Target inhibition networks: predicting selective combinations of druggable targets to block cancer survival pathways

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Target inhibition networks : predicting selective combinations of druggable targets to block cancer survival pathways. / Tang, Jing; Karhinen, Leena; Xu, Tao; Szwajda, Agnieszka; Yadav, Bhagwan; Wennerberg, Krister; Aittokallio, Tero.

In: PLOS Computational Biology, Vol. 9, No. 9, 2013, p. e1003226.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Tang, J, Karhinen, L, Xu, T, Szwajda, A, Yadav, B, Wennerberg, K & Aittokallio, T 2013, 'Target inhibition networks: predicting selective combinations of druggable targets to block cancer survival pathways', PLOS Computational Biology, vol. 9, no. 9, pp. e1003226. https://doi.org/10.1371/journal.pcbi.1003226

APA

Tang, J., Karhinen, L., Xu, T., Szwajda, A., Yadav, B., Wennerberg, K., & Aittokallio, T. (2013). Target inhibition networks: predicting selective combinations of druggable targets to block cancer survival pathways. PLOS Computational Biology, 9(9), e1003226. https://doi.org/10.1371/journal.pcbi.1003226

Vancouver

Tang J, Karhinen L, Xu T, Szwajda A, Yadav B, Wennerberg K et al. Target inhibition networks: predicting selective combinations of druggable targets to block cancer survival pathways. PLOS Computational Biology. 2013;9(9):e1003226. https://doi.org/10.1371/journal.pcbi.1003226

Author

Tang, Jing ; Karhinen, Leena ; Xu, Tao ; Szwajda, Agnieszka ; Yadav, Bhagwan ; Wennerberg, Krister ; Aittokallio, Tero. / Target inhibition networks : predicting selective combinations of druggable targets to block cancer survival pathways. In: PLOS Computational Biology. 2013 ; Vol. 9, No. 9. pp. e1003226.

Bibtex

@article{72d150ed13f844288a4fd9ba840bbee2,
title = "Target inhibition networks: predicting selective combinations of druggable targets to block cancer survival pathways",
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. ",
keywords = "Antineoplastic Agents/pharmacology, Cell Line, Tumor, Cell Survival/drug effects, Humans, Models, Theoretical, Neoplasms/drug therapy",
author = "Jing Tang and Leena Karhinen and Tao Xu and Agnieszka Szwajda and Bhagwan Yadav and Krister Wennerberg and Tero Aittokallio",
year = "2013",
doi = "10.1371/journal.pcbi.1003226",
language = "English",
volume = "9",
pages = "e1003226",
journal = "P L o S Computational Biology (Online)",
issn = "1553-734X",
publisher = "Public Library of Science",
number = "9",

}

RIS

TY - JOUR

T1 - Target inhibition networks

T2 - predicting selective combinations of druggable targets to block cancer survival pathways

AU - Tang, Jing

AU - Karhinen, Leena

AU - Xu, Tao

AU - Szwajda, Agnieszka

AU - Yadav, Bhagwan

AU - Wennerberg, Krister

AU - Aittokallio, Tero

PY - 2013

Y1 - 2013

N2 - 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.

AB - 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.

KW - Antineoplastic Agents/pharmacology

KW - Cell Line, Tumor

KW - Cell Survival/drug effects

KW - Humans

KW - Models, Theoretical

KW - Neoplasms/drug therapy

U2 - 10.1371/journal.pcbi.1003226

DO - 10.1371/journal.pcbi.1003226

M3 - Journal article

C2 - 24068907

VL - 9

SP - e1003226

JO - P L o S Computational Biology (Online)

JF - P L o S Computational Biology (Online)

SN - 1553-734X

IS - 9

ER -

ID: 199431531