Skip to main content
eScholarship
Open Access Publications from the University of California

UCLA

UCLA Electronic Theses and Dissertations bannerUCLA

The Effects of Neoadjuvant anti-Programmed Cell Death Protein 1 (PD-1) Therapy on the Tumor Infiltrating T cell compartment and Tumor Microenvironment Immune Composition in Recurrent Glioblastoma Patients

No data is associated with this publication.
Abstract

Glioblastoma (GBM) is the most common malignant tumor in the central nervous system and has poor patient survival rates. Unlike other cancers, immune checkpoint therapies, such as PD-1 checkpoint blockade, have been largely ineffective in GBMs for several reasons: an immunosuppressive tumor microenvironment, a lack of suitable neoantigens, and poor intratumoral T cell infiltration and activity. However, there is evidence that using anti-PD-1 therapy in the neoadjuvant setting may generate a more robust anti-tumor immune response, though characterizing how the GBM tumor microenvironment changes with such therapy is incomplete. As such, we performed high dimensional analysis using CyTOF mass cytometry and single-cell RNAsequencing to study the intratumoral immune populations in GBM patients treated with or without neoadjuvant anti-PD-1 therapy. Characterizing PD-1 expressing tumor infiltrating T cell populations showed that PD-1 was associated with markers of T cell activation and dysfunction regardless of treatment and that this association existed less strongly in peripheral PD-1 expressing T cells. We studied the effects of neoadjuvant anti-PD-1 therapy on a large patient cohort of tumor infiltrating immune cells and found that neoadjuvant anti-PD-1 therapy significantly increased the proportion of several intratumoral T cell sub-populations, including a TCF7-expressing progenitor exhausted population. Downstream effects of neoadjuvant anti-PD-1 therapy on T cells, namely increased production of IFN-g, included transcriptionally altering the myeloid and dendritic cell populations to be more immune suppressive but also potentially more vulnerable to other immune checkpoint therapies, specifically anti-TIGIT and anti-CTLA-4 therapies. Due to the impact of neoadjuvant anti-PD-1 therapy on intratumoral T cell populations, we examined whether we could detect tumor-reactive T cells by cloning TCRs from transcriptionally defined populations in a patient treated with neoadjuvant anti-PD-1 and testing reactivity to the patient-derived gliomasphereline. Among TCRs cloned, we discovered that T cells arising from the activated and exhausted population showed tumor reactivity, suggesting that utilizing transcriptional phenotypes can guide selection of potential tumor-reactive TCRs in GBM patients treated with neoadjuvant anti-PD-1 therapy. In conclusion, neoadjuvant anti-PD-1 therapies alters the immune landscape in these tumors and can be potentially used in combination with other immunotherapies to more effectively treat this malignancy.

Main Content

This item is under embargo until March 27, 2025.