The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress

Platinum chemotherapies are highly effective cytotoxic agents but often induce resistance when used as monotherapies. Combinatorial strategies limit this risk and provide effective treatment options for many cancers. Here, we repurpose atovaquone (ATQ), a well-tolerated & FDA-approved anti-malar...

Full description

Bibliographic Details
Main Authors: Coates, JTT, Rodriguez-Berriguete, G, Puliyadi, R, Ashton, T, Prevo, R, Wing, A, Granata, G, Pirovano, G, McKenna, GW, Higgins, GS
Format: Journal article
Language:English
Published: Springer Nature 2020
Subjects:
_version_ 1797088940159664128
author Coates, JTT
Rodriguez-Berriguete, G
Puliyadi, R
Ashton, T
Prevo, R
Wing, A
Granata, G
Pirovano, G
McKenna, GW
Higgins, GS
author_facet Coates, JTT
Rodriguez-Berriguete, G
Puliyadi, R
Ashton, T
Prevo, R
Wing, A
Granata, G
Pirovano, G
McKenna, GW
Higgins, GS
author_sort Coates, JTT
collection OXFORD
description Platinum chemotherapies are highly effective cytotoxic agents but often induce resistance when used as monotherapies. Combinatorial strategies limit this risk and provide effective treatment options for many cancers. Here, we repurpose atovaquone (ATQ), a well-tolerated & FDA-approved anti-malarial agent by demonstrating that it potentiates cancer cell death of a subset of platinums. We show that ATQ in combination with carboplatin or cisplatin induces striking and repeatable concentration- and time-dependent cell death sensitization in vitro across a variety of cancer cell lines. ATQ induces mitochondrial reactive oxygen species (mROS), depleting intracellular glutathione (GSH) pools in a concentration-dependent manner. The superoxide dismutase mimetic MnTBAP rescues ATQ-induced mROS production and pre-loading cells with the GSH prodrug N-acetyl cysteine (NAC) abrogates the sensitization. Together, these findings implicate ATQ-induced oxidative stress as key mediator of the sensitizing effect. At physiologically achievable concentrations, ATQ and carboplatin furthermore synergistically delay the growth of three-dimensional avascular spheroids. Clinically, ATQ is a safe and specific inhibitor of the electron transport chain (ETC) and is concurrently being repurposed as a candidate tumor hypoxia modifier. Together, these findings suggest that ATQ is deserving of further study as a candidate platinum sensitizing agent.
first_indexed 2024-03-07T02:57:18Z
format Journal article
id oxford-uuid:afc0f6e2-3b40-4bae-8b58-0269a742ed16
institution University of Oxford
language English
last_indexed 2024-03-07T02:57:18Z
publishDate 2020
publisher Springer Nature
record_format dspace
spelling oxford-uuid:afc0f6e2-3b40-4bae-8b58-0269a742ed162022-03-27T03:51:39ZThe anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stressJournal articlehttp://purl.org/coar/resource_type/c_dcae04bcuuid:afc0f6e2-3b40-4bae-8b58-0269a742ed16ChemotherapyPreclinical researchEnglishSymplectic ElementsSpringer Nature 2020Coates, JTTRodriguez-Berriguete, GPuliyadi, RAshton, TPrevo, RWing, AGranata, GPirovano, GMcKenna, GWHiggins, GSPlatinum chemotherapies are highly effective cytotoxic agents but often induce resistance when used as monotherapies. Combinatorial strategies limit this risk and provide effective treatment options for many cancers. Here, we repurpose atovaquone (ATQ), a well-tolerated & FDA-approved anti-malarial agent by demonstrating that it potentiates cancer cell death of a subset of platinums. We show that ATQ in combination with carboplatin or cisplatin induces striking and repeatable concentration- and time-dependent cell death sensitization in vitro across a variety of cancer cell lines. ATQ induces mitochondrial reactive oxygen species (mROS), depleting intracellular glutathione (GSH) pools in a concentration-dependent manner. The superoxide dismutase mimetic MnTBAP rescues ATQ-induced mROS production and pre-loading cells with the GSH prodrug N-acetyl cysteine (NAC) abrogates the sensitization. Together, these findings implicate ATQ-induced oxidative stress as key mediator of the sensitizing effect. At physiologically achievable concentrations, ATQ and carboplatin furthermore synergistically delay the growth of three-dimensional avascular spheroids. Clinically, ATQ is a safe and specific inhibitor of the electron transport chain (ETC) and is concurrently being repurposed as a candidate tumor hypoxia modifier. Together, these findings suggest that ATQ is deserving of further study as a candidate platinum sensitizing agent.
spellingShingle Chemotherapy
Preclinical research
Coates, JTT
Rodriguez-Berriguete, G
Puliyadi, R
Ashton, T
Prevo, R
Wing, A
Granata, G
Pirovano, G
McKenna, GW
Higgins, GS
The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress
title The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress
title_full The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress
title_fullStr The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress
title_full_unstemmed The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress
title_short The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress
title_sort anti malarial drug atovaquone potentiates platinum mediated cancer cell death by increasing oxidative stress
topic Chemotherapy
Preclinical research
work_keys_str_mv AT coatesjtt theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT rodriguezberrigueteg theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT puliyadir theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT ashtont theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT prevor theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT winga theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT granatag theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT pirovanog theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT mckennagw theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT higginsgs theantimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT coatesjtt antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT rodriguezberrigueteg antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT puliyadir antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT ashtont antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT prevor antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT winga antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT granatag antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT pirovanog antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT mckennagw antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress
AT higginsgs antimalarialdrugatovaquonepotentiatesplatinummediatedcancercelldeathbyincreasingoxidativestress