Abstract

Biomaterial-based scaffolds are promising tools for controlled immunomodulation. They can be applied as three dimensional (3D) culture systems in vitro, whereas in vivo they may be used to dictate cellular localization and exert spatiotemporal control over cues presented to the immune system. As such, scaffolds can be exploited to enhance the efficacy of cancer immunotherapies such as adoptive T cell transfer, in which localization and persistence of tumor-specific T cells dictates treatment outcome. Biomimetic polyisocyanopeptide (PIC) hydrogels are polymeric scaffolds with beneficial characteristics as they display reversible thermally-induced gelation at temperatures above 16°C, which allows for their minimally invasive delivery via injection. Moreover, incorporation of azide-terminated monomers introduces functional handles that can be exploited to include immune cell-modulating cues. Here, we explore the potential of synthetic PIC hydrogels to promote the in vitro expansion and in vivo local delivery of pre-activated T cells. We found that PIC hydrogels support the survival and vigorous expansion of pre-stimulated T cells in vitro even at high cell densities, highlighting their potential as 3D culture systems for efficient expansion of T cells for their adoptive transfer. In particular, the reversible thermo-sensitive behavior of the PIC scaffolds favors straightforward recovery of cells. PIC hydrogels that were injected subcutaneously gelated instantly in vivo, after which a confined 3D structure was formed that remained localized for at least 4 weeks. Importantly, we noticed no signs of inflammation, indicating that PIC hydrogels are non-immunogenic. Cells co-delivered with PIC polymers were encapsulated within the scaffold in vivo. Cells egressed gradually from the PIC gel and migrated into distant organs. This confirms that PIC hydrogels can be used to locally deliver cells within a supportive environment. These results demonstrate that PIC hydrogels are highly promising for both the in vitro expansion and in vivo delivery of pre-activated T cells. Covalent attachment of biomolecules onto azide-functionalized PIC polymers provides the opportunity to steer the phenotype, survival or functional response of the adoptively transferred cells. As such, PIC hydrogels can be used as valuable tools to improve current adoptive T cell therapy strategies.

Highlights

  • Scaffolds produced from a variety of biomaterials are widely applied as engineered microenvironments or delivery vehicles in biomedical applications

  • The use of biomaterial-based scaffolds as 3D culture systems and cellular delivery vehicles is a promising approach to improve the efficacy of immunotherapy for cancer and reduce toxicity

  • We characterize the potential of PIC hydrogels as 3D culture systems for imaging of mice s.c. injected with 100 μl Cy5-labeled 1.5 mg/mL PIC polymers mixed with T cells in the dorsal flank

Read more

Summary

Introduction

Scaffolds produced from a variety of biomaterials are widely applied as engineered microenvironments or delivery vehicles in biomedical applications. These biomaterial-based scaffolds can be used as three-dimensional (3D) culture systems in vitro to more faithfully recapitulate the complex set of cues that cells receive in the body [1]. Biomaterial-based scaffolds can be applied in vivo as delivery vehicles of bioactive molecules or cells, since they can exert spatiotemporal control over the release of bioactive molecules [2, 3] and dictate cellular localization [4, 5]. By acting as molecular and cellular delivery vehicles with high spatiotemporal resolution, biomaterial-based scaffolds can have a clear additive value to current cancer immunotherapeutic strategies

Objectives
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