 inflammasome binds to the N-terminal domain of STING, inhibiting its interaction with TBK1, thereby negatively regulating the cGAS-STING signaling pathway [307]. The NLRC3 protein, which includes the CARD domain, impedes the type I IFN response and IL-1β secretion by competing with ASC for caspase-1 binding, disrupting the formation of ASC specks, and interfering with the assembly and activation of the NLRP3 inflammasome [306].
Diverse inflammasomes are also involved in the regulation of cGAS-STING pathway. AIM2 inhibits the cGAS-STING-mediated production of type I IFN upon stimulation with various DNA forms [304]. Upon exposure to cytosolic DNA, dendritic cells (DCs) and macrophages lacking AIM2, ASC, or caspase-1 exhibited significantly increased cGAMP production, STING aggregation, and phosphorylation of TBK1 and IRF3 [308], demonstrating that AIM2 inhibition of the STING pathway affects upstream STING, thereby diminishing the activation cascade of the entire STING pathway. The NLRP3 inflammasome consists of the cytoplasmic sensor NLRP3, the adaptor ASC, and the effector caspase-1. In one study, mice subjected to whole abdomen radiation through timed exposure to X-rays at a cumulative dose exhibited heightened levels of p-TBK1 and p-IRF3 in colonic tissues, along with increased IFN-β levels following NLRP3 deficiency. The absence of NLRP3 resulted in elevated cGAS-STING-mediated IFN-β production following radiation. NLRP3 deficiency also amplified type I IFN production and bolstered the host's resistance [309].
Apart from those inflammasomes mentioned above, nod-like receptors (NLRs) families also participated in the regulation, including NLRX1, NLRP2, NLRC3, NLRC4, NLRC5, NLRP6, and NLRP12 [310,311,312]. The majority of NLRs have a positive impact on inflammatory responses, especially the inflammasome-forming NLRs. However, recent research has shown that NLRC3 has a negative effect on the type I interferon (IFN) response by sequestering and dampening STING activation [306, 313]. Additionally, NLRC3 interacts with pro-caspase 1 and ASC via its CARD domain, thereby obstructing the assembly of NLRP3 and NLRC4 inflammasomes and subsequently suppressing cell pyroptosis [306]. Similar to NLRC3, NLRX1 engages with STING via its nucleotide-binding domain (NBD), leading to the obstruction of STING-TBK1 interaction, consequently impeding the activation of TBK1 necessary for type I IFN production [314]. NLRP2 interacts directly with TBK1, disrupting the TBK1-IRF3 interaction and interfering with TBK1-induced IRF3 phosphorylation, thereby inhibiting IFN signaling [310]. NLRP4 negatively regulates type I IFN signal transduction by activating TBK1, which is then subjected to K48-linked ubiquitination and degradation by the E3 ubiquitin ligase DTX4 [315]. Furthermore, NLRP11 restricts type I IFN activation by hampering TBK1-induced IFN-β promoter activity, indicating its potential role in the cGAS-STING signaling pathway [316] (Fig. 5).
cGAS-STING pathway regulates NLRP3 inflammasome and pyroptosis
The cGAS-STING-NLRP3 signaling axis is a specific mechanism that enables the activation of the NLRP3 inflammasome and the subsequent secretion of IL-1β. In human myeloid cells, the cGAS-STING pathway was essential for the activation of NLRP3 induced by cytoplasmic DNA during viral and bacterial infections [317]. Additionally, the STING-IRF3 axis could trigger LPS-induced cardiac dysfunction, inflammation and pyroptosis by activating the NLRP3 inflammasome in mice [318]. Furthermore, the cGAS-STING pathway was activated in myelodysplastic syndromes (MDS) to stimulate IFN-stimulated genes (ISG), leading to the activation of the NLRP3 inflammasome [319].
Available studies indicate that the interaction between STING and NLRP3 in response to cytoplasmic DNA stimulation promotes NLRP3 inflammasome activation through several mechanisms [304]. Firstly, STING recruits NLRP3 to facilitate its localization in the endoplasmic reticulum, thereby promoting the formation of the NLRP3 inflammasome [320]. Secondly, the TM5 (151-160aa) domain of STING interacts with the NACHT and LRR domains of NLRP3, attenuating its K48- and K63-linked polyubiquitination, effectively deubiquitinating NLRP3 to activate the inflammasome [320]. Thirdly, in an epistatic regulatory mechanism, the H3K4-specific histone methyltransferase WDR5 and the H3K79 methyltransferase DOT1L were found to significantly reduce STING overexpression-mediated NLRP3 upregulation, suggesting that STING promotes histone methylation in the NLRP3 promoter region via WDR5/DOT1L, thereby recruiting IRF3 to increase NLRP3 transcription [321].
Inflammasome pathway
Canonical pyroptotic death is facilitated by inflammasome assembly, which involves GSDMD cleavage and the release of IL-1β and IL-18 [40, 48]. Currently, the inflammasome sensors NLRP1, NLRP3, NLRC4, AIM2, and pyrin are known to form canonical inflammasomes and have been extensively studied [43].
The NLRP1 inflammasome was the first to be described. NLRP1 possesses a C-terminal extension housing a CARD domain, enabling direct interaction with procaspase-1 and circumventing the need for ASC. However, the inclusion of ASC in the complex enhanced the activity of the human NLRP1 inflammasome [322, 323]. Apart from caspase-1, NLRP1 also engages with caspase-5, potentially playing a role in IL-1β processing within human cells [324].
At present, the NLRP3 inflammasome stands as the most comprehensively characterized inflammasome, comprising the NLRP3 scaffold, the ASC (PYCARD) adaptor, and caspase-1 [324]. Exposure to complete pathogens, along with various structurally diverse PAMPs, DAMPs, and environmental irritants, triggers the activation of NLRP3. NLRP3 oligomerization results in the clustering of PYD domains, allowing for homotypic interaction with the adaptor ASC containing PYD and CARD. The CARD domain of ASC recruits the CARD of procaspase-1. Clustering of procaspase-1 enables autocleavage and the formation of the active caspase-1 p10/p20 tetramer, which subsequently processes cytokine proforms like IL-1β to produce active molecules. NLRC3 can also interact with pro-caspase 1 and ASC through its CARD domain, thus impeding the formation of NLRP3 and NLRC4 inflammasomes and consequently inhibiting cell pyroptosis [306].
AIM2 Inflammasome is the first identification of a non-NLR family member forming an inflammasome scaffold [322]. The AIM2 inflammasome comprises AIM2, ASC, and caspase-1. AIM2 features a PYD domain, which, similar to NLRP3, interacts with ASC through homotypic PYD-PYD interactions, enabling the ASC CARD domain to recruit procaspase-1 to the complex. Upon autoactivation, caspase-1, like in other inflammasomes, facilitates the maturation and secretion of proinflammatory cytokines such as IL-1β and IL-18. The ligand requirements for AIM2 are quite permissive, as cytosolic dsDNA from viruses, bacteria, or the host itself can activate the AIM2 inflammasome [325, 326].
Other pathways in the crosstalk of pyroptosis and cytokine in TME
Inflammation related pathways
The transcription factor NF-κB (nuclear factor kappa B) plays a key role in regulating various functions of the innate and adaptive immune systems and acts as a central mediator of inflammatory responses including the activation of NLRP3 [327]. For example, research shows that Metformin promotes the AMPK/SIRT1/NF-κB signaling pathway, which drives the induction of pyroptosis in cancer cells [328]. The JAK/STAT pathway (Janus kinase/signal transducer and activator of transcription) forms a swift membrane-to-nucleus signaling system and triggers the production of numerous essential regulators of cancer and inflammation [329]. Studies revealed that type I interferon-mediated JAK-STAT signaling pathway facilitates the transition from apoptosis to pyroptosis, potentially through the upregulation of the anti-apoptotic Bcl-xL gene [330].
Granzyme-related signaling cascades
Granzymes are serine proteases released from cytoplasmic granules in CD8+ T cells and NK cells, primarily consisting of granzyme A and B [331]. Granzyme-induced pyroptosis converts non-inflammatory cell death into an inflammatory form, enhancing the inflammatory properties within TME [297, 332]. Recent findings suggest that granzymes are involved in pyroptosis through two pathways. Firstly, after entering the target cell, granzyme A can cleave and activate GSDMB at the Lys229/Lys244 sites, leading to pyroptosis in target cells, resulting in cytoplasmic swelling, membrane rupture, and the release of inflammatory factors, eliciting a robust antitumor immune response [34, 80, 90]. The inactivation of granzyme A with 4-Octyl itaconate can inhibit GSDMB-induced pyroptosis and reduce inflammation [333]. Moreover, granzyme B can activate caspase-independent pyroptosis in target cells by directly cleaving GSDME at the same site as caspase 3, which in turn activates granzymes, establishing a positive feedback loop that amplifies the overall effect [334, 335]. Additionally, Euphohelioscopin A enhances the cleavage of GSDME, promoting granzyme B-induced pyroptosis and granzyme B silencing inhibits the activation of caspase-3 and Gasdermin E [336, 337].
Inspiration for treatments
Therapeutic targets (Fig. 6)
Pyroptosis-targeting agents
Therapeutic targeting of pyroptosis offers a promising strategy for enhancing anti-tumor immunity by inducing pro-inflammatory cell death in tumor cells, disrupting the tumor microenvironment (TME), and activating immune responses [34]. Several potential drug candidates and treatment strategies aim to modulate key components of the pyroptotic pathway, particularly focusing on inflammasomes, caspases, and gasdermin proteins (Table 4).
Disulfiram
Gasdermins are the key executors of pyroptosis. Among them, Gasdermin D (GSDMD) plays a pivotal role in forming membrane pores, leading to cell swelling and eventual lysis [371]. For example, Disulfiram functions as an effective inhibitor of pyroptosis by covalently modifying Cysteine-191 (Cys191) on gasdermin D (GSDMD), preventing its pore formation. This inhibition occurs without affecting the upstream steps of inflammasome assembly or the cleavage of GSDMD and IL-1β, indicating that Disulfiram specifically targets the final stage of pyroptosis [372]. The ability to selectively inhibit GSDMD pore formation while allowing cytokine processing suggests that Disulfiram could be repurposed as a therapeutic agent to manage inflammatory diseases associated with excessive pyroptosis, making it a promising candidate in the context of inflammation-driven conditions [373, 374].
Famotidine
A recent study demonstrated that famotidine (300 μm) induced cell pyroptosis, as confirmed by LDH assay. Additionally, famotidine activated NLPR3 inflammasomes, including ASC, Caspase-1, and NLRP, in gastric cancer cells, promoting the maturation and secretion of IL-18 but not IL-1β. Interestingly, famotidine increased GSDME, not GSDMD, in BGC823 and AGS cells. Mechanistically, famotidine significantly enhanced ERK1/2 phosphorylation, while the ERK1/2 inhibitor U0126 reversed famotidine’s effect on IL-18 secretion [342, 343]. These results uncovered a new role of famotidine in gastric cancer cell pyroptosis, requiring careful consideration for treatment strategies.
Caspase Modulators and Chemotherapy-Induced Pyroptosis
Caspase enzymes, particularly caspase-1, caspase-3, and caspase-11, are essential mediators of pyroptosis. Inhibitors or activators of these caspases offer a targeted approach to control pyroptosis [338]. For example, paclitaxel and cisplatin are chemotherapy drugs known to induce GSDME-mediated pyroptosis by activating caspase-3. This pathway has been particularly effective in cancers that express high levels of GSDME [375]. By shifting the mode of cell death from apoptosis to pyroptosis, these agents can trigger a stronger immune response, attracting immune cells like dendritic cells and T cells to the tumor site and amplifying anti-tumor immunity [139]. Additionally, caspase-1 inhibitors, such as VX-765, have been explored for modulating pyroptosis to reduce excessive inflammation, while maintaining enough immune activation to combat tumors [345, 346].
GSDM proteins
In gastric cancer, decreased expression of GSDMD facilitates tumor growth [376]. GSDMD-mediated pyroptosis may occur during standard anti-tumor therapies. For instance, cisplatin has been shown to trigger the NLRP3/caspase-1/GSDMD pyroptosis pathway in breast cancer cells [344]. Indeed, numerous studies have identified numerous compounds that induce GSDMD-dependent pyroptosis in tumor cells through diverse mechanisms. Metformin, for instance, induces GSDMD-mediated pyroptosis in chemo-refractory esophageal squamous cell carcinoma [159]. Anthocyanin activates pyroptosis in oral squamous cell carcinoma cells by upregulating the expression of NLRP3, caspase-1, and IL-1β [377]. Similarly, 4-hydroxybenzoic acid selectively triggers pyroptosis in the lung cancer cell line A549 by enhancing the transcription of caspase-1, IL-1β, and IL-18, while leaving normal lung epithelial cells unaffected [378]. Simvastatin also induces pyroptosis in A549 and H1299 cells by activating the NLRP3 pathway [128]. Val-boroPro, a DPP8/9 inhibitor, induces caspase-1-dependent pyroptosis in human acute myeloid leukemia [379]. Docosahexaenoic acid triggers caspase-1 activation, GSDMD maturation, and IL-1β secretion in the breast cancer cell line MDA-MB-231 through lysosomal damage and ROS formation [380]. Lysosomal rupture appears to be a common downstream event of various interventions leading to pyroptosis in cancer cells [380,381,382]. In epithelial ovarian cancer cells, a non-canonical inflammasome signal, GSDMD/caspase-4, induced by 2-(α-Naphthoyl) ethyltrimethylammonium iodide, contributes to pyroptosis [383]. Additionally, LPS can evoke non-canonical inflammasome caspase-11-mediated pyroptosis in lung cancer cells [127]. Apart from numerous chemicals, various sophisticated nanoparticles have been designed to promote inflammasome-mediated pyroptosis [382].
GSDMNT, known for triggering pyroptosis and inducing antitumor immune responses, has emerged as a highly promising strategy for anticancer therapy. However, its broad cytotoxicity in mammalian cells poses challenges in the production and delivery of cancer cells. Lu et al. devised a method involving a recombinant adeno-associated virus expressing GSDMNT [384]. They utilized a mammal-specific promoter to drive GSDMNT expression and packaged the virus into insect cells to prevent its expression. Additionally, recombinant adeno-associated virus-Cre was used to restore GSDMNT expression. This approach not only induces pyroptosis but also enhances antitumor responses. Importantly, better therapeutic outcomes have been observed when combined with anti-PD-L1 therapy [384].
Antibody-drug-conjugate (ADC) associated with pyroptosis
As described above, in recent years, the cleavage of GSDME by various chemotherapy drugs has been shown to cause cell pyroptosis. Similarly, many studies have also proved that antibody-drug conjugate (ADC) therapy plays an increasingly important role in the armamentarium of anticancer therapies.
Pyroptosis differs from apoptosis by promoting the release of pro-inflammatory cytokines and causing the rupture of the cell membrane, which can enhance antitumor immune responses [41]. For instance, the study by Wittwer et al. demonstrated that a mesothelin-targeting ADC with a tubulysin payload induced pyroptosis in mouse models of breast and colon cancer. This pyroptotic effect was critical for the ADC’s antitumor efficacy, as the cleavage of GSDME led to tumor cell death and stimulated the immune system by increasing the infiltration of cytotoxic T lymphocytes [350]. The combination of ADC therapy with dendritic cell-expanding agents, such as Fms-like tyrosine kinase-3 ligand (Flt3L), further boosted the immune response, especially in GSDME-silenced tumors, highlighting a potential therapeutic strategy for cancers with suppressed GSDME expression [350].
These studies underscore the potential of ADCs not only to directly kill cancer cells but also to reshape the tumor microenvironment by inducing pyroptosis and enhancing antitumor immunity. The use of ADCs that can trigger pyroptosis provides a promising approach to cancer treatment, particularly for tumors that are resistant to apoptosis.
ICIs
Blocking the interaction between immune checkpoints and their ligands with immune checkpoint inhibitors (ICIs) can relieve immune cells from checkpoint-induced inhibition, thereby reinvigorating them to exert anti-tumor effects. Currently, numerous ICIs have shown remarkable progress in clinical applications, representing a breakthrough in tumor therapy. Inhibitors targeting cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), programmed death-1 (PD-1), and programmed death-ligand 1 (PD-L1) have been successfully approved for the treatment of various malignant tumors such as melanoma and non-small cell lung cancer. The success of CTLA-4 and PD-1/PD-L1 antibodies has sparked a surge of research into ICIs [385]. In 2017 and 2018, two PD-1 inhibitors, nivolumab and pembrolizumab, were approved as second-line treatments for HCC [386]. Significantly, the superior outcomes of atezolizumab plus bevacizumab compared to sorafenib for advanced HCC signaled a new direction in combination therapies [387]. Currently, numerous ongoing clinical trials involve ICIs, either alone or combined with anti-VEGF agents or tyrosine kinase inhibitors (TKIs). A deeper understanding of the TME has sparked considerable interest in ICIs. Therefore, manipulating the TME directly or indirectly holds promise for new breakthroughs in clinical cancers treatment.
Recently, more immune checkpoints like lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), and cluster of differentiation 47 (CD47) have been identified. Consequently, extensive preclinical and clinical research on these proteins is underway [388].
Targeting cytokine and signaling pathways
ILRs and signaling pathways
Prior research has shown that within the TME, IL-6 collaborates with the JAK/STAT3 signaling pathway to engage in activities that significantly inhibit the function of immune effector cells [389]. Tocilizumab, a humanized monoclonal antibody targeting IL-6R, has shown broad antitumor and anti-chemoresistance properties across various cancer types in preclinical investigations [390]. During a phase I clinical trial, administration of high-dose tocilizumab was found to promote CD8 + T cell activation and elevate levels of antitumor effectors like IFN-γ and TNF-α, consequently bolstering anticancer immune responses [391]. Additionally, preclinical data suggests that targeting IL-6/JAK/STAT3 signaling could enhance the effectiveness of immune checkpoint-inhibiting monoclonal antibodies in combating tumors [392].
Inflammatory cytokines released during pyroptosis play vital roles in regulating tumor progression and metastasis. Thus, targeting these cytokines presents potential opportunities for treating different cancers. Canakinumab, a human anti-IL-1β monoclonal antibody widely utilized in inflammatory diseases, has been applied in the treatment of various cancers such as lung cancer [393], breast cancer, colon cancer, and other tumor types [394]. Moreover, Yuan et al. [395]. demonstrated that inhibiting IL-1β with canakinumab notably decreased tumor growth in K-ras-mutant lung adenocarcinoma by restructuring the TME [395]. The anti-IL-1β monoclonal antibody (mAb) facilitated the infiltration and activation of CD8 + T cells while suppressing myeloid-derived suppressor cell function. Hence, blocking IL-1β presents a promising therapeutic avenue for K-ras–mutant lung adenocarcinoma. Similarly, inhibiting the IL-1β pathway using an IL-1 receptor antagonist (IL-1Ra) may play a pivotal role in curtailing tumor progression [396]. Anakinra, an IL-1Ra, has demonstrated its ability to inhibit breast cancer growth by reducing IL-1β and IL-22 secretion [397]. Furthermore, anakinra has proven effective in significantly attenuating cytokine release syndrome during CAR-T therapy, offering a promising strategy to mitigate the severe side effects associated with this treatment [398, 399].
TGF-β
TGF-β assumes a significant role in activating CAFs and mediating their interaction with immune cells, as discussed earlier. This suggests that TGF-β inhibition therapy could potentially restore compromised immune responses within the TME [105]. Presently, numerous preclinical and clinical investigations into TGF-β-based immunotherapies are underway [178]. One such example is galunisertib (LY21577299), a small-molecule inhibitor targeting transforming growth factor beta receptor 1 (TGF-βR1), with infrequent reports of discernible cardiac toxicities during treatment [400]. Phase II clinical trials for pancreatic cancer and hepatocellular carcinoma have demonstrated significant therapeutic efficacy of galunisertib against tumors, whether administered in combination with gemcitabine or as monotherapy [401]. Other studies have shown that combining a therapy directed at TGF-β derived from CAFs with checkpoint inhibitors like anti-PD-L1 antibodies produces more significant immunological impacts on tumors compared to individual treatments [402]. Therefore, Ravi et al. [403] endeavored to create chimeric antibodies by fusing the TGF-βR2 extracellular domain with anti-CTLA4 or anti-PD-L1 antibodies, resulting in anti-CTLA4-TGF-βR2 and anti-PD-L1-TGF-βR2 constructs. In comparison to ipilimumab (an anti-CTLA-4 antibody), the anti-CTLA4-TGF-βR2 molecule demonstrates greater efficacy in reducing tumor-infiltrating Treg cells and inhibiting tumor progression [404].
With ongoing clinical trials for next-generation TGFβRI inhibitors and bifunctional antibodies that combine TGFβ and immune checkpoint inhibition, TGFβ has become an appealing therapeutic target in the era of immunotherapy [405]. Another option is targeting macrophages, as they are a major source of inflammatory factors [406]. Currently, some antibodies and inhibitors have demonstrated anti-tumor activities in preclinical studies, and a few have been explored in early-stage clinical trials. A key challenge in targeting inflammation is achieving selective inhibition of pro-tumor chronic inflammation without compromising anti-tumor immunity [110].
Novel application of pyroptosis in antitumor approaches (Fig. 6)
Recent advancements highlight the potential of pyroptosis in enhancing antitumor immunity. A notable development is the Phe-BF3 desilylation bio-orthogonal system, which effectively transports desilylation catalyzed by Phe-BF3 with NP-GSDMA3-mediated delivery into specific mammary tumor cells in mice. This technique boosts T cell-dependent tumor regression by increasing CD4+, CD8+, NK cell, and M1 macrophage populations while reducing Treg cells, M2 macrophages, monocytes, neutrophils, and MDSCs. This system reveals the antitumor immune potential of pyroptosis, suggesting that a gasdermin agonist may enhance cancer immunotherapy efficacy [407].
Additionally, in patients with extrahepatic cholangiocarcinoma (CCA), tumor-cell-derived microvesicles containing methotrexate can induce pyroptosis in CCA cells via a GSDME-dependent pathway. The intracellular contents released from pyroptotic CCA cells activate macrophages to produce pro-inflammatory cytokines, attracting neutrophils to the tumor site and degrading the stromal barrier in the CCA TME, alleviating biliary obstruction in nearly 25% of patients [408].
Another application involves tumor-targeting nanoliposomes loaded with cisplatin. When combined with decitabine (DAC), these nanodrugs activate and upregulate the caspase-3/GSDME pathway, inducing pyroptosis in tumor cells and enhancing the immunological effect of chemotherapy in a mouse triple-negative breast cancer model. DAC also demethylates the GSDME gene in tumor cells [409].
Finally, a chimeric co-stimulatory converting receptor has been designed to disrupt the PD-1 pathway, enhancing the activity of chimeric antigen receptor (CAR)-NK cells against solid tumors. The antitumor activity of NK92 cells is significantly improved by the neo-complex PD1-NKG2D-41BB receptor, primarily through pyroptosis activation [410].
Emerging research and technologies (Fig. 7)
High-throughput cytokine assays
In recent years, the crosstalk between pyroptosis and cytokines within the TME has garnered significant attention due to its implications in cancer progression and therapy. High-throughput cytokine assays have emerged as invaluable tools in elucidating the intricate interplay between these processes. These assays allow for the simultaneous quantification of multiple cytokines in various biological samples, offering a comprehensive view of the cytokine landscape in the TME [411]. By employing advanced technologies such as multiplex immunoassays and microfluidic platforms, researchers can efficiently profile cytokine expression patterns with high sensitivity and specificity. For instance, a study presents a system that enables the dynamic analysis of cellular interactions, proliferation, and therapeutic effectiveness through spatiotemporal monitoring and secretum profiling [411]. Lenalidomide, an immunomodulatory drug, demonstrated a direct anti-proliferative effect on activated B-cell-like Diffuse large B cell lymphoma (DLBCL) spheroids and decreased several cytokines and other markers (such as CCL2, CCL3, CCL4, CD137, and ANG-1 levels) [411]. Together, this innovative spheroid platform will facilitate high-throughput screening of anti-cancer therapeutics in a semi-automated fashion [411]. Furthermore, these assays enable the exploration of dynamic changes in cytokine levels during pyroptosis induction and its subsequent effects on tumor immune responses. Integrating high-throughput cytokine assays into studies investigating the crosstalk between pyroptosis and cytokines provides valuable insights into the underlying mechanisms driving tumor progression and facilitates the development of novel therapeutic strategies targeting these pathways.
Single-cell sequencing
Single-cell sequencing enables the simultaneous profiling of small quantities of tumor cells and TME cellular constituents, making it valuable for studying the co-evolution of tumor cells and the TME during tumor development. Analysis of pancreatic cancer precursors at single-cell resolution revealed an increase in proinflammatory immune components in the TME at an early stage, which were progressively depleted and replaced by stromal myofibroblast populations during neoplastic progression [412]. Profiling patients’ melanoma at single-cell resolution uncovered two distinct tumor cell states, MITF-dominant and AXL-dominant, corresponding to specific tumor microenvironmental patterns, including specific interactions between cancer cells and their TME [413]. Single-cell sequencing can also track dynamic changes in the TME during therapeutic treatment. Using paired scRNA-seq and T-cell receptor sequencing of cells from patients with basal or squamous cell carcinoma before and after anti-PD-1 therapy, T-cells responding to checkpoint blockade were found to mainly derive from a distinct repertoire of T-cell clones not observed before treatment, rather than pre-existing tumor-infiltrating T lymphocytes [414]. Overall, single-cell sequencing of tumor and TME cells provides immense insights into tumor evolution, patient tumor classification, and guidance for cancer therapies.
Along with scRNA-seq, new methods and technologies for profiling genetic, epigenetic, proteomic, spatial, and lineage information in individual cells have been invented and are advancing rapidly. These single-cell multi-omics technologies can reveal cellular heterogeneity at multiple molecular levels. Integrative analysis of multi-omics data will provide profound novel insights into the fundamental mechanisms driving cellular diversity and help to identify targetable cellular subsets or signaling pathways essential for cancer cell adaptation to the TME [415].
Spatial transcriptomics
Spatial transcriptomics has emerged as a powerful tool for studying the spatial organization of gene expression within the TME [416]. This innovative technology allows researchers to visualize and analyze gene expression patterns in situ, providing insights into the complex interactions between tumor cells, stromal cells, and immune cells [416].
Spatial transcriptomics enables the simultaneous profiling of thousands of genes across spatially defined regions of tissue sections, allowing for the identification of cell types and their functional states within the TME [417]. By integrating spatial information with traditional transcriptomic data, researchers can gain a deeper understanding of the heterogeneity and dynamics of cellular interactions within the TME [418].
One key application of spatial transcriptomics in the study of pyroptosis and cytokine signaling in the TME is the identification of spatially distinct expression patterns of key genes involved in these processes. For example, spatial transcriptomic analysis can reveal the spatial distribution of pyroptosis-related genes within the tumor tissue, providing insights into the spatial heterogeneity of cell death mechanisms and their impact on tumor progression [288].
Furthermore, spatial transcriptomics can elucidate the spatial relationships between cytokine-producing cells, such as tumor-infiltrating immune cells, and their target cells within the TME [419]. This spatial information is crucial for understanding the localized effects of cytokines on neighboring cells and their contribution to immune regulation and tumor immunity.
Overall, spatial transcriptomics represents a promising approach for advancing our understanding of pyroptosis, cytokine signaling, and their roles in shaping the TME. By providing spatial context to gene expression data, this technology offers new opportunities for identifying novel therapeutic targets and developing personalized treatment strategies for cancer patients.
Emerging research and technologies in TME. High-throughput cytokine assays provide a comprehensive view of cytokine profiles in the TME by quantifying multiple cytokines simultaneously. Single-cell sequencing allows for studying the co-evolution of tumor cells and TME components by profiling small quantities of cells. Spatial transcriptomics identifies cell types and their functional states within the TME by profiling thousands of genes across spatially defined tissue regions concurrently
Perspective and conclusion
The complex interplay between pyroptosis and cytokine signaling in the tumor microenvironment represents a crucial frontier in cancer research. This review highlights the essential roles of pyroptosis in triggering inflammatory responses and remodeling the TME, as well as the reciprocal effects of cytokines in either promoting tumor progression or facilitating immune-mediated tumor destruction.
Cytokines influence both immune responses and tumor progression within the TME, operating through pro-inflammatory and anti-inflammatory mechanisms [420,421,422,423]. They can induce pyroptosis in cells directly or indirectly, with significant contributions from both tumor and immune cells [39, 140, 424]. Our focus centers on the crosstalk between these cellular entities, pyroptosis, cytokines, and various signaling pathways. Pyroptosis serves a dual role in tumor development: it influences tumor advancement through gene expression linked to pyroptosis and enhances anti-tumor immune responses by promoting the infiltration of CD8 + T cells, NK cells, and M1 macrophages [407]. Additionally, the impact of effectors like RIPK1/3, inflammasomes, and the cytokines and DAMPs released through ICD on immune cells and immune responses remains debated. These findings point to a more intricate relationship between non-apoptotic PCD and immune responses across various tumor types and settings [32]. It is vital to investigate how diverse components of the TME—including immune, tumor, and stromal cells—interact to either inhibit or promote tumor progression via immune mechanisms and metabolic reprogramming. Therefore, novel therapies targeting pyroptosis and cytokine signaling could represent promising avenues for cancer treatment.
However, the application of pyroptosis as a therapeutic strategy in the development of anticancer agents presents considerable challenges. Designing effective drugs that specifically induce pyroptosis in human cells, while adhering to rigorous safety standards, is a significant obstacle in pharmaceutical research [141]. Combining targeted treatments that activate or suppress pyroptosis with immunotherapy offers substantial potential for advancing cancer therapy [425, 426]. This integrative approach could lead to significant breakthroughs, providing patients with more effective and personalized treatment options. While the combination of chemotherapy and ICIs has shown great promise, further investigation into the role of pyroptosis in chemotherapy-related toxicity is necessary [427]. Additionally, the initiation of pyroptosis by radiotherapy-induced DNA damage through various signaling mechanisms may enhance antitumor effects when combined with immunotherapy [428,429,430]. This strategy leverages the strengths of each modality to achieve optimal therapeutic outcomes. Ultimately, the integration of targeted therapies, radiotherapy, and chemotherapy with immunotherapy holds significant promise for improving cancer treatment. Nevertheless, optimizing the timing and sequence of these therapies is crucial to maximizing their efficacy and enhancing patient outcomes.
Despite the growing understanding of the molecular mechanisms underlying pyroptosis and the potential for new therapeutic targets, challenges remain. Firstly, the relationship between cytokines, pyroptosis, and the TME is complex and multifaceted. While significant progress has been made in defining pyroptosis pathways, further investigations are necessary to clarify the intricate signaling mechanisms, explore additional cytokines, and assess the roles of various GSDM family proteins, as well as their pathological significance. Secondly, pyroptosis does not uniformly exert a beneficial therapeutic effect in cancer treatment. For instance, elevated expression of GSDMC has been linked to poorer prognoses in invasive breast carcinoma, correlating with immune cell infiltration [431]. Similarly, upregulated GSDMB has been shown to enhance the proliferation and invasiveness of bladder cancer cells [432]. These contradictory effects may arise from the distinction between acute activation of pyroptosis, which tends to inhibit tumor formation, and sustained pyroptosis, which may promote tumor progression [34, 87]. Thirdly, attention should be paid to potential tissue damage and cytokine release syndrome (CRS). In certain tumor cell lines, GSDME expression levels are lower than in normal cell lines, risking unintentional damage to healthy tissues during chemotherapy [40, 63]. Research demonstrated that tumor cell pyroptosis initiated CRS during CAR T cell therapy by activating inflammatory pathways through granzyme B-mediated GSDME cleavage [35]. While a novel RGD-anchored curcumin-loaded liposome effectively targets macrophages to inhibit pyroptosis, potentially providing a strategy to mitigate CRS and improve outcomes in sepsis-related organ injuries [36].
In summary, targeting pyroptosis and other components within the TME holds significant promise for novel cancer treatments, and substantial efforts are being directed toward translating these findings into clinical applications. Importantly, pyroptosis has the potential to enhance the efficacy of immunotherapy by modulating tumor immunogenicity and increasing lymphocyte infiltration in the TME. However, it is crucial to acknowledge the possibility of negative outcomes despite promising experimental data suggesting the antitumor effects of pyroptosis. Future research will require more animal models to explore the broader consequences of pyroptosis, as well as more clinical trials investigating the modulation of pyroptosis in cancer patients.
Data availability
No datasets were generated or analysed during the current study.
References
Hanahan D, Monje M. Cancer hallmarks intersect with neuroscience in the tumor microenvironment. Cancer Cell. 2023;41(3):573–80.
Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, et al. Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments. Mol Cancer. 2023;22(1):48.
Passaro A, Al Bakir M, Hamilton EG, Diehn M, André F, Roy-Chowdhuri S, et al. Cancer biomarkers: emerging trends and clinical implications for personalized treatment. Cell. 2024;187(7):1617–35.
Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 2019;575(7782):299–309.
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, et al. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther. 2024;9(1):270.
Lorenzo-Martín LF, Hübscher T, Bowler AD, Broguiere N, Langer J, Tillard L, et al. Spatiotemporally resolved colorectal oncogenesis in mini-colons ex vivo. Nature. 2024;629(8011):450–7.
Maacha S, Bhat AA, Jimenez L, Raza A, Haris M, Uddin S, et al. Extracellular vesicles-mediated intercellular communication: roles in the tumor microenvironment and anti-cancer drug resistance. Mol Cancer. 2019;18(1):55.
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer. 2022;21(1):208.
Hinshaw DC, Shevde LA. The Tumor Microenvironment innately modulates Cancer Progression. Cancer Res. 2019;79(18):4557–66.
Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022;19(4):237–53.
Saxton RA, Glassman CR, Garcia KC. Emerging principles of cytokine pharmacology and therapeutics. Nat Rev Drug Discov. 2023;22(1):21–37.
Studebaker AW, Storci G, Werbeck JL, Sansone P, Sasser AK, Tavolari S, et al. Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 2008;68(21):9087–95.
Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48.
Shi Y, Gao W, Lytle NK, Huang P, Yuan X, Dann AM, et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature. 2019;569(7754):131–5.
Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell. 2010;140(6):883–99.
Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, et al. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004;118(3):285–96.
Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell. 2009;15(2):103–13.
Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, Smith K, et al. IL-23 promotes tumour incidence and growth. Nature. 2006;442(7101):461–5.
Kortylewski M, Xin H, Kujawski M, Lee H, Liu Y, Harris T, et al. Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. Cancer Cell. 2009;15(2):114–23.
Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006;66(23):11238–46.
Cursiefen C, Chen L, Borges LP, Jackson D, Cao J, Radziejewski C, et al. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest. 2004;113(7):1040–50.
Korbecki J, Kupnicka P, Chlubek M, Gorący J, Gutowska I, Baranowska-Bosiacka I. CXCR1 receptor: regulation of expression, Signal Transduction, and involvement in Cancer. Int J Mol Sci. 2022;23(4):2168.
Smith MP, Sanchez-Laorden B, O’Brien K, Brunton H, Ferguson J, Young H, et al. The immune microenvironment confers resistance to MAPK pathway inhibitors through macrophage-derived TNFα. Cancer Discov. 2014;4(10):1214–29.
Loftus LV, Amend SR, Pienta KJ. Interplay between cell death and cell proliferation reveals New Strategies for Cancer Therapy. Int J Mol Sci. 2022;23(9):4723.
Hsu SK, Li CY, Lin IL, Syue WJ, Chen YF, Cheng KC, et al. Inflammation-related pyroptosis, a novel programmed cell death pathway, and its crosstalk with immune therapy in cancer treatment. Theranostics. 2021;11(18):8813–35.
Hänggi K, Ruffell B. Cell death, therapeutics, and the immune response in cancer. Trends Cancer. 2023;9(5):381–96.
Fang Y, Tian S, Pan Y, Li W, Wang Q, Tang Y, et al. Pyroptosis: a new frontier in cancer. Biomed Pharmacother. 2020;121:109595.
Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem Sci. 2017;42(4):245–54.
Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375(6586):1254–61.
Liu X, Nie L, Zhang Y, Yan Y, Wang C, Colic M, et al. Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis. Nat Cell Biol. 2023;25(3):404–14.
Zhang H, Liu J, Yuan W, Zhang Q, Luo X, Li Y, et al. Ammonia-induced lysosomal and mitochondrial damage causes cell death of effector CD8(+) T cells. Nat Cell Biol. 2024;26(11):1892–902.
Gao W, Wang X, Zhou Y, Wang X, Yu Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduct Target Ther. 2022;7(1):196.
Hadian K, Stockwell BR. The therapeutic potential of targeting regulated non-apoptotic cell death. Nat Rev Drug Discov. 2023;22(9):723–42.
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, et al. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol. 2022;15(1):174.
Liu Y, Fang Y, Chen X, Wang Z, Liang X, Zhang T, et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci Immunol. 2020;5(43):eaax7969.
Shi Y, Wu Q, Lu Y, Meng LP, Xu XL, Wang XJ, et al. Arginine-Glycine-aspartic acid-anchored curcumin-based Nanotherapeutics Inhibit pyroptosis-induced Cytokine Release Syndrome for in vivo and in Vitro Sepsis Applications. Curr Pharm Des. 2023;29(4):283–94.
Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541–50.
Baker KJ, Houston A, Brint E. IL-1 family members in Cancer; two sides to every story. Front Immunol. 2019;10:1197.
Liu C, Chu D, Kalantar-Zadeh K, George J, Young HA, Liu G. Cytokines: from clinical significance to quantification. Adv Sci (Weinh). 2021;8(15):e2004433.
Xia X, Wang X, Cheng Z, Qin W, Lei L, Jiang J, et al. The role of pyroptosis in cancer: pro-cancer or pro-host? Cell Death Dis. 2019;10(9):650.
Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, et al. Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 2022;19(9):971–92.
Lee C, Do HTT, Her J, Kim Y, Seo D, Rhee I. Inflammasome as a promising therapeutic target for cancer. Life Sci. 2019;231:116593.
Yu P, Zhang X, Liu N, Tang L, Peng C, Chen X. Pyroptosis: mechanisms and diseases. Signal Transduct Target Ther. 2021;6(1):128.
Du T, Gao J, Li P, Wang Y, Qi Q, Liu X, et al. Pyroptosis, metabolism, and tumor immune microenvironment. Clin Transl Med. 2021;11(8):e492.
Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–4.
Wang H, Zhou X, Li C, Yan S, Feng C, He J, et al. The emerging role of pyroptosis in pediatric cancers: from mechanism to therapy. J Hematol Oncol. 2022;15(1):140.
Wang S, Wang H, Feng C, Li C, Li Z, He J, et al. The regulatory role and therapeutic application of pyroptosis in musculoskeletal diseases. Cell Death Discov. 2022;8(1):492.
Frank D, Vince JE. Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 2019;26(1):99–114.
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.
Jiang L, Wang Z, Xu T, Zhang L. When pyro(ptosis) meets palm(itoylation). Cytokine Growth Factor Rev. 2024;77:30–38.
Rao Z, Zhu Y, Yang P, Chen Z, Xia Y, Qiao C, et al. Pyroptosis in inflammatory diseases and cancer. Theranostics. 2022;12(9):4310–29.
Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science. 2002;296(5573):1634–5.
Gerl R, Vaux DL. Apoptosis in the development and treatment of cancer. Carcinogenesis. 2005;26(2):263–70.
Matarrese P, Mattia G, Pagano MT, Pontecorvi G, Ortona E, Malorni W, et al. The sex-related interplay between TME and Cancer: on the critical role of Estrogen, MicroRNAs and Autophagy. Cancers (Basel). 2021;13:13.
Wang P, Long M, Zhang S, Cheng Z, Zhao X, He F, et al. Hypoxia inducible factor-1α regulates autophagy via the p27-E2F1 signaling pathway. Mol Med Rep. 2017;16(2):2107–12.
Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 2014;15(2):135–47.
Qin X, Ma D, Tan YX, Wang HY, Cai Z. The role of necroptosis in cancer: a double-edged sword? Biochim Biophys Acta Rev Cancer. 2019;1871(2):259–66.
Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, et al. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020;16(11):2069–83.
Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18(5):280–96.
Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer. 2019;19(7):405–14.
Song Q, Zhou R, Shu F, Fu W. Cuproptosis scoring system to predict the clinical outcome and immune response in bladder cancer. Front Immunol. 2022;13: 958368.
Zhou Y, Qin X, Hu Q, Qin S, Xu R, Gu K, et al. Cross-talk between disulfidptosis and immune check point genes defines the tumor microenvironment for the prediction of prognosis and immunotherapies in glioblastoma. Sci Rep. 2024;14(1):3901.
Newton K, Dixit VM, Kayagaki N. Dying cells fan the flames of inflammation. Science. 2021;374(6571):1076–80.
Su P, Mao X, Ma J, Huang L, Yu L, Tang S, et al. ERRα promotes glycolytic metabolism and targets the NLRP3/caspase-1/GSDMD pathway to regulate pyroptosis in endometrial cancer. J Exp Clin Cancer Res. 2023;42(1):274.
Rathinam VA, Fitzgerald KA. Inflammasome complexes: emerging mechanisms and Effector functions. Cell. 2016;165(4):792–800.
Ma ZY, Jiang C, Xu LL. Protein-protein interactions and related inhibitors involved in the NLRP3 inflammasome pathway. Cytokine Growth Factor Rev. 2023;74:14–28.
Boucher D, Monteleone M, Coll RC, Chen KW, Ross CM, Teo JL, et al. Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity. J Exp Med. 2018;215(3):827–40.
Ross C, Chan AH, von Pein JB, Maddugoda MP, Boucher D, Schroder K. Inflammatory caspases: toward a Unified Model for Caspase activation by Inflammasomes. Annu Rev Immunol. 2022;40:249–69.
Chen X, He WT, Hu L, Li J, Fang Y, Wang X, et al. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis. Cell Res. 2016;26(9):1007–20.
Xia S, Zhang Z, Magupalli VG, Pablo JL, Dong Y, Vora SM, et al. Gasdermin D pore structure reveals preferential release of mature interleukin-1. Nature. 2021;593(7860):607–11.
Deets KA, Vance RE. Inflammasomes and adaptive immune responses. Nat Immunol. 2021;22(4):412–22.
Ebata T, Terkawi MA, Kitahara K, Yokota S, Shiota J, Nishida Y, et al. Noncanonical pyroptosis triggered by macrophage-derived extracellular vesicles in chondrocytes leading to cartilage catabolism in Osteoarthritis. Arthritis Rheumatol. 2023;75(8):1358–69.
Pang J, Vince JE. The role of caspase-8 in inflammatory signalling and pyroptotic cell death. Semin Immunol. 2023;70:101832.
Kayagaki N, Wong MT, Stowe IB, Ramani SR, Gonzalez LC, Akashi-Takamura S, et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science. 2013;341(6151):1246–9.
Kumari P, Vasudevan SO, Russo AJ, Wright SS, Fraile-Ágreda V, Krajewski D, et al. Host extracellular vesicles confer cytosolic access to systemic LPS licensing non-canonical inflammasome sensing and pyroptosis. Nat Cell Biol. 2023;25(12):1860–72.
Khan MM, Ernst O, Sun J, Fraser IDC, Ernst RK, Goodlett DR, et al. Mass Spectrometry-based Structural Analysis and systems Immunoproteomics strategies for deciphering the Host Response to Endotoxin. J Mol Biol. 2018;430(17):2641–60.
Lo TH, Chen HL, Yao CI, Weng IC, Li CS, Huang CC, et al. Galectin-3 promotes noncanonical inflammasome activation through intracellular binding to lipopolysaccharide glycans. Proc Natl Acad Sci U S A. 2021;118:30.
Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014;514(7521):187–92.
Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016;535(7610):111–6.
Zhong X, Zeng H, Zhou Z, Su Y, Cheng H, Hou Y, et al. Structural mechanisms for regulation of GSDMB pore-forming activity. Nature. 2023;616(7957):598–605.
Shi X, Sun Q, Hou Y, Zeng H, Cao Y, Dong M, et al. Recognition and maturation of IL-18 by caspase-4 noncanonical inflammasome. Nature. 2023;624(7991):442–50.
Bhat AA, Thapa R, Afzal O, Agrawal N, Almalki WH, Kazmi I, et al. The pyroptotic role of Caspase-3/GSDME signalling pathway among various cancer: a review. Int J Biol Macromol. 2023;242(Pt 2):124832.
Hu L, Chen M, Chen X, Zhao C, Fang Z, Wang H, et al. Chemotherapy-induced pyroptosis is mediated by BAK/BAX-caspase-3-GSDME pathway and inhibited by 2-bromopalmitate. Cell Death Dis. 2020;11(4):281.
Wei Y, Lan B, Zheng T, Yang L, Zhang X, Cheng L, et al. GSDME-mediated pyroptosis promotes the progression and associated inflammation of atherosclerosis. Nat Commun. 2023;14(1):929.
Yang F, Bettadapura SN, Smeltzer MS, Zhu H, Wang S. Pyroptosis and pyroptosis-inducing cancer drugs. Acta Pharmacol Sin. 2022;43(10):2462–73.
Zhang JY, Zhou B, Sun RY, Ai YL, Cheng K, Li FN, et al. The metabolite α-KG induces GSDMC-dependent pyroptosis through death receptor 6-activated caspase-8. Cell Res. 2021;31(9):980–97.
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.
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.
Hansen JM, de Jong MF, Wu Q, Zhang LS, Heisler DB, Alto LT, et al. Pathogenic ubiquitination of GSDMB inhibits NK cell bactericidal functions. Cell. 2021;184(12):3178–91.e18.
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. 2020;368(6494):eaaz7548.
Ribeiro AB, de Barcellos-Filho PC, Franci CR, Menescal-de-Oliveira L, Saia RS. Pro-inflammatory cytokines, IL-1β and TNF-α, produce persistent compromise in tonic immobility defensive behaviour in endotoxemia guinea-pigs. Acta Physiol (Oxf). 2016;218(2):123–35.
Thomas JM, Huuskes BM, Sobey CG, Drummond GR, Vinh A. The IL-18/IL-18R1 signalling axis: diagnostic and therapeutic potential in hypertension and chronic kidney disease. Pharmacol Ther. 2022;239:108191.
Karki R, Sharma BR, Tuladhar S, Williams EP, Zalduondo L, Samir P, et al. Synergism of TNF-α and IFN-γ triggers inflammatory cell death, tissue damage, and Mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell. 2021;184(1):149–68.e17.
Liu X, Xia S, Zhang Z, Wu H, Lieberman J. Channelling inflammation: gasdermins in physiology and disease. Nat Rev Drug Discov. 2021;20(5):384–405.
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.
Faria SS, Costantini S, de Lima VCC, de Andrade VP, Rialland M, Cedric R, et al. NLRP3 inflammasome-mediated cytokine production and pyroptosis cell death in breast cancer. J Biomed Sci. 2021;28(1):26.
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 Exp Clin Cancer Res. 2021;40(1):153.
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol. 2022;86(Pt 2):280–95.
Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12(12):860–75.
Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in immunity and sterile inflammation. Annu Rev Pathol. 2020;15:493–518.
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. The impact of obesity, adipose tissue, and Tumor Microenvironment on Macrophage polarization and metastasis. Biology (Basel). 2022;11(2):339.
Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS ONE. 2009;4(4):e4992.
Fukumura D, Kloepper J, Amoozgar Z, Duda DG, Jain RK. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15(5):325–40.
Bejarano L, Jordāo MJC, Joyce JA. Therapeutic targeting of the Tumor Microenvironment. Cancer Discov. 2021;11(4):933–59.
Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–98.
Olumi AF, Grossfeld GD, Hayward SW, Carroll PR, Tlsty TD, Cunha GR. Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res. 1999;59(19):5002–11.
Shekhar MP, Werdell J, Santner SJ, Pauley RJ, Tait L. Breast stroma plays a dominant regulatory role in breast epithelial growth and differentiation: implications for tumor development and progression. Cancer Res. 2001;61(4):1320–6.
Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, et al. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68(3):918–26.
Henriksson ML, Edin S, Dahlin AM, Oldenborg PA, Öberg Å, Van Guelpen B, et al. Colorectal cancer cells activate adjacent fibroblasts resulting in FGF1/FGFR3 signaling and increased invasion. Am J Pathol. 2011;178(3):1387–94.
Cassetta L, Pollard JW. Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discov. 2018;17(12):887–904.
Frame RJ, Wahed S, Mohiuddin MK, Katory M. Right lateral position for laparoscopic splenic flexure mobilization. Colorectal Dis. 2011;13(7):e178–80.
Grecian R, Whyte MKB, Walmsley SR. The role of neutrophils in cancer. Br Med Bull. 2018;128(1):5–14.
Kovacs SB, Miao EA, Gasdermins. Effectors of Pyroptosis. Trends Cell Biol. 2017;27(9):673–84.
de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41(3):374–403.
Liu Y, Zhang Q, Xing B, Luo N, Gao R, Yu K, et al. Immune phenotypic linkage between colorectal cancer and liver metastasis. Cancer Cell. 2022;40(4):424–37.e5.
Jin MZ, Jin WL. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct Target Ther. 2020;5(1):166.
Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677–87.
Isazadeh A, Heris JA, Shahabi P, Mohammadinasab R, Shomali N, Nasiri H, et al. Pattern-recognition receptors (PRRs) in SARS-CoV-2. Life Sci. 2023;329:121940.
Lee-Kirsch MA. The type I interferonopathies. Annu Rev Med. 2017;68:297–315.
Minter MR, Taylor JM, Crack PJ. The contribution of neuroinflammation to amyloid toxicity in Alzheimer’s disease. J Neurochem. 2016;136(3):457–74.
Broz P, Dixit VM. Inflammasomes: mechanism of assembly, regulation and signalling. Nat Rev Immunol. 2016;16(7):407–20.
Zhai Z, Liu W, Kaur M, Luo Y, Domenico J, Samson JM, et al. NLRP1 promotes tumor growth by enhancing inflammasome activation and suppressing apoptosis in metastatic melanoma. Oncogene. 2017;36(27):3820–30.
Chow MT, Sceneay J, Paget C, Wong CS, Duret H, Tschopp J, et al. NLRP3 suppresses NK cell-mediated responses to carcinogen-induced tumors and metastases. Cancer Res. 2012;72(22):5721–32.
He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012–21.
Wei Q, Zhu R, Zhu J, Zhao R, Li M. E2-Induced activation of the NLRP3 inflammasome triggers pyroptosis and inhibits autophagy in HCC cells. Oncol Res. 2019;27(7):827–34.
Ikuta T, Kobayashi Y, Kitazawa M, Shiizaki K, Itano N, Noda T, et al. ASC-associated inflammation promotes cecal tumorigenesis in aryl hydrocarbon receptor-deficient mice. Carcinogenesis. 2013;34(7):1620–7.
Yokoyama S, Cai Y, Murata M, Tomita T, Yoneda M, Xu L, et al. A novel pathway of LPS uptake through syndecan-1 leading to pyroptotic cell death. Elife. 2018;7:7.
Wang F, Liu W, Ning J, Wang J, Lang Y, Jin X, et al. Simvastatin suppresses Proliferation and Migration in Non-small Cell Lung Cancer via Pyroptosis. Int J Biol Sci. 2018;14(4):406–17.
Vincenzi A, Goettert MI, Volken de Souza CF. An evaluation of the effects of probiotics on tumoral necrosis factor (TNF-α) signaling and gene expression. Cytokine Growth Factor Rev. 2021;57:27–38.
Mocellin S, Rossi CR, Pilati P, Nitti D. Tumor necrosis factor, cancer and anticancer therapy. Cytokine Growth Factor Rev. 2005;16(1):35–53.
Gao H, Zhong Y, Zhou L, Lin S, Hou X, Ding Z, et al. Kindlin-2 inhibits TNF/NF-κB-Caspase 8 pathway in hepatocytes to maintain liver development and function. Elife. 2023;12:12.
Mandal R, Barrón JC, Kostova I, Becker S, Strebhardt K. Caspase-8: the double-edged sword. Biochim Biophys Acta Rev Cancer. 2020;1873(2):188357.
Pollock TY, Vázquez Marrero VR, Brodsky IE, Shin S. TNF licenses macrophages to undergo rapid caspase-1, -11, and – 8-mediated cell death that restricts Legionella pneumophila infection. PLoS Pathog. 2023;19(6):e1010767.
Fu J, Wu H. Structural mechanisms of NLRP3 Inflammasome Assembly and Activation. Annu Rev Immunol. 2023;41:301–16.
Huang Y, Xu W, Zhou R. NLRP3 inflammasome activation and cell death. Cell Mol Immunol. 2021;18(9):2114–27.
Phillips FC, Gurung P, Kanneganti TD. Microbiota and caspase-1/caspase-8 regulate IL-1β-mediated bone disease. Gut Microbes. 2016;7(4):334–41.
Bai R, Li Y, Jian L, Yang Y, Zhao L, Wei M. The hypoxia-driven crosstalk between tumor and tumor-associated macrophages: mechanisms and clinical treatment strategies. Mol Cancer. 2022;21(1):177.
Ding B, Chen H, Tan J, Meng Q, Zheng P, Ma P, et al. ZIF-8 nanoparticles evoke pyroptosis for high-efficiency Cancer Immunotherapy. Angew Chem Int Ed Engl. 2023;62(10):e202215307.
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.
Hou J, Hsu JM, Hung MC. Molecular mechanisms and functions of pyroptosis in inflammation and antitumor immunity. Mol Cell. 2021;81(22):4579–90.
Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, et al. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 2020;13(1):110.
Kappelhoff S, Margheritis EG, Cosentino K. New insights into gasdermin D pore formation. Biochem Soc Trans. 2024;52(2):681–92.
Barnett KC, Li S, Liang K, Ting JP. A 360° view of the inflammasome: mechanisms of activation, cell death, and diseases. Cell. 2023;186(11):2288–312.
Yan J, Xie Y, Si J, Gan L, Li H, Sun C, et al. Crosstalk of the Caspase Family and mammalian target of Rapamycin Signaling. Int J Mol Sci. 2021;22(2):817.
Kesavardhana S, Malireddi RKS, Kanneganti TD. Caspases in cell death, inflammation, and Pyroptosis. Annu Rev Immunol. 2020;38:567–95.
Khan M, Ai M, Du K, Song J, Wang B, Lin J, et al. Pyroptosis relates to tumor microenvironment remodeling and prognosis: a pan-cancer perspective. Front Immunol. 2022;13:1062225.
Wang S, Gao S, Shan L, Qian X, Luan J, Lv X. Comprehensive genomic signature of pyroptosis-related genes and relevant characterization in hepatocellular carcinoma. PeerJ. 2023;11:e14691.
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 Discov. 2020;10(2):254–69.
Hatscher L, Amon L, Heger L, Dudziak D. Inflammasomes in dendritic cells: friend or foe? Immunol Lett. 2021;234:16–32.
Zhang M, Shi Z, Peng X, Cai D, Peng R, Lin Y, et al. NLRP3 inflammasome-mediated pyroptosis induce Notch signal activation in endometriosis angiogenesis. Mol Cell Endocrinol. 2023;574:111952.
Hong Y, Wei C, Fu M, Li X, Zhang H, Yao B. MCC950 alleviates seizure severity and angiogenesis by inhibiting NLRP3/ IL-1β signaling pathway-mediated pyroptosis in mouse model of epilepsy. Int Immunopharmacol. 2024;126:111236.
Drummer Ct, Saaoud F, Jhala NC, Cueto R, Sun Y, Xu K, et al. Caspase-11 promotes high-fat diet-induced NAFLD by increasing glycolysis, OXPHOS, and pyroptosis in macrophages. Front Immunol. 2023;14:1113883.
Neel DV, Basu H, Gunner G, Bergstresser MD, Giadone RM, Chung H, et al. Gasdermin-E mediates mitochondrial damage in axons and neurodegeneration. Neuron. 2023;111(8):1222–e409.
Shangguan F, Zhou H, Ma N, Wu S, Huang H, Jin G, et al. A novel mechanism of Cannabidiol in suppressing Hepatocellular Carcinoma by Inducing GSDME Dependent pyroptosis. Front Cell Dev Biol. 2021;9:697832.
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.
Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–45.
Bourne CM, Taabazuing CY. Harnessing pyroptosis for Cancer Immunotherapy. Cells. 2024;13(4):346.
Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA, Kobold S. Interleukins in cancer: from biology to therapy. Nat Rev Cancer. 2021;21(8):481–99.
Shalapour S, Karin M. Pas de deux: control of anti-tumor immunity by Cancer-Associated inflammation. Immunity. 2019;51(1):15–26.
Waldmann TA. Cytokines in Cancer Immunotherapy. Cold Spring Harb Perspect Biol. 2018;10(12):a028472.
Ozga AJ, Chow MT, Luster AD. Chemokines and the immune response to cancer. Immunity. 2021;54(5):859–74.
Greten FR, Grivennikov SI. Inflammation and Cancer: triggers, mechanisms, and consequences. Immunity. 2019;51(1):27–41.
Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–89.
Nakao S, Noda K, Zandi S, Sun D, Taher M, Schering A, et al. VAP-1-mediated M2 macrophage infiltration underlies IL-1β- but not VEGF-A-induced lymph- and angiogenesis. Am J Pathol. 2011;178(4):1913–21.
Wu L, Lu H, Pan Y, Liu C, Wang J, Chen B, et al. The role of pyroptosis and its crosstalk with immune therapy in breast cancer. Front Immunol. 2022;13:973935.
Cheng F, He L, Wang J, Lai L, Ma L, Qu K, et al. Synergistic immunotherapy with a calcium-based nanoinducer: evoking pyroptosis and remodeling tumor-associated macrophages for enhanced antitumor immune response. Nanoscale. 2024.
Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021;20(1):131.
Mestrallet G, Sone K, Bhardwaj N. Strategies to overcome DC dysregulation in the tumor microenvironment. Front Immunol. 2022;13: 980709.
Tengesdal IW, Dinarello A, Powers NE, Burchill MA, Joosten LAB, Marchetti C, et al. Tumor NLRP3-Derived IL-1β drives the IL-6/STAT3 Axis resulting in sustained MDSC-Mediated immunosuppression. Front Immunol. 2021;12:661323.
Cheng C, Hsu SK, Chen YC, Liu W, Shu ED, Chien CM, et al. Burning down the house: pyroptosis in the tumor microenvironment of hepatocellular carcinoma. Life Sci. 2024;347:122627.
Sheikpranbabu S, Kalishwaralal K, Venkataraman D, Eom SH, Park J, Gurunathan S. Silver nanoparticles inhibit VEGF-and IL-1beta-induced vascular permeability via src dependent pathway in porcine retinal endothelial cells. J Nanobiotechnol. 2009;7:8.
He Q, Liu M, Huang W, Chen X, Zhang B, Zhang T, et al. IL-1β-Induced Elevation of Solute Carrier Family 7 Member 11 promotes Hepatocellular Carcinoma Metastasis through Up-regulating programmed death Ligand 1 and colony-stimulating factor 1. Hepatology. 2021;74(6):3174–93.
Wei L, Wang X, Zhou H. Interaction among inflammasome, PANoptosise, and innate immune cells in infection of influenza virus: updated review. Immun Inflamm Dis. 2023;11(9):e997.
Lee YS, Radford KJ. The role of dendritic cells in cancer. Int Rev Cell Mol Biol. 2019;348:123–78.
Wang J, Matosevic S. Functional and metabolic targeting of natural killer cells to solid tumors. Cell Oncol (Dordr). 2020;43(4):577–600.
Hu M, Deng F, Song X, Zhao H, Yan F. The crosstalk between immune cells and tumor pyroptosis: advancing cancer immunotherapy strategies. J Exp Clin Cancer Res. 2024;43(1):190.
Stojanovic A, Cerwenka A. Natural killer cells and solid tumors. J Innate Immun. 2011;3(4):355–64.
Batlle E, Massagué J. Transforming growth Factor-β signaling in immunity and Cancer. Immunity. 2019;50(4):924–40.
Trotta R, Dal Col J, Yu J, Ciarlariello D, Thomas B, Zhang X, et al. TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J Immunol. 2008;181(6):3784–92.
Donatelli SS, Zhou JM, Gilvary DL, Eksioglu EA, Chen X, Cress WD, et al. TGF-β-inducible microRNA-183 silences tumor-associated natural killer cells. Proc Natl Acad Sci U S A. 2014;111(11):4203–8.
Viel S, Marçais A, Guimaraes FS, Loftus R, Rabilloud J, Grau M, et al. TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci Signal. 2016;9(415):ra19.
Bian Y, Li W, Kremer DM, Sajjakulnukit P, Li S, Crespo J, et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature. 2020;585(7824):277–82.
Hu ZC, Wang B, Zhou XG, Liang HF, Liang B, Lu HW, et al. Golgi apparatus-targeted photodynamic therapy for enhancing Tumor Immunogenicity by eliciting NLRP3 protein-dependent pyroptosis. ACS Nano. 2023;17(21):21153–69.
Zhou B, Zhang JY, Liu XS, Chen HZ, Ai YL, Cheng K, et al. Tom20 senses iron-activated ROS signaling to promote melanoma cell pyroptosis. Cell Res. 2018;28(12):1171–85.
Zhou JY, Wang WJ, Zhang CY, Ling YY, Hong XJ, Su Q, et al. Ru(II)-modified TiO(2) nanoparticles for hypoxia-adaptive photo-immunotherapy of oral squamous cell carcinoma. Biomaterials. 2022;289:121757.
Senda N, Yanai H, Hibino S, Li L, Mizushima Y, Miyagaki T, et al. HMGB1-mediated chromatin remodeling attenuates Il24 gene expression for the protection from allergic contact dermatitis. Proc Natl Acad Sci U S A. 2021;118(1):e2022343118.
Eble JA, Niland S. The extracellular matrix in tumor progression and metastasis. Clin Exp Metastasis. 2019;36(3):171–98.
Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891–906.
Gong Y, Qiu J, Jiang T, Li Z, Zhang W, Zheng X, et al. Maltol ameliorates intervertebral disc degeneration through inhibiting PI3K/AKT/NF-κB pathway and regulating NLRP3 inflammasome-mediated pyroptosis. Inflammopharmacology. 2023;31(1):369–84.
Serrels A, Lund T, Serrels B, Byron A, McPherson RC, von Kriegsheim A, et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell. 2015;163(1):160–73.
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol. 2023;16(1):24.
Jia X, Xi J, Tian B, Zhang Y, Wang Z, Wang F, et al. The tautomerase activity of Tumor Exosomal MIF promotes pancreatic Cancer Progression by modulating MDSC differentiation. Cancer Immunol Res. 2024;12(1):72–90.
Otani Y, Yoo JY, Lewis CT, Chao S, Swanner J, Shimizu T, et al. NOTCH-Induced MDSC Recruitment after oHSV virotherapy in CNS Cancer models modulates Antitumor Immunotherapy. Clin Cancer Res. 2022;28(7):1460–73.
Li Y, Zhang Q, Wu M, Zhang P, Huang L, Ai X, et al. Suppressing MDSC infiltration in Tumor Microenvironment serves as an option for treating Ovarian Cancer Metastasis. Int J Biol Sci. 2022;18(9):3697–713.
O’Byrne PM, Metev H, Puu M, Richter K, Keen C, Uddin M, et al. Efficacy and safety of a CXCR2 antagonist, AZD5069, in patients with uncontrolled persistent asthma: a randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2016;4(10):797–806.
Piwocka O, Piotrowski I, Suchorska WM, Kulcenty K. Dynamic interactions in the tumor niche: how the cross-talk between CAFs and the tumor microenvironment impacts resistance to therapy. Front Mol Biosci. 2024;11: 1343523.
Pei L, Liu Y, Liu L, Gao S, Gao X, Feng Y, et al. Roles of cancer-associated fibroblasts (CAFs) in anti- PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer. 2023;22(1):29.
Saw PE, Chen J, Song E. Targeting CAFs to overcome anticancer therapeutic resistance. Trends Cancer. 2022;8(7):527–55.
Kochetkova M, Samuel MS. Differentiation of the tumor microenvironment: are CAFs the organizer? Trends Cell Biol. 2022;32(4):285–94.
Zhang G, Zheng G, Zhang H, Qiu L. MUC1 induces the accumulation of Foxp3(+) Treg cells in the tumor microenvironment to promote the growth and metastasis of cholangiocarcinoma through the EGFR/PI3K/Akt signaling pathway. Int Immunopharmacol. 2023;118:110091.
Marangoni F, Zhakyp A, Corsini M, Geels SN, Carrizosa E, Thelen M, et al. Expansion of tumor-associated Treg cells upon disruption of a CTLA-4-dependent feedback loop. Cell. 2021;184(15):3998–4015.e19.
Ohue Y, Nishikawa H. Regulatory T (Treg) cells in cancer: can Treg cells be a new therapeutic target? Cancer Sci. 2019;110(7):2080–9.
Liu C, Chikina M, Deshpande R, Menk AV, Wang T, Tabib T, et al. Treg cells promote the SREBP1-Dependent metabolic fitness of Tumor-promoting macrophages via repression of CD8(+) T cell-derived Interferon-γ. Immunity. 2019;51(2):381 – 97.e6.
Yerolatsite M, Torounidou N, Gogadis A, Kapoulitsa F, Ntellas P, Lampri E, et al. TAMs and PD-1 networking in gastric Cancer: a review of the literature. Cancers (Basel). 2023;16(1):196.
Liu J, Cao X. Glucose metabolism of TAMs in tumor chemoresistance and metastasis. Trends Cell Biol. 2023;33(11):967–78.
Tajaldini M, Saeedi M, Amiriani T, Amiriani AH, Sedighi S, Mohammad Zadeh F, et al. Cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs); where do they stand in tumorigenesis and how they can change the face of cancer therapy? Eur J Pharmacol. 2022;928:175087.
Yang Y, Guo J, Huang L. Tackling TAMs for Cancer Immunotherapy: it’s Nano Time. Trends Pharmacol Sci. 2020;41(10):701–14.
Dayoub AS, Brekken RA. TIMs, TAMs, and PS- antibody targeting: implications for cancer immunotherapy. Cell Commun Signal. 2020;18(1):29.
Tian S, Chu Y, Hu J, Ding X, Liu Z, Fu D, et al. Tumour-associated neutrophils secrete AGR2 to promote colorectal cancer metastasis via its receptor CD98hc-xCT. Gut. 2022;71(12):2489–501.
Yamazaki T, Gunderson AJ, Gilchrist M, Whiteford M, Kiely MX, Hayman A, et al. Galunisertib plus neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer: a single-arm, phase 2 trial. Lancet Oncol. 2022;23(9):1189–200.
Wu L, Zhang XH. Tumor-Associated neutrophils and macrophages-heterogenous but not chaotic. Front Immunol. 2020;11:553967.
Lin YJ, Wei KC, Chen PY, Lim M, Hwang TL. Roles of neutrophils in Glioma and Brain metastases. Front Immunol. 2021;12:701383.
Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015;348(6230):74–80.
Kapanadze T, Gamrekelashvili J, Ma C, Chan C, Zhao F, Hewitt S, et al. Regulation of accumulation and function of myeloid derived suppressor cells in different murine models of hepatocellular carcinoma. J Hepatol. 2013;59(5):1007–13.
Faria SS, Fernando AJ, de Lima VCC, Rossi AG, de Carvalho JMA, Magalhães KG. Induction of pyroptotic cell death as a potential tool for cancer treatment. J Inflamm (Lond). 2022;19(1):19.
Zhou J, Liu M, Sun H, Feng Y, Xu L, Chan AWH, et al. Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut. 2018;67(5):931–44.
Chiu DK, Xu IM, Lai RK, Tse AP, Wei LL, Koh HY, et al. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology. 2016;64(3):797–813.
Kretzschmar K, Weber C, Driskell RR, Calonje E, Watt FM. Compartmentalized epidermal activation of β-Catenin differentially affects lineage reprogramming and underlies Tumor Heterogeneity. Cell Rep. 2016;14(2):269–81.
Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Invest. 2007;117(1):50–9.
Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 2013;123(5):1902–10.
Quante M, Tu SP, Tomita H, Gonda T, Wang SS, Takashi S, et al. Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell. 2011;19(2):257–72.
Ziani L, Chouaib S, Thiery J. Alteration of the Antitumor Immune response by Cancer-Associated fibroblasts. Front Immunol. 2018;9:414.
Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-Associated fibroblasts are activated in Incipient Neoplasia to Orchestrate Tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17(2):135–47.
Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells-An overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology. 2015;4(1):e954829.
Fang Y, Wang YJ, Zhao HL, Huang X, Fang YN, Chen WY, et al. Development of FAP-Targeted Chimeric Antigen Receptor NK-92 cells for Non-small Cell Lung Cancer. Discov Med. 2023;35(176):405–17.
Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007;25(18):2586–93.
Chen KJ, Lin SZ, Zhou L, Xie HY, Zhou WH, Taki-Eldin A, et al. Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS ONE. 2011;6(9):e24671.
Fu YP, Yi Y, Cai XY, Sun J, Ni XC, He HW, et al. Overexpression of interleukin-35 associates with hepatocellular carcinoma aggressiveness and recurrence after curative resection. Br J Cancer. 2016;114(7):767–76.
Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–86.
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233(9):6425–40.
Allavena P, Sica A, Garlanda C, Mantovani A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol Rev. 2008;222:155–61.
Yeung OW, Lo CM, Ling CC, Qi X, Geng W, Li CX, et al. Alternatively activated (M2) macrophages promote tumour growth and invasiveness in hepatocellular carcinoma. J Hepatol. 2015;62(3):607–16.
Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41(1):49–61.
Chen C, Wang Z, Ding Y, Qin Y. Tumor microenvironment-mediated immune evasion in hepatocellular carcinoma. Front Immunol. 2023;14:14.
Yan W, Liu X, Ma H, Zhang H, Song X, Gao L, et al. Tim-3 fosters HCC development by enhancing TGF-β-mediated alternative activation of macrophages. Gut. 2015;64(10):1593–604.
Zhou J, Ding T, Pan W, Zhu LY, Li L, Zheng L. Increased intratumoral regulatory T cells are related to intratumoral macrophages and poor prognosis in hepatocellular carcinoma patients. Int J Cancer. 2009;125(7):1640–8.
Kuang DM, Peng C, Zhao Q, Wu Y, Chen MS, Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma promote expansion of memory T helper 17 cells. Hepatology. 2010;51(1):154–64.
Zhao F, Hoechst B, Gamrekelashvili J, Ormandy LA, Voigtländer T, Wedemeyer H, et al. Human CCR4 + CCR6 + Th17 cells suppress autologous CD8 + T cell responses. J Immunol. 2012;188(12):6055–62.
Han Y, Chen Z, Yang Y, Jiang Z, Gu Y, Liu Y, et al. Human CD14 + CTLA-4 + regulatory dendritic cells suppress T-cell response by cytotoxic T-lymphocyte antigen-4-dependent IL-10 and indoleamine-2,3-dioxygenase production in hepatocellular carcinoma. Hepatology. 2014;59(2):567–79.
Xiao X, Lao XM, Chen MM, Liu RX, Wei Y, Ouyang FZ, et al. PD-1hi identifies a Novel Regulatory B-cell Population in Human Hepatoma that promotes Disease Progression. Cancer Discov. 2016;6(5):546–59.
Liu K, Zan P, Li Z, Lu H, Liu P, Zhang L, et al. Engineering bimetallic polyphenol for mild photothermal osteosarcoma therapy and immune microenvironment remodeling by activating pyroptosis and cGAS-STING pathway. Adv Healthc Mater. 2024;13(22):e2400623.
Ghaffari S, Rezaei N. Eosinophils in the tumor microenvironment: implications for cancer immunotherapy. J Transl Med. 2023;21(1):551.
Van Bruggen S, Jarrot PA, Thomas E, Sheehy CE, Silva CMS, Hsu AY, et al. NLRP3 is essential for neutrophil polarization and chemotaxis in response to leukotriene B4 gradient. Proc Natl Acad Sci U S A. 2023;120(35):e2303814120.
Wang Y, Lyu Z, Qin Y, Wang X, Sun L, Zhang Y, et al. FOXO1 promotes tumor progression by increased M2 macrophage infiltration in esophageal squamous cell carcinoma. Theranostics. 2020;10(25):11535–48.
Ouyang FZ, Wu RQ, Wei Y, Liu RX, Yang D, Xiao X, et al. Dendritic cell-elicited B-cell activation fosters immune privilege via IL-10 signals in hepatocellular carcinoma. Nat Commun. 2016;7:13453.
Katsuno Y, Derynck R. Epithelial plasticity, epithelial-mesenchymal transition, and the TGF-β family. Dev Cell. 2021;56(6):726–46.
Ning Y, Lenz HJ. Targeting IL-8 in colorectal cancer. Expert Opin Ther Targets. 2012;16(5):491–7.
Zhang YE, Stuelten CH. Alternative splicing in EMT and TGF-β signaling during cancer progression. Semin Cancer Biol. 2024;101:1–11.
Darvishi B, Majidzadeh AK, Ghadirian R, Mosayebzadeh M, Farahmand L. Recruited bone marrow derived cells, local stromal cells and IL-17 at the front line of resistance development to anti-VEGF targeted therapies. Life Sci. 2019;217:34–40.
Ribatti D. Mast cells and macrophages exert beneficial and detrimental effects on tumor progression and angiogenesis. Immunol Lett. 2013;152(2):83–8.
Giansanti M, Theinert T, Boeing SK, Haas D, Schlegel PG, Vacca P, et al. Exploiting autophagy balance in T and NK cells as a new strategy to implement adoptive cell therapies. Mol Cancer. 2023;22(1):201.
Shyh-Chang N. Metabolic changes during Cancer Cachexia Pathogenesis. Adv Exp Med Biol. 2017;1026:233–49.
Zhang H, Liu Y, Liu J, Chen J, Wang J, Hua H, et al. cAMP-PKA/EPAC signaling and cancer: the interplay in tumor microenvironment. J Hematol Oncol. 2024;17(1):5.
Nishida N, Kudo M. Genetic/Epigenetic alteration and Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: transforming the Immune Microenvironment with Molecular-targeted agents. Liver Cancer. 2024;13(2):136–49.
Cai X, Chen J, Xu H, Liu S, Jiang QX, Halfmann R, et al. Prion-like polymerization underlies signal transduction in antiviral immune defense and inflammasome activation. Cell. 2014;156(6):1207–22.
Afonina IS, Müller C, Martin SJ, Beyaert R. Proteolytic Processing of Interleukin-1 Family cytokines: variations on a common theme. Immunity. 2015;42(6):991–1004.
Sborgi L, Rühl S, Mulvihill E, Pipercevic J, Heilig R, Stahlberg H, et al. GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. Embo j. 2016;35(16):1766–78.
Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H, et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 2016;535(7610):153–8.
Wiggins KA, Parry AJ, Cassidy LD, Humphry M, Webster SJ, Goodall JC, et al. IL-1α cleavage by inflammatory caspases of the noncanonical inflammasome controls the senescence-associated secretory phenotype. Aging Cell. 2019;18(3):e12946.
Gringhuis SI, Kaptein TM, Wevers BA, Theelen B, van der Vlist M, Boekhout T, et al. Dectin-1 is an extracellular pathogen sensor for the induction and processing of IL-1β via a noncanonical caspase-8 inflammasome. Nat Immunol. 2012;13(3):246–54.
Ketelut-Carneiro N, Ghosh S, Levitz SM, Fitzgerald KA, da Silva JS. A dectin-1-Caspase-8 pathway licenses canonical Caspase-1 inflammasome activation and Interleukin-1β release in response to a pathogenic fungus. J Infect Dis. 2018;217(2):329–39.
Vajjhala PR, Lu A, Brown DL, Pang SW, Sagulenko V, Sester DP, et al. The Inflammasome adaptor ASC induces Procaspase-8 death Effector Domain filaments. J Biol Chem. 2015;290(49):29217–30.
Antonopoulos C, Russo HM, El Sanadi C, Martin BN, Li X, Kaiser WJ, et al. Caspase-8 as an Effector and Regulator of NLRP3 Inflammasome Signaling. J Biol Chem. 2015;290(33):20167–84.
Kudo Y, Sugimoto M, Arias E, Kasashima H, Cordes T, Linares JF, et al. PKCλ/ι loss induces Autophagy, oxidative phosphorylation, and NRF2 to promote Liver Cancer Progression. Cancer Cell. 2020;38(2):247–62.e11.
Zhang Y, Yang H, Sun M, He T, Liu Y, Yang X, et al. Alpinumisoflavone suppresses hepatocellular carcinoma cell growth and metastasis via NLRP3 inflammasome-mediated pyroptosis. Pharmacol Rep. 2020;72(5):1370–82.
Ding F, Liu J, Ai K, Xu C, Mao X, Liu Z, et al. Simultaneous activation of Pyroptosis and cGAS-STING pathway with Epigenetic/ photodynamic nanotheranostic for enhanced Tumor Photoimmunotherapy. Adv Mater. 2024;36(7):e2306419.
Ling YY, Xia XY, Hao L, Wang WJ, Zhang H, Liu LY, et al. Simultaneous photoactivation of cGAS-STING pathway and pyroptosis by platinum(II) triphenylamine complexes for Cancer Immunotherapy. Angew Chem Int Ed Engl. 2022;61(43):e202210988.
Xuzhang W, Lu T, Jin W, Yu Y, Li Z, Shen L, et al. Cisplatin-induced pyroptosis enhances the efficacy of PD-L1 inhibitor in small-cell Lung Cancer via GSDME/IL12/CD4Tem Axis. Int J Biol Sci. 2024;20(2):537–53.
Karmakar M, Minns M, Greenberg EN, Diaz-Aponte J, Pestonjamasp K, Johnson JL, et al. N-GSDMD trafficking to neutrophil organelles facilitates IL-1β release independently of plasma membrane pores and pyroptosis. Nat Commun. 2020;11(1):2212.
Esmailbeig M, Ghaderi A. Interleukin-18: a regulator of cancer and autoimmune diseases. Eur Cytokine Netw. 2017;28(4):127–40.
Greenberg AH. Granzyme B-induced apoptosis. Adv Exp Med Biol. 1996;406:219–28.
Zhuo L, Chen X, Sun Y, Wang Y, Shi Y, Bu L, et al. Rapamycin Inhibited pyroptosis and reduced the release of IL-1β and IL-18 in the septic response. Biomed Res Int. 2020;2020:5960375.
Sutton C, Brereton C, Keogh B, Mills KH, Lavelle EC. A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis. J Exp Med. 2006;203(7):1685–91.
Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol. 2007;8(9):942–9.
Deng J, Yu XQ, Wang PH. Inflammasome activation and Th17 responses. Mol Immunol. 2019;107:142–64.
Van Den Eeckhout B, Tavernier J, Gerlo S. Interleukin-1 as Innate Mediator of T Cell Immunity. Front Immunol. 2020;11:621931.
Ikeda S, Saijo S, Murayama MA, Shimizu K, Akitsu A, Iwakura Y. Excess IL-1 signaling enhances the development of Th17 cells by downregulating TGF-β-induced Foxp3 expression. J Immunol. 2014;192(4):1449–58.
Chan BCL, Lam CWK, Tam LS, Wong CK. IL33: roles in allergic inflammation and therapeutic perspectives. Front Immunol. 2019;10: 364.
Hu G, Wu P, Cheng P, Zhang Z, Wang Z, Yu X, et al. Tumor-infiltrating CD39(+)γδTregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology. 2017;6(2):e1277305.
Feng WQ, Zhang YC, Xu ZQ, Yu SY, Huo JT, Tuersun A, et al. IL-17A-mediated mitochondrial dysfunction induces pyroptosis in colorectal cancer cells and promotes CD8 + T-cell tumour infiltration. J Transl Med. 2023;21(1):335.
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, et al. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther. 2024;9(1):61.
Kang H, Seo E, Oh YS, Jun HS. TGF-β activates NLRP3 inflammasome by an autocrine production of TGF-β in LX-2 human hepatic stellate cells. Mol Cell Biochem. 2022;477(5):1329–38.
Tu CE, Hu Y, Zhou P, Guo X, Gu C, Zhang Y, et al. Lactate and TGF-β antagonistically regulate inflammasome activation in the tumor microenvironment. J Cell Physiol. 2021;236(6):4528–37.
Huang Y, Yang W, Yang L, Wang T, Li C, Yu J, et al. Nrf2 inhibition increases sensitivity to chemotherapy of colorectal cancer by promoting ferroptosis and pyroptosis. Sci Rep. 2023;13(1):14359.
Shen W, Ma X, Shao D, Wu X, Wang S, Zheng J, et al. Neutrophil Extracellular traps mediate bovine endometrial epithelial cell pyroptosis in dairy cows with Endometritis. Int J Mol Sci. 2022;23(22):14013.
Wang W, Prokopec JS, Zhang Y, Sukhoplyasova M, Shinglot H, Wang MT, et al. Sensing plasma membrane pore formation induces chemokine production in survivors of regulated necrosis. Dev Cell. 2022;57(2):228 – 45.e6.
Anderton H, Wicks IP, Silke J. Cell death in chronic inflammation: breaking the cycle to treat rheumatic disease. Nat Rev Rheumatol. 2020;16(9):496–513.
Chen W, He Y, Zhou G, Chen X, Ye Y, Zhang G, et al. Multiomics characterization of pyroptosis in the tumor microenvironment and therapeutic relevance in metastatic melanoma. BMC Med. 2024;22(1):24.
Bruchard M, Rebé C, Derangère V, Togbé D, Ryffel B, Boidot R, et al. The receptor NLRP3 is a transcriptional regulator of TH2 differentiation. Nat Immunol. 2015;16(8):859–70.
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.
Tengesdal IW, Menon DR, Osborne DG, Neff CP, Powers NE, Gamboni F, et al. Targeting tumor-derived NLRP3 reduces melanoma progression by limiting MDSCs expansion. Proc Natl Acad Sci U S A. 2021;118(10):e2000915118.
Markwardt F. Human P2X7 receptors - Properties of single ATP-gated ion channels. Biochem Pharmacol. 2021;187:114307.
Sharif H, Wang L, Wang WL, Magupalli VG, Andreeva L, Qiao Q, et al. Structural mechanism for NEK7-licensed activation of NLRP3 inflammasome. Nature. 2019;570(7761):338–43.
Wang Q, Imamura R, Motani K, Kushiyama H, Nagata S, Suda T. Pyroptotic cells externalize eat-me and release find-me signals and are efficiently engulfed by macrophages. Int Immunol. 2013;25(6):363–72.
Tan G, Huang C, Chen J, Zhi F. HMGB1 released from GSDME-mediated pyroptotic epithelial cells participates in the tumorigenesis of colitis-associated colorectal cancer through the ERK1/2 pathway. J Hematol Oncol. 2020;13(1):149.
McKenzie BA, Mamik MK, Saito LB, Boghozian R, Monaco MC, Major EO, et al. Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc Natl Acad Sci U S A. 2018;115(26):E6065–74.
Li M, Jiang P, Yang Y, Xiong L, Wei S, Wang J, et al. The role of pyroptosis and gasdermin family in tumor progression and immune microenvironment. Exp Hematol Oncol. 2023;12(1):103.
Burdette DL, Vance RE. STING and the innate immune response to nucleic acids in the cytosol. Nat Immunol. 2013;14(1):19–26.
Dhanwani R, Takahashi M, Sharma S. Cytosolic sensing of immuno-stimulatory DNA, the enemy within. Curr Opin Immunol. 2018;50:82–7.
Jiang M, Chen P, Wang L, Li W, Chen B, Liu Y, et al. cGAS-STING, an important pathway in cancer immunotherapy. J Hematol Oncol. 2020;13(1):81.
Hopfner KP, Hornung V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat Rev Mol Cell Biol. 2020;21(9):501–21.
Manes NP, Nita-Lazar A. Molecular mechanisms of the toll-like receptor, STING, MAVS, Inflammasome, and Interferon pathways. mSystems. 2021;6(3):e0033621.
Wang Y, Ning X, Gao P, Wu S, Sha M, Lv M, et al. Inflammasome activation triggers caspase-1-Mediated cleavage of cGAS to regulate responses to DNA virus infection. Immunity. 2017;46(3):393–404.
Liu J, Zhou J, Luan Y, Li X, Meng X, Liao W, et al. cGAS-STING, inflammasomes and pyroptosis: an overview of crosstalk mechanism of activation and regulation. Cell Commun Signal. 2024;22(1):22.
Zheng Y, Liu Q, Wu Y, Ma L, Zhang Z, Liu T, et al. Zika virus elicits inflammation to evade antiviral response by cleaving cGAS via NS1-caspase-1 axis. Embo j. 2018;37(18):e99347.
Eren E, Berber M, Özören N. NLRC3 protein inhibits inflammation by disrupting NALP3 inflammasome assembly via competition with the adaptor protein ASC for pro-caspase-1 binding. J Biol Chem. 2017;292(30):12691–701.
Yan S, Shen H, Lian Q, Jin W, Zhang R, Lin X, et al. Deficiency of the AIM2-ASC Signal uncovers the STING-Driven overreactive response of type I IFN and reciprocal depression of protective IFN-γ immunity in mycobacterial infection. J Immunol. 2018;200(3):1016–26.
Corrales L, Woo SR, Williams JB, McWhirter SM, Dubensky TW Jr, Gajewski TF. Antagonism of the STING pathway via activation of the AIM2 inflammasome by intracellular DNA. J Immunol. 2016;196(7):3191–8.
Wu T, Gao J, Liu W, Cui J, Yang M, Guo W, et al. NLRP3 protects mice from radiation-induced colon and skin damage via attenuating cGAS-STING signaling. Toxicol Appl Pharmacol. 2021;418:115495.
Yang Y, Lang X, Sun S, Gao C, Hu J, Ding S, et al. NLRP2 negatively regulates antiviral immunity by interacting with TBK1. Eur J Immunol. 2018;48(11):1817–25.
Li X, Deng M, Petrucelli AS, Zhu C, Mo J, Zhang L, et al. Viral DNA binding to NLRC3, an inhibitory nucleic acid Sensor, unleashes STING, a cyclic dinucleotide receptor that activates type I Interferon. Immunity. 2019;50(3.e6):591–9.
Chui AJ, Okondo MC, Rao SD, Gai K, Griswold AR, Johnson DC, et al. N-terminal degradation activates the NLRP1B inflammasome. Science. 2019;364(6435):82–5.
Zhang L, Mo J, Swanson KV, Wen H, Petrucelli A, Gregory SM, et al. NLRC3, a member of the NLR family of proteins, is a negative regulator of innate immune signaling induced by the DNA sensor STING. Immunity. 2014;40(3):329–41.
Guo H, König R, Deng M, Riess M, Mo J, Zhang L, et al. NLRX1 sequesters STING to negatively regulate the Interferon Response, thereby facilitating the replication of HIV-1 and DNA viruses. Cell Host Microbe. 2016;19(4):515–28.
Cui J, Li Y, Zhu L, Liu D, Songyang Z, Wang HY, et al. NLRP4 negatively regulates type I interferon signaling by targeting the kinase TBK1 for degradation via the ubiquitin ligase DTX4. Nat Immunol. 2012;13(4):387–95.
Ellwanger K, Becker E, Kienes I, Sowa A, Postma Y, Cardona Gloria Y, et al. The NLR family pyrin domain-containing 11 protein contributes to the regulation of inflammatory signaling. J Biol Chem. 2018;293(8):2701–10.
Gaidt MM, Ebert TS, Chauhan D, Ramshorn K, Pinci F, Zuber S, et al. The DNA inflammasome in human myeloid cells is initiated by a STING-Cell death program Upstream of NLRP3. Cell. 2017;171(5):1110–24.e18.
Li N, Zhou H, Wu H, Wu Q, Duan M, Deng W, et al. STING-IRF3 contributes to lipopolysaccharide-induced cardiac dysfunction, inflammation, apoptosis and pyroptosis by activating NLRP3. Redox Biol. 2019;24:101215.
McLemore AF, Hou HA, Meyer BS, Lam NB, Ward GA, Aldrich AL, et al. Somatic gene mutations expose cytoplasmic DNA to co-opt the cGAS/STING/NLRP3 axis in myelodysplastic syndromes. JCI Insight. 2022;7(15):e159430.
Wang W, Hu D, Wu C, Feng Y, Li A, Liu W, et al. STING promotes NLRP3 localization in ER and facilitates NLRP3 deubiquitination to activate the inflammasome upon HSV-1 infection. PLoS Pathog. 2020;16(3):e1008335.
Xiao Y, Zhao C, Tai Y, Li B, Lan T, Lai E, et al. STING mediates hepatocyte pyroptosis in liver fibrosis by Epigenetically activating the NLRP3 inflammasome. Redox Biol. 2023;62:102691.
Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–32.
Faustin B, Lartigue L, Bruey JM, Luciano F, Sergienko E, Bailly-Maitre B, et al. Reconstituted NALP1 inflammasome reveals two-step mechanism of caspase-1 activation. Mol Cell. 2007;25(5):713–24.
Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–26.
Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009;458(7237):514–8.
Muruve DA, Pétrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452(7183):103–7.
Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.
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. 2020;19(10):1089–104.
Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target Ther. 2021;6(1):402.
Lee S, Hirohama M, Noguchi M, Nagata K, Kawaguchi A. Influenza A virus infection triggers pyroptosis and apoptosis of respiratory epithelial cells through the type I Interferon Signaling Pathway in a mutually exclusive manner. J Virol. 2018;92(14):e00396.
Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol. 2015;15(6):388–400.
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.
Gong W, Fu H, Yang K, Zheng T, Guo K, Zhao W. 4-Octyl itaconate blocks GSDMB-mediated pyroptosis and restricts inflammation by inactivating granzyme A. Cell Prolif. 2024:e13711. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cpr.13711.
Li L, Jiang M, Qi L, Wu Y, Song D, Gan J, et al. Pyroptosis, a new bridge to tumor immunity. Cancer Sci. 2021;112(10):3979–94.
Long Y, Jia X, Chu L. Insight into the structure, function and the tumor suppression effect of gasdermin E. Biochem Pharmacol. 2024;226:116348.
Gong C, Mu H, Luo J, Zhang R, Hu D, Chen Z, et al. Euphohelioscopin a enhances NK cell antitumor immunity through GSDME-triggered pyroptosis. J Leukoc Biol. 2024;116(3):621–31.
Zhang Y, Cai X, Wang B, Zhang B, Xu Y. Exploring the molecular mechanisms of the involvement of GZMB-Caspase-3-GSDME pathway in the progression of rheumatoid arthritis. Mol Immunol. 2023;161:82–90.
Zhang L, Bai H, Zhou J, Ye L, Gao L. Role of tumor cell pyroptosis in anti-tumor immunotherapy. Cell Insight. 2024;3(3):100153.
Meybodi SM, Ejlalidiz M, Manshadi MR, Raeisi M, Zarin M, Kalhor Z, et al. Crosstalk between hypoxia-induced pyroptosis and immune escape in cancer: from mechanisms to therapy. Crit Rev Oncol Hematol. 2024;197:104340.
Fang Y, Tang Y, Huang B. Pyroptosis: A road to next-generation cancer immunotherapy. Semin Immunol. 2023;68:101782.
Zheng Y, Yuan D, Zhang F, Tang R. A systematic pan-cancer analysis of the gasdermin (GSDM) family of genes and their correlation with prognosis, the tumor microenvironment, and drug sensitivity. Front Genet. 2022;13:926796.
Huang J, Fan P, Liu M, Weng C, Fan G, Zhang T, et al. Famotidine promotes inflammation by triggering cell pyroptosis in gastric cancer cells. BMC Pharmacol Toxicol. 2021;22(1):62.
Tang YL, Tao Y, Zhu L, Shen JL, Cheng H. Role of NLRP3 inflammasome in hepatocellular carcinoma: a double-edged sword. Int Immunopharmacol. 2023;118:110107.
Yan H, Luo B, Wu X, Guan F, Yu X, Zhao L, et al. Cisplatin induces pyroptosis via activation of MEG3/NLRP3/caspase-1/GSDMD pathway in Triple-negative breast Cancer. Int J Biol Sci. 2021;17(10):2606–21.
Lyu H, Ni H, Huang J, Yu G, Zhang Z, Zhang Q. VX-765 prevents intestinal ischemia-reperfusion injury by inhibiting NLRP3 inflammasome. Tissue Cell. 2022;75:101718.
Wen S, Deng F, Li L, Xu L, Li X, Fan Q. VX-765 ameliorates renal injury and fibrosis in diabetes by regulating caspase-1-mediated pyroptosis and inflammation. J Diabetes Investig. 2022;13(1):22–33.
Colunga AG, Laing JM, Aurelian L. The HSV-2 mutant DeltaPK induces melanoma oncolysis through nonredundant death programs and associated with autophagy and pyroptosis proteins. Gene Ther. 2010;17(3):315–27.
Brumatti G, Ma C, Lalaoui N, Nguyen NY, Navarro M, Tanzer MC, et al. The caspase-8 inhibitor emricasan combines with the SMAC mimetic birinapant to induce necroptosis and treat acute myeloid leukemia. Sci Transl Med. 2016;8(339):339ra69.
Alphonse MP, Rubens JH, Ortines RV, Orlando NA, Patel AM, Dikeman D, et al. Pan-caspase inhibition as a potential host-directed immunotherapy against MRSA and other bacterial skin infections. Sci Transl Med. 2021;13:601.
Wittwer NL, Staudacher AH, Liapis V, Cardarelli P, Warren H, Brown MP. An anti-mesothelin targeting antibody drug conjugate induces pyroptosis and ignites antitumor immunity in mouse models of cancer. J Immunother Cancer. 2023;11(3):e006274.
Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et al. DS-8201a, A Novel HER2-Targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a Promising Antitumor Efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22(20):5097–108.
Horwitz S, O’Connor OA, Pro B, Trümper L, Iyer S, Advani R, et al. The ECHELON-2 trial: 5-year results of a randomized, phase III study of brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma. Ann Oncol. 2022;33(3):288–98.
Goldenberg DM, Sharkey RM. Sacituzumab Govitecan, a novel, third-generation, antibody-drug conjugate (ADC) for cancer therapy. Expert Opin Biol Ther. 2020;20(8):871–85.
Vafaei S, Zekiy AO, Khanamir RA, Zaman BA, Ghayourvahdat A, Azimizonuzi H, et al. Combination therapy with immune checkpoint inhibitors (ICIs); a new frontier. Cancer Cell Int. 2022;22(1):2.
Nguyen HM, Bommareddy PK, Silk AW, Saha D. Optimal timing of PD-1 blockade in combination with oncolytic virus therapy. Semin Cancer Biol. 2022;86(Pt 3):971–80.
Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in Cancer Immunotherapy. Comput Struct Biotechnol J. 2019;17:661–74.
Mathew M, Enzler T, Shu CA, Rizvi NA. Combining chemotherapy with PD-1 blockade in NSCLC. Pharmacol Ther. 2018;186:130–7.
Zhang J, Fu L, Yasuda-Yoshihara N, Yonemura A, Wei F, Bu L, et al. IL-1β derived from mixed-polarized macrophages activates fibroblasts and synergistically forms a cancer-promoting microenvironment. Gastric Cancer. 2023;26(2):187–202.
Diwanji R, O’Brien NA, Choi JE, Nguyen B, Laszewski T, Grauel AL, et al. Targeting the IL1β pathway for Cancer Immunotherapy remodels the Tumor Microenvironment and enhances Antitumor Immune responses. Cancer Immunol Res. 2023;11(6):777–91.
Hsieh CY, Lin CC, Huang YW, Chen JH, Tsou YA, Chang LC, et al. Macrophage secretory IL-1β promotes docetaxel resistance in head and neck squamous carcinoma via SOD2/CAT-ICAM1 signaling. JCI Insight. 2022;7(23):e157285.
Soler MF, Abaurrea A, Azcoaga P, Araujo AM, Caffarel MM. New perspectives in cancer immunotherapy: targeting IL-6 cytokine family. J Immunother Cancer. 2023;11(11):e007530.
Browning L, Patel MR, Horvath EB, Tawara K, Jorcyk CL. IL-6 and ovarian cancer: inflammatory cytokines in promotion of metastasis. Cancer Manag Res. 2018;10:6685–93.
Waldner MJ, Neurath MF. TGFβ and the Tumor Microenvironment in Colorectal Cancer. Cells. 2023;12(8):1139.
Rastogi S, Mishra SS, Arora MK, Kaithwas G, Banerjee S, Ravichandiran V, et al. Lactate acidosis and simultaneous recruitment of TGF-β leads to alter plasticity of hypoxic cancer cells in tumor microenvironment. Pharmacol Ther. 2023;250:108519.
Yi M, Li T, Niu M, Wu Y, Zhao Z, Wu K. TGF-β: a novel predictor and target for anti-PD-1/PD-L1 therapy. Front Immunol. 2022;13: 1061394.
Tschernia NP, Gulley JL. Tumor in the Crossfire: inhibiting TGF-β to Enhance Cancer Immunotherapy. BioDrugs. 2022;36(2):153–80.
Shi X, Yang J, Deng S, Xu H, Wu D, Zeng Q, et al. TGF-β signaling in the tumor metabolic microenvironment and targeted therapies. J Hematol Oncol. 2022;15(1):135.
Li L, Wen Q, Ding R. Therapeutic targeting of VEGF and/or TGF-β to enhance anti-PD-(L)1 therapy: the evidence from clinical trials. Front Oncol. 2022;12:905520.
Gulley JL, Schlom J, Barcellos-Hoff MH, Wang XJ, Seoane J, Audhuy F, et al. Dual inhibition of TGF-β and PD-L1: a novel approach to cancer treatment. Mol Oncol. 2022;16(11):2117–34.
Chan MK, Chung JY, Tang PC, Chan AS, Ho JY, Lin TP, et al. TGF-β signaling networks in the tumor microenvironment. Cancer Lett. 2022;550:215925.
Shao R, Lou X, Xue J, Ning D, Chen G, Jiang L. Review: the role of GSDMD in sepsis. Inflamm Res. 2022;71(10–11):1191–202.
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.
Yu F, Tan W, Chen Z, Shen X, Mo X, Mo X, et al. Nitidine chloride induces caspase 3/GSDME-dependent pyroptosis by inhibting PI3K/Akt pathway in lung cancer. Chin Med. 2022;17(1):115.
Yao F, Jin Z, Zheng Z, Lv X, Ren L, Yang J, et al. HDAC11 promotes both NLRP3/caspase-1/GSDMD and caspase-3/GSDME pathways causing pyroptosis via ERG in vascular endothelial cells. Cell Death Discov. 2022;8(1):112.
Gielecińska A, Kciuk M, Yahya EB, Ainane T, Mujwar S, Kontek R. Apoptosis, necroptosis, and pyroptosis as alternative cell death pathways induced by chemotherapeutic agents? Biochim Biophys Acta Rev Cancer. 2023;1878(6):189024.
Wang WJ, Chen D, Jiang MZ, Xu B, Li XW, Chu Y, et al. Downregulation of gasdermin D promotes gastric cancer proliferation by regulating cell cycle-related proteins. J Dig Dis. 2018;19(2):74–83.
Yue E, Tuguzbaeva G, Chen X, Qin Y, Li A, Sun X, et al. Anthocyanin is involved in the activation of pyroptosis in oral squamous cell carcinoma. Phytomedicine. 2019;56:286–94.
Sannino F, Sansone C, Galasso C, Kildgaard S, Tedesco P, Fani R, et al. Pseudoalteromonas haloplanktis TAC125 produces 4-hydroxybenzoic acid that induces pyroptosis in human A459 lung adenocarcinoma cells. Sci Rep. 2018;8(1):1190.
Johnson DC, Taabazuing CY, Okondo MC, Chui AJ, Rao SD, Brown FC, et al. DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia. Nat Med. 2018;24(8):1151–6.
Pizato N, Luzete BC, Kiffer L, Corrêa LH, de Oliveira Santos I, Assumpção JAF, et al. Omega-3 docosahexaenoic acid induces pyroptosis cell death in triple-negative breast cancer cells. Sci Rep. 2018;8(1):1952.
Nadeem S, Yang C, Du Y, Li F, Chen Z, Zhou Y, et al. A virus-spike tumor-activatable Pyroptotic Agent. Small. 2021;17(8):e2006599.
Ploetz E, Zimpel A, Cauda V, Bauer D, Lamb DC, Haisch C, et al. Metal-Organic Framework nanoparticles induce pyroptosis in cells controlled by the Extracellular pH. Adv Mater. 2020;32(19):e1907267.
Qiao L, Wu X, Zhang J, Liu L, Sui X, Zhang R, et al. α-NETA induces pyroptosis of epithelial ovarian cancer cells through the GSDMD/caspase-4 pathway. Faseb j. 2019;33(11):12760–7.
Lu Y, He W, Huang X, He Y, Gou X, Liu X, et al. Strategies to package recombinant Adeno-Associated Virus expressing the N-terminal gasdermin domain for tumor treatment. Nat Commun. 2021;12(1):7155.
Jiang X, Wang J, Deng X, Xiong F, Ge J, Xiang B, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18(1):10.
Nivolumab Approved for Liver Cancer. Cancer Discov. 2017;7(11):Of3.
Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020;382(20):1894–905.
Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. 2011;332(6029):600–3.
Heichler C, Scheibe K, Schmied A, Geppert CI, Schmid B, Wirtz S, et al. STAT3 activation through IL-6/IL-11 in cancer-associated fibroblasts promotes colorectal tumour development and correlates with poor prognosis. Gut. 2020;69(7):1269–82.
Ham IH, Oh HJ, Jin H, Bae CA, Jeon SM, Choi KS, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 2019;18(1):68.
Dijkgraaf EM, Santegoets SJ, Reyners AK, Goedemans R, Wouters MC, Kenter GG, et al. A phase I trial combining carboplatin/doxorubicin with tocilizumab, an anti-IL-6R monoclonal antibody, and interferon-α2b in patients with recurrent epithelial ovarian cancer. Ann Oncol. 2015;26(10):2141–9.
Liu H, Shen J, Lu K. IL-6 and PD-L1 blockade combination inhibits hepatocellular carcinoma cancer development in mouse model. Biochem Biophys Res Commun. 2017;486(2):239–44.
Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10105):1833–42.
Wong CC, Baum J, Silvestro A, Beste MT, Bharani-Dharan B, Xu S, et al. Inhibition of IL1β by Canakinumab May be effective against Diverse Molecular subtypes of Lung Cancer: an exploratory analysis of the CANTOS Trial. Cancer Res. 2020;80(24):5597–605.
Yuan B, Clowers MJ, Velasco WV, Peng S, Peng Q, Shi Y, et al. Targeting IL-1β as an immunopreventive and therapeutic modality for K-ras-mutant lung cancer. JCI Insight. 2022;7(11):e157788.
Guo B, Fu S, Zhang J, Liu B, Li Z. Targeting inflammasome/IL-1 pathways for cancer immunotherapy. Sci Rep. 2016;6: 36107.
Voigt C, May P, Gottschlich A, Markota A, Wenk D, Gerlach I, et al. Cancer cells induce interleukin-22 production from memory CD4(+) T cells via interleukin-1 to promote tumor growth. Proc Natl Acad Sci U S A. 2017;114(49):12994–9.
Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med. 2018;24(6):731–8.
Gazeau N, Liang EC, Wu QV, Voutsinas JM, Barba P, Iacoboni G, et al. Anakinra for Refractory Cytokine Release Syndrome or Immune Effector Cell-Associated Neurotoxicity Syndrome after Chimeric Antigen Receptor T Cell Therapy. Transpl Cell Ther. 2023;29(7):430–7.
Kovacs RJ, Maldonado G, Azaro A, Fernández MS, Romero FL, Sepulveda-Sánchez JM, et al. Cardiac Safety of TGF-β receptor I kinase inhibitor LY2157299 monohydrate in Cancer patients in a first-in-human dose study. Cardiovasc Toxicol. 2015;15(4):309–23.
Faivre S, Santoro A, Kelley RK, Gane E, Costentin CE, Gueorguieva I, et al. Novel transforming growth factor beta receptor I kinase inhibitor galunisertib (LY2157299) in advanced hepatocellular carcinoma. Liver Int. 2019;39(8):1468–77.
Mariathasan S, Turley SJ, Nickles D, Castiglioni A, Yuen K, Wang Y, et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554(7693):544–8.
Ravi R, Noonan KA, Pham V, Bedi R, Zhavoronkov A, Ozerov IV, et al. Bifunctional immune checkpoint-targeted antibody-ligand traps that simultaneously disable TGFβ enhance the efficacy of cancer immunotherapy. Nat Commun. 2018;9(1):741.
Xia Q, Zhang FF, Geng F, Liu CL, Xu P, Lu ZZ, et al. Anti-tumor effects of DNA vaccine targeting human fibroblast activation protein α by producing specific immune responses and altering tumor microenvironment in the 4T1 murine breast cancer model. Cancer Immunol Immunother. 2016;65(5):613–24.
Ciardiello D, Elez E, Tabernero J, Seoane J. Clinical development of therapies targeting TGFβ: current knowledge and future perspectives. Ann Oncol. 2020;31(10):1336–49.
Leon-Cabrera S, Schwertfeger KL, Terrazas LI. Inflammation as a target in Cancer Therapy. Mediators Inflamm. 2019;2019:1971698.
Wang Q, Wang Y, Ding J, Wang C, Zhou X, Gao W, et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature. 2020;579(7799):421–6.
Gao Y, Zhang H, Zhou N, Xu P, Wang J, Gao Y, et al. Methotrexate-loaded tumour-cell-derived microvesicles can relieve biliary obstruction in patients with extrahepatic cholangiocarcinoma. Nat Biomed Eng. 2020;4(7):743–53.
Fan JX, Deng RH, Wang H, Liu XH, Wang XN, Qin R, et al. Epigenetics-based Tumor cells pyroptosis for enhancing the Immunological Effect of Chemotherapeutic Nanocarriers. Nano Lett. 2019;19(11):8049–58.
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.
Berger Fridman I, Kostas J, Gregus M, Ray S, Sullivan MR, Ivanov AR, et al. High-throughput microfluidic 3D biomimetic model enabling quantitative description of the human breast tumor microenvironment. Acta Biomater. 2021;132:473–88.
Bernard V, Semaan A, Huang J, San Lucas FA, Mulu FC, Stephens BM, et al. Single-cell transcriptomics of pancreatic Cancer precursors demonstrates epithelial and microenvironmental heterogeneity as an early event in neoplastic progression. Clin Cancer Res. 2019;25(7):2194–205.
Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2, Treacy D, Trombetta JJ, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352(6282):189–96.
Yost KE, Satpathy AT, Wells DK, Qi Y, Wang C, Kageyama R, et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat Med. 2019;25(8):1251–9.
Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221:107753.
Elhanani O, Ben-Uri R, Keren L. Spatial profiling technologies illuminate the tumor microenvironment. Cancer Cell. 2023;41(3):404–20.
Nasir I, McGuinness C, Poh AR, Ernst M, Darcy PK, Britt KL. Tumor macrophage functional heterogeneity can inform the development of novel cancer therapies. Trends Immunol. 2023;44(12):971–85.
Wu K, Lin K, Li X, Yuan X, Xu P, Ni P, et al. Redefining Tumor-Associated macrophage subpopulations and functions in the Tumor Microenvironment. Front Immunol. 2020;11:1731.
Dhainaut M, Rose SA, Akturk G, Wroblewska A, Nielsen SR, Park ES, et al. Spatial CRISPR genomics identifies regulators of the tumor microenvironment. Cell. 2022;185(7):1223–39.e20.
Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 2016;27(8):1482–92.
Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, et al. The cancer metabolic reprogramming and immune response. Mol Cancer. 2021;20(1):28.
Denk D, Greten FR. Inflammation: the incubator of the tumor microenvironment. Trends Cancer. 2022;8(11):901–14.
Lasry A, Nadorp B, Fornerod M, Nicolet D, Wu H, Walker CJ, et al. An inflammatory state remodels the immune microenvironment and improves risk stratification in acute myeloid leukemia. Nat Cancer. 2023;4(1):27–42.
Sharma BR, Kanneganti TD. NLRP3 inflammasome in cancer and metabolic diseases. Nat Immunol. 2021;22(5):550–9.
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, et al. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther. 2022;7(1):286.
Zhou S, Liu J, Wan A, Zhang Y, Qi X. Epigenetic regulation of diverse cell death modalities in cancer: a focus on pyroptosis, ferroptosis, cuproptosis, and disulfidptosis. J Hematol Oncol. 2024;17(1):22.
Zhang Z, Zhang Y, Lieberman J. Lighting a fire: can we harness pyroptosis to Ignite Antitumor Immunity? Cancer Immunol Res. 2021;9(1):2–7.
Philippou Y, Sjoberg H, Lamb AD, Camilleri P, Bryant RJ. Harnessing the potential of multimodal radiotherapy in prostate cancer. Nat Rev Urol. 2020;17(6):321–38.
McLaughlin M, Patin EC, Pedersen M, Wilkins A, Dillon MT, Melcher AA, et al. Inflammatory microenvironment remodelling by tumour cells after radiotherapy. Nat Rev Cancer. 2020;20(4):203–17.
Liu YG, Chen JK, Zhang ZT, Ma XJ, Chen YC, Du XM, et al. NLRP3 inflammasome activation mediates radiation-induced pyroptosis in bone marrow-derived macrophages. Cell Death Dis. 2017;8(2):e2579.
Sun K, Chen RX, Li JZ, Luo ZX. LINC00511/hsa-miR-573 axis-mediated high expression of Gasdermin C associates with dismal prognosis and tumor immune infiltration of breast cancer. Sci Rep. 2022;12(1):14788.
He H, Yi L, Zhang B, Yan B, Xiao M, Ren J, et al. USP24-GSDMB complex promotes bladder cancer proliferation via activation of the STAT3 pathway. Int J Biol Sci. 2021;17(10):2417–29.
Acknowledgements
We would like to express our gratitude to BioRender (https://app.biorender.com/) for assistance in creating the figures.
Funding
This work was supported by the National Natural Science Foundation of China (82272664, 82172500, 32300528), the Science and Technology Innovation Program of Hunan Province (2023RC3085, 2023RC3080), Hunan Provincial Health High-Level Talent Scientific Research Project (R2023054), Hunan Provincial Natural Science Foundation of China (2022JJ30843), the Scientific Research Fund of Hunan Provincial Education Department (23B0023), the Excellent Youth Foundation of Hunan Scientific Committee (2024JJ2084), the Science and Technology Development Fund Guided by Central Government (2021Szvup169), Hunan Provincial Administration of Traditional Chinese Medicine Project (D2022117), the Scientific Research Program of Hunan Provincial Health Commission (B202304077077), the Fundamental Research Funds for the Central Universities of Central South University (2024ZZTS0961).
Author information
Authors and Affiliations
Contributions
L.W., C.T. and H.X. participated in the conception and design of the study; H.W., T.W. and S.Y. wrote the manuscript; J.T., Y.Z. and L.W. reviewed and revised the manuscript; H.W., T.W. and S.Y. contributed equally to this work. C.T. and H.X. jointly supervised this work. All authors reviewed the manuscript.
Corresponding authors
Ethics declarations
Ethical approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Wang, H., Wang, T., Yan, S. et al. Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication. Mol Cancer 23, 268 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-024-02183-9
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-024-02183-9