Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer

Abstract Background Studying the intracellular distribution of pharmacological agents, including anticancer compounds, is of central importance in biomedical research. It constitutes a prerequisite for a better understanding of the molecular mechanisms underlying drug action and resistance developme...

Full description

Bibliographic Details
Main Authors: Bernhard Englinger, Sebastian Kallus, Julia Senkiv, Daniela Heilos, Lisa Gabler, Sushilla van Schoonhoven, Alessio Terenzi, Patrick Moser, Christine Pirker, Gerald Timelthaler, Walter Jäger, Christian R. Kowol, Petra Heffeter, Michael Grusch, Walter Berger
Format: Article
Language:English
Published: BMC 2017-09-01
Series:Journal of Experimental & Clinical Cancer Research
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13046-017-0592-3
id doaj-ab5f3166b8d14e12903480ffb0754129
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Bernhard Englinger
Sebastian Kallus
Julia Senkiv
Daniela Heilos
Lisa Gabler
Sushilla van Schoonhoven
Alessio Terenzi
Patrick Moser
Christine Pirker
Gerald Timelthaler
Walter Jäger
Christian R. Kowol
Petra Heffeter
Michael Grusch
Walter Berger
spellingShingle Bernhard Englinger
Sebastian Kallus
Julia Senkiv
Daniela Heilos
Lisa Gabler
Sushilla van Schoonhoven
Alessio Terenzi
Patrick Moser
Christine Pirker
Gerald Timelthaler
Walter Jäger
Christian R. Kowol
Petra Heffeter
Michael Grusch
Walter Berger
Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer
Journal of Experimental & Clinical Cancer Research
FGFR1
Nintedanib
Fluorescence
Lysosomes
Resistance
author_facet Bernhard Englinger
Sebastian Kallus
Julia Senkiv
Daniela Heilos
Lisa Gabler
Sushilla van Schoonhoven
Alessio Terenzi
Patrick Moser
Christine Pirker
Gerald Timelthaler
Walter Jäger
Christian R. Kowol
Petra Heffeter
Michael Grusch
Walter Berger
author_sort Bernhard Englinger
title Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer
title_short Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer
title_full Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer
title_fullStr Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer
title_full_unstemmed Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancer
title_sort intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in fgfr-driven lung cancer
publisher BMC
series Journal of Experimental & Clinical Cancer Research
issn 1756-9966
publishDate 2017-09-01
description Abstract Background Studying the intracellular distribution of pharmacological agents, including anticancer compounds, is of central importance in biomedical research. It constitutes a prerequisite for a better understanding of the molecular mechanisms underlying drug action and resistance development. Hyperactivated fibroblast growth factor receptors (FGFRs) constitute a promising therapy target in several types of malignancies including lung cancer. The clinically approved small-molecule FGFR inhibitor nintedanib exerts strong cytotoxicity in FGFR-driven lung cancer cells. However, subcellular pharmacokinetics of this compound and its impact on therapeutic efficacy remain obscure. Methods 3-dimensional fluorescence spectroscopy was conducted to asses cell-free nintedanib fluorescence properties. MTT assay was used to determine the impact of the lysosome-targeting agents bafilomycin A1 and chloroquine combined with nintedanib on lung cancer cell viability. Flow cytometry and live cell as well as confocal microscopy were performed to analyze uptake kinetics as well as subcellular distribution of nintedanib. Western blot was conducted to investigate protein expression. Cryosections of subcutaneous tumor allografts were generated to detect intratumoral nintedanib in mice after oral drug administration. Results Here, we report for the first time drug-intrinsic fluorescence properties of nintedanib in living and fixed cancer cells as well as in cryosections derived from allograft tumors of orally treated mice. Using this feature in conjunction with flow cytometry and confocal microscopy allowed to determine cellular drug accumulation levels, impact of the ABCB1 efflux pump and to uncover nintedanib trapping into lysosomes. Lysosomal sequestration - resulting in an organelle-specific and pH-dependent nintedanib fluorescence - was identified as an intrinsic resistance mechanism in FGFR-driven lung cancer cells. Accordingly, combination of nintedanib with agents compromising lysosomal acidification (bafilomycin A1, chloroquine) exerted distinctly synergistic growth inhibitory effects. Conclusion Our findings provide a powerful tool to dissect molecular factors impacting organismal and intracellular pharmacokinetics of nintedanib. Regarding clinical application, prevention of lysosomal trapping via lysosome-alkalization might represent a promising strategy to circumvent cancer cell-intrinsic nintedanib resistance.
topic FGFR1
Nintedanib
Fluorescence
Lysosomes
Resistance
url http://link.springer.com/article/10.1186/s13046-017-0592-3
work_keys_str_mv AT bernhardenglinger intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT sebastiankallus intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT juliasenkiv intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT danielaheilos intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT lisagabler intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT sushillavanschoonhoven intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT alessioterenzi intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT patrickmoser intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT christinepirker intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT geraldtimelthaler intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT walterjager intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT christianrkowol intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT petraheffeter intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT michaelgrusch intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
AT walterberger intrinsicfluorescenceoftheclinicallyapprovedmultikinaseinhibitornintedanibrevealslysosomalsequestrationasresistancemechanisminfgfrdrivenlungcancer
_version_ 1726006620608004096
spelling doaj-ab5f3166b8d14e12903480ffb07541292020-11-24T21:19:11ZengBMCJournal of Experimental & Clinical Cancer Research1756-99662017-09-0136111310.1186/s13046-017-0592-3Intrinsic fluorescence of the clinically approved multikinase inhibitor nintedanib reveals lysosomal sequestration as resistance mechanism in FGFR-driven lung cancerBernhard Englinger0Sebastian Kallus1Julia Senkiv2Daniela Heilos3Lisa Gabler4Sushilla van Schoonhoven5Alessio Terenzi6Patrick Moser7Christine Pirker8Gerald Timelthaler9Walter Jäger10Christian R. Kowol11Petra Heffeter12Michael Grusch13Walter Berger14Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Inorganic Chemistry, University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Inorganic Chemistry, University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaDepartment of Pharmaceutical Chemistry, Division of Clinical Pharmacy and Diagnostics, University of ViennaInstitute of Inorganic Chemistry, University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaInstitute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of ViennaAbstract Background Studying the intracellular distribution of pharmacological agents, including anticancer compounds, is of central importance in biomedical research. It constitutes a prerequisite for a better understanding of the molecular mechanisms underlying drug action and resistance development. Hyperactivated fibroblast growth factor receptors (FGFRs) constitute a promising therapy target in several types of malignancies including lung cancer. The clinically approved small-molecule FGFR inhibitor nintedanib exerts strong cytotoxicity in FGFR-driven lung cancer cells. However, subcellular pharmacokinetics of this compound and its impact on therapeutic efficacy remain obscure. Methods 3-dimensional fluorescence spectroscopy was conducted to asses cell-free nintedanib fluorescence properties. MTT assay was used to determine the impact of the lysosome-targeting agents bafilomycin A1 and chloroquine combined with nintedanib on lung cancer cell viability. Flow cytometry and live cell as well as confocal microscopy were performed to analyze uptake kinetics as well as subcellular distribution of nintedanib. Western blot was conducted to investigate protein expression. Cryosections of subcutaneous tumor allografts were generated to detect intratumoral nintedanib in mice after oral drug administration. Results Here, we report for the first time drug-intrinsic fluorescence properties of nintedanib in living and fixed cancer cells as well as in cryosections derived from allograft tumors of orally treated mice. Using this feature in conjunction with flow cytometry and confocal microscopy allowed to determine cellular drug accumulation levels, impact of the ABCB1 efflux pump and to uncover nintedanib trapping into lysosomes. Lysosomal sequestration - resulting in an organelle-specific and pH-dependent nintedanib fluorescence - was identified as an intrinsic resistance mechanism in FGFR-driven lung cancer cells. Accordingly, combination of nintedanib with agents compromising lysosomal acidification (bafilomycin A1, chloroquine) exerted distinctly synergistic growth inhibitory effects. Conclusion Our findings provide a powerful tool to dissect molecular factors impacting organismal and intracellular pharmacokinetics of nintedanib. Regarding clinical application, prevention of lysosomal trapping via lysosome-alkalization might represent a promising strategy to circumvent cancer cell-intrinsic nintedanib resistance.http://link.springer.com/article/10.1186/s13046-017-0592-3FGFR1NintedanibFluorescenceLysosomesResistance