Skip to main content

Programmed cell death disrupts inflammatory tumor microenvironment (TME) and promotes glioblastoma evolution

Abstract

Glioblastoma (GBM) is the most common malignant brain tumor and has a dismal prognosis even under the current first-line treatment, with a 5-year survival rate less than 7%. Therefore, it is important to understand the mechanism of treatment resistance and develop new anti-tumor strategies. Induction of programmed cell death (PCD) has become a promising anti-tumor strategy, but its effectiveness in treating GBM remains controversial. On the one hand, PCD triggers tumor cell death and then release mediators to draw in immune cells, creating a pro-inflammatory tumor microenvironment (TME). One the other hand, mounting evidence suggests that PCD and inflammatory TME will force tumor cells to evolve under survival stress, leading to tumor recurrence. The purpose of this review is to summarize the role of PCD and inflammatory TME in the tumor evolution of GBM and promising methods to overcome tumor evolution.

Background

Glioblastoma (GBM), the most common type of malignant glioma and primary brain tumor, is still one of the most fatal tumors, with a 5-year survival rate of 6.9% [1, 2]. Due to its late diagnosis, aggressive infiltration, and higher inter- and intra-tumoral genetic heterogeneity, GBM has limited treatment choices [3]. Tumor evolution, defined by genetic and biological adaptations, is a crucial approach for tumor cells to cope with the complicated living environment, especially when tumor cells are under the survival stress imposed by various anti-tumor treatment. It is worth noting that selective pressure might force tumor cells to evolve along different pathways. Using both bulk and single-cell data, many studies on GBM have shown different evolution models of the GBM genome, including gradualism and punctuated evolution [4, 5]. Therefore, tumor evolution can result in the heterogeneity and therapeutic resistance in GBM [6]. For instance, the proteomic landscape of 134 primary GBM (pGBM) and recurrent GBM (rGBM) samples, including 40 paired pGBM – rGBM ones, reveals that tumor recurrence is associated with post-treatment tumor evolution, by activating conventional tumor-promoting pathways such as epithelial-mesenchymal transition (EMT) [7]. Another study by Piao et al. shows that anti-vascular endothelial growth factor treatment can induce hypoxic tumor microenvironment (TME) and stimulate myeloid cell infiltration, as well as tumor mesenchymal transition, promoting tumor progression [8]. Therefore, confirming the drivers of tumor evolution is essential for understanding how GBM tumor cells evade cancer treatment.

Programmed cell death (PCD) eliminates unwanted cells to maintain the physiological homeostasis through several distinctive pathways, with the major types including apoptosis, autophagy, ferroptosis and pyroptosis [9]. Apoptosis, the classical and extensively studied type of PCD, is characterized by release of cytochrome C from injured mitochondria [10]. Apoptotic cell death is regulated by the delicate balance between pro-apoptotic and anti-apoptotic proteins to activate downstream caspases, initiating the well-characterized process including cell shrinkage, chromatin condensation and DNA fragmentation [11]. Autophagy (PCD type II), on the contrary, is critical for sustaining cell viability under stress rather than triggering cell death as other forms of PCD [12]. With the administration of chemotherapy or other anti-tumor treatment, PI3K/Akt/mTOR signaling is suppressed, forcing GBM cells to activate autophagy as a protective response [12,13,14]. Autophagy is also enhanced when Wnt signaling is activated to regulate cell proliferation and migration [12]. Ferroptosis, a relatively newly discovered form of PCD, occurs due to the intracellular accumulation of iron and reactive oxygen species (ROS) when poly unsaturated fatty acids undergo lipid peroxidation, thus disrupting the intracellular redox balance [12]. Lastly, pyroptosis, another recently discovered form of PCD, is involved in immune activation, contrasting with the immunosuppressive nature of apoptosis [12]. It is marked by cell swelling and subsequent membrane rupture, releasing intracellular components to attract inflammatory cells and activate immune responses [10, 12]. Many studies have shown the powerful anti-tumor properties of PCD in GBM. For instance, Apatinib, a tyrosine kinase inhibitor, can induce ferroptosis in glioblastoma cell lines and consequently inhibit tumor proliferation [15]. Furthermore, another antineoplastic therapy, Roxadustat, amplifies hypoxia-inducible factor (HIF) signaling to stimulate ferroptosis and suppress the growth of chemoresistant GBM cells [16]. Zhibo Liu and colleagues developed a biorthogonal system delivering gasdermin to tumor cells, which suggests gasdermin-induced pyroptosis may provoke robust anti-tumor immunity and improve the effectiveness of anti-PD-1 therapy in pan-cancer [17]. However, as more evidence uncovers the therapeutic effects of PCD, the role of PCD in tumor evolution, particularly its interaction with TME, starts to draw renewed attention in the scientific community. For example, tumor cells undergoing PCD secrete cytokines to prompt the immune cell infiltration and thus reshape the TME [18,19,20]. Consequently, the reshaped TME can transform the infiltrated immune cells into immune-suppressive types to support tumor development. Additionally, local immune cells in the TME can also undergo PCD themselves, leading to intratumoral immune suppression and tumor progression [21].

As mentioned above, the relationship between TME and tumor progression is intriguing and PCD can function as a double-edged sword in glioblastoma development. This article aims to summarize the contribution of PCD in the tumorigenic TME and subsequently tumor evolution in GBM. Furthermore, we summarize the dual function of PCD in TME formation, providing the basis of designing innovative therapeutic strategies to overcome treatment resistance in glioblastoma.

PCD supports the cancer-immunity cycle and reshapes inflammatory TME

PCD not only eliminates tumor cells directly through various types of cell death, but also allows GBM cells to release tumor antigens that will drain into cervical lymph nodes, provoking immunological responses. Afterwards, antigen-presenting cells (APCs) present the tumor antigens to and activate effector immune cells. These activated effector cells will then be trafficked to the brain tumor and initiate immunological destruction of tumor cells upon recognition of tumor antigens. As a result of immune infiltration, more tumor cells will undergo PCD and release increasing amount of tumor antigens, completing the cancer-immunity cycle (Fig. 1A). This can be illustrated by an H&E staining of the tumor tissue from a GBM patient, showing abundant immune cell infiltration, including lymphocytes and macrophages, around the central necrotic region (Fig. 1B). In recent years, many treatments for GBM have been shown to involve PCD as their anti-tumor actions. As one of the few treatments for GBM that have demonstrated substantial survival benefit in clinical trials, tumor treating fields (TTFields) can activate stimulator of interferon genes (STING) and absent in melanoma 2 (AIM2) inflammasomes to trigger pyroptosis in tumor cells, which will then release tumor antigens and recruit dendritic cell (DCs) as well as tumor-specific cytotoxic T lymphocytes (CTLs), thus converting the TME into a pro-inflammatory environment [20]. Unexpectedly, immune recruitment and infiltration in GBM are not necessarily associated with improved prognosis. In fact, PCD and immune infiltration might be predictive of worse clinical outcome as shown in bioinformatics analysis, in vitro and in vivo studies [22,23,24,25]. The seemingly contradictory evidence piques our curiosity in the exact role of PCD in GBM.

Fig. 1
figure 1

The overview of the cancer-immunity cycle. A: PCD can support and sustain the cancer-immunity cycle. PCD can induce immune system to recognize destroyed tumor cells, and recruit numerous immune cells into TME. The infiltrative immune cells can communicate with tumor cells. B: H&E-staining of GBM patient presents numerous lymphocytes (red arrow) and macrophages (black arrow) infiltration around necrotic region (white arrow)

Good-to-bad inflammatory microenvironment caused by PCD

Ever since the nineteenth century, there is ongoing debate about the possible link between inflammation and malignancies [26]. Early activation of acute inflammatory response can boost cytotoxic lymphocyte responses and elicit immune-mediated cancer cell death [27]. However, as the evidence accumulates, inflammation can be either pro-tumor or anti-tumor, depending on whether its presence is acute or persistent.

Acute inflammation caused by PCD kills the tumor

As previously discussed, the anti-tumor immune response triggered by PCD is comprised of pyroptosis, ferroptosis, and autophagy, all of which induce tumor cell death and then causes acute inflammation [28, 29]. Ferroptosis is a subtype of PCD characterized by iron-dependent lipid peroxidation [30]. Ferroptosis primarily limit glioblastoma cell survival through ROS generation to activate acute inflammation and enhance tumor cell apoptosis [31]. Its effectiveness in tumor suppression is exemplified by the fact that numerous cancer therapies act via ferroptosis, including preclinical medications (Erastin, RSL3), approved medications (sorafenib, sulfasalazine, statins, artemisinin) and radiation therapy [32, 33]. Pyroptosis, another subtype of PCD, is a newly discovered controlled cell death. Similar to ferroptosis, it also promotes cancer cell death and the subsequent release of inflammatory molecules, eliciting robust cytotoxic lymphocyte responses to prevent tumor progression [34, 35].

Long-term chronic inflammation modulates the TME

Although PCD-based immune-stimulating therapies are meant to detect and kill tumor cells, the anti-tumor effect of PCD is debatable in the actual clinical setting. GBM patients with enhanced ferroptosis have a higher risk of developing the disease, having a worse prognosis, and experiencing worse immunosuppression [36]. Clinical data also show that glioma patients expressing high level of pyroptosis-related genes(PRGs) have worse outcomes and are at greater risk of metastasis [37]. Moreover, compared to low-grade gliomas, high-grade gliomas have higher expression of PRGs probably associated with their malignant progression [38].

The reason behind PCD’s inadequacy in the clinical setting lies in the duration of immune stimulation. While preclinical studies prove that the short-term proinflammatory and immune-stimulating effect of PCD can suppress tumor proliferation, persisting proinflammatory immune response may alter the immunological microenvironment to favor tumor growth. Firstly, PCD induced inflammation drives M2 polarization of the infiltrative macrophages, a pro-tumor phenotype of macrophages. As demonstrated by Dai et al., the oxidative stress caused by ferroptosis in tumor cells will trigger the release of oncogenic KRAS protein, which is then taken up by local tumor-associated macrophages, causing them to switch to an M2 phenotype [39]. Zheng et al. develop a pyroptosis-related gene based prognostic index (PRGPI) and discover that patients with high PRGPI exhibit an extensively immune-suppressed TME, specifically higher infiltration of M2-type macrophages, lower infiltration of CD8 T cells and activated NK cells, and higher expression of immune checkpoints [38]. In addition to macrophages, ferroptosis also inhibit CD36+ T cells from releasing cytotoxic cytokines and tune down T cell antitumor activity, especially when paired with anti-PD-1 [40]. Inflammatory factors such as IL-18 may stimulate Th2 responses and angiogenesis, leading to enhanced tumor migration and invasion, and IL-1β can attract monocytes in the TME and operate as a master cytokine in cancer growth, as well as dampen T cell responses in the TME [41,42,43,44]. Furthermore, IL-6 can influence other cells within the TME to create a favorable growing environment for tumor cells, allowing for easier angiogenesis and tumor escape from immune surveillance [45]. Glioblastoma-derived IL-33 also promotes tumor growth by orchestrating an inflammatory TME [46]. Studies in many different tumors have shown that human regulatory B (Breg) cells secrete many cytokines (including IL-6, IL-10, IL-35, and TGF-β) to support the expansion of regulatory T cells (Tregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) around the tumor bed, driving these cells toward immune-suppressive phenotypes to enhance the tumor-promoting microenvironment [47]. The prolonged immune stimulation brought on by PCD and inflammatory cytokines reshapes the TME into an immunosuppressive one and help tumor cells evade host immune surveillance. The long-term effect of PCD on promoting the development of an immunosuppressive milieu underscores the necessity to harness the tumor-killing effect of acute inflammation while impede the transformation into immunosuppressive TME for effective cancer treatment (Fig. 2).

Fig. 2
figure 2

Treatments cause PCD and inflammatory TME to promote tumor evolution. PCD-based therapies such as chemotherapy, immune checkpoint inhibition and personalized immunotherapy can induce cancer cell death, exposing intracellular contents as immunogenic signals and transforming immunologically “cold tumor” into “hot tumor” with inflammatory TME. However, prolonged PCD may lead to functional exhaustion among infiltrating immune cells due to a sustained influx of inflammatory cytokines, resulting in the emergence of immune-suppressive cell types like N2 neutrophils, regulatory T cells (Treg), and the M2 subtype of tumor-associated macrophages (TAM). Under the selection pressure of chronic PCD and the resultant enduring inflammatory TME, tumor cells will adapt to the harsh environment through natural selection. The epithelial-mesenchymal transition (EMT) of GBM cells in the inflammatory TME leads to glioma stem cell (GSC) accumulation, driving tumor evolution and treatment resistance. Therefore, indiscriminate treatments, including TTFields, LITT and HFRT, can kill tumor cells intensively and inhibit tumor evolution. (ILC2: type 2 innate lymphoid cells; MDSC: myeloid-derived suppressor cells; NK2: type 2 natural killer cells; NKT2: type 2 natural killer T cells; Th2: type 2 T helper cells; Treg: regulatory T cells)

Tumor cells evolve to adapt to the PCD and inflammatory TME

The TME can influence tumor sensitivity to treatment by facilitating immune evasion and cancer progression [48]. PCD-induced proinflammatory TME is an independent predictor of poor prognosis in glioma patients and there is mounting evidence that the TME of GBM, the most malignant type of gliomas, has higher rates of immune cell infiltration and programmed cell death (PCD) scores compared to lower grade gliomas [49]. As the hallmark of malignant transformation, tumor evolution might be the reason behind the contradicting effect of PCD on the TME (Fig. 2). Under the pressure of tumor elimination, tumor cells will experience genomic alterations to adapt to the harsh living environment. For instance, PD-1/PD-L1 blockade can increase the amount of IFN-γ secreted by CD8+ T cells, which in turn trigger tumor cells to upregulate the Wnt/β-catenin signaling and accelerate the progression of malignant cancers [50]. Similarly, using in vivo CRISPR screens in the GBM mouse model, Stephen J. Elledge et al. discover that tumor cells only evolve to downregulate genes in antigen presenting pathway and upregulate inhibitory immune checkpoint genes in immunocompetent mice, but not in immunocompromised mice [51]. This study suggests that the survival pressure imposed by the pro-inflammatory immunity in TME will select and expand tumor cell clones with the capability of evading adaptive immune surveillance, thus provoking tumor evolution [51]. Furthermore, tumor destruction early on at the course of disease will recruit neutrophils and subsequently cause neutrophil-mediated ferroptosis in GBM cells, forming a positive feedback cycle [21]. However, this PCD-amplified immune infiltration educates neutrophils to deliver myeloperoxidase into GBM cells and as a result, tumor cells evolve by increasing the expression of mesenchymal-transition related genes [21]. Meanwhile, overexpression of ferroptosis-inhibiting molecules, such as glutathione peroxidase 4 (GPX4) which is an essential phospholipid peroxidase, can significantly alleviate PCD-mediated necrosis and diminish tumor aggressiveness [21]. All in all, tumor cells interact with the TME to drive malignant progression through evolution.

PCD-induced inflammatory TME drives tumor evolution by facilitating EMT transformation and glioma stem cells (GSCs) accumulation

GSCs, a small and uncommon subset of glioma tissues, are characterized by their self-renewal and multi-lineage differentiation abilities and can be identified by their unique markers, such as CD133, CD44, VIM, and N-cadherin [52]. The tumor-initiating characteristics of GSCs lead to intratumoral heterogeneity, immunosuppressive TME, and treatment resistance [52]. Notably, GSCs may be responsible for tumor evolution and further reinforce the treatment resistance brought about by PCD. A growing body of research suggests that inflammatory TME and GSCs have a mutually beneficial relationship, implying that inflammatory TME might cause GSC conversion and tumor progression. For example, PCD can produce IFN-γ, a classical proinflammatory cytokine, to reshape the TME. The proinflammatory TME will then prompt GSCs to enter an immunological-insensitive state via the IDO-kynurenine-AHR pathway, resulting in chemotherapy resistance [53]. In addition to promote GSC accumulation through EMT signaling activation, the IFN-γ release upon PCD induction can upregulate the expression of immune checkpoints, such as PD-L1 in cancers, thus worsening the immune suppression in PCD-reshaped TME [54]. Likewise, other pro-inflammatory cytokines such as IL-6, IL-1, and TNF-α also can drive stem-cell like transition via EMT pathway in high-grade gliomas, leading to GSC-related tumor progression and therapy resistance.

GSCs are often resistant to conventional first-line treatments, such as radiation and chemo-therapies, due to the fact that stem-like cells in inflammatory TME can temporarily enter the quiescent G0 state in cell cycle, whereas conventional therapies only eradicate actively proliferating cells [55]. Additionally, GSCs have a high expression level of ATP binding cassette transporter G2 (ABCG2) that will potentially increase the chemotherapeutic drug efflux, diminishing treatment efficacy [56]. Immunotherapies also have poor efficacy on GSC elimination, partially because GSCs increase PD-L1 expression to exhaust T cells and secrete numerous chemokines as well as cytokines to induce M2 TAM polarization and accumulation of anti-inflammatory immune cells, such as Treg and MDSCs [57].

Therefore, PCD-induced inflammatory cell recruitment assists in GSC expansion. In turn, GSCs reinforce the immune suppression in TME while ensuring PCD to entice more immune cell infiltration and maintain the suppressive TME to transform incoming immune cells into anti-inflammatory pro-tumor subtypes. GSCs are the source for tumor adaptation under various treatment-induced hostile environment and eradicating GSCs is a critical target for innovative therapy model development (Fig. 2).

Overall, most anti-cancer modalities for GBM may result in PCD of tumor cells and an altered inflammatory TME, triggering tumor evolution and treatment resistance in a long-term exposure. Hence, it is of great importance to examine the few remaining highly effective treatment modalities for glioblastomas through the lens of PCD-tumor evolution theory.

Lessons from treatment for GBM

Lessons from first-line standard treatment

Currently, the first-line standard management for GBM is the “Stupp’s Regimen” that includes radiation combined with concomitant and adjuvant temozolomide (TMZ) [58]. It has been reported that both radiotherapy and TMZ chemotherapy trigger PCD to eliminate tumor cells. As an alkylating agent, TMZ is able to methylate DNA at guanine residues to initiate mismatch repair as well as the resultant double strand breaks, and Ca2+-dependent caspase 3 overexpression with elevated Bax/Bcl-2 ratio, inducing apoptotic cell death [59,60,61]. In addition to apoptosis, autophagy is also activated upon radiation and TMZ in GBM cells and act to halt tumor growth via the degradation of oncogenic proteins, stimulation of AMPK and inhibition of Akt/mTOR signaling [59, 62, 63]. The application of autophagy inhibitor can abolish the tumor-killing effect of TMZ in in vitro experiments [64]. Moreover, radiotherapy and TMZ promote ferroptosis, the most common type of PCD in malignant gliomas, to eliminate GBM cells [22, 65, 66]. Some studies have discovered that ferroptosis can in turn improve TMZ’s cytotoxic activity [67, 68]. As proved by Hanjie Liu and colleagues, GBM cell lines that have acquired TMZ resistance are more susceptible to the ferroptosis inducer, erastin, than TMZ-sensitive GBM cells [69].

However, accumulating evidence indicates that various types of PCD contribute to TMZ resistance and tumor evolution. Autophagy serves a dual function, with both tumor-promoting and tumor-suppressing characteristics depending on the stages of tumorigenesis [59]. An analysis of expression data from 467 GBM samples reveals that GBM patients with higher expression of autophagy genes have worse prognosis and damage-regulated autophagy modulator (DRAM1) gene upregulation is linked to the activation of mitogen-activated protein kinase (MAPK) in GSCs, playing a role in TMZ resistance [70]. In preclinical models, DNA injuries induced by TMZ force glioma cells to increase ATP production as a stress response, protecting tumors from chemotherapy and maintaining the survival of GSCs [70, 71]. Apart from autophagy, several investigations have shown that ferroptosis can affect iron metabolism and enhance GSC growth, resulting in TMZ resistance [72]. In U87 and U251 GBM cell lines, although erastin can stall tumor proliferation, it also strengthens the migratory ability of tumor cells [69]. The conflict results in these recent researches, in our opinion, are due to the timing of PCD induction. In the early phase of tumor evolution, PCD can inhibit tumor cell proliferation and act synergistically with TMZ. Nevertheless, tumor progression is a chronic event, and enduring PCD of tumor cells will prompt tumor evolution as well as TMZ resistance. As a result, the PCD risk scores designed from tumor tissues of GBM patients and real-world GBM database are found to be inversely associated to patient survival time, contrary to the results from in vitro experiments [37, 69]. Other than directly assisting in tumor adaptation, PCD also sustains tumor growth and promote TMZ insensitivity by reshaping the TME into a highly immune suppressed environment. With machine learning, it is discovered that GBM patients who have little response to TMZ therapy are associated with higher expression levels of immune checkpoints (PD1, PD-L1, PD-L2) [73]. In TMZ resistant glioblastoma cell lines, expression levels of pyroptosis-related genes, genes associated with regulatory T cells and immune checkpoints are significantly higher than TMZ sensitive GBM cells [74]. These findings suggest that PCD and the long-term immune suppression in TME it induced might lead to TMZ resistance.

Lessons from immunotherapies

Immune checkpoint inhibitors (ICIs)

Great breakthrough in GBM immunotherapy has been observed over the previous decades, and ICIs have gained a lot of attention [75]. Well-known ICIs, such as pembrolizumab and nivolumab, have altered the clinical management regimen for a variety of malignancies, including NSCLC and melanoma [76, 77]. The promising results in other cancers pique our curiosity in the role of ICIs in GBMs. Given that chemotherapy-resistant GBM cells have typically evolved to be insensitive to the intrinsic apoptotic pathway triggered by TMZ-induced DNA damage, the mechanism by which ICIs eradicate cancers involves activating extrinsic cell death pathway via enhancing the release of death signals from surveilling immune cells, as shown in preclinical studies [78, 79]. In addition, CD47-targeting immune checkpoint inhibition can induce GBM cell death via Akt/mTOR inactivation and elevated autophagic flux in glioblastoma cell lines [79, 80]. Nonetheless, ICIs failed to demonstrate efficacy in the clinical studies. The Keynote-028 clinical trial explores pembrolizumab monotherapy in 26 recurrent GBM patients and finds that it provides only marginal survival advantages [81]. Furthermore, the results of the first large-scale phase 3 clinical study investigating the effectiveness of ICIs for GBM are likewise unsatisfactory, with nivolumab monotherapy failing to extend survival time when compared to the bevacizumab-treated control group [82]. Afterwards, researchers try to combine ICIs with first-line therapies in pGBM (Checkmate 498 and Checkmate 548), but neither trial demonstrates significant survival improvement [83, 84]. Currently, ICI application before surgery can increase chemokine release, immune cell infiltration, and clonal diversity among tumor-infiltrating T lymphocytes, indicating a bright future for neoadjuvant immunotherapies in GBMs [85]. Interestingly, the scientists observe that neoadjuvant PD-1 checkpoint blockade can successfully enhance the infiltration and clonocal expansion of T cells and conventional type 1 dendritic cells (cDC1s), but it cannot overcome the immunosuppressive TAMs in recurrent GBM [86]. The difficulty is primarily caused by the co-evolution of tumor cells and TME. ICIs may activate T cells to secrete numerous pro-inflammation cytokines to eliminate malignant tumor cells, but at the same time ICIs may stimulate the production of immunosuppressive molecules such as PD-L1, IDO1, and IL4I1, resulting in M2 macrophage polarization and T-cell activity inhibition [86]. Therefore, future studies should target TAMs to complement the efficacy of neoadjuvant ICIs.

Adoptive immunotherapy

Chimeric antigen receptor (CAR) T-cell therapy is a promising new therapeutic option for GBM. CAR-T cells are created from genetically modifying T cells armed with CARs targeting particular antigens on GBM cells, such as EGFRvIII, IL13Ra2, and B7-H3, ushering in a new age of individualized cancer therapy [87]. Despite some encouraging results, each CAR-T therapy only aims at a single tumor antigen, imposing the survival pressure on tumor cells. Overtime, GBM cells will evolve by partial or total loss of target antigen expression, leading to therapeutic resistance, a phenomenon known as antigen escape. For example, in 2017, the first clinical trial (NCT02209376) of CAR-T EGFRvIII in 10 recurrent GBM patients with EGFRvIII-positive tumors is conducted and five of the seven patients who received post-CAR-T reoperation has reduced EGFRvIII in resected tumor tissues [88]. Based on this phenomenon, ongoing clinical investigations focus on the development of CAR-T therapies that target multiple tumor antigens.

The prospects of relatively indiscriminate treatments

It is true that the induction of PCD and inflammatory TME can hinder tumor proliferation, but we should consider treatment efficacy in terms of tumor evolution. Specifically, the intensity and duration of therapy are crucial since they are closely associated with tumor adaptation and treatment resistance. Due to the selectivity of blood brain barrier (BBB), many chemotherapies and immunotherapies cannot reach adequate concentration in CNS and BBB also hampers immune cell infiltration. As a consequence, these treatments are not concentrated enough to kill tumor cells and will instead reshape the TME to which tumor cells can slowly adapt under the selective pressure. Therefore, it is necessary to combine numerous physically indistinguishable therapeutic modalities, like Tumor-treating fields (TTFields), hypofractionated radiotherapy (HFRT) and laser interstitial thermotherapy (LITT) to improve effectiveness and prevent tumor progression (Fig. 2). These localized physical treatments exert a potent tumor cell destruction effect and suppress tumor evolution due to their indiscriminate killing mechanisms, unlike the targeted killing actions of targeted therapies. Table 1 summarizes therapies with promising prospects and the multi-modal combinations that are currently under clinical investigation.

Table 1 Clinical trials of relatively indiscriminate treatments for GBM. Combination treatments amplifies the effect for GBM
Tumor-treating fields (TTFields)

As a novel anti-tumor treatment, TTFields inhibits cell division via alternating electric fields of intermediate frequency (100–500 kHz) and low intensity (1–3 V/cm) [89]. TTFields not only increases the BBB permeability, but also enhance various anti-tumor signaling pathways, such as anti-tumor immune response, anti-mitotic signaling, and DNA damage repair pathway [90]. When treating patient-derived glioma stem-like cells with TTFields, TTFields therapy disrupts DNA damage repair system and the functioning of replication fork, significantly increasing level of apoptotic cell death in previously treatment-resistant GSCs [91]. Apart from apoptosis, studies demonstrate that TTFields-treated GBM cells have elevated autophagic flux, stimulating ATP production that function as a signal to attract immune cells [92, 93]. In addition to its direct inhibitory effects on GBM cells, TTFields alters the TME of GBM cells, particularly the immunological TME through pyroptosis, to boost antitumor immune response, indirectly limiting tumor growth [20]. In murine lung and colon cancer models, it is verified that TTFields cause immunogenic cell death via pyroptosis and can improve antitumor efficacy when combined with anti-PD-1 therapy [92]. For GBM, ongoing phase II clinical trial(NCT03405792), the mOS in combination of TTFields, TMZ and anti-PD-1 therapy group is 24.8 months [94]. Preclinical studies demonstrate that TTFields can increase chemotherapy sensitivity in human glioblastoma cell lines and animal models, providing the theoretical basis for combination therapies with other GBM treatment (Table 1). TTFields as an add-on to traditional radiotherapy and chemotherapy has emerged as a breakthrough in the clinical management of GBM [95]. In a phase 3 clinical trial, adding TTFields to maintenance temozolomide chemotherapy significantly prolonged progression-free and overall survival in GBM patients [96]. Preclinical research supports the use of TTFields treatment immediately following radiation therapy (RT) as a feasible regimen for improving RT outcome [97]. TTFields combined with targeted therapy, such as bevacizumab, an inhibitor of VEGF, and dabrafenib, an inhibitor of BRAFV600E, can prolong survival [98]. Additionally, in preclinical research, it is reported that TTFields can downregulate stem cell markers, and promote the efficacy of proton beam [99]. All of this evidence has shown that TTFields has enormous potential in GBM treatment.

Hypofractionated radiotherapy (HFRT)

Hypofractionated radiation therapy (HFRT) offers several advantages. HFRT is now widely accepted for treating patients with poor physical conditions. HFRT is preferred and recommended RT modality for patients with astrocytoma, oligodendroglioma, or glioblastoma who have a Karnofsky performance status (KPS) score less than 60, according to the 2022 National Comprehensive Cancer Network (NCCN) recommendations [100]. Additionally, due to the safety and tolerability of HFRT, even senior glioblastoma patients in good physical condition might consider it as a therapy option [101]. HFRT has improved tumor-killing capacity by giving a larger radiation dosage each time and shorten the overall treatment course [102, 103]. Radiation has been proven in vivo and in vitro to promote glioma cell death by oxidative stress, DNA damage, and apoptosis [104]. However, in vitro studies suggest that irradiation at lower doses, despite capable of initiating autophagy initially, failed to kill cancer stem cells and the increased autophagic flux provide energy as well as metabolic building blocks for GSCs, thus leading to GSC proliferation and tumor evolution [105, 106]. On the contrary, HFRT deliver high doses of radiation during each session, eliminating glioma stem cells and preventing tumor resistance [107, 108]. In addition, HFRT drastically shortens the treatment time, while conventional radiation (cRT) takes longer. Patients in poorer health state may drop out midway through treatment process, and tumor cells may re-grow during treatment [109]. The goal of HFRT is to complete the entire therapy process in three weeks [110]. Clinical evidence suggests that HFRT may slow the pace of GBM cell repopulation and improve patient compliance [111]. Following HFRT, the transcriptomes of glioma stem cells also change dramatically, perhaps contributing to the improved long-term clinical outcomes [112]. Lastly, HFRT may modulate GBM microenvironment to boost the immune response, thereby improving the effectiveness of immunotherapy [113]. At low doses of radiotherapy delivered by conventional radiotherapy, GBM TME exhibits a buildup of radioresistant immunosuppressive cells, including M2 type of tumor-associated macrophages, myeloid-derived suppressor cells and regulatory T cells [114,115,116]. However, high dosages of radiation as in HFRT shift suppressive TME into a supportive one, with increased infiltration of pro-inflammatory immune cells and APCs [114, 117]. Hence, anti-PD1 antibodies, when combined with HFRT, appear to provide a longer lasting anti-tumor action, potentially doubling survival time [118]. These findings highlight that HFRT as a treatment modality can modulate TME and hinder tumor evolution of malignant gliomas.

Laser interstitial thermotherapy (LITT)

As a minimally invasive technique, LITT has gained a lot of attention in treating CNS malignant tumors [119]. LITT works by introducing an optical fiber into the tumor under MRI navigation followed by laser heating tumors to increase local hyperthermia and anticancer activity [119]. Currently, intraoperative MR thermometry can achieve precision tumor targeting and accurately deliver therapeutic heat doses with real-time observation of tissue damage [120]. LITT, as a physical treatment mode, has been used to treat epilepsy, metastatic brain cancers, and gliomas for decades [121]. In glioblastoma, LITT has equivalent efficacy as surgical resection. In a multicenter prospective study of LITT in IDH wild-type glioblastoma, de Groot JF et al. find that patients with GBM treated with LITT and postoperative chemoradiotherapy has an OS of 16.14 months and a PFS of 11.93 months, comparable to conventional surgical resection, indicating that LITT can be a good option for those not suitable for surgery [122]. At ablation temperature near the tip of laser, the heat trigger mitochondria damage and dysfunctional DNA repair, activating intrinsic pathway of apoptosis [123,124,125]. In addition to the direct killing of tumor cells through thermal effects, LITT also acts synergistically with chemotherapy and immunotherapy by increasing BBB permeability and inducing immunogenic cell death, while preventing tumor evolution through the timely elimination of residual GBM cells [126]. At a further distance from the laser tip, tumor tissues are exposed to hyperthermia, a temperature range that does not induce apoptosis but rather triggers immune activation [125]. Hyperthermia can enhance antigen presentation by APCs, partially because of accelerated maturation of dendritic cells, and facilitate activation as well as migration of T cells [125, 127]. The current ongoing clinical trials involving LITT in GBM are summarized in Table 1.

Conclusion

Glioblastoma is a subtype of glioma characterized by a dismal prognosis and limited treatment options. Because high-grade gliomas have extraordinary plasticity, these tumor cells can evolve to avoid destruction imposed by anti-tumor therapies. Inflammatory TME, EMT signaling, hypoxia, and angiogenesis all contribute to survival pressure, which in turn aids tumor evolution. This review aims to address the controversies regarding the efficacy of PCD-based therapies. Short-term and intense PCD can indeed cause tumor cell death by activating apoptosis, autophagy, ferroptosis pyroptosis and other forms of PCD. However, due to the constraints imposed by the permeability of blood brain barrier, long-term systemic administration of PCD-based therapeutics often immerse GBM cells in sub-optimal concentrations. As a result, chronic and inadequate PCD eventually leads to epithelial-mesenchymal transformation and accumulation of GSCs, thereby promoting tumor evolution and treatment resistance. Targeted treatment modalities, including temozolomide and immunotherapies, have limited effectiveness in GBM due to tumor adaptation upon PCD induction. In contrast, indiscriminate therapeutic methods such as TTFields, HFRT and LITT can deliver intensified stimulation of PCD while simultaneously halting the EMT transformation of GBM cells. Therefore, these indiscriminate therapies not only induce strong and localized PCD-related tumor cell death to prevent tumor evolution, but also inhibit the formation of glioma stem cells, averting tumor progression. The combination of these indiscriminate therapies with other regimens is expected to be the future trend in GBM management.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ABCG2:

ATP binding cassette transporter G2

APCs:

antigen-presenting cells

Bev:

Bevacizumab

CAR:

Chimeric antigen receptor

CNS:

Central nervous system

GBM:

Glioblastoma

HFRT:

Hypofractionated radiotherapy

ICIs:

Immune checkpoint inhibitors

LGG:

low-grade glioma

LITT:

Laser interstitial thermotherapy

NES:

Natural evolution signature

PCD:

Programmed cell death

TME:

Tumor microenvironment

TMZ:

Temozolomide

TTFields:

Tumor-treating fields

NCCN:

National Comprehensive Cancer Network

References

  1. Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: primary brain and other Central Nervous System tumors diagnosed in the United States in 2016–2020. Neuro Oncol. 2023;25:iv1–99.

    Article  PubMed  Google Scholar 

  2. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:459–66.

    Article  CAS  PubMed  Google Scholar 

  3. Gangoso E, Southgate B, Bradley L, Rus S, Galvez-Cancino F, McGivern N, Guc E, Kapourani CA, Byron A, Ferguson KM, et al. Glioblastomas acquire myeloid-affiliated transcriptional programs via epigenetic immunoediting to elicit immune evasion. Cell. 2021;184:2454–e24702426.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Bao Z, Wang Y, Wang Q, Fang S, Shan X, Wang J, Jiang T. Intratumor heterogeneity, microenvironment, and mechanisms of drug resistance in glioma recurrence and evolution. Front Med. 2021;15:551–61.

    Article  PubMed  Google Scholar 

  5. Kim J, Lee IH, Cho HJ, Park CK, Jung YS, Kim Y, Nam SH, Kim BS, Johnson MD, Kong DS, et al. Spatiotemporal evolution of the primary glioblastoma genome. Cancer Cell. 2015;28:318–28.

    Article  CAS  PubMed  Google Scholar 

  6. Varn FS, Johnson KC, Martinek J, Huse JT, Nasrallah MP, Wesseling P, Cooper LAD, Malta TM, Wade TE, Sabedot TS, et al. Glioma progression is shaped by genetic evolution and microenvironment interactions. Cell. 2022;185:2184–e21992116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tatari N, Khan S, Livingstone J, Zhai K, McKenna D, Ignatchenko V, Chokshi C, Gwynne WD, Singh M, Revill S, et al. The proteomic landscape of glioblastoma recurrence reveals novel and targetable immunoregulatory drivers. Acta Neuropathol. 2022;144:1127–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Piao Y, Liang J, Holmes L, Zurita AJ, Henry V, Heymach JV, de Groot JF. Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro Oncol. 2012;14:1379–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu J, Hong M, Li Y, Chen D, Wu Y, Hu Y. Programmed cell death tunes Tumor Immunity. Front Immunol. 2022;13:847345.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cell Mol Immunol. 2021;18:1106–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fraser A, Evan G. A license to kill. Cell. 1996;85:781–4.

    Article  CAS  PubMed  Google Scholar 

  12. Wan S, Zhang G, Liu R, Abbas MN, Cui H. Pyroptosis, ferroptosis, and autophagy cross-talk in glioblastoma opens up new avenues for glioblastoma treatment. Cell Commun Signal. 2023;21:115.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Eskilsson E, Rosland GV, Solecki G, Wang Q, Harter PN, Graziani G, Verhaak RGW, Winkler F, Bjerkvig R, Miletic H. EGFR heterogeneity and implications for therapeutic intervention in glioblastoma. Neuro Oncol. 2018;20:743–52.

    Article  CAS  PubMed  Google Scholar 

  14. Koul D, Shen R, Kim YW, Kondo Y, Lu Y, Bankson J, Ronen SM, Kirkpatrick DL, Powis G, Yung WK. Cellular and in vivo activity of a novel PI3K inhibitor, PX-866, against human glioblastoma. Neuro Oncol. 2010;12:559–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Xia L, Gong M, Zou Y, Wang Z, Wu B, Zhang S, Li L, Jin K, Sun C. Apatinib Induces Ferroptosis of Glioma Cells through Modulation of the VEGFR2/Nrf2 Pathway. Oxid Med Cell Longev 2022, 2022:9925919.

  16. Su X, Xie Y, Zhang J, Li M, Zhang Q, Jin G, Liu F. HIF-alpha activation by the prolyl hydroxylase inhibitor roxadustat suppresses chemoresistant glioblastoma growth by inducing ferroptosis. Cell Death Dis. 2022;13:861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang Q, Wang Y, Ding J, Wang C, Zhou X, Gao W, Huang H, Shao F, Liu Z. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature. 2020;579:421–6.

    Article  CAS  PubMed  Google Scholar 

  18. Wang K, Wang J, Zhang J, Zhang A, Liu Y, Zhou J, Wang X, Zhang J. Ferroptosis in Glioma Immune Microenvironment: opportunity and challenge. Front Oncol. 2022;12:917634.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang Y, Zhang C, Yang Y, Wang G, Wang Z, Liu J, Zhang L, Yu Y. Pyroptosis-related gene signature predicts prognosis and indicates Immune Microenvironment Infiltration in Glioma. Front Cell Dev Biol. 2022;10:862493.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chen D, Le SB, Hutchinson TE, Calinescu AA, Sebastian M, Jin D, Liu T, Ghiaseddin A, Rahman M, Tran DD. Tumor Treating fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma. J Clin Invest 2022, 132.

  21. Yee PP, Wei Y, Kim SY, Lu T, Chih SY, Lawson C, Tang M, Liu Z, Anderson B, Thamburaj K, et al. Neutrophil-induced ferroptosis promotes tumor necrosis in glioblastoma progression. Nat Commun. 2020;11:5424.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Liu T, Zhu C, Chen X, Guan G, Zou C, Shen S, Wu J, Wang Y, Lin Z, Chen L, et al. Ferroptosis, as the most enriched programmed cell death process in glioma, induces immunosuppression and immunotherapy resistance. Neuro Oncol. 2022;24:1113–25.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Zheng XJ, Chen WL, Yi J, Li W, Liu JY, Fu WQ, Ren LW, Li S, Ge BB, Yang YH, et al. Apolipoprotein C1 promotes glioblastoma tumorigenesis by reducing KEAP1/NRF2 and CBS-regulated ferroptosis. Acta Pharmacol Sin. 2022;43:2977–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang Z, Dai Z, Zhang H, Zhang N, Liang X, Peng L, Zhang J, Liu Z, Peng Y, Cheng Q, Liu Z. Comprehensive analysis of pyroptosis-related gene signatures for glioblastoma immune microenvironment and target therapy. Cell Prolif. 2023;56:e13376.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Markwell SM, Ross JL, Olson CL, Brat DJ. Necrotic reshaping of the glioma microenvironment drives disease progression. Acta Neuropathol. 2022;143:291–310.

    Article  CAS  PubMed  Google Scholar 

  26. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357:539–45.

    Article  CAS  PubMed  Google Scholar 

  27. Minton K. Pyroptosis heats tumour immunity. Nat Rev Immunol. 2020;20:274–5.

    Article  CAS  PubMed  Google Scholar 

  28. Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, Huang J, Wang F, Zhou F, Zhang L. Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 2022;19:971–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kolb R, Liu GH, Janowski AM, Sutterwala FS, Zhang W. Inflammasomes in cancer: a double-edged sword. Protein Cell. 2014;5:12–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266–82.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zhang J, Fu M, Wu J, Fan F, Zhang X, Li C, Yang H, Wu Y, Yin Y, Hua W. The Anti-glioma Effect of Juglone Derivatives through ROS Generation. Front Pharmacol. 2022;13:911760.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Xu H, Ye D, Ren M, Zhang H, Bi F. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med. 2021;27:856–67.

    Article  CAS  PubMed  Google Scholar 

  33. Zhao N, Huang Y, Wang YH, Muir RK, Chen YC, Wei J, Hooshdaran N, Viswanath P, Seo Y, Ruggero D, et al. Ferronostics: measuring Tumoral Ferrous Iron with PET to Predict Sensitivity to Iron-targeted Cancer therapies. J Nucl Med. 2021;62:949–55.

    Article  CAS  PubMed  Google Scholar 

  34. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Broz P, Pelegrin P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 2020;20:143–57.

    Article  CAS  PubMed  Google Scholar 

  36. Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18:280–96.

    Article  CAS  PubMed  Google Scholar 

  37. Zheng J, Zhou Z, Qiu Y, Wang M, Yu H, Wu Z, Wang X, Jiang X. A pyroptosis-related gene Prognostic Index correlated with survival and Immune Microenvironment in Glioma. J Inflamm Res. 2022;15:17–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen J, Chen S, Li B, Zhou S, Lin H. A pyroptosis-related signature predicts prognosis and indicates immune microenvironment infiltration in glioma. Cancer Med 2022.

  39. Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, Zeh HJ, Kang R, Wang J, Tang D. Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 2020;16:2069–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ma X, Xiao L, Liu L, Ye L, Su P, Bi E, Wang Q, Yang M, Qian J, Yi Q. CD36-mediated ferroptosis dampens intratumoral CD8(+) T cell effector function and impairs their antitumor ability. Cell Metab. 2021;33:1001–e10121005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kaplanov I, Carmi Y, Kornetsky R, Shemesh A, Shurin GV, Shurin MR, Dinarello CA, Voronov E, Apte RN. Blocking IL-1beta reverses the immunosuppression in mouse breast cancer and synergizes with anti-PD-1 for tumor abrogation. Proc Natl Acad Sci U S A. 2019;116:1361–9.

    Article  CAS  PubMed  Google Scholar 

  42. Pachathundikandi SK, Blaser N, Bruns H, Backert S. Helicobacter pylori avoids the critical activation of NLRP3 inflammasome-mediated production of oncogenic mature IL-1beta in Human Immune cells. Cancers (Basel) 2020, 12.

  43. Daley D, Mani VR, Mohan N, Akkad N, Pandian G, Savadkar S, Lee KB, Torres-Hernandez A, Aykut B, Diskin B, et al. NLRP3 signaling drives macrophage-induced adaptive immune suppression in pancreatic carcinoma. J Exp Med. 2017;214:1711–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ershaid N, Sharon Y, Doron H, Raz Y, Shani O, Cohen N, Monteran L, Leider-Trejo L, Ben-Shmuel A, Yassin M, et al. NLRP3 inflammasome in fibroblasts links tissue damage with inflammation in breast cancer progression and metastasis. Nat Commun. 2019;10:4375.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Fisher DT, Appenheimer MM, Evans SS. The two faces of IL-6 in the tumor microenvironment. Semin Immunol. 2014;26:38–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. De Boeck A, Ahn BY, D’Mello C, Lun X, Menon SV, Alshehri MM, Szulzewsky F, Shen Y, Khan L, Dang NH, et al. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat Commun. 2020;11:4997.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Blair PA, Norena LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, Mauri C. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity. 2010;32:129–40.

    Article  CAS  PubMed  Google Scholar 

  48. Bagaev A, Kotlov N, Nomie K, Svekolkin V, Gafurov A, Isaeva O, Osokin N, Kozlov I, Frenkel F, Gancharova O, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021;39:845–e865847.

    Article  CAS  PubMed  Google Scholar 

  49. Zhang C, Cheng W, Ren X, Wang Z, Liu X, Li G, Han S, Jiang T, Wu A. Tumor Purity as an underlying key factor in Glioma. Clin Cancer Res. 2017;23:6279–91.

    Article  CAS  PubMed  Google Scholar 

  50. Li G, Choi JE, Kryczek I, Sun Y, Liao P, Li S, Wei S, Grove S, Vatan L, Nelson R, et al. Intersection of immune and oncometabolic pathways drives cancer hyperprogression during immunotherapy. Cancer Cell. 2023;41:304–e322307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martin TD, Patel RS, Cook DR, Choi MY, Patil A, Liang AC, Li MZ, Haigis KM, Elledge SJ. The adaptive immune system is a major driver of selection for tumor suppressor gene inactivation. Science. 2021;373:1327–35.

    Article  CAS  PubMed  Google Scholar 

  52. Ma Q, Long W, Xing C, Chu J, Luo M, Wang HY, Liu Q, Wang RF. Cancer Stem cells and immunosuppressive microenvironment in Glioma. Front Immunol. 2018;9:2924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liu Y, Liang X, Yin X, Lv J, Tang K, Ma J, Ji T, Zhang H, Dong W, Jin X, et al. Blockade of IDO-kynurenine-AhR metabolic circuitry abrogates IFN-gamma-induced immunologic dormancy of tumor-repopulating cells. Nat Commun. 2017;8:15207.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Imai D, Yoshizumi T, Okano S, Itoh S, Ikegami T, Harada N, Aishima S, Oda Y, Maehara Y. IFN-gamma promotes epithelial-mesenchymal transition and the expression of PD-L1 in pancreatic Cancer. J Surg Res. 2019;240:115–23.

    Article  CAS  PubMed  Google Scholar 

  55. O’Connor SA, Feldman HM, Arora S, Hoellerbauer P, Toledo CM, Corrin P, Carter L, Kufeld M, Bolouri H, Basom R et al. Neural G0: a quiescent-like state found in neuroepithelial‐derived cells and glioma. Mol Syst Biol 2021, 17.

  56. Chen L, Shi L, Wang W, Zhou Y. ABCG2 downregulation in glioma stem cells enhances the therapeutic efficacy of demethoxycurcumin. Oncotarget. 2017;8:43237–47.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Zheng Y, Liu L, Wang Y, Xiao S, Mai R, Zhu Z, Cao Y. Glioblastoma stem cell (GSC)-derived PD-L1-containing exosomes activates AMPK/ULK1 pathway mediated autophagy to increase temozolomide-resistance in glioblastoma. Cell Biosci. 2021;11:63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997–1003.

    Article  CAS  PubMed  Google Scholar 

  59. Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, Siemianowicz K, Likus W, Wiechec E, Toyota BD, et al. Glioblastoma and chemoresistance to alkylating agents: involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther. 2018;184:13–41.

    Article  CAS  PubMed  Google Scholar 

  60. Mojas N, Lopes M, Jiricny J. Mismatch repair-dependent processing of methylation damage gives rise to persistent single-stranded gaps in newly replicated DNA. Genes Dev. 2007;21:3342–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Das A, Banik NL, Patel SJ, Ray SK. Dexamethasone protected human glioblastoma U87MG cells from temozolomide induced apoptosis by maintaining Bax:Bcl-2 ratio and preventing proteolytic activities. Mol Cancer. 2004;3:36.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Filippi-Chiela EC, Bueno e Silva MM, Thome MP, Lenz G. Single-cell analysis challenges the connection between autophagy and senescence induced by DNA damage. Autophagy. 2015;11:1099–113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chen CM, Syu JP, Way TD, Huang LJ, Kuo SC, Lin CT, Lin CL. BC3EE2,9B, a synthetic carbazole derivative, upregulates autophagy and synergistically sensitizes human GBM8901 glioblastoma cells to temozolomide. Int J Mol Med. 2015;36:1244–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kanzawa T, Germano IM, Komata T, Ito H, Kondo Y, Kondo S. Role of autophagy in temozolomide-induced cytotoxicity for malignant glioma cells. Cell Death Differ. 2004;11:448–57.

    Article  CAS  PubMed  Google Scholar 

  65. Song Q, Peng S, Sun Z, Heng X, Zhu X. Temozolomide drives ferroptosis via a DMT1-Dependent pathway in Glioblastoma Cells. Yonsei Med J. 2021;62:843–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ye LF, Chaudhary KR, Zandkarimi F, Harken AD, Kinslow CJ, Upadhyayula PS, Dovas A, Higgins DM, Tan H, Zhang Y, et al. Radiation-Induced lipid peroxidation triggers Ferroptosis and synergizes with Ferroptosis Inducers. ACS Chem Biol. 2020;15:469–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Buccarelli M, Marconi M, Pacioni S, De Pascalis I, D’Alessandris QG, Martini M, Ascione B, Malorni W, Larocca LM, Pallini R, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018;9:841.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Tong S, Hong Y, Xu Y, Sun Q, Ye L, Cai J, Ye Z, Chen Q, Tian D. TFR2 regulates ferroptosis and enhances temozolomide chemo-sensitization in gliomas. Exp Cell Res. 2023;424:113474.

    Article  CAS  PubMed  Google Scholar 

  69. Liu HJ, Hu HM, Li GZ, Zhang Y, Wu F, Liu X, Wang KY, Zhang CB, Jiang T. Ferroptosis-related gene signature predicts Glioma Cell Death and Glioma Patient Progression. Front Cell Dev Biol. 2020;8:538.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Galavotti S, Bartesaghi S, Faccenda D, Shaked-Rabi M, Sanzone S, McEvoy A, Dinsdale D, Condorelli F, Brandner S, Campanella M, et al. The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene. 2013;32:699–712.

    Article  CAS  PubMed  Google Scholar 

  71. Katayama M, Kawaguchi T, Berger MS, Pieper RO. DNA damaging agent-induced autophagy produces a cytoprotective adenosine triphosphate surge in malignant glioma cells. Cell Death Differ. 2007;14:548–58.

    Article  CAS  PubMed  Google Scholar 

  72. Hu Z, Mi Y, Qian H, Guo N, Yan A, Zhang Y, Gao X. A potential mechanism of Temozolomide Resistance in Glioma-Ferroptosis. Front Oncol. 2020;10:897.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wang Z, Wang Y, Yang T, Xing H, Wang Y, Gao L, Guo X, Xing B, Wang Y, Ma W. Machine learning revealed stemness features and a novel stemness-based classification with appealing implications in discriminating the prognosis, immunotherapy and temozolomide responses of 906 glioblastoma patients. Brief Bioinform 2021, 22.

  74. Liang L, Yan B, Liu Y, Jiang S, He H, Huang J, Liu W, Xie L. FOXP3 Contributes to TMZ Resistance, Prognosis, and Immune Infiltration in GBM from a Novel Pyroptosis-Associated Risk Signature. Dis Markers 2022, 2022:4534080.

  75. Yang T, Kong Z, Ma W. PD-1/PD-L1 immune checkpoint inhibitors in glioblastoma: clinical studies, challenges and potential. Hum Vaccin Immunother. 2021;17:546–53.

    Article  CAS  PubMed  Google Scholar 

  76. Suresh K, Naidoo J, Lin CT, Danoff S. Immune Checkpoint Immunotherapy for Non-small Cell Lung Cancer: benefits and pulmonary toxicities. Chest. 2018;154:1416–23.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in melanoma. Lancet. 2021;398:1002–14.

    Article  CAS  PubMed  Google Scholar 

  78. Beug ST, Beauregard CE, Healy C, Sanda T, St-Jean M, Chabot J, Walker DE, Mohan A, Earl N, Lun X et al. Smac mimetics synergize with immune checkpoint inhibitors to promote tumour immunity against glioblastoma. Nat Commun 2017, 8.

  79. Lee MJ, Ye AS, Gardino AK, Heijink AM, Sorger PK, MacBeath G, Yaffe MB. Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks. Cell. 2012;149:780–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zhang X, Chen W, Fan J, Wang S, Xian Z, Luan J, Li Y, Wang Y, Nan Y, Luo M, et al. Disrupting CD47-SIRPalpha axis alone or combined with autophagy depletion for the therapy of glioblastoma. Carcinogenesis. 2018;39:689–99.

    Article  CAS  PubMed  Google Scholar 

  81. Ott PA, Bang YJ, Piha-Paul SA, Razak ARA, Bennouna J, Soria JC, Rugo HS, Cohen RB, O’Neil BH, Mehnert JM, et al. T-Cell-inflamed gene-expression Profile, programmed death Ligand 1 expression, and Tumor Mutational Burden Predict Efficacy in patients treated with Pembrolizumab Across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37:318–27.

    Article  PubMed  Google Scholar 

  82. Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A, Baehring J, Ahluwalia MS, Roth P, Bahr O, et al. Effect of Nivolumab vs Bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 phase 3 Randomized Clinical Trial. JAMA Oncol. 2020;6:1003–10.

    Article  PubMed  Google Scholar 

  83. Lim M, Weller M, Idbaih A, Steinbach J, Finocchiaro G, Raval RR, Ansstas G, Baehring J, Taylor JW, Honnorat J, et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. 2022;24:1935–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Omuro A, Brandes AA, Carpentier AF, Idbaih A, Reardon DA, Cloughesy T, Sumrall A, Baehring J, van den Bent M, Bahr O, et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: an international randomized phase III trial. Neuro Oncol. 2023;25:123–34.

    Article  CAS  PubMed  Google Scholar 

  85. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, Lopez-Janeiro A, Porciuncula A, Idoate MA, Inoges S, de Andrea C, de Lopez-Diaz A, Tejada S, et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 2019;25:470–6.

    Article  CAS  PubMed  Google Scholar 

  86. Lee AH, Sun L, Mochizuki AY, Reynoso JG, Orpilla J, Chow F, Kienzler JC, Everson RG, Nathanson DA, Bensinger SJ, et al. Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma. Nat Commun. 2021;12:6938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Bagley SJ, Desai AS, Linette GP, June CH, O’Rourke DM. CAR T-cell therapy for glioblastoma: recent clinical advances and future challenges. Neuro Oncol. 2018;20:1429–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. O’Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, Martinez-Lage M, Brem S, Maloney E, Shen A et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med 2017, 9.

  89. Wenger C, Miranda PC, Salvador R, Thielscher A, Bomzon Z, Giladi M, Mrugala MM, Korshoej AR. A review on Tumor-Treating fields (TTFields): clinical implications inferred from computational modeling. IEEE Rev Biomed Eng. 2018;11:195–207.

    Article  PubMed  Google Scholar 

  90. Rominiyi O, Vanderlinden A, Clenton SJ, Bridgewater C, Al-Tamimi Y, Collis SJ. Tumour treating fields therapy for glioblastoma: current advances and future directions. Br J Cancer. 2021;124:697–709.

    Article  PubMed  Google Scholar 

  91. Vanderlinden A, Jones CG, Myers KN, Rominiyi O, Collis SJ. DNA damage response inhibitors enhance tumour treating fields (TTFields) potency in glioma stem-like cells. Br J Cancer. 2023;129:1829–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Voloshin T, Kaynan N, Davidi S, Porat Y, Shteingauz A, Schneiderman RS, Zeevi E, Munster M, Blat R, Tempel Brami C, et al. Tumor-treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti-PD-1 therapy. Cancer Immunol Immunother. 2020;69:1191–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Stingele S, Stoehr G, Storchova Z. Activation of autophagy in cells with abnormal karyotype. Autophagy. 2013;9:246–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Tran DD, Ghiaseddin AP, Chen DD, Le SB. Final analysis of 2-THE-TOP: a phase 2 study of TTFields (Optune) plus pembrolizumab plus maintenance temozolomide (TMZ) in patients with newly diagnosed glioblastoma. J Clin Oncol. 2023;41:2024–2024.

    Article  Google Scholar 

  95. Wang M, Zhang C, Wang X, Yu H, Zhang H, Xu J, Zhao J, Jiang X. Tumor-treating fields (TTFields)-based cocktail therapy: a novel blueprint for glioblastoma treatment. Am J Cancer Res. 2021;11:1069–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Stupp R, Taillibert S, Kanner AA, Kesari S, Steinberg DM, Toms SA, Taylor LP, Lieberman F, Silvani A, Fink KL, et al. Maintenance Therapy with Tumor-Treating Fields Plus Temozolomide vs Temozolomide alone for Glioblastoma: a Randomized Clinical Trial. JAMA. 2015;314:2535–43.

    Article  CAS  PubMed  Google Scholar 

  97. Giladi M, Munster M, Schneiderman RS, Voloshin T, Porat Y, Blat R, Zielinska-Chomej K, Haag P, Bomzon Z, Kirson ED, et al. Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cells. Radiat Oncol. 2017;12:206.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Lu G, Rao M, Zhu P, Liang B, El-Nazer RT, Fonkem E, Bhattacharjee MB, Zhu JJ. Triple-drug Therapy with Bevacizumab, Irinotecan, and Temozolomide Plus Tumor Treating fields for recurrent glioblastoma: a retrospective study. Front Neurol. 2019;10:42.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Lee WS, Seo SJ, Chung HK, Park JW, Kim JK, Kim EH. Tumor-treating fields as a proton beam-sensitizer for glioblastoma therapy. Am J Cancer Res. 2021;11:4582–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Horbinski C, Nabors LB, Portnow J, Baehring J, Bhatia A, Bloch O, Brem S, Butowski N, Cannon DM, Chao S, et al. NCCN Guidelines(R) insights: Central Nervous System Cancers, Version 2.2022. J Natl Compr Canc Netw. 2023;21:12–20.

    Article  CAS  PubMed  Google Scholar 

  101. Horbinski C, Nabors LB, Portnow J, Baehring J, Bhatia A, Bloch O, Brem S, Butowski N, Cannon DM, Chao S, et al. NCCN Guidelines® insights: Central Nervous System Cancers, Version 2.2022. J Natl Compr Canc Netw. 2023;21:12–20.

    Article  CAS  PubMed  Google Scholar 

  102. Floyd NS, Woo SY, Teh BS, Prado C, Mai WY, Trask T, Gildenberg PL, Holoye P, Augspurger ME, Carpenter LS, et al. Hypofractionated intensity-modulated radiotherapy for primary glioblastoma multiforme. Int J Radiat Oncol Biol Phys. 2004;58:721–6.

    Article  PubMed  Google Scholar 

  103. Hingorani M, Colley WP, Dixit S, Beavis AM. Hypofractionated radiotherapy for glioblastoma: strategy for poor-risk patients or hope for the future? Br J Radiol. 2012;85:e770–781.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. McKelvey KJ, Hudson AL, Donaghy H, Stoner SP, Wheeler HR, Diakos CI, Howell VM. Differential effects of radiation fractionation regimens on glioblastoma. Radiat Oncol. 2022;17:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Khan I, Baig MH, Mahfooz S, Rahim M, Karacam B, Elbasan EB, Ulasov I, Dong JJ, Hatiboglu MA. Deciphering the role of Autophagy in Treatment of Resistance mechanisms in Glioblastoma. Int J Mol Sci 2021, 22.

  106. Koukourakis MI, Mitrakas AG, Giatromanolaki A. Therapeutic interactions of autophagy with radiation and temozolomide in glioblastoma: evidence and issues to resolve. Br J Cancer. 2016;114:485–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Kotecha R, Mehta MP. Extreme hypofractionation for newly diagnosed glioblastoma: rationale, dose, techniques, and outcomes. Neuro Oncol. 2020;22:1062–4.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Fu J, Liu ZG, Liu XM, Chen FR, Shi HL, Pangjesse CS, Ng HK, Chen ZP. Glioblastoma stem cells resistant to temozolomide-induced autophagy. Chin Med J (Engl). 2009;122:1255–9.

    CAS  PubMed  Google Scholar 

  109. Liao G, Zhao Z, Yang H, Li X. Efficacy and safety of Hypofractionated Radiotherapy for the Treatment of Newly Diagnosed Glioblastoma Multiforme: a systematic review and Meta-analysis. Front Oncol. 2019;9:1017.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Edmonston D, Vanderwalde N, Ballo M. Hypofractionation in older adults with Glioblastoma. Semin Radiat Oncol. 2022;32:168–71.

    Article  PubMed  Google Scholar 

  111. Qi XS, Schultz CJ, Li XA. An estimation of radiobiologic parameters from clinical outcomes for radiation treatment planning of brain tumor. Int J Radiat Oncol Biol Phys. 2006;64:1570–80.

    Article  PubMed  Google Scholar 

  112. Kalasauskas D, Sorokin M, Sprang B, Elmasri A, Viehweg S, Salinas G, Opitz L, Rave-Fraenk M, Schulz-Schaeffer W, Kantelhardt SR et al. Diversity of clinically relevant Outcomes resulting from Hypofractionated Radiation in Human Glioma stem cells mirrors distinct patterns of transcriptomic changes. Cancers (Basel) 2020, 12.

  113. Sahebjam S, Forsyth PA, Tran ND, Arrington JA, Macaulay R, Etame AB, Walko CM, Boyle T, Peguero EN, Jaglal M, et al. Hypofractionated stereotactic re-irradiation with pembrolizumab and bevacizumab in patients with recurrent high-grade gliomas: results from a phase I study. Neuro Oncol. 2021;23:677–86.

    Article  CAS  PubMed  Google Scholar 

  114. Jarosz-Biej M, Smolarczyk R, Cichon T, Kulach N. Tumor Microenvironment as a game changer in Cancer Radiotherapy. Int J Mol Sci 2019, 20.

  115. Gandhi S, Chandna S. Radiation-induced inflammatory cascade and its reverberating crosstalks as potential cause of post-radiotherapy second malignancies. Cancer Metastasis Rev. 2017;36:375–93.

    Article  CAS  PubMed  Google Scholar 

  116. Ostrand-Rosenberg S, Horn LA, Ciavattone NG. Radiotherapy both promotes and inhibits myeloid-derived suppressor cell function: novel strategies for preventing the Tumor-Protective effects of Radiotherapy. Front Oncol. 2019;9:215.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Wang Y, Liu ZG, Yuan H, Deng W, Li J, Huang Y, Kim BYS, Story MD, Jiang W. The reciprocity between Radiotherapy and Cancer Immunotherapy. Clin Cancer Res. 2019;25:1709–17.

    Article  CAS  PubMed  Google Scholar 

  118. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J, Durham N, Meyer C, Harris TJ, Albesiano E, et al. Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys. 2013;86:343–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Chen C, Lee I, Tatsui C, Elder T, Sloan AE. Laser interstitial thermotherapy (LITT) for the treatment of tumors of the brain and spine: a brief review. J Neurooncol. 2021;151:429–42.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Pang S, Kapur A, Zhou K, Anastasiadis P, Ballirano N, Kim AJ, Winkles JA, Woodworth GF, Huang HC. Nanoparticle-assisted, image-guided laser interstitial thermal therapy for cancer treatment. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2022;14:e1826.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Bozinov O, Yang Y, Oertel MF, Neidert MC, Nakaji P. Laser interstitial thermal therapy in gliomas. Cancer Lett. 2020;474:151–7.

    Article  CAS  PubMed  Google Scholar 

  122. de Groot JF, Kim AH, Prabhu S, Rao G, Laxton AW, Fecci PE, O’Brien BJ, Sloan A, Chiang V, Tatter SB, et al. Efficacy of laser interstitial thermal therapy (LITT) for newly diagnosed and recurrent IDH wild-type glioblastoma. Neurooncol Adv. 2022;4:vdac040.

    PubMed  PubMed Central  Google Scholar 

  123. Takaki H, Cornelis F, Kako Y, Kobayashi K, Kamikonya N, Yamakado K. Thermal ablation and immunomodulation: from preclinical experiments to clinical trials. Diagn Interv Imaging. 2017;98:651–9.

    Article  CAS  PubMed  Google Scholar 

  124. Chu KF, Dupuy DE. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat Rev Cancer. 2014;14:199–208.

    Article  CAS  PubMed  Google Scholar 

  125. Lerner EC, Edwards RM, Wilkinson DS, Fecci PE. Laser ablation: heating up the anti-tumor response in the intracranial compartment. Adv Drug Deliv Rev. 2022;185:114311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Liu EK, Sulman EP, Wen PY, Kurz SC. Novel therapies for Glioblastoma. Curr Neurol Neurosci Rep. 2020;20:19.

    Article  PubMed  Google Scholar 

  127. Ostberg JR, Dayanc BE, Yuan M, Oflazoglu E, Repasky EA. Enhancement of natural killer (NK) cell cytotoxicity by fever-range thermal stress is dependent on NKG2D function and is associated with plasma membrane NKG2D clustering and increased expression of MICA on target cells. J Leukoc Biol. 2007;82:1322–31.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National High Level Hospital Clinical Research Funding (2022-PUMCH-A-019) and the Beijing Municipal Natural Science Foundation (7202150) for Yu Wang, and by the National High Level Hospital Clinical Research Funding (2022-PUMCH-B-113), the Tsinghua University-Peking Union Medical College Hospital Initiative Scientific Research Program (2019ZLH101) and the Beijing Municipal Natural Science Foundation (19JCZDJC64200[Z]) for Wenbin Ma.

Author information

Authors and Affiliations

Authors

Contributions

T.L., L.G. and X.K. drafted the work. J.L. and Y.S. analyzed the table. L.G. prepared Figs. 1 and 1. Y.W. and W.M. revised the work. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yu Wang or Wenbin Ma.

Ethics declarations

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liang, T., Gu, L., Kang, X. et al. Programmed cell death disrupts inflammatory tumor microenvironment (TME) and promotes glioblastoma evolution. Cell Commun Signal 22, 333 (2024). https://doi.org/10.1186/s12964-024-01602-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-024-01602-0

Keywords