Abstract

Near-infrared (NIR)-activable liposomes containing photosensitizer (PS)-lipid conjugates are emerging as tunable, high-payload, and tumor-selective platforms for photodynamic therapy (PDT)-based theranostics. To date, the impact that the membrane composition of a NIR-activable liposome (the chemical nature and subsequent conformation of PS-lipid conjugates) has on their in vitro and in vivo functionality has not been fully investigated. While their chemical nature is critical, the resultant physical conformation dictates their interactions with the immediate biological environments. Here, we evaluate NIR-activable liposomes containing lipid conjugates of the clinically-used PSs benzoporphyrin derivative (BPD; hydrophobic, membrane-inserting conformation) or IRDye 700DX (hydrophilic, membrane-protruding conformation) and demonstrate that membrane composition is critical for their function as tumor-selective PDT-based platforms. The PS-lipid conformations were primarily dictated by the varying solubilities of the two PSs and assisted by their lipid conjugation sites. Conformation was further validated by photophysical analysis and computational predictions of PS membrane partitioning (topological polar surface area [tPSA], calculated octanol/water partition [cLogP], and apparent biomembrane permeability coefficient [Papp]). Results show that the membrane-protruding lipo-IRDye700DX exhibits 5-fold more efficient photodynamic generation of reactive molecular species (RMS), 12-fold expedited phototriggered burst release of entrap-ped agents, and 15-fold brighter fluorescence intensity as compared to the membrane-inserting lipo-BPD-PC (phosphatidylcholine conjugate). Although the membrane-inserting lipo-BPD-PC exhibits less efficient photo-dynamic generation of RMS, it allows for more sustained phototriggered release, 10-fold greater FaDu cancer cell phototoxicity, and 7.16-fold higher tumor-selective delivery in orthotopic mouse FaDu head and neck tumors. These critical insights pave the path for the rational design of emerging NIR-activable liposomes, whereby functional consequences of membrane composition can be tailored toward a specific therapeutic purpose.

Highlights

  • Mina Guirguis and Chanda Bhandari contributed to this study.Photonanomedicines (PNMs) are light-activable nanoscale drug delivery systems that facilitate photodynamic therapy (PDT)

  • Our results enable the rational design of NIR-activable liposomes by introducing membrane composition as a critical component that can be selected in a purpose-specific manner for optimal therapeutic and theranostic functionality

  • Computational simulations predicted that DSPE-PEG-IRDye700DX was membrane protruding, with a high topological polar surface area of 471.72, a low cLogP of −14.2 and a low biomembrane permeability coefficient of Papp = 0.071 × 10−6 cm/s

Read more

Summary

Introduction

Mina Guirguis and Chanda Bhandari contributed to this study. Photonanomedicines (PNMs) are light-activable nanoscale drug delivery systems that facilitate photodynamic therapy (PDT). PDT destroys disease tissue through photochemistry, a light-initiated reaction between a light activable molecule – the photosensitizer (PS) – and oftentimes molecular oxygen [1, 2]. Common lipid-based PNM platforms that are frequently used for PDT-based regimens include liposomes, liposome bilayer coated nanoconstructs, and monolayer lipid-coated nanoconstructs. VisudyneTM, a near-infrared (NIR)-activable liposome containing the PS benzoporphyrin derivative (BPD), was the first PNM to gain Food and Drug Administration approval in 2000 for PDT of Age-related Macular Degeneration [3]. NIR-activable PNMs, especially those based on nanometric liposomal

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