↓ Skip to main content

PLOS

Target Inhibition Networks: Predicting Selective Combinations of Druggable Targets to Block Cancer Survival Pathways

Overview of attention for article published in PLoS Computational Biology, September 2013
Altmetric Badge

Mentioned by

twitter
8 X users
patent
3 patents

Citations

dimensions_citation
73 Dimensions

Readers on

mendeley
170 Mendeley
citeulike
6 CiteULike
Title
Target Inhibition Networks: Predicting Selective Combinations of Druggable Targets to Block Cancer Survival Pathways
Published in
PLoS Computational Biology, September 2013
DOI 10.1371/journal.pcbi.1003226
Pubmed ID
Authors

Jing Tang, Leena Karhinen, Tao Xu, Agnieszka Szwajda, Bhagwan Yadav, Krister Wennerberg, Tero Aittokallio

Abstract

A recent trend in drug development is to identify drug combinations or multi-target agents that effectively modify multiple nodes of disease-associated networks. Such polypharmacological effects may reduce the risk of emerging drug resistance by means of attacking the disease networks through synergistic and synthetic lethal interactions. However, due to the exponentially increasing number of potential drug and target combinations, systematic approaches are needed for prioritizing the most potent multi-target alternatives on a global network level. We took a functional systems pharmacology approach toward the identification of selective target combinations for specific cancer cells by combining large-scale screening data on drug treatment efficacies and drug-target binding affinities. Our model-based prediction approach, named TIMMA, takes advantage of the polypharmacological effects of drugs and infers combinatorial drug efficacies through system-level target inhibition networks. Case studies in MCF-7 and MDA-MB-231 breast cancer and BxPC-3 pancreatic cancer cells demonstrated how the target inhibition modeling allows systematic exploration of functional interactions between drugs and their targets to maximally inhibit multiple survival pathways in a given cancer type. The TIMMA prediction results were experimentally validated by means of systematic siRNA-mediated silencing of the selected targets and their pairwise combinations, showing increased ability to identify not only such druggable kinase targets that are essential for cancer survival either individually or in combination, but also synergistic interactions indicative of non-additive drug efficacies. These system-level analyses were enabled by a novel model construction method utilizing maximization and minimization rules, as well as a model selection algorithm based on sequential forward floating search. Compared with an existing computational solution, TIMMA showed both enhanced prediction accuracies in cross validation as well as significant reduction in computation times. Such cost-effective computational-experimental design strategies have the potential to greatly speed-up the drug testing efforts by prioritizing those interventions and interactions warranting further study in individual cancer cases.

X Demographics

X Demographics

The data shown below were collected from the profiles of 8 X users who shared this research output. Click here to find out more about how the information was compiled.
Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 170 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
United States 10 6%
Finland 2 1%
France 1 <1%
Korea, Republic of 1 <1%
Brazil 1 <1%
Switzerland 1 <1%
India 1 <1%
Netherlands 1 <1%
Qatar 1 <1%
Other 1 <1%
Unknown 150 88%

Demographic breakdown

Readers by professional status Count As %
Student > Ph. D. Student 52 31%
Researcher 37 22%
Student > Master 14 8%
Student > Doctoral Student 9 5%
Professor 9 5%
Other 26 15%
Unknown 23 14%
Readers by discipline Count As %
Agricultural and Biological Sciences 57 34%
Computer Science 22 13%
Biochemistry, Genetics and Molecular Biology 18 11%
Medicine and Dentistry 17 10%
Chemistry 7 4%
Other 19 11%
Unknown 30 18%