Abstract

Glioblastoma, one of the most aggressive primary brain tumors, is characterized by highly immunosuppressive microenvironment. This contributes to glioblastoma resistance to standard treatment modalities and allows tumor growth and recurrence. Several immune-targeted approaches have been recently developed and are currently under preclinical and clinical investigation. Oncolytic viruses, including the autonomous protoparvovirus H-1 (H-1PV), show great promise as novel immunotherapeutic tools. In a first phase I/IIa clinical trial (ParvOryx01), H-1PV was safe and well tolerated when locally or systemically administered to recurrent glioblastoma patients. The virus was able to cross the blood–brain (tumor) barrier after intravenous infusion. Importantly, H-1PV treatment of glioblastoma patients was associated with immunogenic changes in the tumor microenvironment. Tumor infiltration with activated cytotoxic T cells, induction of cathepsin B and inducible nitric oxide (NO) synthase (iNOS) expression in tumor-associated microglia/macrophages (TAM), and accumulation of activated TAM in cluster of differentiation (CD) 40 ligand (CD40L)-positive glioblastoma regions was detected. These are the first-in-human observations of H-1PV capacity to switch the immunosuppressed tumor microenvironment towards immunogenicity. Based on this pilot study, we present a tentative model of H-1PV-mediated modulation of glioblastoma microenvironment and propose a combinatorial therapeutic approach taking advantage of H-1PV-induced microglia/macrophage activation for further (pre)clinical testing.

Highlights

  • GlioblastomaGlioblastoma is the most common and aggressive primary brain tumor

  • The ability of H-1 protoparvovirus (H-1PV)-infected tumor cells to activate antitumor immune responses has been demonstrated in several preclinical cancer models, in particular hepatoma [34], melanoma [35], pancreatic [36], colorectal [37,38] and nasopharyngeal [39] carcinomas

  • In line with the above, CD 40 ligand (CD40L) expression was detected in non-macrophage, EGFR-positive, i.e., most likely glioblastoma cells

Read more

Summary

Glioblastoma

Glioblastoma is the most common and aggressive primary brain tumor It has a dismal prognosis and is typically characterized by largely inevitable recurrence within six months to one year after initial treatment [1,2]. Standard cytotoxic agents, corticosteroids, TMZ rechallenge, carboplatin and irinotecan are applied, among other agents, to palliate symptoms and improve quality of life, but fail to prolong the time to progression [6]. In 2009, the humanized monoclonal antibody bevacizumab targeting tumor angiogenesis (through the vascular endothelial growth factor (VEGF)), was approved for the treatment of recurrent glioblastoma patients on the basis of its ability to achieve superior progression-free survival (PFS), yet in the lamentable absence of meaningful overall survival (OS) improvement [7]

Immune Suppression in Glioblastoma Patients
Immune-Targeted Therapeutic Approaches for Glioblastoma
Tumor Vaccination
Immune Checkpoint Inhibition
Adoptive T Cell Transfer
Need for Novel Immunotherapeutic Strategies
Oncolytic H-1 Parvovirus
In Tumor Models Other than Glioma
In Glioma Models
Immunotherapeutic Potential of H-1PV in Glioblastoma Patients
Glioblastoma Infiltration with Immune Cells
Activation Status of Glioblastoma-Infiltrating Immune Cells
CD40L Expression by Glioblastoma Cells
Proinflammatory Cytokine Production in Glioblastoma Microenvironment
Induction of Cathepsin B Expression
Other Oncolytic Viruses and Tumor Microenvironment Modulation
Preclinical Studies
Clinical Studies
Tumor Microenvironment Modulation by H-1PV
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