Abstract

Intratumoral (IT) injections of Glucopyranosyl lipid A (G100), a synthetic toll-like receptor 4 (TLR4) agonist formulated in a stable emulsion, resulted in T-cell inflammation of the tumor microenvironment (TME) and complete cure of 60% of mice with large established A20 lymphomas. Strong abscopal effects on un-injected lesions were observed in a bilateral tumor model and surviving mice resisted a secondary tumor challenge. Depletion of CD8 T-cells, but not CD4 or NK cells, abrogated the anti-tumor effect. Unexpectedly, TLR4 knock-out rendered A20 tumors completely non-responsive to G100. In vitro studies showed that GLA has direct effect on A20 cells, but not on A20 cells deficient for TLR4. As shown by genotyping and phenotyping analysis, G100 strongly activated antigen presentation functions in A20 cells in vitro and in vivo and induced their apoptosis in a dose dependent manner. Similarly, the TLR4 positive human mantle cell lymphoma line Mino showed in vitro activation with G100 that was blocked with an anti-TLR4 antibody. In the A20 model, direct activation of B-lymphoma cells with G100 is sufficient to induce protective CD8 T-cell responses and TLR4 expressing human B-cell lymphomas may be amenable to this therapy as well.

Highlights

  • A minority of cancer patients currently benefits from immunotherapeutic interventions aimed at rescuing functional T-cells responses through modulation of immune check points, and this resistance is at least partly be due to the immunosuppressive nature of the tumor microenvironment (TME) [1]

  • Using the murine A20 B-cell lymphoma, we investigated the effect of IT G100 on the TME and its dependency on Toll-like receptor 4 (TLR4) expression of the tumor cells

  • Since A20 is derived from murine B cells which are known to express TLR4, we investigated the importance of TLR4 expression on tumor cells by generating a biallelic TLR4 knockout A20 cell line

Read more

Summary

Introduction

A minority of cancer patients currently benefits from immunotherapeutic interventions aimed at rescuing functional T-cells responses through modulation of immune check points, and this resistance is at least partly be due to the immunosuppressive nature of the tumor microenvironment (TME) [1]. Therapies that are capable of inducing proinflammatory changes in the TME, activating antigen presenting cells and inducing immune responses should have significant therapeutic potential on their own and in combination with other immunotherapies [4, 5] To this end, several agents designed to modify the TME are currently under clinical investigation, including cytokines, oncolytic viruses, toll-like receptor (TLR) 3, 7/8 and 9 agonists, RIG-I agonists and others [6, 7]. Several agents designed to modify the TME are currently under clinical investigation, including cytokines, oncolytic viruses, toll-like receptor (TLR) 3, 7/8 and 9 agonists, RIG-I agonists and others [6, 7] While some of these treatment modalities have shown impressive levels of efficacy in murine. A recent clinical trial showed that intratumoral injection of GLA-SE (termed G100) inflamed the TME of Merkel cell carcinoma patients, with antitumor immune responses and objective tumor responses observed [17]

Methods
Results
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