Abstract

Glioblastoma (GB) is the most common high-grade intracranial malignant tumor with highly malignant biological behavior and a high recurrence rate. Although anti-PD-1/PD-L1 antibodies have achieved significant survival benefits in several kinds of solid tumors, the phase III clinical trial Checkmate 143 demonstrated that nivolumab, which targets PD-1, did not achieve survival benefits compared with bevacizumab in recurrent glioblastoma (rGB) patients. Nevertheless, neoadjuvant anti-PD-1 therapy followed by surgery and adjuvant anti-PD-1 therapy could effectively activate local and systemic immune responses and significantly improve the OS of rGB patients. Furthermore, several studies have also confirmed the progress made in applying tumor-specific peptide vaccination or chimeric antigen receptor-T (CAR-T) cell therapy to treat rGB patients, and successes with antibodies targeting other inhibitory checkpoints or costimulatory molecules have also been reported. These successes inspired us to explore candidate combination treatments based on anti-PD-1/PD-L1 antibodies. However, effective predictive biomarkers for clinical efficacy are urgently needed to avoid economic waste and treatment delay. Attempts to prolong the CAR-T cell lifespan and increase T cell infiltration through engineering techniques are addressing the challenge of strengthening T cell function. In this review, we describe the immunosuppressive molecular characteristics of rGB; clinical trials exploring anti-PD-1/PD-L1 therapy, tumor-specific peptide vaccination, and CAR-T cell therapy; candidate combination strategies; and issues related to strengthening T cell function.

Highlights

  • Glioblastoma (GB) has an incidence of 0.59–3.69/100,000 people worldwide, with a median onset age of 63.0 years

  • From current phase I/II clinical trials, we conclude that chimeric antigen receptor-T (CAR-T) cell therapy has achieved only limited clinical efficacy in recurrent glioblastoma (rGB) patients and that the future of CAR-T cell therapy will depend on the effective recognition of tumor-specific antigens with sufficient and stable expression

  • GB is a tumor with highly malignant biological behavior, and more than 90% of GB patients will experience recurrence and progression

Read more

Summary

Introduction

100,000 people worldwide, with a median onset age of 63.0 years. The age-adjusted morbidity is 3.97/100,000 for males and 2.53/100,000 for females[1,2,3]. When the tumor burden is reduced by surgery and combined neoadjuvant therapy and adjuvant PD-1 checkpoint blockade are administered, the “sandwich treatment strategy” can effectively activate local and systemic immune responses and significantly improve the OS of rGB patients. Schalper and colleagues confirmed that the underlying mechanism by which neoadjuvant anti-PD-1 therapy plays a critical role may involve increased chemokine transcript expression, enhanced infiltration, and T cell receptor (TCR) clonal diversity in the effector T lymphocyte population They demonstrated that nivolumab did not change the immune cell distribution in the GB microenvironment, based on an immunofluorescence assay comparing before and after therapy. Bloch et al investigated the clinical efficacy and safety of heat-shock protein peptide complex-96 (HSPPC-96) vaccination in rGB patients following surgical resection and reported that the median OS time was 42.6 weeks (Table 2). A phase II clinical study was conducted to investigate

Phase Results
20 ID 74 IV 10 IV 41 ID 20 ID 17 ID 88 IH
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call