Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma

RV Lukas, J Rodon, K Becker, ET Wong, K Shih… - Journal of neuro …, 2018 - Springer
RV Lukas, J Rodon, K Becker, ET Wong, K Shih, M Touat, M Fassò, S Osborne, L Molinero
Journal of neuro-oncology, 2018Springer
Purpose Glioblastoma is the most common primary malignant brain tumor. No standard
treatment exists for recurrent disease. Glioblastoma is associated with an
immunosuppressive tumor microenvironment. Immune checkpoint inhibitors, including
atezolizumab (anti-programmed death-ligand 1), have demonstrated clinical activity in
various cancers. Here, we present the safety and efficacy of atezolizumab in patients with
glioblastoma from the phase 1a PCD4989g clinical trial (NCT01375842). Methods Eligible …
Purpose
Glioblastoma is the most common primary malignant brain tumor. No standard treatment exists for recurrent disease. Glioblastoma is associated with an immunosuppressive tumor microenvironment. Immune checkpoint inhibitors, including atezolizumab (anti-programmed death-ligand 1), have demonstrated clinical activity in various cancers. Here, we present the safety and efficacy of atezolizumab in patients with glioblastoma from the phase 1a PCD4989g clinical trial (NCT01375842).
Methods
Eligible patients had confirmed recurrent glioblastoma and measurable disease per RANO criteria. Atezolizumab (1200 mg) was administered intravenously every 3 weeks until progression or unacceptable toxicity. Patients were monitored for safety; response was evaluated at least every 6 weeks. Baseline biomarkers were evaluated.
Results
All 16 patients enrolled had received prior chemotherapy, and 50% prior bevacizumab. Ten patients (63%) experienced a treatment-related event. No treatment-related grade 4–5 events were reported. All deaths occurred due to progression or during follow-up. One patient experienced a partial response (5.3 months); 3 experienced disease stabilization. The median overall survival was 4.2 months (range 1.2 to 18.8+ months). Association between peripheral CD4+ T cells and efficacy was observed. Two patients with IDH1-mutant tumors and 1 with a POLE-mutant tumor experienced ≥ 16 months survival.
Conclusions
Atezolizumab was safe and well tolerated in this group of patients with recurrent glioblastoma. Our preliminary findings suggest that biomarkers, including peripheral CD4+ T cells and hypermutated tumor status, may help guide selection of patients with recurrent glioblastoma who might receive most benefit from atezolizumab therapy, supporting further atezolizumab combination studies in glioblastoma.
Springer