Cargando…

Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization

ABSTRACT: Target therapies based on BRAF and MEK inhibitors (MAPKi) have changed the therapeutic landscape for metastatic melanoma patients bearing mutations in the BRAF kinase. However, the emergence of drug resistance imposes the necessity to conceive novel therapeutic strategies capable to achiev...

Descripción completa

Detalles Bibliográficos
Autores principales: Fattore, Luigi, Malpicci, Debora, Milite, Ciro, Castellano, Sabrina, Sbardella, Gianluca, Botti, Gerardo, Ascierto, Paolo A., Mancini, Rita, Ciliberto, Gennaro
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7493390/
https://www.ncbi.nlm.nih.gov/pubmed/32933538
http://dx.doi.org/10.1186/s12964-020-00633-7
_version_ 1783582558928240640
author Fattore, Luigi
Malpicci, Debora
Milite, Ciro
Castellano, Sabrina
Sbardella, Gianluca
Botti, Gerardo
Ascierto, Paolo A.
Mancini, Rita
Ciliberto, Gennaro
author_facet Fattore, Luigi
Malpicci, Debora
Milite, Ciro
Castellano, Sabrina
Sbardella, Gianluca
Botti, Gerardo
Ascierto, Paolo A.
Mancini, Rita
Ciliberto, Gennaro
author_sort Fattore, Luigi
collection PubMed
description ABSTRACT: Target therapies based on BRAF and MEK inhibitors (MAPKi) have changed the therapeutic landscape for metastatic melanoma patients bearing mutations in the BRAF kinase. However, the emergence of drug resistance imposes the necessity to conceive novel therapeutic strategies capable to achieve a more durable disease control. In the last years, retrotransposons laying in human genome have been shown to undergo activation during tumorigenesis, where they contribute to genomic instability. Their activation can be efficiently controlled with reverse transcriptase inhibitors (RTIs) frequently used in the treatment of AIDS. These drugs have demonstrated anti-proliferative effects in several cancer models, including also metastatic melanoma. However, to our knowledge no previous study investigated the capability of RTIs to mitigate drug resistance to target therapy in BRAF-mutant melanomas. In this short report we show that the non-nucleoside RTI, SPV122 in combination with MAPKi strongly inhibits BRAF-mutant melanoma cell growth, induces apoptosis, and delays the emergence of resistance to target therapy in vitro. Mechanistically, this combination strongly induces DNA double-strand breaks, mitochondrial membrane depolarization and increased ROS levels. Our results shed further light on the molecular activity of RTI in melanoma and pave the way to their use as a novel therapeutic option to improve the efficacy of target therapy. GRAPHICAL ABSTRACT: [Image: see text]
format Online
Article
Text
id pubmed-7493390
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-74933902020-09-16 Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization Fattore, Luigi Malpicci, Debora Milite, Ciro Castellano, Sabrina Sbardella, Gianluca Botti, Gerardo Ascierto, Paolo A. Mancini, Rita Ciliberto, Gennaro Cell Commun Signal Short Report ABSTRACT: Target therapies based on BRAF and MEK inhibitors (MAPKi) have changed the therapeutic landscape for metastatic melanoma patients bearing mutations in the BRAF kinase. However, the emergence of drug resistance imposes the necessity to conceive novel therapeutic strategies capable to achieve a more durable disease control. In the last years, retrotransposons laying in human genome have been shown to undergo activation during tumorigenesis, where they contribute to genomic instability. Their activation can be efficiently controlled with reverse transcriptase inhibitors (RTIs) frequently used in the treatment of AIDS. These drugs have demonstrated anti-proliferative effects in several cancer models, including also metastatic melanoma. However, to our knowledge no previous study investigated the capability of RTIs to mitigate drug resistance to target therapy in BRAF-mutant melanomas. In this short report we show that the non-nucleoside RTI, SPV122 in combination with MAPKi strongly inhibits BRAF-mutant melanoma cell growth, induces apoptosis, and delays the emergence of resistance to target therapy in vitro. Mechanistically, this combination strongly induces DNA double-strand breaks, mitochondrial membrane depolarization and increased ROS levels. Our results shed further light on the molecular activity of RTI in melanoma and pave the way to their use as a novel therapeutic option to improve the efficacy of target therapy. GRAPHICAL ABSTRACT: [Image: see text] BioMed Central 2020-09-15 /pmc/articles/PMC7493390/ /pubmed/32933538 http://dx.doi.org/10.1186/s12964-020-00633-7 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Short Report
Fattore, Luigi
Malpicci, Debora
Milite, Ciro
Castellano, Sabrina
Sbardella, Gianluca
Botti, Gerardo
Ascierto, Paolo A.
Mancini, Rita
Ciliberto, Gennaro
Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization
title Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization
title_full Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization
title_fullStr Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization
title_full_unstemmed Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization
title_short Reverse transcriptase inhibition potentiates target therapy in BRAF-mutant melanomas: effects on cell proliferation, apoptosis, DNA-damage, ROS induction and mitochondrial membrane depolarization
title_sort reverse transcriptase inhibition potentiates target therapy in braf-mutant melanomas: effects on cell proliferation, apoptosis, dna-damage, ros induction and mitochondrial membrane depolarization
topic Short Report
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7493390/
https://www.ncbi.nlm.nih.gov/pubmed/32933538
http://dx.doi.org/10.1186/s12964-020-00633-7
work_keys_str_mv AT fattoreluigi reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT malpiccidebora reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT militeciro reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT castellanosabrina reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT sbardellagianluca reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT bottigerardo reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT asciertopaoloa reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT mancinirita reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization
AT cilibertogennaro reversetranscriptaseinhibitionpotentiatestargettherapyinbrafmutantmelanomaseffectsoncellproliferationapoptosisdnadamagerosinductionandmitochondrialmembranedepolarization