Abstract

Abstract Introduction: In vivo multiphoton autofluorescence microscopy provides live, label free, single cell imaging of metabolic changes. These metabolic changes are quantified via the metabolic coenzymes NAD(P)H and FAD which are autofluorescent molecules endogenous to all cells. Metabolic reprogramming of tumor and immune cells is closely associated with cancer progression and cell phenotype. We aim to study metabolic changes during administration of an effective, triple-combination immunotherapy within murine melanoma tumors. This therapy includes external beam radiation, intratumoral hu14.18-IL2 immunocytokine (anti-GD2 mAb fused to IL2, provided by Anyxis Immuno-Oncology GmbH of Vienna, Austria), and intraperitoneal anti-CTLA-4 leading to in situ vaccination and cure of GD2+ murine tumors. Previous work has shown that a T cell response is critical to the efficacy of this therapy, so we created an mCherry-labeled T cell mouse model to study this response. Methods: We implanted syngeneic B78 (GD2+) melanoma cells into the flanks of mCherry-labeled CD8 T cell reporter mice (C57Bl/6 background) to induce tumors. Under anesthesia, skin flap surgery was performed and tumors were imaged at several time points during therapy. Multiphoton imaging was performed to collect NAD(P)H, FAD, mCherry, and collagen signal with a 40X objective. Fluorescence lifetime data was collected using time correlated single photon counting electronics. Tissues were harvested and analyzed via flow cytometry and multiplex immunofluorescence to corroborate intravital imaging findings and characterize the immune infiltrate. Results: Here we demonstrate that our in vivo imaging is sensitive to metabolic changes in both B78 melanoma and CD8 T cells from immunotherapy treated versus control tumors. These metabolic differences include changes in protein binding, redox balance, and fluorescence lifetime. We also show remodeling of collagen, a major component of the extracellular matrix, during immunotherapy that may be explained by an observed decrease in macrophages. Flow cytometry and multiplex immunofluorescence illustrate changes in the immune infiltrate composition, activation, cytotoxicity, and spatial distribution during therapy. We are also currently investigating metabolic changes in an MC38 mouse model - a hot tumor model - that we anticipate will provide excellent metabolic contrast to our B78 immunologically cold tumor model. Conclusions: These results show that in vivo metabolic imaging enables single cell quantification of metabolic changes in tumor and immune cells during therapy. Combined with other traditional assays, we can elucidate key immune cell populations and the crucial timepoints during therapy where changes are occurring. With continued efforts, this imaging platform may be leveraged to develop new combinations of immunotherapies. Citation Format: Alexa R. Heaton, Anna Hoefges, Peter R. Rehani, Nathaniel J. Burkard, Arika S. Feils, Dan V. Spiegelman, Noah W. Tsarovsky, Alina A. Hampton, Amy K. Erbe Gurel, Alexander L. Rakhmilevich, Paul M. Sondel, Melissa C. Skala. In vivo multiphoton autofluorescence imaging is sensitive to CD8 T cell and tumor cell metabolic changes during immunotherapy in a murine melanoma model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2389.

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