Cargando…

LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures

Pediatric low-grade glioma (pLGG), the most common brain tumors in children, are driven by alterations in the MAPK pathway. Several clinical trials have shown the potential for MAPK inhibitor (MAPKi) treatment in pLGG. However, the range of response to MAPKi is heterogeneous, even between tumors sha...

Descripción completa

Detalles Bibliográficos
Autores principales: Sigaud, Romain, Heß, Caroline, Hielscher, Thomas, Winkler, Nadine, Selt, Florian, Kocher, Daniela, Nonnenbroich, Leo, Usta, Diren, Sommerkamp, Alex, Büdenbender, Isabel, Capper, David, Thomale, Ulrich W, Driever, Pablo Hernáiz, le Simon, Michè, Koch, Arend, Jabado, Nada, Andrade, Augusto F, van Meeteren, Netteke Schouten-, Hoving, Eelco, van Tilburg, Cornelis M, Pfister, Stefan M, Witt, Olaf, Jones, David T W, Milde, Till
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Oxford University Press 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9165112/
http://dx.doi.org/10.1093/neuonc/noac079.340
_version_ 1784720310539386880
author Sigaud, Romain
Heß, Caroline
Hielscher, Thomas
Winkler, Nadine
Selt, Florian
Kocher, Daniela
Nonnenbroich, Leo
Usta, Diren
Sommerkamp, Alex
Büdenbender, Isabel
Capper, David
Thomale, Ulrich W
Driever, Pablo Hernáiz
le Simon, Michè
Koch, Arend
Jabado, Nada
Andrade, Augusto F
van Meeteren, Netteke Schouten-
Hoving, Eelco
van Tilburg, Cornelis M
Pfister, Stefan M
Witt, Olaf
Jones, David T W
Milde, Till
author_facet Sigaud, Romain
Heß, Caroline
Hielscher, Thomas
Winkler, Nadine
Selt, Florian
Kocher, Daniela
Nonnenbroich, Leo
Usta, Diren
Sommerkamp, Alex
Büdenbender, Isabel
Capper, David
Thomale, Ulrich W
Driever, Pablo Hernáiz
le Simon, Michè
Koch, Arend
Jabado, Nada
Andrade, Augusto F
van Meeteren, Netteke Schouten-
Hoving, Eelco
van Tilburg, Cornelis M
Pfister, Stefan M
Witt, Olaf
Jones, David T W
Milde, Till
author_sort Sigaud, Romain
collection PubMed
description Pediatric low-grade glioma (pLGG), the most common brain tumors in children, are driven by alterations in the MAPK pathway. Several clinical trials have shown the potential for MAPK inhibitor (MAPKi) treatment in pLGG. However, the range of response to MAPKi is heterogeneous, even between tumors sharing the same driving MAPK alteration. A predictive stratification tool is needed to identify tumors that will be sensitive to MAPK inhibition. We generated sensitivity gene signatures for each MAPKi class (BRAFi, MEKi, ERKi), based on MAPK-related genes differentially regulated between MAPKi sensitive and non-sensitive cell lines from the Genomics of Drug Sensitivity in Cancer (GDSC) dataset. Single sample Gene Set Enrichment Analysis was used to measure and validate the MAPKi predictive sensitivity scores in the GDSC dataset and an independent patient-derived xenograft (PDX) dataset (XevaDB). The validated signatures were tested in a pLGG-specific background, using gene expression data from pLGG cell lines and primary pLGG samples. Our MAPKi sensitivity signatures discriminated MAPKi sensitive and non-sensitive cells in the GDSC dataset, and significantly correlated with MAPKi response in the PDX dataset. The sensitivity scores discerned gliomas with varying MAPK alterations from those without MAPK alterations, and showed higher scores in pLGG compared to high-grade gliomas and normal brain tissue. MAPKi-predicted sensitivity was heterogeneous within pLGG groups with a common MAPK alteration, as observed in MAPKi clinical trials. Intriguingly, we observed a strong positive correlation between our MAPKi sensitivity signature scores and the predicted immune cell infiltration rate as determined by the ESTIMATE score. These data demonstrate the potential relevance of gene-expression signatures to predict response to MAPKi treatment in pLGG patients, worth of further investigation in a prospective manner in upcoming clinical trials. In addition, our data could support a role of immune cell infiltration in the response to MAPKi in pLGG, warranting further validation.
format Online
Article
Text
id pubmed-9165112
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Oxford University Press
record_format MEDLINE/PubMed
spelling pubmed-91651122022-06-05 LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures Sigaud, Romain Heß, Caroline Hielscher, Thomas Winkler, Nadine Selt, Florian Kocher, Daniela Nonnenbroich, Leo Usta, Diren Sommerkamp, Alex Büdenbender, Isabel Capper, David Thomale, Ulrich W Driever, Pablo Hernáiz le Simon, Michè Koch, Arend Jabado, Nada Andrade, Augusto F van Meeteren, Netteke Schouten- Hoving, Eelco van Tilburg, Cornelis M Pfister, Stefan M Witt, Olaf Jones, David T W Milde, Till Neuro Oncol Low Grade Glioma Pediatric low-grade glioma (pLGG), the most common brain tumors in children, are driven by alterations in the MAPK pathway. Several clinical trials have shown the potential for MAPK inhibitor (MAPKi) treatment in pLGG. However, the range of response to MAPKi is heterogeneous, even between tumors sharing the same driving MAPK alteration. A predictive stratification tool is needed to identify tumors that will be sensitive to MAPK inhibition. We generated sensitivity gene signatures for each MAPKi class (BRAFi, MEKi, ERKi), based on MAPK-related genes differentially regulated between MAPKi sensitive and non-sensitive cell lines from the Genomics of Drug Sensitivity in Cancer (GDSC) dataset. Single sample Gene Set Enrichment Analysis was used to measure and validate the MAPKi predictive sensitivity scores in the GDSC dataset and an independent patient-derived xenograft (PDX) dataset (XevaDB). The validated signatures were tested in a pLGG-specific background, using gene expression data from pLGG cell lines and primary pLGG samples. Our MAPKi sensitivity signatures discriminated MAPKi sensitive and non-sensitive cells in the GDSC dataset, and significantly correlated with MAPKi response in the PDX dataset. The sensitivity scores discerned gliomas with varying MAPK alterations from those without MAPK alterations, and showed higher scores in pLGG compared to high-grade gliomas and normal brain tissue. MAPKi-predicted sensitivity was heterogeneous within pLGG groups with a common MAPK alteration, as observed in MAPKi clinical trials. Intriguingly, we observed a strong positive correlation between our MAPKi sensitivity signature scores and the predicted immune cell infiltration rate as determined by the ESTIMATE score. These data demonstrate the potential relevance of gene-expression signatures to predict response to MAPKi treatment in pLGG patients, worth of further investigation in a prospective manner in upcoming clinical trials. In addition, our data could support a role of immune cell infiltration in the response to MAPKi in pLGG, warranting further validation. Oxford University Press 2022-06-03 /pmc/articles/PMC9165112/ http://dx.doi.org/10.1093/neuonc/noac079.340 Text en © The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. https://creativecommons.org/licenses/by-nc/4.0/This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
spellingShingle Low Grade Glioma
Sigaud, Romain
Heß, Caroline
Hielscher, Thomas
Winkler, Nadine
Selt, Florian
Kocher, Daniela
Nonnenbroich, Leo
Usta, Diren
Sommerkamp, Alex
Büdenbender, Isabel
Capper, David
Thomale, Ulrich W
Driever, Pablo Hernáiz
le Simon, Michè
Koch, Arend
Jabado, Nada
Andrade, Augusto F
van Meeteren, Netteke Schouten-
Hoving, Eelco
van Tilburg, Cornelis M
Pfister, Stefan M
Witt, Olaf
Jones, David T W
Milde, Till
LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
title LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
title_full LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
title_fullStr LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
title_full_unstemmed LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
title_short LGG-26. Predicting MAPK inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
title_sort lgg-26. predicting mapk inhibitor sensitivity in pediatric low-grade gliomas with novel gene expression-derived signatures
topic Low Grade Glioma
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9165112/
http://dx.doi.org/10.1093/neuonc/noac079.340
work_keys_str_mv AT sigaudromain lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT heßcaroline lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT hielscherthomas lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT winklernadine lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT seltflorian lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT kocherdaniela lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT nonnenbroichleo lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT ustadiren lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT sommerkampalex lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT budenbenderisabel lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT capperdavid lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT thomaleulrichw lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT drieverpablohernaiz lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT lesimonmiche lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT kocharend lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT jabadonada lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT andradeaugustof lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT vanmeeterennettekeschouten lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT hovingeelco lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT vantilburgcornelism lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT pfisterstefanm lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT wittolaf lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT jonesdavidtw lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures
AT mildetill lgg26predictingmapkinhibitorsensitivityinpediatriclowgradegliomaswithnovelgeneexpressionderivedsignatures