Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis

Tyrosine kinase inhibitors (TKIs) are anticancer drugs with a lesser toxicity than classical chemotherapeutic agents but still with a narrow therapeutic window. While hepatotoxicity is known for most TKIs, underlying mechanisms remain mostly unclear. We therefore aimed at investigating mechanisms of...

Full description

Bibliographic Details
Main Authors: Franziska Paech, Jamal Bouitbir, Stephan Krähenbühl
Format: Article
Language:English
Published: Frontiers Media S.A. 2017-06-01
Series:Frontiers in Pharmacology
Subjects:
ROS
Online Access:http://journal.frontiersin.org/article/10.3389/fphar.2017.00367/full
id doaj-0b10a7289b494c778ece0d1ba486b805
record_format Article
spelling doaj-0b10a7289b494c778ece0d1ba486b8052020-11-24T22:47:14ZengFrontiers Media S.A.Frontiers in Pharmacology1663-98122017-06-01810.3389/fphar.2017.00367239357Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of GlycolysisFranziska Paech0Franziska Paech1Jamal Bouitbir2Jamal Bouitbir3Jamal Bouitbir4Stephan Krähenbühl5Stephan Krähenbühl6Stephan Krähenbühl7Division of Clinical Pharmacology and Toxicology, University Hospital BaselBasel, SwitzerlandDepartment of Biomedicine, University of BaselBasel, SwitzerlandDivision of Clinical Pharmacology and Toxicology, University Hospital BaselBasel, SwitzerlandDepartment of Biomedicine, University of BaselBasel, SwitzerlandSwiss Centre of Applied Human ToxicologyBasel, SwitzerlandDivision of Clinical Pharmacology and Toxicology, University Hospital BaselBasel, SwitzerlandDepartment of Biomedicine, University of BaselBasel, SwitzerlandSwiss Centre of Applied Human ToxicologyBasel, SwitzerlandTyrosine kinase inhibitors (TKIs) are anticancer drugs with a lesser toxicity than classical chemotherapeutic agents but still with a narrow therapeutic window. While hepatotoxicity is known for most TKIs, underlying mechanisms remain mostly unclear. We therefore aimed at investigating mechanisms of hepatotoxicity for imatinib, sunitinib, lapatinib and erlotinib in vitro. We treated HepG2 cells, HepaRG cells and mouse liver mitochondria with TKIs (concentrations 1–100 μM) for different periods of time and assessed toxicity. In HepG2 cells maintained with glucose (favoring glycolysis), all TKIs showed a time- and concentration-dependent cytotoxicity and, except erlotinib, a drop in intracellular ATP. In the presence of galactose (favoring mitochondrial metabolism), imatinib, sunitinib and erlotinib showed a similar toxicity profile as for glucose whereas lapatinib was less toxic. For imatinib, lapatinib and sunitinib, cytotoxicity increased in HepaRG cells induced with rifampicin, suggesting formation of toxic metabolites. In contrast, erlotinib was more toxic in HepaRG cells under basal than CYP-induced conditions. Imatinib, sunitinib and lapatinib reduced the mitochondrial membrane potential in HepG2 cells and in mouse liver mitochondria. In HepG2 cells, these compounds increased reactive oxygen species production, impaired glycolysis, and induced apoptosis. In addition, imatinib and sunitinib impaired oxygen consumption and activities of complex I and III (only imatinib), and reduced the cellular GSH pool. In conclusion, imatinib and sunitinib are mitochondrial toxicants after acute and long-term exposure and inhibit glycolysis. Lapatinib affected mitochondria only weakly and inhibited glycolysis, whereas the cytotoxicity of erlotinib could not be explained by a mitochondrial mechanism.http://journal.frontiersin.org/article/10.3389/fphar.2017.00367/fulltyrosine kinase inhibitorhepatotoxicitymitochondrial toxicityglycolysisROSapoptosis
collection DOAJ
language English
format Article
sources DOAJ
author Franziska Paech
Franziska Paech
Jamal Bouitbir
Jamal Bouitbir
Jamal Bouitbir
Stephan Krähenbühl
Stephan Krähenbühl
Stephan Krähenbühl
spellingShingle Franziska Paech
Franziska Paech
Jamal Bouitbir
Jamal Bouitbir
Jamal Bouitbir
Stephan Krähenbühl
Stephan Krähenbühl
Stephan Krähenbühl
Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis
Frontiers in Pharmacology
tyrosine kinase inhibitor
hepatotoxicity
mitochondrial toxicity
glycolysis
ROS
apoptosis
author_facet Franziska Paech
Franziska Paech
Jamal Bouitbir
Jamal Bouitbir
Jamal Bouitbir
Stephan Krähenbühl
Stephan Krähenbühl
Stephan Krähenbühl
author_sort Franziska Paech
title Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis
title_short Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis
title_full Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis
title_fullStr Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis
title_full_unstemmed Hepatocellular Toxicity Associated with Tyrosine Kinase Inhibitors: Mitochondrial Damage and Inhibition of Glycolysis
title_sort hepatocellular toxicity associated with tyrosine kinase inhibitors: mitochondrial damage and inhibition of glycolysis
publisher Frontiers Media S.A.
series Frontiers in Pharmacology
issn 1663-9812
publishDate 2017-06-01
description Tyrosine kinase inhibitors (TKIs) are anticancer drugs with a lesser toxicity than classical chemotherapeutic agents but still with a narrow therapeutic window. While hepatotoxicity is known for most TKIs, underlying mechanisms remain mostly unclear. We therefore aimed at investigating mechanisms of hepatotoxicity for imatinib, sunitinib, lapatinib and erlotinib in vitro. We treated HepG2 cells, HepaRG cells and mouse liver mitochondria with TKIs (concentrations 1–100 μM) for different periods of time and assessed toxicity. In HepG2 cells maintained with glucose (favoring glycolysis), all TKIs showed a time- and concentration-dependent cytotoxicity and, except erlotinib, a drop in intracellular ATP. In the presence of galactose (favoring mitochondrial metabolism), imatinib, sunitinib and erlotinib showed a similar toxicity profile as for glucose whereas lapatinib was less toxic. For imatinib, lapatinib and sunitinib, cytotoxicity increased in HepaRG cells induced with rifampicin, suggesting formation of toxic metabolites. In contrast, erlotinib was more toxic in HepaRG cells under basal than CYP-induced conditions. Imatinib, sunitinib and lapatinib reduced the mitochondrial membrane potential in HepG2 cells and in mouse liver mitochondria. In HepG2 cells, these compounds increased reactive oxygen species production, impaired glycolysis, and induced apoptosis. In addition, imatinib and sunitinib impaired oxygen consumption and activities of complex I and III (only imatinib), and reduced the cellular GSH pool. In conclusion, imatinib and sunitinib are mitochondrial toxicants after acute and long-term exposure and inhibit glycolysis. Lapatinib affected mitochondria only weakly and inhibited glycolysis, whereas the cytotoxicity of erlotinib could not be explained by a mitochondrial mechanism.
topic tyrosine kinase inhibitor
hepatotoxicity
mitochondrial toxicity
glycolysis
ROS
apoptosis
url http://journal.frontiersin.org/article/10.3389/fphar.2017.00367/full
work_keys_str_mv AT franziskapaech hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT franziskapaech hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT jamalbouitbir hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT jamalbouitbir hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT jamalbouitbir hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT stephankrahenbuhl hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT stephankrahenbuhl hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
AT stephankrahenbuhl hepatocellulartoxicityassociatedwithtyrosinekinaseinhibitorsmitochondrialdamageandinhibitionofglycolysis
_version_ 1725682360099274752