Cargando…

Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion

BACKGROUND: The mitogen-activated protein kinase (MAPK) signaling pathway is frequently hyperactivated in malignant melanoma and its inhibition has proved to be an efficient treatment option for cases harboring BRAF(V600) mutations (BRAF(Mut)). However, there is still a significant need for effectiv...

Descripción completa

Detalles Bibliográficos
Autores principales: Kosnopfel, Corinna, Wendlinger, Simone, Niessner, Heike, Siewert, Johannes, Sinnberg, Tobias, Hofmann, Angelika, Wohlfarth, Jonas, Schrama, David, Berthold, Marion, Siedel, Claudia, Sauer, Birgit, Jayanthan, Aarthi, Lenz, Georg, Dunn, Sandra E., Schilling, Bastian, Schittek, Birgit
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10354913/
https://www.ncbi.nlm.nih.gov/pubmed/37464364
http://dx.doi.org/10.1186/s13046-023-02755-5
_version_ 1785075024353296384
author Kosnopfel, Corinna
Wendlinger, Simone
Niessner, Heike
Siewert, Johannes
Sinnberg, Tobias
Hofmann, Angelika
Wohlfarth, Jonas
Schrama, David
Berthold, Marion
Siedel, Claudia
Sauer, Birgit
Jayanthan, Aarthi
Lenz, Georg
Dunn, Sandra E.
Schilling, Bastian
Schittek, Birgit
author_facet Kosnopfel, Corinna
Wendlinger, Simone
Niessner, Heike
Siewert, Johannes
Sinnberg, Tobias
Hofmann, Angelika
Wohlfarth, Jonas
Schrama, David
Berthold, Marion
Siedel, Claudia
Sauer, Birgit
Jayanthan, Aarthi
Lenz, Georg
Dunn, Sandra E.
Schilling, Bastian
Schittek, Birgit
author_sort Kosnopfel, Corinna
collection PubMed
description BACKGROUND: The mitogen-activated protein kinase (MAPK) signaling pathway is frequently hyperactivated in malignant melanoma and its inhibition has proved to be an efficient treatment option for cases harboring BRAF(V600) mutations (BRAF(Mut)). However, there is still a significant need for effective targeted therapies for patients with other melanoma subgroups characterized by constitutive MAPK activation, such as tumors with NRAS or NF-1 alterations (NRAS(Mut), NF-1(LOF)), as well as for patients with MAPK pathway inhibitor-resistant BRAF(Mut) melanomas, which commonly exhibit a reactivation of this pathway. p90 ribosomal S6 kinases (RSKs) represent central effectors of MAPK signaling, regulating cell cycle progression and survival. METHODS: RSK activity and the functional effects of its inhibition by specific small molecule inhibitors were investigated in established melanoma cell lines and patient-derived short-term cultures from different MAPK pathway-hyperactivated genomic subgroups (NRAS(Mut), BRAF(Mut), NF-1(LOF)). Real-time qPCR, immunoblots and flow cytometric cell surface staining were used to explore the molecular changes following RSK inhibition. The effect on melanoma cell growth was evaluated by various two- and three-dimensional in vitro assays as well as with melanoma xenograft mouse models. Co-cultures with gp100- or Melan-A-specific cytotoxic T cells were used to assess immunogenicity of melanoma cells and associated T-cell responses. RESULTS: In line with elevated activity of the MAPK/RSK signaling axis, growth and survival of not only BRAF(Mut) but also NRAS(Mut) and NF-1(LOF) melanoma cells were significantly impaired by RSK inhibitors. Intriguingly, RSK inhibition was particularly effective in three-dimensional growth settings with long-term chronic drug exposure and suppressed tumor cell growth of in vivo melanoma models. Additionally, our study revealed that RSK inhibition simultaneously promoted differentiation and immunogenicity of the tumor cells leading to enhanced T-cell activation and melanoma cell killing. CONCLUSIONS: Collectively, RSK inhibitors exhibited both multi-layered anti-tumor efficacy and broad applicability across different genomic melanoma subgroups. RSK inhibition may therefore represent a promising novel therapeutic strategy for malignant melanoma with hyperactivated MAPK signaling. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02755-5.
format Online
Article
Text
id pubmed-10354913
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103549132023-07-20 Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion Kosnopfel, Corinna Wendlinger, Simone Niessner, Heike Siewert, Johannes Sinnberg, Tobias Hofmann, Angelika Wohlfarth, Jonas Schrama, David Berthold, Marion Siedel, Claudia Sauer, Birgit Jayanthan, Aarthi Lenz, Georg Dunn, Sandra E. Schilling, Bastian Schittek, Birgit J Exp Clin Cancer Res Research BACKGROUND: The mitogen-activated protein kinase (MAPK) signaling pathway is frequently hyperactivated in malignant melanoma and its inhibition has proved to be an efficient treatment option for cases harboring BRAF(V600) mutations (BRAF(Mut)). However, there is still a significant need for effective targeted therapies for patients with other melanoma subgroups characterized by constitutive MAPK activation, such as tumors with NRAS or NF-1 alterations (NRAS(Mut), NF-1(LOF)), as well as for patients with MAPK pathway inhibitor-resistant BRAF(Mut) melanomas, which commonly exhibit a reactivation of this pathway. p90 ribosomal S6 kinases (RSKs) represent central effectors of MAPK signaling, regulating cell cycle progression and survival. METHODS: RSK activity and the functional effects of its inhibition by specific small molecule inhibitors were investigated in established melanoma cell lines and patient-derived short-term cultures from different MAPK pathway-hyperactivated genomic subgroups (NRAS(Mut), BRAF(Mut), NF-1(LOF)). Real-time qPCR, immunoblots and flow cytometric cell surface staining were used to explore the molecular changes following RSK inhibition. The effect on melanoma cell growth was evaluated by various two- and three-dimensional in vitro assays as well as with melanoma xenograft mouse models. Co-cultures with gp100- or Melan-A-specific cytotoxic T cells were used to assess immunogenicity of melanoma cells and associated T-cell responses. RESULTS: In line with elevated activity of the MAPK/RSK signaling axis, growth and survival of not only BRAF(Mut) but also NRAS(Mut) and NF-1(LOF) melanoma cells were significantly impaired by RSK inhibitors. Intriguingly, RSK inhibition was particularly effective in three-dimensional growth settings with long-term chronic drug exposure and suppressed tumor cell growth of in vivo melanoma models. Additionally, our study revealed that RSK inhibition simultaneously promoted differentiation and immunogenicity of the tumor cells leading to enhanced T-cell activation and melanoma cell killing. CONCLUSIONS: Collectively, RSK inhibitors exhibited both multi-layered anti-tumor efficacy and broad applicability across different genomic melanoma subgroups. RSK inhibition may therefore represent a promising novel therapeutic strategy for malignant melanoma with hyperactivated MAPK signaling. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02755-5. BioMed Central 2023-07-19 /pmc/articles/PMC10354913/ /pubmed/37464364 http://dx.doi.org/10.1186/s13046-023-02755-5 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This 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/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://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 Research
Kosnopfel, Corinna
Wendlinger, Simone
Niessner, Heike
Siewert, Johannes
Sinnberg, Tobias
Hofmann, Angelika
Wohlfarth, Jonas
Schrama, David
Berthold, Marion
Siedel, Claudia
Sauer, Birgit
Jayanthan, Aarthi
Lenz, Georg
Dunn, Sandra E.
Schilling, Bastian
Schittek, Birgit
Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion
title Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion
title_full Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion
title_fullStr Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion
title_full_unstemmed Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion
title_short Inhibition of p90 ribosomal S6 kinases disrupts melanoma cell growth and immune evasion
title_sort inhibition of p90 ribosomal s6 kinases disrupts melanoma cell growth and immune evasion
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10354913/
https://www.ncbi.nlm.nih.gov/pubmed/37464364
http://dx.doi.org/10.1186/s13046-023-02755-5
work_keys_str_mv AT kosnopfelcorinna inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT wendlingersimone inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT niessnerheike inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT siewertjohannes inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT sinnbergtobias inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT hofmannangelika inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT wohlfarthjonas inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT schramadavid inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT bertholdmarion inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT siedelclaudia inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT sauerbirgit inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT jayanthanaarthi inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT lenzgeorg inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT dunnsandrae inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT schillingbastian inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion
AT schittekbirgit inhibitionofp90ribosomals6kinasesdisruptsmelanomacellgrowthandimmuneevasion