Abstract

Targeting mitotic regulators as a strategy to fight cancer implies the development of drugs against key proteins, such as Aurora-A and -B. Current drugs, which target mitosis through a general mechanism of action (stabilization/destabilization of microtubules), have several side effects (neutropenia, alopecia, and emesis). Pharmaceutical companies aim at avoiding these unwanted effects by generating improved and selective drugs that increase the quality of life of the patients. However, the development of these drugs is an ambitious task that involves testing thousands of compounds through biochemical and cell-based assays. In addition, molecules usually target complex biological processes, involving several proteins and different molecular pathways, further emphasizing the need for high-throughput screening techniques and multiplexing technologies in order to identify drugs with the desired phenotype. We will briefly describe two multiplexing technologies [high-content imaging (HCI) and flow cytometry] and two key processes for drug discovery research (assay development and validation) following our own published industry quality standards. We will further focus on HCI as a useful tool for phenotypic screening and will provide a concrete example of HCI assay to detect Aurora-A or -B selective inhibitors discriminating the off-target effects related to the inhibition of other cell cycle or non-cell cycle key regulators. Finally, we will describe other assays that can help to characterize the in vitro pharmacology of the inhibitors.

Highlights

  • Aurora-A and Aurora-B are two serine threonine kinases that regulate cell cycle progression from G2 through to cytokinesis in a coordinated manner even though their localization and activation timing during the cell cycle varies

  • Aurora-A is required for mitotic entry, centrosome maturation and separation, and chromosome alignment [1], whereas Aurora-B is involved in chromosome condensation, segregation, and cytokinesis by regulating microtubule kinetochore associations [2]

  • The same concept applies for molecules against Aurora kinases, where most clinical trials have focused on Aurora-A selective agents (53%) as opposed to Pan-Aurora (32%) and Aurora-B-specific compounds (15%) [9]

Read more

Summary

INTRODUCTION

Aurora-A and Aurora-B are two serine threonine kinases that regulate cell cycle progression from G2 through to cytokinesis in a coordinated manner even though their localization and activation timing during the cell cycle varies. Aurora-A is required for mitotic entry, centrosome maturation and separation, and chromosome alignment [1], whereas Aurora-B is involved in chromosome condensation, segregation, and cytokinesis by regulating microtubule kinetochore associations [2]. Inhibition of any of these two kinases will produce a different phenotype, while Aurora-A inhibition delays mitotic entry and progression and accumulates cells in G2/M phase [3, 4], Aurora-B inhibition prevents proper alignment of chromosomes to the spindle plate, inhibits cytokinesis, and results in the formation of multinucleated cells [5]. The fact that human Aurora-A and -B share 71% identity in its carboxy-terminal catalytic domain is critical for evaluating the specificity of inhibitors [6]

Phenotypic Screening for Aurora Inhibitors
MULTIPLEXING TECHNOLOGIES
Flow Cytometry
Setting Up a Flow Scheme
Assay Validation
PRACTICAL EXAMPLES APPLIED TO AURORA INHIBITORS
Phenotypic Screening for Aurora Inhibitors C
Compound Treatment
Assay Performance
Interpretation and Phenotype Deconvolution
Phenotypic Screening for Aurora Inhibitors B
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