Multiplexed non-invasive tumor imaging of glucose metabolism and receptor-ligand engagement using dark quencher FRET acceptor

Theranostics. 2020 Aug 15;10(22):10309-10325. doi: 10.7150/thno.45825. eCollection 2020.

Abstract

Rationale: Following an ever-increased focus on personalized medicine, there is a continuing need to develop preclinical molecular imaging modalities to guide the development and optimization of targeted therapies. Near-Infrared (NIR) Macroscopic Fluorescence Lifetime Förster Resonance Energy Transfer (MFLI-FRET) imaging offers a unique method to robustly quantify receptor-ligand engagement in live intact animals, which is critical to assess the delivery efficacy of therapeutics. However, to date, non-invasive imaging approaches that can simultaneously measure cellular drug delivery efficacy and metabolic response are lacking. A major challenge for the implementation of concurrent optical and MFLI-FRET in vivo whole-body preclinical imaging is the spectral crowding and cross-contamination between fluorescent probes. Methods: We report on a strategy that relies on a dark quencher enabling simultaneous assessment of receptor-ligand engagement and tumor metabolism in intact live mice. Several optical imaging approaches, such as in vitro NIR FLI microscopy (FLIM) and in vivo wide-field MFLI, were used to validate a novel donor-dark quencher FRET pair. IRDye 800CW 2-deoxyglucose (2-DG) imaging was multiplexed with MFLI-FRET of NIR-labeled transferrin FRET pair (Tf-AF700/Tf-QC-1) to monitor tumor metabolism and probe uptake in breast tumor xenografts in intact live nude mice. Immunohistochemistry was used to validate in vivo imaging results. Results: First, we establish that IRDye QC-1 (QC-1) is an effective NIR dark acceptor for the FRET-induced quenching of donor Alexa Fluor 700 (AF700). Second, we report on simultaneous in vivo imaging of the metabolic probe 2-DG and MFLI-FRET imaging of Tf-AF700/Tf-QC-1 uptake in tumors. Such multiplexed imaging revealed an inverse relationship between 2-DG uptake and Tf intracellular delivery, suggesting that 2-DG signal may predict the efficacy of intracellular targeted delivery. Conclusions: Overall, our methodology enables for the first time simultaneous non-invasive monitoring of intracellular drug delivery and metabolic response in preclinical studies.

Keywords: FRET; breast cancer; lifetime imaging; metabolism; target engagement.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Benzenesulfonates / metabolism
  • Cell Line
  • Cell Line, Tumor
  • Drug Delivery Systems / methods
  • Fluorescence
  • Fluorescence Resonance Energy Transfer / methods*
  • Fluorescent Dyes / metabolism
  • Glucose / metabolism*
  • Humans
  • Indoles / metabolism
  • Ligands
  • Mice
  • Mice, Nude
  • Optical Imaging / methods*
  • Transferrin / metabolism

Substances

  • Benzenesulfonates
  • Fluorescent Dyes
  • IRDye 800CW
  • Indoles
  • Ligands
  • Transferrin
  • Glucose