Modeling Oncolytic Viral Therapy, Immune Checkpoint Inhibition, and the Complex Dynamics of Innate and Adaptive Immunity in Glioblastoma Treatment

29Citations
Citations of this article
29Readers
Mendeley users who have this article in their library.

Abstract

Oncolytic viruses are of growing interest to cancer researchers and clinicians, due to their selectivity for tumor cells over healthy cells and their immunostimulatory properties. The immune response to an oncolytic virus plays a critical role in treatment efficacy. However, uncertainty remains regarding the circumstances under which the immune system either assists in eliminating tumor cells or inhibits treatment via rapid viral clearance, leading to the cessation of the immune response. In this work, we develop an ordinary differential equation model of treatment for a lethal brain tumor, glioblastoma, using an oncolytic Herpes Simplex Virus. We use a mechanistic approach to model the interactions between distinct populations of immune cells, incorporating both innate and adaptive immune responses to oncolytic viral therapy (OVT), and including a mechanism of adaptive immune suppression via the PD-1/PD-L1 checkpoint pathway. We focus on the tradeoff between viral clearance by innate immune cells and the innate immune cell-mediated recruitment of antiviral and antitumor adaptive immune cells. Our model suggests that when a tumor is treated with OVT alone, the innate immune cells' ability to clear the virus quickly after administration has a much larger impact on the treatment outcome than the adaptive immune cells' antitumor activity. Even in a highly antigenic tumor with a strong innate immune response, the faster recruitment of antitumor adaptive immune cells is not sufficient to offset the rapid viral clearance. This motivates our subsequent incorporation of an immunotherapy that inhibits the PD-1/PD-L1 checkpoint pathway by blocking PD-1, which we combine with OVT within the model. The combination therapy is most effective for a highly antigenic tumor or for intermediate levels of innate immune localization. Extreme levels of innate immune cell activity either clear the virus too quickly or fail to activate a sufficiently strong adaptive response, yielding ineffective combination therapy of GBM. Hence, we show that the innate and adaptive immune interactions significantly influence treatment response and that combining OVT with an immune checkpoint inhibitor expands the range of immune conditions that allow for tumor size reduction or clearance.

Cite

CITATION STYLE

APA

Storey, K. M., Lawler, S. E., & Jackson, T. L. (2020). Modeling Oncolytic Viral Therapy, Immune Checkpoint Inhibition, and the Complex Dynamics of Innate and Adaptive Immunity in Glioblastoma Treatment. Frontiers in Physiology, 11. https://doi.org/10.3389/fphys.2020.00151

Register to see more suggestions

Mendeley helps you to discover research relevant for your work.

Already have an account?

Save time finding and organizing research with Mendeley

Sign up for free