Product Citations: 307

2 images found

Modulating the Immunosuppressive Tumor Microenvironment and Inhibiting Growth in Mutp53-Driven CRPC via STAT3 Pathway Blockade.

In International Journal of Biological Sciences on 19 May 2025 by Bian, Z., Chen, J., et al.

Mutations in TP53, particularly the p.R248Q variant, contribute to the progression of castration-resistant prostate cancer (CRPC) by reshaping the tumor microenvironment (TME). This study examined the impact of p.R248Q (mutp53) on immune suppression and CRPC progression. We introduced the Trp53 p.R245Q mutation into RM-1 mouse prostate cancer (PCa) cells via CRISPR/Cas9, which mimics human TP53 p.R248Q. These cells were implanted into C57BL/6 mice to model tumor progression and immune interactions. Mice were treated with JAK2 and STAT3 inhibitors to assess immune and tumor responses. Tumor behavior and immune responses were analyzed via histology, immunofluorescence, flow cytometry, Enzyme-linked immunosorbent assay (ELISA), and bioinformatics. Findings were validated in the C4-2 human PCa cell line. Compared with wild-type p53, TP53 mutations were present in 27% of PCa patients and were significantly correlated with reduced overall survival (p < 0.001, HR = 1.97) and recurrence-free survival (p = 0.02, HR = 1.62). The p.R248Q mutation was most prevalent. Gene-edited mutp53 cells exhibited increased proliferation and tumorigenicity. Screening and validation confirmed that IL6/JAK2/STAT3 pathway activation in mutp53 tumors led to immune microenvironment alterations. Flow cytometry and immunofluorescence revealed an immunosuppressive profile, with decreased proinflammatory cytokines and elevated anti-inflammatory factors. Coimmunoprecipitation revealed that mutp53 competes with SHP1 for STAT3 binding, sustaining its activation. Inhibition of STAT3 reduced mutp53-driven immune suppression and tumor progression. Mutp53 promotes an immunosuppressive TME and facilitates CRPC progression through the STAT3 pathway, underscoring its potential as a therapeutic target.
© The author(s).

  • Cancer Research

PD-1 blockade employed at the time CD8+ T cells are activated enhances their antitumor efficacy.

In Journal for Immunotherapy of Cancer on 7 May 2025 by Moseman, J. E., Rastogi, I., et al.

We have previously shown that immune checkpoint receptors, including PD-1, are upregulated on T cells at the time of their activation, and that blockade of these receptors can improve the efficacy of antitumor vaccines. In the present study, we sought to determine whether, and by what mechanisms, the timing of PD-1 blockade with respect to vaccination affects antitumor T cell function.
TRAMP-C1 or E.G7-OVA tumor-bearing mice received PD-1 blockade at different timing intervals with a tumor-associated antigen vaccine. Tumor growth, survival, and immune-infiltrating populations were assessed. In vitro models of T cell activation using OT-I T cells and PD-(L)1 axis disruption with a PD-1 blocking antibody or PD-L1KO dendritic cells were used.
Mice receiving PD-1 blockade at the time of T cell activation with vaccine had better antitumor outcomes in comparison to mice receiving PD-1 blockade before or after immunization. T cells activated in vitro in the presence of PD-(L)1 axis disruption had a more differentiated, functional phenotype with decreased CD28 and CCR7 expression and increased production of the Tc1 cytokines IL-2, TNFα, and IFNγ. Intriguingly, a small subset of undifferentiated cells (CD28+) was of a stem-like Tc17 phenotype (IL-17α+, TCF1+). Tumor-bearing mice receiving T cells activated in the presence of PD-(L)1-axis disruption had better antitumor outcomes and a greater number of complete responses.
These data indicate that PD-1 blockade, when used with antitumor vaccines, acts primarily at the time of T cell activation, not exclusively within the tumor microenvironment. Consequently, PD-1 blockade may be best used when delivered concurrently with T cell activating agents such as vaccines.
© Author(s) (or their employer(s)) 2025. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ Group.

  • Immunology and Microbiology

Asparagine drives immune evasion in bladder cancer via RIG-I stability and type I IFN signaling.

In The Journal of Clinical Investigation on 15 April 2025 by Wei, W., Li, H., et al.

Tumor cells often employ many ways to restrain type I IFN signaling to evade immune surveillance. However, whether cellular amino acid metabolism regulates this process remains unclear, and its effects on antitumor immunity are relatively unexplored. Here, we found that asparagine inhibited IFN-I signaling and promoted immune escape in bladder cancer. Depletion of asparagine synthetase (ASNS) strongly limited in vivo tumor growth in a CD8+ T cell-dependent manner and boosted immunotherapy efficacy. Moreover, clinically approved L-asparaginase (ASNase),synergized with anti-PD-1 therapy in suppressing tumor growth. Mechanistically, asparagine can directly bind to RIG-I and facilitate CBL-mediated RIG-I degradation, thereby suppressing IFN signaling and antitumor immune responses. Clinically, tumors with higher ASNS expression show decreased responsiveness to immune checkpoint inhibitor therapy. Together, our findings uncover asparagine as a natural metabolite to modulate RIG-I-mediated IFN-I signaling, providing the basis for developing the combinatorial use of ASNase and anti-PD-1 for bladder cancer.

  • Cancer Research
  • Immunology and Microbiology

The epigenetic modification of histone H3 lysine 27 trimethylation (H3K27me3) by the embryonic ectoderm development (EED) protein is closely associated with the regulation of transcriptional programs and is implicated in autoimmune diseases. However, the efficacy of targeting H3K27me3 for the treatment of neuroinflammation remains unclear. In this study, we demonstrate that systemic administration of an EED inhibitor diminishes the inflammatory response mediated by dendritic cells (DCs), thereby alleviating experimental autoimmune encephalitis (EAE), a representative mouse model of autoimmune diseases in the central nervous system (CNS). Our findings indicate that EED inhibitors suppress DC migration by upregulating genes in the WNT signaling pathway that are epigenetically marked by H3K27me3. Conversely, inhibiting the WNT pathway partially reverses the impaired DC migration caused by EED inhibitors. Additionally, the genetic deletion of Eed inhibits DC migration and effectively mitigates autoimmune symptoms and inflammatory infiltration into the CNS in EAE. These results highlight EED as a critical regulator of DC migration and suggest its potential as a therapeutic target for autoimmune disorders.
© 2025. The Author(s).

  • FC/FACS
  • Mus musculus (House mouse)
  • Genetics
  • Immunology and Microbiology

Dendritic cell (DC)-derived extracellular vesicles (DEVs) are promising candidates for cancer vaccines, but their therapeutic effects still need further optimization. In this study, we utilized neoantigens, lipopolysaccharide and IFN-γ to induce the maturation of DCs, and then isolated DEVs derived from these mature DCs. We showed that the immune checkpoint inhibitor (anti-CTLA-4 antibody, aCTLA-4) can improve the immunostimulatory function of DEVs by directly activating T cells through immune checkpoint signal blockade. The cytokine interleukin-12 (IL-12), as one of the third signals for T cell activation, can also enhance the capability of DEVs to activate T cells directly. Based on these findings, we designed the engineered DEVs conjugated with IL-12 and aCTLA-4 (DEV@IL-12-aCTLA-4) to improve the therapeutic potential of DEVs by providing sufficient immune regulatory signals. Moreover, the carrier property of DEVs also contributes to the delivery of IL-12 and aCTLA-4 to lymph nodes. This indicates that the conjugation of DEVs with IL-12 and aCTLA-4 constitutes a complementary approach, where IL-12 and aCTLA-4 help to enhance the T cell activation effect of DEVs, and DEVs facilitate the delivery of IL-12 and aCTLA-4. Our results showed that DEV@IL-12-aCTLA-4 can enhance the Th1 immune response and reverse exhausted CD8+ T cells in the tumour microenvironment, effectively inducing robust T cell immune responses and inhibiting tumour growth in tumour-bearing mice. Overall, this study expands the theoretical foundation of DEVs and provides a universal strategy for optimizing cancer combination immunotherapy by reprogramming DEVs.
© 2025 The Author(s). Journal of Extracellular Vesicles published by Wiley Periodicals, LLC on behalf of the International Society for Extracellular Vesicles.

  • Cancer Research
  • Immunology and Microbiology
View this product on CiteAb