Abstract

Targeted therapy has been widely adopted as an effective treatment strategy to battle against cancer. However, cancers are not single disease entities, but comprising multiple molecularly distinct subtypes, and the heterogeneity nature prevents precise selection of patients for optimized therapy. Dissecting cancer subtype-specific signaling pathways is crucial to pinpointing dysregulated genes for the prioritization of novel therapeutic targets. Nested effects models (NEMs) are a group of graphical models that encode subset relations between observed downstream effects under perturbations to upstream signaling genes, providing a prototype for mapping the inner workings of the cell. In this study, we developed NEM-Tar, which extends the original NEMs to predict drug targets by incorporating causal information of (epi)genetic aberrations for signaling pathway inference. An information theory-based score, weighted information gain (WIG), was proposed to assess the impact of signaling genes on a specific downstream biological process of interest. Subsequently, we conducted simulation studies to compare three inference methods and found that the greedy hill-climbing algorithm demonstrated the highest accuracy and robustness to noise. Furthermore, two case studies were conducted using multi-omics data for colorectal cancer (CRC) and gastric cancer (GC) in the TCGA database. Using NEM-Tar, we inferred signaling networks driving the poor-prognosis subtypes of CRC and GC, respectively. Our model prioritized not only potential individual drug targets such as HER2, for which FDA-approved inhibitors are available but also the combinations of multiple targets potentially useful for the design of combination therapies.

Highlights

  • Cancers are always discovered with diverse molecular properties and heterogeneous clinical outcomes, even when occurring in the same tissues or organs

  • We model copy number variations or mutations, hyper/hypo methylation as ‘natural’ perturbations in tumors, which are different from experimental perturbations such as RNA interference and CRISPR-Cas9 knockout modeled in the classic Nested effects models (NEMs)

  • For the choice of the regulatory elements, we focused on the signature genes of the maximum a posteriori (MAP)-kinase pathway (KRAS, BRAF), frequently mutated kinases/TFs (TP53, ARID1A, CDH1, and ERBB2) (Gastric Adenocarcinoma - My Cancer Genome) and significantly upregulated TFs as well as downregulated miRNAs in the epithelial-mesenchymal transition (EMT) subtype

Read more

Summary

Introduction

Cancers are always discovered with diverse molecular properties and heterogeneous clinical outcomes, even when occurring in the same tissues or organs. The last decade has witnessed tremendous progress in the emerging field of precision medicine for more accurate patient stratification for more optimized therapeutic treatment. It remains challenging to Network Inference Predicting Drug Targets dissect the mechanism underlying cancer heterogeneity to identify novel drug targets for further development of targeted therapies. Pathway redundancies, complex feedback, and crosstalk present in cancer cells often result in drug resistance, leading to treatment failure (Bernards, 2012; Yamaguchi et al, 2014). A key task of precision medicine is excavating the causally wired relationship among the regulatory elements contributing to specific cancer molecular subtypes

Objectives
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