Cargando…

Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology

Many complex diseases such as cancer are associated with multiple pathological manifestations. Moreover, the therapeutics for their treatments often lead to serious side effects. Thus, it is needed to develop multi-indication therapeutics that can simultaneously target multiple clinical indications...

Descripción completa

Detalles Bibliográficos
Autores principales: Lim, Hansaim, He, Di, Qiu, Yue, Krawczuk, Patrycja, Sun, Xiaoru, Xie, Lei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6576746/
https://www.ncbi.nlm.nih.gov/pubmed/31206508
http://dx.doi.org/10.1371/journal.pcbi.1006619
_version_ 1783427833952993280
author Lim, Hansaim
He, Di
Qiu, Yue
Krawczuk, Patrycja
Sun, Xiaoru
Xie, Lei
author_facet Lim, Hansaim
He, Di
Qiu, Yue
Krawczuk, Patrycja
Sun, Xiaoru
Xie, Lei
author_sort Lim, Hansaim
collection PubMed
description Many complex diseases such as cancer are associated with multiple pathological manifestations. Moreover, the therapeutics for their treatments often lead to serious side effects. Thus, it is needed to develop multi-indication therapeutics that can simultaneously target multiple clinical indications of interest and mitigate the side effects. However, conventional one-drug-one-gene drug discovery paradigm and emerging polypharmacology approach rarely tackle the challenge of multi-indication drug design. For the first time, we propose a one-drug-multi-target-multi-indication strategy. We develop a novel structural systems pharmacology platform 3D-REMAP that uses ligand binding site comparison and protein-ligand docking to augment sparse chemical genomics data for the machine learning model of genome-scale chemical-protein interaction prediction. Experimentally validated predictions systematically show that 3D-REMAP outperforms state-of-the-art ligand-based, receptor-based, and machine learning methods alone. As a proof-of-concept, we utilize the concept of drug repurposing that is enabled by 3D-REMAP to design dual-indication anti-cancer therapy. The repurposed drug can demonstrate anti-cancer activity for cancers that do not have effective treatment as well as reduce the risk of heart failure that is associated with all types of existing anti-cancer therapies. We predict that levosimendan, a PDE inhibitor for heart failure, inhibits serine/threonine-protein kinase RIOK1 and other kinases. Subsequent experiments and systems biology analyses confirm this prediction, and suggest that levosimendan is active against multiple cancers, notably lymphoma, through the direct inhibition of RIOK1 and RNA processing pathway. We further develop machine learning models to predict cancer cell-line’s and a patient’s response to levosimendan. Our findings suggest that levosimendan can be a promising novel lead compound for the development of safe, effective, and precision multi-indication anti-cancer therapy. This study demonstrates the potential of structural systems pharmacology in designing polypharmacology for precision medicine. It may facilitate transforming the conventional one-drug-one-gene-one-disease drug discovery process and single-indication polypharmacology approach into a new one-drug-multi-target-multi-indication paradigm for complex diseases.
format Online
Article
Text
id pubmed-6576746
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-65767462019-06-28 Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology Lim, Hansaim He, Di Qiu, Yue Krawczuk, Patrycja Sun, Xiaoru Xie, Lei PLoS Comput Biol Research Article Many complex diseases such as cancer are associated with multiple pathological manifestations. Moreover, the therapeutics for their treatments often lead to serious side effects. Thus, it is needed to develop multi-indication therapeutics that can simultaneously target multiple clinical indications of interest and mitigate the side effects. However, conventional one-drug-one-gene drug discovery paradigm and emerging polypharmacology approach rarely tackle the challenge of multi-indication drug design. For the first time, we propose a one-drug-multi-target-multi-indication strategy. We develop a novel structural systems pharmacology platform 3D-REMAP that uses ligand binding site comparison and protein-ligand docking to augment sparse chemical genomics data for the machine learning model of genome-scale chemical-protein interaction prediction. Experimentally validated predictions systematically show that 3D-REMAP outperforms state-of-the-art ligand-based, receptor-based, and machine learning methods alone. As a proof-of-concept, we utilize the concept of drug repurposing that is enabled by 3D-REMAP to design dual-indication anti-cancer therapy. The repurposed drug can demonstrate anti-cancer activity for cancers that do not have effective treatment as well as reduce the risk of heart failure that is associated with all types of existing anti-cancer therapies. We predict that levosimendan, a PDE inhibitor for heart failure, inhibits serine/threonine-protein kinase RIOK1 and other kinases. Subsequent experiments and systems biology analyses confirm this prediction, and suggest that levosimendan is active against multiple cancers, notably lymphoma, through the direct inhibition of RIOK1 and RNA processing pathway. We further develop machine learning models to predict cancer cell-line’s and a patient’s response to levosimendan. Our findings suggest that levosimendan can be a promising novel lead compound for the development of safe, effective, and precision multi-indication anti-cancer therapy. This study demonstrates the potential of structural systems pharmacology in designing polypharmacology for precision medicine. It may facilitate transforming the conventional one-drug-one-gene-one-disease drug discovery process and single-indication polypharmacology approach into a new one-drug-multi-target-multi-indication paradigm for complex diseases. Public Library of Science 2019-06-17 /pmc/articles/PMC6576746/ /pubmed/31206508 http://dx.doi.org/10.1371/journal.pcbi.1006619 Text en © 2019 Lim et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Lim, Hansaim
He, Di
Qiu, Yue
Krawczuk, Patrycja
Sun, Xiaoru
Xie, Lei
Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
title Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
title_full Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
title_fullStr Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
title_full_unstemmed Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
title_short Rational discovery of dual-indication multi-target PDE/Kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
title_sort rational discovery of dual-indication multi-target pde/kinase inhibitor for precision anti-cancer therapy using structural systems pharmacology
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6576746/
https://www.ncbi.nlm.nih.gov/pubmed/31206508
http://dx.doi.org/10.1371/journal.pcbi.1006619
work_keys_str_mv AT limhansaim rationaldiscoveryofdualindicationmultitargetpdekinaseinhibitorforprecisionanticancertherapyusingstructuralsystemspharmacology
AT hedi rationaldiscoveryofdualindicationmultitargetpdekinaseinhibitorforprecisionanticancertherapyusingstructuralsystemspharmacology
AT qiuyue rationaldiscoveryofdualindicationmultitargetpdekinaseinhibitorforprecisionanticancertherapyusingstructuralsystemspharmacology
AT krawczukpatrycja rationaldiscoveryofdualindicationmultitargetpdekinaseinhibitorforprecisionanticancertherapyusingstructuralsystemspharmacology
AT sunxiaoru rationaldiscoveryofdualindicationmultitargetpdekinaseinhibitorforprecisionanticancertherapyusingstructuralsystemspharmacology
AT xielei rationaldiscoveryofdualindicationmultitargetpdekinaseinhibitorforprecisionanticancertherapyusingstructuralsystemspharmacology