Limited inhibition of multiple nodes in a driver network blocks metastasis

Metastasis suppression by high-dose, multi-drug targeting is unsuccessful due to network heterogeneity and compensatory network activation. Here, we show that targeting driver network signaling capacity by limited inhibition of core pathways is a more effective anti-metastatic strategy. This princip...

Full description

Bibliographic Details
Main Authors: Ali Ekrem Yesilkanal, Dongbo Yang, Andrea Valdespino, Payal Tiwari, Alan U Sabino, Long Chi Nguyen, Jiyoung Lee, Xiao-He Xie, Siqi Sun, Christopher Dann, Lydia Robinson-Mailman, Ethan Steinberg, Timothy Stuhlmiller, Casey Frankenberger, Elizabeth Goldsmith, Gary L Johnson, Alexandre F Ramos, Marsha R Rosner
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2021-05-01
Series:eLife
Subjects:
Online Access:https://elifesciences.org/articles/59696
_version_ 1811180407661527040
author Ali Ekrem Yesilkanal
Dongbo Yang
Andrea Valdespino
Payal Tiwari
Alan U Sabino
Long Chi Nguyen
Jiyoung Lee
Xiao-He Xie
Siqi Sun
Christopher Dann
Lydia Robinson-Mailman
Ethan Steinberg
Timothy Stuhlmiller
Casey Frankenberger
Elizabeth Goldsmith
Gary L Johnson
Alexandre F Ramos
Marsha R Rosner
author_facet Ali Ekrem Yesilkanal
Dongbo Yang
Andrea Valdespino
Payal Tiwari
Alan U Sabino
Long Chi Nguyen
Jiyoung Lee
Xiao-He Xie
Siqi Sun
Christopher Dann
Lydia Robinson-Mailman
Ethan Steinberg
Timothy Stuhlmiller
Casey Frankenberger
Elizabeth Goldsmith
Gary L Johnson
Alexandre F Ramos
Marsha R Rosner
author_sort Ali Ekrem Yesilkanal
collection DOAJ
description Metastasis suppression by high-dose, multi-drug targeting is unsuccessful due to network heterogeneity and compensatory network activation. Here, we show that targeting driver network signaling capacity by limited inhibition of core pathways is a more effective anti-metastatic strategy. This principle underlies the action of a physiological metastasis suppressor, Raf Kinase Inhibitory Protein (RKIP), that moderately decreases stress-regulated MAP kinase network activity, reducing output to transcription factors such as pro-metastastic BACH1 and motility-related target genes. We developed a low-dose four-drug mimic that blocks metastatic colonization in mouse breast cancer models and increases survival. Experiments and network flow modeling show limited inhibition of multiple pathways is required to overcome variation in MAPK network topology and suppress signaling output across heterogeneous tumor cells. Restricting inhibition of individual kinases dissipates surplus signal, preventing threshold activation of compensatory kinase networks. This low-dose multi-drug approach to decrease signaling capacity of driver networks represents a transformative, clinically relevant strategy for anti-metastatic treatment.
first_indexed 2024-04-11T09:01:38Z
format Article
id doaj.art-4ff20943f44747458fc45b3108f69f89
institution Directory Open Access Journal
issn 2050-084X
language English
last_indexed 2024-04-11T09:01:38Z
publishDate 2021-05-01
publisher eLife Sciences Publications Ltd
record_format Article
series eLife
spelling doaj.art-4ff20943f44747458fc45b3108f69f892022-12-22T04:32:45ZengeLife Sciences Publications LtdeLife2050-084X2021-05-011010.7554/eLife.59696Limited inhibition of multiple nodes in a driver network blocks metastasisAli Ekrem Yesilkanal0https://orcid.org/0000-0003-4988-1294Dongbo Yang1Andrea Valdespino2Payal Tiwari3Alan U Sabino4https://orcid.org/0000-0003-1094-5078Long Chi Nguyen5Jiyoung Lee6https://orcid.org/0000-0001-8503-4805Xiao-He Xie7Siqi Sun8Christopher Dann9Lydia Robinson-Mailman10Ethan Steinberg11Timothy Stuhlmiller12Casey Frankenberger13Elizabeth Goldsmith14Gary L Johnson15https://orcid.org/0000-0003-2867-0551Alexandre F Ramos16Marsha R Rosner17https://orcid.org/0000-0001-6586-8335Ben May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesInstituto do Câncer do Estado de São Paulo, Faculdade de Medicina and Escola de Artes, Ciências e Humanidades; University of São Paulo, São Paulo, BrazilBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesDepartment of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United StatesBen May Department for Cancer Research, University of Chicago, Chicago, United StatesUT Southwestern, Dallas, United StatesDepartment of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, United StatesInstituto do Câncer do Estado de São Paulo, Faculdade de Medicina and Escola de Artes, Ciências e Humanidades; University of São Paulo, São Paulo, BrazilBen May Department for Cancer Research, University of Chicago, Chicago, United StatesMetastasis suppression by high-dose, multi-drug targeting is unsuccessful due to network heterogeneity and compensatory network activation. Here, we show that targeting driver network signaling capacity by limited inhibition of core pathways is a more effective anti-metastatic strategy. This principle underlies the action of a physiological metastasis suppressor, Raf Kinase Inhibitory Protein (RKIP), that moderately decreases stress-regulated MAP kinase network activity, reducing output to transcription factors such as pro-metastastic BACH1 and motility-related target genes. We developed a low-dose four-drug mimic that blocks metastatic colonization in mouse breast cancer models and increases survival. Experiments and network flow modeling show limited inhibition of multiple pathways is required to overcome variation in MAPK network topology and suppress signaling output across heterogeneous tumor cells. Restricting inhibition of individual kinases dissipates surplus signal, preventing threshold activation of compensatory kinase networks. This low-dose multi-drug approach to decrease signaling capacity of driver networks represents a transformative, clinically relevant strategy for anti-metastatic treatment.https://elifesciences.org/articles/59696metastasisMAPKraf kinase inhibitory proteinBACH1drug combinationsmathematical modeling
spellingShingle Ali Ekrem Yesilkanal
Dongbo Yang
Andrea Valdespino
Payal Tiwari
Alan U Sabino
Long Chi Nguyen
Jiyoung Lee
Xiao-He Xie
Siqi Sun
Christopher Dann
Lydia Robinson-Mailman
Ethan Steinberg
Timothy Stuhlmiller
Casey Frankenberger
Elizabeth Goldsmith
Gary L Johnson
Alexandre F Ramos
Marsha R Rosner
Limited inhibition of multiple nodes in a driver network blocks metastasis
eLife
metastasis
MAPK
raf kinase inhibitory protein
BACH1
drug combinations
mathematical modeling
title Limited inhibition of multiple nodes in a driver network blocks metastasis
title_full Limited inhibition of multiple nodes in a driver network blocks metastasis
title_fullStr Limited inhibition of multiple nodes in a driver network blocks metastasis
title_full_unstemmed Limited inhibition of multiple nodes in a driver network blocks metastasis
title_short Limited inhibition of multiple nodes in a driver network blocks metastasis
title_sort limited inhibition of multiple nodes in a driver network blocks metastasis
topic metastasis
MAPK
raf kinase inhibitory protein
BACH1
drug combinations
mathematical modeling
url https://elifesciences.org/articles/59696
work_keys_str_mv AT aliekremyesilkanal limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT dongboyang limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT andreavaldespino limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT payaltiwari limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT alanusabino limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT longchinguyen limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT jiyounglee limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT xiaohexie limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT siqisun limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT christopherdann limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT lydiarobinsonmailman limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT ethansteinberg limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT timothystuhlmiller limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT caseyfrankenberger limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT elizabethgoldsmith limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT garyljohnson limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT alexandreframos limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis
AT marsharrosner limitedinhibitionofmultiplenodesinadrivernetworkblocksmetastasis