Crosslink between Temozolomide and PD-L1 immune-checkpoint inhibition in glioblastoma multiforme.

Department of Neurosurgery, Medical Center University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany. Faculty of Medicine, University of Freiburg, Freiburg, Germany. Department of Neuroradiology, Medical Center - University of Freiburg, Freiburg, Germany. Clinic for Neuropediatrics and Neurorehabilitation, Epilepsy Center for Children and Adolescents, Schön Klinik, Vogtareuth, Germany. Department of Neurosurgery, University of Aachen, Aachen, Germany. Department of Neurosurgery, Medical Center University of Freiburg, Breisacher Straße 64, 79106, Freiburg, Germany. dieter.henrik.heiland@uniklinik-freiburg.de. Faculty of Medicine, University of Freiburg, Freiburg, Germany. dieter.henrik.heiland@uniklinik-freiburg.de.

BMC cancer. 2019;(1):117
Full text from:

Abstract

BACKGROUND In recent years, PD-1/PD-L1 immune checkpoint inhibitors have improved cancer therapy in many tumor types, but no benefit of immune checkpoint therapy has been found in glioblastoma multiforme (GBM). Based on the results of our earlier work, which showed a reduction of PD-L1 expression in patients treated with temozolomide (TMZ), we aimed to investigate the link between TMZ therapy and the immune control point target PD-L1. METHODS RNA-sequencing data from de-novo and recurrent glioblastoma were analyzed by AutoPipe algorithm. Results were confirmed either in a cell model by two primary and one established GBM cell line and specimens of de-novo and recurrent GBM. PD-L1 and pathway activation of the JAK/STAT pathway was analyzed by quantitative real-time PCR and western blot. RESULTS We found a significant downregulation of the JAK/STAT pathway and immune response in recurrent tumors. The cell model showed an upregulation of PD-L1 after IFNγ treatment, while additional TMZ treatment lead to a reduction of PD-L1 expression and JAK/STAT pathway activation. These findings were confirmed in specimens of de-novo and recurrent glioblastoma. CONCLUSIONS Our results suggest that TMZ therapy leads to a down-regulation of PD-L1 in primary GBM cells. These results support the clinical findings where PD-L1 is significantly reduced in recurrent GBMs. If the target is diminished, it may also lead to impaired efficacy of PD-1/PD-L1 inhibitors such as nivolumab.