Open Access

Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review)

  • Authors:
    • Jianheng Wu
    • Nannan Wang
  • View Affiliations

  • Published online on: September 30, 2024     https://doi.org/10.3892/mmr.2024.13344
  • Article Number: 221
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Glioblastoma (GBM) is the most common central nervous system malignancy in adults. GBM may be classified as grade IV diffuse astrocytoma according to the 2021 World Health Organization revised classification of central nervous system tumors, which means it is the most aggressive, invasive, undifferentiated type of tumor. Immune checkpoint blockade (ICB), particularly anti‑programmed cell death protein‑1 (PD‑1)/PD‑1 ligand‑1 immunotherapy, has been confirmed to be successful across several tumor types. However, in GBM, this treatment is still uncommon and the efficacy is unpredictable, and <10% of patients show long‑term responses. Recently, numerous studies have been conducted to explore what factors may indicate or affect the ICB response rate in GBM, including molecular alterations, immune expression signatures and immune infiltration. The present review aimed to summarize the current progress to improve the understanding of immunotherapy for GBM.

Introduction

Glioblastoma (GBM) is the most common central nervous system malignancy in adults (1,2). GBM may be classified as grade IV diffuse astrocytoma according to the 2021 World Health Organization revised classification of central nervous system tumors, which indicates the most aggressive, invasive, undifferentiated type of tumor (3,4). The median survival after diagnosis is only 14–16 months, with a 2-year survival rate of 26.5% and a 5-year survival rate of 5% (1). GBM arises from astrocytes and belongs to a heterogeneous collection of GBM brain tumors. Most GBM is found in the supratentorial brain (frontal, temporal, parietal, occipital lobes, cerebellum), brainstem and spinal cord, but manifestation in the spinal cord is rarer (1,5,6). The annual incidence of GBM is 3–5 per 100,000 individuals, and the median age at diagnosis is 65 years. Furthermore, men are ~1.6 times more likely than women to develop the disease (5,7,8). Risk factors for GBM development have not been well-identified and rare familial cancer syndromes, including neurofibroma type I, tuberous sclerosis, Lynch syndrome and Li-Fraumeni syndrome, are associated with an increased risk of GBM (9). In addition, exposure to ionizing radiation has been suggested as a risk factor for developing GBM (10).

GBM carries a range of mutations that provide a selective growth advantage for cells to promote survival and proliferation in a hostile and hypoxic environment (11), and 30–40% of GBM cases have amplification of epidermal growth factor receptor (EGFR), a tyrosine kinase receptor that activates MAPK and PI3K signaling (12,13). In addition, a subset of GBM expresses EGFRVIII variants in which the receptor's extracellular domain is absent, leading to constitutive activation (14). In addition, the extracellular matrix also participates in the migration of GBM cells and is associated with altering the microenvironmental composition (15). Furthermore, dysregulation of intracellular Ca2+ signaling contributes to the motility of GBM cells via activating numerous cell membrane channels such as inositol 1,4,5-triphosphate receptor channels and store-operated channels (16). Previous studies by our group also showed that aberrant expression of certain genes and long noncoding RNAs facilitates GBM cell malignancy and progression (1720).

Neuroimaging, surgical resection, radiotherapy and chemotherapy for malignant gliomas remain the standard of care (2,10,21). Surgical resection is the most frequently used approach in all grades of glioma, yet maximizing safe resection and protecting the patient's neurological function remains challenging (7,10). However, resection becomes more difficult if the tumor is located in a critical, unresectable location, such as the brain. Currently, the treatment of GBM is mainly performed by maximum and safest resection of the lesion in combination with radiotherapy and adjuvant chemotherapy in the form of oral temozolomide (22,23). Despite this, the median survival of patients with GBM after treatment is ~15 months, with a 2-year life expectancy of <30% (24). The prognosis is even worse for unresectable GBM (up to 35–40% of patients) (25,26). The microscopic infiltration of GBM cells and the tumor's location make it difficult to complete the lesion resection and markedly increase the chance of recurrence (27). Therefore, searching for new GBM therapies is an urgent and pressing need. The prognosis of GBM is usually poor because of its genetic instability, highly aggressive nature, high vascular proliferation and resistance to chemotherapeutic agents (27). There is still no effective treatment for progressive or relapsed GBM, which occurs invariably in most patients (28). GBM represents one of the greatest therapeutic challenges of the modern era, with a low cure rate, high recurrence rate and limited survival time. Therapeutic options are limited at the time of new diagnosis and relapse (10,29).

Anti-programmed cell death protein-1 (PD-1)/PD-1 ligand-1 (PD-L1) immunotherapy

The idea of tumor immunotherapy is to deploy the immune system as a tool to treat cancer (30). Immunotherapy offers great hope for a cure in cancer treatment by re-educating and harnessing the patient's immune response to the tumor (29). Current immunotherapeutic strategies used to treat cancer are mainly based on immune checkpoint blockers (ICB) (29,31), therapeutic vaccines (29,32), adoptive cell therapy (33,34), monoclonal antibodies (35) and oncolytic viruses (36). The current review just focuses on ICB, anti-PD-1/PDL1 immunotherapy in particular.

The PD-1 receptor is expressed primarily on activated T cells, and binding of PD-1 to PD-L1 leads to the inactivation of these T cells (Fig. 1). The normal function of the pair of receptor and ligand is to ensure that T-cell responses preserve self-tolerance while effectively protecting the body from pathogens and neoplasia (30). However, in cancer, overexpression of PD-L1 was found to constrain the CD8+ T-cell cytotoxic antitumour response, which is dependent on PD-1 (37,38). Based on this, monoclonal antibodies targeting PD-1 and PD-L1 are developed to treat cancer.

In the last decade, anti-PD-1/PDL1 has shown remarkable success in treating a variety of tumors, including advanced melanoma (39), non-small-cell lung cancer (40), Hodgkin's lymphoma (41,42) and other solid tumors (41,43,44).

Anti-PD-1/PDL1 immunotherapy in GBM

While the brain was previously thought to be an ‘immune privileged’ organ because of the blood-brain barrier (BBB), an increasing amount of research has shown that T cells and antigen-presenting cells (APCs, i.e. microglia, macrophages and dendritic cells) may transfer through cerebrospinal fluid-filled channels and the perivascular space to recognize neoplastic cells (45). Studies have indicated that the PD-1 receptor is expressed particularly on brain activated T cells (45) and PD-L1 is also overexpressed by GBM and APC cells, leading to the effective binding of PD-L1 to PD-1, thereby suppressing the immune response (46). While it has remained elusive how anti-PD-1/PDL1 penetrates the BBB in brain tumors, certain studies proved that blocking PD-1/PDL1 provides a survival benefit with intratumoral and systemic immune responses in recurrent GBM (47,48), which indicates that at least part of the anti-PD-1/PDL1 is able to penetrate the BBB to reach brain tumors. One potential mechanism is equilibrative nucleoside transporter 2, a transporter that regulates nucleoside flux at the BBB, which may offer an unexpected path for anti-PD-1/PDL1 to brain tumors (48).

There has been considerable interest in utilizing immunotherapy in GBM; however, most clinical studies of anti-PD-1/PD-L1 monotherapy for GBM have shown limited efficacy (28,49). As an illustration, pembrolizumab, a Food and Drug Administration-approved anti-PD1 treatment for non-small cell lung cancer, melanoma, hepatocellular carcinoma and various other cancers, did not extend the lifespan of individuals with GBM multiforme when administered in isolation (50). The Keynote-0289 clinical trial explored the use of pembrolizumab as a standalone treatment in 26 patients with recurrent GBM. The results indicated relatively modest survival advantages, with a median duration of stable disease (SD) at 39.4 weeks (range, 7.1-85.9 weeks), a median progression-free survival (PFS) of 2.8 months (range, 1.9-9.1 months) and a median overall survival (OS) of 14.4 months (range, 10.3 months to not reached) (51). Furthermore, multiple studies focusing on high-grade gliomas (classified as World Health Organization grade 3 and 4) have demonstrated that pembrolizumab as a sole therapy exhibits limited effectiveness compared to control groups (52). Nivolumab is another widely approved PD-1 targeting antibody. The Checkmate 143 trial, which is the first large-scale phase 3 clinical trial involving the use of ICB in the treatment of GBM, initially assessed the combination of ipilimumab and nivolumab. However, compared to the control group treated with bevacizumab, nivolumab did not result in any significant improvement in survival (53). The 12-month OS rate was 42% in both the nivolumab and bevacizumab arms, while the median OS was 9.8 months in the nivolumab group and 10.0 months in the bevacizumab group. The median PFS was 1.5 months for the nivolumab group and 3.5 months for the bevacizumab group (53). As to anti-PD-L1 antibodies, atezolizumab and durvalumab have been approved for several cancers. A phase 1 clinical trial of atezolizumab monotherapy involving 16 patients with recurrent GBM15 demonstrated unextended survival, except for 3 patients with isocitrate dehydrogenase (IDH) or DNA polymerase ε (POLE) mutations who survived for >16 months. A phase 2 trial evaluating durvalumab in 5 GBM cohorts published preliminary results. Data from subgroups indicated partial clinical benefits. In the arm involving 30 patients with recurrent GBM who received durvalumab monotherapy, the overall disease control rate was 60.0% (4 with a partial response and 14 with SD). The median OS was 28.9 weeks (22.9-not reached) and the median PFS was 13.9 weeks (range, 8.1-24.0 weeks). As to anti-PD-L1 antibodies, both atezolizumab and durvalumab have obtained approval for treating various cancers (54,55). In a phase I clinical trial of atezolizumab as a single-agent therapy, which included 16 patients with recurrent GBM, it was observed that the treatment did not significantly extend OS (51). In one arm of the trial, which involved 30 patients with recurrent GBM treated with durvalumab as a monotherapy, the overall disease control rate was 60.0%, with four patients exhibiting partial responses and 14 showing SD. The median OS was 28.9 weeks (range, 22.9 weeks to not reached) and the median PFS was 13.9 weeks (range, 8.1 to 24.0 weeks) (56). In conclusion, the effectiveness of anti-PD-1/PDL1 monotherapy in the treatment of GBM is not deemed satisfactory. There is a need for in-depth analysis of the underlying factors contributing to this outcome and the development of novel treatment approaches. Below, emerging studies on the elements affecting the response rate of anti-PD-1/PDL1 immunotherapy in GBM are summarized.

Common causes of anti-PD-1/PDL1 immunotherapy resistance

A recent clinical trial of PD-1 immune checkpoint inhibitors in recurrent GBM showed that only a small subset of patients (8%) demonstrated objective responses (49). Patients with specific GBM subtypes may benefit more from anti-PD-1/PDL1 immunotherapy therapies. A higher tumor mutation burden (TMB), higher microsatellite instability (MSI), neoantigen, MMR system deficiency (MMRD) and germline POLE mutation suggest better efficacy, which has been confirmed in clinical practice (50). The MMR genes, including PMS1 homolog 2, mismatch repair system component, mutL homolog 1, mutS homolog 2 and mutS homolog 6, take care of DNA mismatches during replication and maintain DNA stability (50,57), and POLE acts as the major leading-strand DNA polymerase for mismatch repair during genome replication (58). MMRD and POLE mutation lead to increases in the TMB, higher MSI and more neoantigens (50,58). Studies have indicated that in GBM, the factors of MMRD, POLE mutation and higher mutational burden are associated with better anti-PD-1/PDL1 efficacy (50,59). In addition, high PD-L1 expression is associated with poor prognosis in patients with GBM (60), while they show a better response to PD-1-based immunotherapy in GBM (61,62). This is expected, as PD-L1 suppresses the T cells' function (47), while it may be targeted by anti-PD-1/PDL1. In addition, a recent study in an in situ GBM mouse model found reduced cytotoxic capacity of tumor-infiltrating lymphocytes (TIL) and a more polyclonal TCR library in anti-PD-1/PDL1-resistant mice (63), indicating that T-cell infiltration and dysfunction markers may also be important in accurately predicting anti-PD-1/PDL1 response (31,64). The above evidence shows that PD-L1 expression, tumor mutational load, neoantigen load and tumor-infiltrating T-lymphocytes may also affect the response rate of anti-PD-1/PDL1 immunotherapy in GBM.

Genomic and transcriptomic associations of response to anti-PD-1/PDL1 immunotherapy in GBM

In GBM, numerous mutations occur in suppressor genes, such as p53, p21, p16 and phosphatase and tensin homolog (PTEN), indicating a high degree of cellular instability (65,66). This high instability has led to a lack of effective therapeutic approaches. To further identify molecular determinants of immunotherapeutic response in GBM, an emerging clinical study by Zhao et al (28) carried out a retrospective analysis of individuals with recurrent GBM, 17 of whom were ‘long-term’ responders who had SD for at least 6 months and 49 of whom were non-responders to anti-PD-1 therapy. Genomic and transcriptomic analysis revealed noteworthy mutations significantly enriched in either responsive or non-responsive tumors.

PTEN

In the study by Zhao et al (28), 23 PTEN mutations were found among the 32 non-responders but only three mutations among the 13 responders (28). Various existing studies on melanoma have proved that PTEN loss or mutation may lead to increasing immunosuppressive cytokine expression, resulting in decreased T-cell infiltration in tumors and inhibition of autophagy, which decreases T cell-mediated cell death (63,67), indicating that PTEN mutations are associated with poor response to anti-PD-1. In addition, loss of PTEN was also found to be associated with resistance to anti-PD-1 therapy in metastatic uterine leiomyosarcoma and prostate cancer (68,69). These studies indicate that PTEN mutation or expression screen may need to be performed prior to anti-PD1 treatment.

MAPK pathway

The MAPK pathway genes were also found to be frequently mutated in the aforementioned study (28). Among the 13 responders, four mutations in the MAPK pathway were found, while only one mutation was found among the 32 non-responders (28). Considering the high prevalence of mutations in BRAF, which encodes MAPK kinases and induces activation of the entire MAPK pathway, observed in various cancer types such as melanoma (70,71), colorectal cancer (72) and non-small cell lung cancer (72), performing a screen for MAPK pathway mutations or expression levels may help predict the response rate to anti-PD1 treatment (73). More importantly, pretreatment with anti-PD-1 has been demonstrated to mitigate therapeutic resistance to MAPK inhibitor treatment (74).

B-cell lymphoma 3 (BCL-3)

While certain genes have been identified to affect the response to anti-PD1, more biomarkers need to be further explored. Mechanistic studies have identified IDH1 mutations as a common type of GBM (3,4,75). However, no big difference in mutated IDH1 was found in the non-responsive tumors compared to the responsive one (28). Numerous studies have already shown that IDH1 may be involved in the pathogenesis of GBM through altered expression or mutations of genes, and IDH1 mutations may improve prognosis by suppressing the expression of BCL-3 (17). BCL-3 is a B-cell chronic lymphocytic leukemia-associated gene whose members include BCL1-11, an atypical inhibitor of κB protein. Functionally, BCL-3 both activates and inhibits NF-κB signaling (76). NF-κB is important in promoting GBM resistance to DNA-damaging agents (77,78). In clinical studies, positive expression of BCL3 in gliomas was found to correlate directly with poor clinical features. Positive expression of BCL3 is associated with reduced 5-year OS in patients with glioma (17). A recent study suggests that BCL3 may be a promising biomarker and prognostic indicator for GBM (21).

A study has identified that BCL3 may exhibit immunomodulatory functions as an upstream transcription factor of IDH1 (79). Functionally, BCL-3 can regulate clone formation and cell cycle progression by regulating ubiquitination-mediated c-Myc degradation in colorectal cancer, leading to upregulation of PD-L1 expression, which in turn promotes apoptosis and tolerance of T cells and suppresses tumor immunity (80). These data indicate that the BCL3 expression is a promising predictor of the anti-PD-1/PDL1 responses.

Complex immunosuppressive microenvironment

While improved responses to anti-PD-1/PDL1 therapy are associated with higher mutational burdens in tumors across multiple cancer types and with levels of T-cell infiltration in the tumor microenvironment (TME) (31,43,44), compared with melanomas or non-small-cell lung cancer, GBM harbors a lower burden of somatic mutations and a more immunosuppressive TME (81). In GBM, the TME consists of multiple cell types: Infiltrating tumor cells and cancer stem cells, as well as non-cancerous cells, such as bone marrow cells, resident histiocytes and lymphocytes, all of which may interact with each other, and the cells secrete chemokines, growth factors and cytokines into the TME and the release of these molecules attracts and stimulates immunosuppressive cells (81,82). These immune cells contribute to the efficacy of anti-PD-1 therapy. For instance, tumor-associated macrophages (TAMs) drive anti-PD-1/PDL1 immunotherapy resistance through PD-L1/CD80-mediated CD4+ T-cell suppression and Treg amplification (83). Whether PD-L1 expression on TAMs is a preexisting or acquired anti-PD-1/PDL1 immunotherapy resistance mechanism and whether it may be used as a biomarker for patient stratification remains to be investigated (21).

In addition, analysis of peripheral blood from patients with primary and recurrent GBM and central and borderline tumor areas indicated an increased presence of hematogenous MΦs in both tumor areas, a higher frequency of infiltrating lymphocytes and high levels of depletion markers in recurrent GBM (84). A significant negative correlation between infiltrating T cells and myeloid-derived suppressor cell subpopulations was also confirmed, again suggesting that immune cell composition affects the clinical outcome (84,85). These immunosuppressive cells interact with GBM cells through different immunosuppressive receptors or compete to consume the PD-1 antibody, leading to anti-PD-1/PDL1 immunotherapy resistance (28,44).

Combination of anti-PD-1/PDL1 therapy and other treatments

Numerous factors could be mentioned that may predict or enhance the response of anti-PD-1/L1 therapy; however, of note, GBM is largely resistant to anti-PD-1/PDL1 immunotherapy, except for rare hypermutated GBM. As the efficacy of anti-PD-1/PDL1 monotherapy has not met expectations, combination therapies with anti-PD-1/PDL1 and other treatments are being pursued. Preclinical animal studies have indicated the partial potential of the combination of standard therapy, targeted therapy or other immunotherapies (28,44). Anti-PD-1/PDL1 immunotherapy is currently well tolerated in the presence of an immune response in ongoing ICB clinical studies. In recent prospective, relatively early-phase clinical trials in which anti-PD-1 therapy was administered in a neoadjuvant setting (anti-PD-1 was administered before and after surgical resection of recurrent GBM) (49), it was found that neoadjuvant nivolumab could enhance the expression of chemokine transcripts, leading to higher immune cell infiltration and augmenting TCR clonal diversity among tumor-infiltrating T lymphocytes, supporting a local immunomodulatory effect of treatment (86). In another randomized prospective trial comparing two schedules of pembrolizumab [before and after (neoadjuvant) surgery vs. only after (adjuvant) surgery for 35 recurrent GBMs], neoadjuvant PD-1 (pembrolizumab) was found to significantly improve both overall (13.7 vs. 7.5 months, P=0.04) and PFS (3.3 vs. 2.4 months, P=0.03) (86).

Furthermore, neoadjuvant PD-1 showed a significant increase in gene signatures related to interferon-γ responsiveness and a significant decrease in the number of tumors with cell cycle gene expression signatures. It was also evaluated whether there was evidence of ICB treatment prompting immune evasion by GBMs, indicating that more neoadjuvant tumors showed spatially focal induction of PD-L1 expression when compared with adjuvant tumors (48,86). In addition, blockade of T cell immunoreceptor with Ig and ITIM domains has also been confirmed to improve the effectiveness of anti-PD-1/PD-L1 monotherapy via augmented CD8+ TIL accumulation and functions in a murine GBM model (87). This indicates that combination with other treatments will be the future of anti-PD-1/PDL1 treatment. An increasing number of clinical trials on the combination of anti-PD-1/PD-L1 monotherapy with other treatments to promote the response rate are being performed. Information on the current trials of the combination treatments is provided in Table I.

Table I.

Current clinical trials investigating the combination of anti-programmed cell death protein-1/programmed cell death protein-1 ligand-1 monotherapy with other treatments in glioblastoma.

Table I.

Current clinical trials investigating the combination of anti-programmed cell death protein-1/programmed cell death protein-1 ligand-1 monotherapy with other treatments in glioblastoma.

NCT numberDatesPopulationOutcome measuresDesignPhaseCheckpoint inhibitorCombination
NCT037074572019.3-2024.230PFS, OS, toxicityNon-randomized1Nivolumab + ipilimumabMK-4166 (anti-GITR) INCB024360 (IDO1 inhibitor)
NCT03422094 2018.10-2023.1030Toxicity, personalized neoantigen peptide vaccine generationNon-randomized1Nivolumab + ipilimumabNeovax
NCT037265152019.3-2034.127OS, PFS, toxicity, ORRSingle group1PembrolizumabEgfrviii-CAR-T
NCT037223422019.1-2020.420OS, PFS, toxicity, ORR, DCR, immunogenicitySequential assignment1PembrolizumabTTAC-0001 (Anti-VEGF2)
NCT040036492019.8-2022.160OS, OS-9, PFS, T cell levels, cytokine levels, tumor response, toxicityNon-randomized1Nivolumab + ipilimumab Il13ralpha2-CAR-T
NCT04015700 2019.10-2023.1030Toxicity, personalized neoantigen peptide vaccine generation, candidate tumor-specific neoantigen identificationNon-randomized1Nivolumab + ipilimumabPersonalized neoantigen vaccine
NCT039619712019.8-2023.615OS, PFS, toxicity, ORRSingle group1SpartalizumabMbg453 (anti-TIM3)
NCT040136722019.9-2021.1251PFS, toxicityNon-randomized2PembrolizumabSurvaxm sargramostim
NCT040477062019.9-2023.630OS, PFS-6, toxicity, ORR, RR, MDRNon-randomized1NivolumabBms-986205 RT TMZ
NCT023359182015.1-2018.12175ORR, OS-12Single group assignment1/2NivolumabVarlilumab
NCT032913142017.3- 2019.152OS, PFS-6, ORR, neurocognitive functionParallel assignment2AvelumabAxitinib
NCT029689402017.3-2020.843Toxicity, PFS-6, PFS, OS, ORR, MDRSingle group2AvelumabHypofractionated radiation therapy
NCT034916832018.5- 2021.152Toxicity, OS-18, molecular changesNon-randomized1/2CemiplimabIno-5401 Ino-9012
NCT022874282014.11-2021.846Toxicity, PFS-8, T cell responseRandomized1PembrolizumabRT, personalized neoantigen vaccine

[i] DCR, disease control rate; Egfrviii-CAR-T, chimeric antigen receptor T cells targeting epidermal growth factor receptor viii mutation (deletion of exons 2–7); GITR, humanized IgG1 agonist mAb targeting glucocorticoid-induced TNF receptor; IDO1, indoleamine 2,3-dioxigenase 1; Il13ralpha2-CAR-T, Chimeric antigen receptor T cells targeting Il13ralpha2; Ino-5401, synthetic DNA plasmid encoding telomerase, Wilms Tumor-1 and prostate specific membrane antigen; Ino-9012, synthetic DNA plasmid encoding IL-12; Mbg453, sabatolimab; MDR, medical device reporting; NCT, National Clinical Trial; ORR, overall response rate; OS, overall survival; OS-9, 9-month OS; OS-12, 12-month OS; OS-18, 18-month OS; RR, relative risk; RT, radiation therapy; PFS, progression-free survival; PFS-6, 6-month PFS; PFS-8, 8-month PFS; TIM3, T cell immunoglobulin and mucin domain-containing protein 3; TMZ, temozolomide; VEGF2, vascular endothelial growth factor 2.

Conclusion

The treatment of GBM has been based on standard surgical resection combined with radiotherapy or chemotherapy, with poor patient prognosis. Although immunotherapy has revolutionized the treatment of numerous cancers, treatment outcomes remain suboptimal in GBM (4,70). The growth location, heterogeneity and widespread immunosuppression of GBM pose significant challenges to its treatment and no approved immunotherapies are available. Predictive biomarkers are even more important for the personalized treatment of GBM and it has now been demonstrated that the impact of the immune system is associated with therapeutic efficacy and clinical benefit. Immune checkpoint molecule (PD1/PDL1) expression has an important role and additional clinical studies should confirm the clinical therapeutic effect of the PD-1/PD-L1 axis in GBM. A series of studies have indicated that various factors influence the response rate of anti-PD-1/PD-L1, encompassing endogenetic factors within the GBM and immune cell infiltrates in the microenvironment (Fig. 2) (45,50,59,60), which will help to predict or enhance the response rate of anti-PD-1/PDL1. In addition, combining other therapies (surgical resection, radiotherapy or chemotherapy) will improve anti-PD-1/PDL1 treatments. In particular, the present review intends to emphasize that BCL-3 should receive more extensive attention as an important predictor in diseases where they can provide improved information for disease treatment or even new therapeutic targets (17,80). Of note, BCL-3 is reported to induce expression via tamoxifen (88) and IL-6 (89), so pretreating with these drugs to induce higher BCL-3 expression may offer a promising way to promote the efficacy of anti-PD-1 treatment. In addition, further characterization of tumor-infiltrating immune cell subpopulations may lead to the identification of new markers and targets. However, all of the above factors need to be systematically validated in larger patient cohorts to speculate on the relationship between prognostic predictors and the immune correlates of therapeutic efficacy to provide more data to support the treatment of GBM.

Acknowledgements

Not applicable.

Funding

The study was supported by the Medical Scientific Research Foundation of Guangdong Province, China (grant no. A2021298).

Availability of data and materials

Not applicable.

Author's contributions

JW and NW conceived and designed the study. JW and NW performed the literature search/selection. JW drafted the manuscript and NW reviewed and edited it. Data authentication is not applicable. Both authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Oronsky B, Reid TR, Oronsky A, Sandhu N and Knox SJ: A review of newly diagnosed glioblastoma. Front Oncol. 10:5740122021. View Article : Google Scholar : PubMed/NCBI

2 

Wang LB, Karpova A, Gritsenko MA, Kyle JE, Cao S, Li Y, Rykunov D, Colaprico A, Rothstein JH, Hong R, et al: Proteogenomic and metabolomic characterization of human glioblastoma. Cancer Cell. 39:509–528.e20. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Osborn AG, Louis DN, Poussaint TY, Linscott LL and Salzman KL: The 2021 World health organization classification of tumors of the central nervous System: What neuroradiologists need to know. AJNR Am J Neuroradiol. 43:928–937. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, et al: The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 23:1231–1251. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Miller KD, Ostrom QT, Kruchko C, Patil N, Tihan T, Cioffi G, Fuchs HE, Waite KA, Jemal A, Siegel RL, et al: Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 71:381–406. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Miller KE, Cassady KA, Roth JC, Clements J, Schieffer KM, Leraas K, Miller AR, Prasad N, Leavenworth JW, Aban IB, et al: Immune activity and response differences of oncolytic viral therapy in recurrent glioblastoma: Gene expression analyses of a phase IB study. Clin Cancer Res. 28:498–506. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Hanif F, Muzaffar K, Perveen K, Malhi SM and Simjee ShU: Glioblastoma Multiforme: A review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pac J Cancer Prev. 18:3–9. 2017.PubMed/NCBI

8 

Tadipatri R, Lyon K, Azadi A and Fonkem E: A view of the epidemiologic landscape: How population-based studies can lend novel insights regarding the pathophysiology of glioblastoma. Chin Clin Oncol. 10:352021. View Article : Google Scholar : PubMed/NCBI

9 

Ostrom QT, Adel Fahmideh M, Cote DJ, Muskens IS, Schraw JM, Scheurer ME and Bondy ML: Risk factors for childhood and adult primary brain tumors. Neuro Oncol. 21:1357–1375. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Vogelbaum MA, Brown PD, Messersmith H, Brastianos PK, Burri S, Cahill D, Dunn IF, Gaspar LE, Gatson NTN, Gondi V, et al: Treatment for brain metastases: ASCO-SNO-ASTRO guideline. J Clin Oncol. 40:492–516. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Cai X and Sughrue ME: Glioblastoma: New therapeutic strategies to address cellular and genomic complexity. Oncotarget. 9:9540–9554. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Ayati A, Moghimi S, Salarinejad S, Safavi M, Pouramiri B and Foroumadi A: A review on progression of epidermal growth factor receptor (EGFR) inhibitors as an efficient approach in cancer targeted therapy. Bioorg Chem. 99:1038112020. View Article : Google Scholar : PubMed/NCBI

13 

An Z, Aksoy O, Zheng T, Fan QW and Weiss WA: Epidermal growth factor receptor and EGFRvIII in glioblastoma: Signaling pathways and targeted therapies. Oncogene. 37:1561–1575. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Nozawa T, Okada M, Natsumeda M, Eda T, Abe H, Tsukamoto Y, Okamoto K, Oishi M, Takahashi H, Fujii Y and Kakita A: EGFRvIII is expressed in cellular areas of tumor in a subset of glioblastoma. Neurol Med Chir (Tokyo). 59:89–97. 2019. View Article : Google Scholar : PubMed/NCBI

15 

So JS, Kim H and Han KS: Mechanisms of invasion in glioblastoma:. Extracellular Matrix; Ca2+ signaling, and glutamate: Front Cell Neurosci. 15. pp. 6630922015, View Article : Google Scholar : PubMed/NCBI

16 

Kang SS, Han KS, Ku BM, Lee YK, Hong J, Shin HY, Almonte AG, Woo DH, Brat DJ, Hwang EM, et al: Caffeine-mediated inhibition of calcium release channel inositol 1,4,5-trisphosphate receptor subtype 3 blocks glioblastoma invasion and extends survival. Cancer Res. 70:1173–1183. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Wu J, Li L, Jiang G, Zhan H and Wang N: B-cell CLL/lymphoma 3 promotes glioma cell proliferation and inhibits apoptosis through the oncogenic STAT3 pathway. Int J Oncol. 49:2471–2479. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Wu J, Li L, Jiang G, Zhan H, Zhu X and Yang W: NCAPG2 facilitates glioblastoma cells' malignancy and xenograft tumor growth via HBO1 activation by phosphorylation. Cell Tissue Res. 383:693–706. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Wu J, Li R, Li L, Gu Y, Zhan H, Zhou C and Zhong C: MYC-activated lncRNA HNF1A-AS1 overexpression facilitates glioma progression via cooperating with miR-32-5p/SOX4 axis. Cancer Med. 9:6387–6398. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Wu J, Wang N, Yang Y, Jiang G, Zhan H and Li F: LINC01152 upregulates MAML2 expression to modulate the progression of glioblastoma multiforme via Notch signaling pathway. Cell Death Dis. 12:1152021. View Article : Google Scholar : PubMed/NCBI

21 

Aslan K, Turco V, Blobner J, Sonner JK, Liuzzi AR, Núñez NG, De Feo D, Kickingereder P, Fischer M, Green E, et al: Heterogeneity of response to immune checkpoint blockade in hypermutated experimental gliomas. Nat Commun. 11:9312020. View Article : Google Scholar : PubMed/NCBI

22 

Chang SM, Lamborn KR, Malec M, Larson D, Wara W, Sneed P, Rabbitt J, Page M, Nicholas MK and Prados MD: Phase II study of temozolomide and thalidomide with radiation therapy for newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys. 60:353–357. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Fisher JP and Adamson DC: Current FDA-Approved therapies for high-grade malignant gliomas. Biomedicines. 9:3242021. View Article : Google Scholar : PubMed/NCBI

24 

Hanna C, Kurian KM, Williams K, Watts C, Jackson A, Carruthers R, Strathdee K, Cruickshank G, Dunn L, Erridge S, et al: Pharmacokinetics, safety, and tolerability of olaparib and temozolomide for recurrent glioblastoma: Results of the phase I OPARATIC trial. Neuro Oncol. 22:1840–1850. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Löber-Handwerker R, Döring K, Bock C, Rohde V and Malinova V: Defining the impact of adjuvant treatment on the prognosis of patients with inoperable glioblastoma undergoing biopsy only: Does the survival benefit outweigh the treatment effort? Neurosurg Rev. 45:2339–2347. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Dumas AA, Pomella N, Rosser G, Guglielmi L, Vinel C, Millner TO, Rees J, Aley N, Sheer D, Wei J, et al: Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J. 39:e1037902020. View Article : Google Scholar : PubMed/NCBI

27 

Lara-Velazquez M, Al-Kharboosh R, Jeanneret S, Vazquez-Ramos C, Mahato D, Tavanaiepour D, Rahmathulla G and Quinones-Hinojosa A: Advances in brain tumor surgery for glioblastoma in adults. Brain Sci. 7:1662017. View Article : Google Scholar : PubMed/NCBI

28 

Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T, Bordbar D, Shan D, Samanamud J, Mahajan A, et al: Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med. 25:462–469. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Tan AC, Ashley DM, López GY, Malinzak M, Friedman HS and Khasraw M: Management of glioblastoma: State of the art and future directions. CA Cancer J Clin. 70:299–312. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Waldman AD, Fritz JM and Lenardo MJ: A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat Rev Immunol. 20:651–668. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P, Ho TS, et al: Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 348:124–128. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Vansteenkiste JF, Cho BC, Vanakesa T, De Pas T, Zielinski M, Kim MS, Jassem J, Yoshimura M, Dahabreh J, Nakayama H, et al: Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 17:822–835. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Hirabayashi K, Du H, Xu Y, Shou P, Zhou X, Fucá G, Landoni E, Sun C, Chen Y, Savoldo B and Dotti G: Dual-targeting CAR-T cells with optimal co-stimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors. Nat Cancer. 2:904–918. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Wang G, Zhou X, Fucà G, Dukhovlinova E, Shou P, Li H, Johnston C, Mcguinness B, Dotti G and Du H: Fully human antibody VH domains to generate mono and bispecific CAR to target solid tumors. J Immunother Cancer. 9:e0021732021. View Article : Google Scholar : PubMed/NCBI

35 

Adams GP and Weiner LM: Monoclonal antibody therapy of cancer. Nat Biotechnol. 23:1147–1157. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, et al: Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 33:2780–2788. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T and Minato N: Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 99:12293–12297. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G, et al: Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 65:1089–1096. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Kirkwood JM, Tarhini AA, Panelli MC, Moschos SJ, Zarour HM, Butterfield LH and Gogas HJ: Next Generation of Immunotherapy for Melanoma. Clin Oncol. 26:3445–3455. 2008.PubMed/NCBI

40 

Vitale I, Shema E, Loi S and Galluzzi L: Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med. 27:212–224. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al: PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 372:311–319. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Azad NS, Gray RJ, Overman MJ, Schoenfeld JD, Mitchell EP, Zwiebel JA, Sharon E, Streicher H, Li S, McShane LM, et al: Nivolumab is effective in mismatch repair-deficient noncolorectal cancers: Results From Arm Z1D-A Subprotocol of the NCI-MATCH (EAY131) Study. J Clin Oncol. 38:214–222. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al: PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 372:2509–2520. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, et al: PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 515:568–571. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Litak J, Mazurek M, Grochowski C, Kamieniak P and Roliński J: PD-L1/PD-1 axis in glioblastoma multiforme. Int J Mol Sci. 20:53472019. View Article : Google Scholar : PubMed/NCBI

46 

Scheffel TB, Grave N, Vargas P, Diz FM, Rockenbach L and Morrone FB: Immunosuppression in gliomas via PD-1/PD-L1 axis and adenosine pathway. Front Oncol. 10:6173852021. View Article : Google Scholar : PubMed/NCBI

47 

Caccese M, Indraccolo S, Zagonel V and Lombardi G: PD-1/PD-L1 immune-checkpoint inhibitors in glioblastoma: A concise review. Crit Rev Oncol Hematol. 135:128–134. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Cloughesy TF, Mochizuki AY, Orpilla JR, Hugo W, Lee AH, Davidson TB, Wang AC, Ellingson BM, Rytlewski JA, Sanders CM, et al: Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 25:477–486. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, López-Janeiro A, Porciuncula A, Idoate MA, Inogés S, de Andrea C, López-Diaz de Cerio A, Tejada S, et al: Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 25:470–476. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Yang T, Kong Z and Ma W: PD-1/PD-L1 immune checkpoint inhibitors in glioblastoma: Clinical studies, challenges and potential. Hum Vaccin Immunother. 17:546–553. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Lukas RV, Rodon J, Becker K, Wong ET, Shih K, Touat M, Fassò M, Osborne S, Molinero L, O'Hear C, et al: Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J Neurooncol. 140:317–328. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Kurz SC, Cabrera LP, Hastie D, Huang R, Unadkat P, Rinne M, Nayak L, Lee EQ, Reardon DA and Wen PY: PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology. 91:e1355–e1359. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Reardon DA, Omuro A, Brandes AA, Rieger J, Wick A, Sepulveda J, Phuphanich S, de Souza P, Ahluwalia MS, Lim M, et al: OS10.3 Randomized phase 3 study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent glioblastoma: CheckMate 143. Neuro Oncol. 19 (Suppl_3):iii212017. View Article : Google Scholar

54 

Horn L, Mansfield AS, Szczęsna A, Havel L, Krzakowski M, Hochmair MJ, Huemer F, Losonczy G, Johnson ML, Nishio M, et al: First-Line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med. 379:2220–2229. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KH, de Wit M, et al: Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 377:1919–1929. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Reardon DA, Kaley TJ, Dietrich J, Clarke JL, Dunn GP, Lim M, Cloughesy TM, Gan HK, Park AJ, Schwarzenberger P, et al: Phase 2 study to evaluate safety and efficacy of MEDI4736 (durvalumab [DUR]) in glioblastoma (GBM) patients: An update. J Clin Oncol. 35:2042017. View Article : Google Scholar

57 

Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M, Durno C, Krueger J, Cabric V, Ramaswamy V, et al: Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol. 34:2206–2211. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Logan CV, Murray JE, Parry DA, Robertson A, Bellelli R, Tarnauskaitė Ž, Challis R, Cleal L, Borel V, Fluteau A, et al: DNA polymerase epsilon deficiency causes IMAGe syndrome with variable immunodeficiency. Am J Hum Genet. 103:1038–1044. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Yarchoan M, Hopkins A and Jaffee EM: Tumor mutational burden and response Rate to PD-1 Inhibition. N Engl J Med. 377:2500–2501. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Guo X, Zhang Y, Jiao H and Miao X: The prognostic significance of PD-L1 expression in patients with glioblastoma: A meta-analysis. Front Oncol. 12:9255602022. View Article : Google Scholar : PubMed/NCBI

61 

Galon J and Bruni D: Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 18:197–218. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Raphael I, Kumar R, McCarl LH, Shoger K, Wang L, Sandlesh P, Sneiderman CT, Allen J, Zhai S, Campagna ML, et al: TIGIT and PD-1 immune checkpoint pathways are associated with patient outcome and anti-tumor immunity in glioblastoma. Front Immunol. 12:6371462021. View Article : Google Scholar : PubMed/NCBI

63 

Jan CI, Tsai WC, Harn HJ, Shyu WC, Liu MC, Lu HM, Chiu SC and Cho DY: Predictors of response to autologous dendritic cell therapy in glioblastoma multiforme. Front Immunol. 9:7272018. View Article : Google Scholar : PubMed/NCBI

64 

Fanelli GN, Grassini D, Ortenzi V, Pasqualetti F, Montemurro N, Perrini P, Naccarato AG and Scatena C: Decipher the glioblastoma microenvironment: The first milestone for new groundbreaking therapeutic strategies. Genes (Basel). 12:4452021. View Article : Google Scholar : PubMed/NCBI

65 

Ohgaki H and Kleihues P: Genetic pathways to primary and secondary glioblastoma. Am J Pathol. 170:1445–1453. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Qi X, Jha SK, Jha NK, Dewanjee S, Dey A, Deka R, Pritam P, Ramgopal K, Liu W and Hou K: Antioxidants in brain tumors: Current therapeutic significance and future prospects. Mol Cancer. 21:2042022. View Article : Google Scholar : PubMed/NCBI

67 

Peng W, Chen JQ, Liu C, Malu S, Creasy C, Tetzlaff MT, Xu C, McKenzie JA, Zhang C, Liang X, et al: Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 6:202–216. 2016. View Article : Google Scholar : PubMed/NCBI

68 

George S, Miao D, Demetri GD, Adeegbe D, Rodig SJ, Shukla S, Lipschitz M, Amin-Mansour A, Raut CP, Carter SL, et al: Loss of PTEN Is Associated with Resistance to Anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity. 46:197–204. 2017. View Article : Google Scholar : PubMed/NCBI

69 

de Bono JS, De Giorgi U, Rodrigues DN, Massard C, Bracarda S, Font A, Arranz Arija JA, Shih KC, Radavoi GD, Xu N, et al: Randomized phase II study evaluating akt blockade with ipatasertib, in combination with abiraterone, in patients with metastatic prostate cancer with and without PTEN Loss. Clin Cancer Res. 25:928–936. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Ascierto PA, Capone M, Grimaldi AM, Mallardo D, Simeone E, Madonna G, Roder H, Meyer K, Asmellash S, Oliveira C, et al: Proteomic test for anti-PD-1 checkpoint blockade treatment of metastatic melanoma with and without BRAF mutations. J Immunother Cancer. 7:912019. View Article : Google Scholar : PubMed/NCBI

71 

Sumimoto H, Imabayashi F, Iwata T and Kawakami Y: The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med. 203:1651–1656. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Caputo F, Santini C, Bardasi C, Cerma K, Casadei-Gardini A, Spallanzani A, Andrikou K, Cascinu S and Gelsomino F: BRAF-Mutated colorectal cancer: Clinical and molecular insights. Int J Mol Sci. 20:53692019. View Article : Google Scholar : PubMed/NCBI

73 

Dong C, Davis RJ and Flavell RA: MAP kinases in the immune response. Annu Rev Immunol. 20:55–72. 2002. View Article : Google Scholar : PubMed/NCBI

74 

Wang Y, Liu S, Yang Z, Algazi AP, Lomeli SH, Wang Y, Othus M, Hong A, Wang X, Randolph CE, et al: Anti-PD-1/L1 lead-in before MAPK inhibitor combination maximizes antitumor immunity and efficacy. Cancer Cell. 39:1375–1387.e6. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Wang J, Cazzato E, Ladewig E, Frattini V, Rosenbloom DI, Zairis S, Abate F, Liu Z, Elliott O, Shin YJ, et al: Clonal evolution of glioblastoma under therapy. Nat Genet. 48:768–776. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Collins PE, Kiely PA and Carmody RJ: Inhibition of Transcription by B Cell Leukemia 3 (Bcl-3) Protein Requires Interaction with Nuclear Factor κB (NF-κB) p50. J Biol Chem. 289:7059–7067. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Cahill KE, Morshed RA and Yamini B: Nuclear factor-κB in glioblastoma: Insights into regulators and targeted therapy. Neuro Oncol. 18:329–339. 2015. View Article : Google Scholar : PubMed/NCBI

78 

Hu YH, Jiao BH, Wang CY and Wu JL: Regulation of temozolomide resistance in glioma cells via the RIP2/NF-κB/MGMT pathway. CNS Neurosci Ther. 27:552–563. 2021. View Article : Google Scholar : PubMed/NCBI

79 

Fan S, Wu N, Chang S, Chen L and Sun X: The immune regulation of BCL3 in glioblastoma with mutated IDH1. Aging (Albany NY). 14:3856–3873. 2022. View Article : Google Scholar : PubMed/NCBI

80 

Zou Y, Uddin M, Padmanabhan S, Zhu Y, Bu P, Vancura A and Vancurova I: The proto-oncogene Bcl3 induces immune checkpoint PD-L1 expression, mediating proliferation of ovarian cancer cells. J Biol Chem. 293:18483–15496. 2018. View Article : Google Scholar

81 

Tomaszewski W, Sanchez-Perez L, Gajewski TF and Sampson JH: Brain tumor microenvironment and host state: Implications for immunotherapy. Clin Cancer Res. 25:4202–4210. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Pombo Antunes AR, Scheyltjens I, Duerinck J, Neyns B, Movahedi K and Van Ginderachter JA: Understanding the glioblastoma immune microenvironment as basis for the development of new immunotherapeutic strategies. ELife. 9:e521762020. View Article : Google Scholar : PubMed/NCBI

83 

Hansen LJ, Yang R, Roso K, Wang W, Chen L, Yang Q, Pirozzi CJ and He Y: MTAP loss correlates with an immunosuppressive profile in GBM and its substrate MTA stimulates alternative macrophage polarization. Sci Rep. 12:41832022. View Article : Google Scholar : PubMed/NCBI

84 

Zhang H and Chen Y: Identification of glioblastoma immune subtypes and immune landscape based on a large cohort. Hereditas. 158:302021. View Article : Google Scholar : PubMed/NCBI

85 

Bird L: MDSC metabolite stuns T cells. Nat Rev Immunol. 20:352–353. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Ito H, Nakashima H and Chiocca EA: Molecular responses to immune checkpoint blockade in glioblastoma. Nat Med. 25:359–361. 2019. View Article : Google Scholar : PubMed/NCBI

87 

Hung AL, Maxwell R, Theodros D, Belcaid Z, Mathios D, Luksik AS, Kim E, Wu A, Xia Y, Garzon-Muvdi T, et al: TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM. Oncoimmunology. 7:e14667692018. View Article : Google Scholar : PubMed/NCBI

88 

Czapiewski P, Cornelius M, Hartig R, Kalinski T, Haybaeck J, Dittmer A, Dittmer J, Ignatov A and Nass N: BCL3 expression is strongly associated with the occurrence of breast cancer relapse under tamoxifen treatment in a retrospective cohort study. Virchows Arch. 480:529–541. 2022. View Article : Google Scholar : PubMed/NCBI

89 

Brocke-Heidrich K, Ge B, Cvijic H, Pfeifer G, Löffler D, Henze C, McKeithan TW and Horn F: BCL3 is induced by IL-6 via Stat3 binding to intronic enhancer HS4 and represses its own transcription. Oncogene. 25:7297–7304. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 30 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu J and Wang N: Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review). Mol Med Rep 30: 221, 2024.
APA
Wu, J., & Wang, N. (2024). Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review). Molecular Medicine Reports, 30, 221. https://doi.org/10.3892/mmr.2024.13344
MLA
Wu, J., Wang, N."Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review)". Molecular Medicine Reports 30.6 (2024): 221.
Chicago
Wu, J., Wang, N."Current progress of anti‑PD‑1/PDL1 immunotherapy for glioblastoma (Review)". Molecular Medicine Reports 30, no. 6 (2024): 221. https://doi.org/10.3892/mmr.2024.13344