Skip to main content
  • Cross talk between CD133+ glioma cells and macrophages/microglia increases B7-H4 expression that limits therapeutic success of dendritic cell based immunotherapy in patients with malignant glioma

    Final Number:
    1157

    Authors:
    Yu Yao; Hongxing Ye; Lianjie Mo; Qi Yue; Lingchao Chen MD; Aparajita Baral; Juan Carlos Vera; Xingxing Zang; John D. Heiss; Clark C. Chen; Kunlin Jin; Ying Wang BS; Ying Mao MD; Liangfu Zhou

    Study Design:
    Laboratory Investigation

    Subject Category:

    Meeting: Congress of Neurological Surgeons 2013 Annual Meeting

    Introduction: Cancer-induced immunosuppression, both local and systemic forms, may play an important role in tumor progression. Currently, this pathology poses a significant challenge to the viability of immunotherapy as a treatment for brain malignancy. Investigations into immune regulatory molecules, in particular B7-H4 (B7X, B7S1), have revealed some extent of their immunomodulatory capacity, especially in inciting immunosuppression, and have linked increasing levels of B7-H4 with poor clinical prognosis in various cancers.

    Methods: Mfs /microglia was isolatied from peripheral blood and tissues by microbeads. Cell migration was done by using Transwell assay. Flow cytometry was done for detecting surface B7-H4, CD11b or CD133 expression. Subcutaneous glioma model was done using U87 cells injected into the right flank of NOD/SCID mice.

    Results: We first illustrate an association of B7-H4 with the notorious biological features of intracranial gliomas. Next, we show that CD133+ glioma cells induce normal human macrophages (Mfs) to express B7-H4 via IL-6 and IL-10 in tumor environment. Interestingly, we observed that glioma infiltrating macrophages/microglia (GIMs) also stimulate CD133+ cells to induce B7-H4 expression and auto-regulation via IL-6. Given the historical emphasis on a pivotal role of human CD133+ glioma stem-like cells (GSCs) in tumor progression and recurrence, along with the designation of Mfs/microglia as the principal tumor-in?ltrating immune cells in glioblastoma, we sought to determine if B7-H4 localization to GIMs is perhaps a required featured for sustaining the tumor microenvironment. We go on to elaborate on the effects of B7-H4 expression, depicting that CD133+ cells contribute to immunopathology through suppressive B7-H4+ Mfs in vitro and in vivo.

    Conclusions: Our data supports the utility of B7-H4 as a novel biomarker of glioma and presents previously unrecognized evidence that increased B7-H4 expression due to suppressive cross talk between CD133+ cells and GIMs hinders dendritic cells (DCs)-based immunotherapy.

    Patient Care: We demonstrated that B7-H4 may be clinically useful as a biomarker for human gliomas because its tumor concentration given its clinical implications correlated negatively with prognostic function and positively with malignancy grading. Moreover, we demonstrate that the increase of B7-H4 in GIMs, mediated by GSCs and GIMs themselves, constitutes a previously unrecognized and relevant target for improving therapeutic outcomes of DCs-based active immunotherapy in patients with malignant gliomas.

    Learning Objectives: Investigate te role of immune regulatory molecules B7-H4 between CD133+ glioma cells and macrophages/microglia.

    References:

We use cookies to improve the performance of our site, to analyze the traffic to our site, and to personalize your experience of the site. You can control cookies through your browser settings. Please find more information on the cookies used on our site. Privacy Policy