Abstract

BackgroundThe selection and prioritization of drug targets is a central problem in drug discovery. Computational approaches can leverage the growing number of large-scale human genomics and proteomics data to make in-silico target identification, reducing the cost and the time needed.ResultsWe developed a machine learning approach to score proteins to generate a druggability score of novel targets. In our model we incorporated 70 protein features which included properties derived from the sequence, features characterizing protein functions as well as network properties derived from the protein-protein interaction network. The advantage of this approach is that it is unbiased and even less studied proteins with limited information about their function can score well as most of the features are independent of the accumulated literature. We build models on a training set which consist of targets with approved drugs and a negative set of non-drug targets. The machine learning techniques help to identify the most important combination of features differentiating validated targets from non-targets. We validated our predictions on an independent set of clinical trial drug targets, achieving a high accuracy characterized by an Area Under the Curve (AUC) of 0.89. Our most predictive features included biological function of proteins, network centrality measures, protein essentiality, tissue specificity, localization and solvent accessibility. Our predictions, based on a small set of 102 validated oncology targets, recovered the majority of known drug targets and identifies a novel set of proteins as drug target candidates.ConclusionsWe developed a machine learning approach to prioritize proteins according to their similarity to approved drug targets. We have shown that the method proposed is highly predictive on a validation dataset consisting of 277 targets of clinical trial drug confirming that our computational approach is an efficient and cost-effective tool for drug target discovery and prioritization. Our predictions were based on oncology targets and cancer relevant biological functions, resulting in significantly higher scores for targets of oncology clinical trial drugs compared to the scores of targets of trial drugs for other indications. Our approach can be used to make indication specific drug-target prediction by combining generic druggability features with indication specific biological functions.

Highlights

  • The selection and prioritization of drug targets is a central problem in drug discovery

  • With the accumulation of approved and clinical trial drugs, it has become clear that successful drug targets share several important features, which include having a disease relevant biological function and certain properties that would favor the existence of binding sites, making the protein capable of binding to small molecules

  • We determined a comprehensive list of 70 properties for all human proteins by combining the manually curated literature captured in the Swiss-prot database, the computational predictions for missing features and network centrality properties calculated based on the protein-protein interaction network (Methods)

Read more

Summary

Introduction

The selection and prioritization of drug targets is a central problem in drug discovery. Computational approaches on the other hand can leverage the growing number of large-scale human genomics and proteomics data sets to make in-silico target identification, potentially reducing substantially the cost and the time needed to assess a target. A targeted computational approaches for example is the structure-based drug discovery where protein druggability is determined through molecular docking methods which can predict binding sites and binding affinity of the target proteins [3]. These methods are limited in finding novel targets because the three-dimensional structure of most proteins is not readily available

Methods
Results
Discussion
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