Abstract

Over the past decades there have been exciting and rapid developments of highly specific molecules to bind cancer antigens that are overexpressed on the surfaces of malignant cells. Nanomedicine aims to exploit these ligands to generate nanoscale platforms for targeted cancer therapy, and to do so with negligible off-target effects. Aptamers are structured nucleic acids that bind to defined molecular targets ranging from small molecules and proteins to whole cells or viruses. They are selected through an iterative process of amplification and enrichment called SELEX (systematic evolution of ligands by exponential enrichment), in which a combinatorial oligonucleotide library is exposed to the target of interest for several repetitive rounds. Nucleic acid ligands able to bind and internalize into malignant cells have been extensively used as tools for targeted delivery of therapeutic payloads both in vitro and in vivo. However, current cell targeting aptamer platforms suffer from limitations that have slowed their translation to the clinic. This is especially true for applications in which the cargo must reach the cytosol to exert its biological activity, as only a small percentage of the endocytosed cargo is typically able to translocate into the cytosol. Innovative technologies and selection strategies are required to enhance cytoplasmic delivery. In this review, we describe current selection methods used to generate aptamers that target cancer cells, and we highlight some of the factors that affect productive endosomal escape of cargoes. We also give an overview of the most promising strategies utilized to improve and monitor endosomal escape of therapeutic cargoes. The methods we highlight exploit tools and technologies that can potentially be incorporated in the SELEX process. Innovative selection protocols may identify aptamers with extended biological functionalities that allow effective cytosolic translocation of therapeutics. This in turn may facilitate successful translation of these platforms into clinical applications.

Highlights

  • Cancer remains one of the leading causes of death worldwide, with high morbidity and mortality

  • A combined strategy that utilizes key features of fluorescent-activated cell sorting (FACS)- and cell-internalization-SELEX could represent an intriguing strategy of selection because it would simultaneously reduce limitations associated to cell-based SELEX and allow for direct monitoring of aptamer enrichment at each round of the selection via a dye-labeled RNA pool

  • Ligand-Guided Selection (LIGS) exploits natural ligands or other high-affinity reagents such as monoclonal antibodies that recognize the same target receptor to displace and elute specific aptamers. When this displacing ligand binds its cognate antigen on a certain cell-surface marker, it outcompetes and displaces aptamer sequences that interact at that site

Read more

Summary

Introduction

Cancer remains one of the leading causes of death worldwide, with high morbidity and mortality. Advances in nanotechnology might overcome these constraints, additional limitations need to be addressed to allow for an efficient transition to clinical application Key among these is the productive engagement of biological mechanisms that govern the processing of these aptamer-based delivery platforms upon internalization into target cells. The amount of therapeutic cargo that can successfully access the cytosol appears to depend mainly on three factors: the number of targeted receptor molecules on the cell surface, the rates of their endocytic uptake, and the endosomal escape efficiency over the course of a cycle of internalization. Extending rapidly (15min) [20,22,23] These examples are, to some extent, the low-hanging fruit of targeted delivery to cells that display surface proteins at more typical levels will require technological the field. An exciting frontier could be to incorporate these strategies into aptamer selection protocols to identify cell targeting aptamers that ensure improved cytoplasmic accessibility of their cargoes

In Vitro Selection of Cell Targeting Aptamers
Recent Advances in Cell-internalization SELEX
Schematic illustration of the cell-internalization
Strategies to Enhance and Monitor Endosomal Escape of Cargoes
Traditional and Innovative Methods to Monitor Endosomal Escape of Cargoes
Conclusions
Findings
Acknowledgments:
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