Abstract

BackgroundTreatments that generate T cell-mediated immunity to a patient’s unique neoantigens are the current holy grail of cancer immunotherapy. In particular, treatments that do not require cumbersome and individualized ex vivo processing or manufacturing processes are especially sought after. Here we report that AGI-134, a glycolipid-like small molecule, can be used for coating tumor cells with the xenoantigen Galα1-3Galβ1-4GlcNAc (α-Gal) in situ leading to opsonization with pre-existing natural anti-α-Gal antibodies (in short anti-Gal), which triggers immune cascades resulting in T cell mediated anti-tumor immunity.MethodsVarious immunological effects of coating tumor cells with α-Gal via AGI-134 in vitro were measured by flow cytometry: (1) opsonization with anti-Gal and complement, (2) antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells, and (3) phagocytosis and antigen cross-presentation by antigen presenting cells (APCs). A viability kit was used to test AGI-134 mediated complement dependent cytotoxicity (CDC) in cancer cells. The anti-tumoral activity of AGI-134 alone or in combination with an anti-programmed death-1 (anti-PD-1) antibody was tested in melanoma models in anti-Gal expressing galactosyltransferase knockout (α1,3GT−/−) mice. CDC and phagocytosis data were analyzed by one-way ANOVA, ADCC results by paired t-test, distal tumor growth by Mantel–Cox test, C5a data by Mann–Whitney test, and single tumor regression by repeated measures analysis.ResultsIn vitro, α-Gal labelling of tumor cells via AGI-134 incorporation into the cell membrane leads to anti-Gal binding and complement activation. Through the effects of complement and ADCC, tumor cells are lysed and tumor antigen uptake by APCs increased. Antigen associated with lysed cells is cross-presented by CD8α+ dendritic cells leading to activation of antigen-specific CD8+ T cells. In B16-F10 or JB/RH melanoma models in α1,3GT−/− mice, intratumoral AGI-134 administration leads to primary tumor regression and has a robust abscopal effect, i.e., it protects from the development of distal, uninjected lesions. Combinations of AGI-134 and anti-PD-1 antibody shows a synergistic benefit in protection from secondary tumor growth.ConclusionsWe have identified AGI-134 as an immunotherapeutic drug candidate, which could be an excellent combination partner for anti-PD-1 therapy, by facilitating tumor antigen processing and increasing the repertoire of tumor-specific T cells prior to anti-PD-1 treatment.

Highlights

  • Treatments that generate T cell-mediated immunity to a patient’s unique neoantigens are the current holy grail of cancer immunotherapy

  • Human anti‐Gal antibodies bind to AGI‐134‐treated human cancer cells To initiate the immunological cascade that results in antitumor immunity, AGI-134 must first incorporate into tumor cell plasma membranes and present the α-Gal antigen for binding by anti-Gal antibodies

  • (See figure on page.) Fig. 1 Anti-Gal binds to AGI-134-treated human cancer cells and activates complement dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). a Human SW480 and A549 cancer cells were treated with phosphate buffered saline (PBS) or the indicated concentrations of AGI-134

Read more

Summary

Introduction

Treatments that generate T cell-mediated immunity to a patient’s unique neoantigens are the current holy grail of cancer immunotherapy. To boost the efficacy of the immune checkpoint inhibitors in refractory patient populations, there are several approaches that aim to either: (1) produce intratumoral inflammation, (2) increase tumor antigen processing and prime naïve T cells against tumor-specific antigens (TSAs) or (3) relieve immunosuppression in the tumor microenvironment [3, 4]. One method that has been explored to boost tumor tissue processing and produce activation of TSA-specific T cells harnesses the ability of pre-existing natural antibodies to mediate rejection of xenogeneic tissue bearing Galα1-3Galβ1-4GlcNAc (α-Gal) epitopes. When anti-Gal antibodies bind to α-Gal-bearing tissue, they activate the complement cascade and initiate antibody-dependent cellular cytotoxicity (ADCC) [9,10,11], resulting in the release of inflammatory mediators and lysis of the tissue It is the activation of complement and ADCC by anti-Gal that drives the hyperacute rejection of xenotransplants in humans

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.