108 results on '"Wellbrock C"'
Search Results
2. Judging A Magazine by Its Cover – A Conceptual Framework to Understand Sales Through Content and Design Interaction
- Author
-
Püchel, Lea, primary and Wellbrock, C.-M., additional
- Published
- 2022
- Full Text
- View/download PDF
3. Combination of MEK and SRC inhibition suppresses melanoma cell growth and invasion
- Author
-
Ferguson, J, Arozarena, I, Ehrhardt, M, and Wellbrock, C
- Published
- 2013
- Full Text
- View/download PDF
4. In melanoma, beta-catenin is a suppressor of invasion
- Author
-
Arozarena, I, Bischof, H, Gilby, D, Belloni, B, Dummer, R, and Wellbrock, C
- Published
- 2011
- Full Text
- View/download PDF
5. OC-042 - IDENTIFICACIÓN DE UN MECANISMO DE RADIOPROTECCIÓN MEDIADO POR DEXAMETASONA QUE MUESTRA NUEVAS VULNERABILIDADES TERAPÉUTICAS EN EL GLIOBLASTOMA
- Author
-
Zazpe, I., Aldaz, P., Auzmendi, J., Lasheras, I., Sempron, N., Armendariz, M., Carrasco, E., Olias, A., Morilla, I., Redondo, M., Squatrito, M., Wellbrock, C., Matheu, A., and Arozarena, I.
- Published
- 2023
- Full Text
- View/download PDF
6. Judging A Magazine by Its Cover – A Conceptual Framework to Understand Sales Through Content and Design Interaction.
- Author
-
Püchel, Lea and Wellbrock, C.-M.
- Subjects
- *
MAGAZINE covers , *CONSUMER attitudes , *HEDONISTIC consumption , *CONTENT analysis , *MEDIA consumption , *MAGAZINE design - Abstract
Content Design is advocated as one of the key factors influencing consumers' attitude and buying behavior. However, its real effects on sales performance remain largely unknown. We propose a research framework to evaluate the effect of content design on sales and empirically test its employability within the German magazine retail sector. To develop the research framework, we first derive a set of related success factors from several literature streams. We then conduct a content analysis of over 500 magazine covers and relate this data to real-life retail sales figures via linear mixed model analysis to investigate which aspects of content design affect sales. The findings point toward the usefulness of our proposed framework. They further indicate that in hedonic media consumption, the colors purple and blue, text-image congruence, wording that leads to ease of cognitive processing and promotional activities are pivotal in sales. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. BRAF, AN ONCOGENE AND THERAPEUTIC TARGET IN MELANOMA
- Author
-
Marais, R., Garnett, M., Wellbrock, C., Karasarides, M., Wan, P., Roe, M., Niculescu-Duvaz, D., Barford, D., and Springer, C.
- Published
- 2004
8. Report from the II Melanoma Translational Meeting of the Spanish Melanoma Group (GEM)
- Author
-
Gonzalez-Cao, M, Berrocal, A, Puig, S, Karachaliou, N, De Matos-Arruda, L, Seoane, J, Escors, D, Alvarez, C, Vaque, JP, Prat, A, Wellbrock, C, Arozarena, I, Marquez-Rodas, I, Espinosa, E, Molina, MA, Puertolas, T, Juan-Otero, M, Malagrida, R, Jantus-Lewintre, E, Soriano, V, Arance, A, Manzano, JL, Lorigan, P, Gajewski, TF, Rosell, R, and Martin-Algarra, S
- Subjects
medicine.medical_specialty ,Medical education ,Research groups ,business.industry ,translational ,Translational research ,General Medicine ,meeting ,Meeting Report ,Spanish ,Preclinical data ,Basic research ,Ophthalmology ,medicine ,business ,Melanoma - Abstract
The II melanoma translational meeting of the Spanish Melanoma Group (GEM) was held in Barcelona, Spain, at the Hospital Universitari Quiron-Dexeus on November 3 rd , 2016. The conference featured leaders in the fields of oncology, immunology and dermatology, all working at both national and international levels on basic research with direct applications to the clinical setting. The objective was to present cutting edge research on melanoma, mainly from Spanish research groups, but also from some international groups, with the aim of generating a network for future national and international collaborations. During a single day, fifteen speakers, seven biologists and eight clinicians, with a special focus on translational research, outlined the main findings of their ongoing studies. The moderators in every session were recognized GEM oncologists who guided discussion from the clinical point of view regarding the preclinical data presented. Herein, we summarize the main topics discussed during the meeting.
- Published
- 2017
9. The role of tumour heterogeneity in melanoma progression
- Author
-
Rowling, E., primary, Chapman, A., additional, Telfer, B., additional, Hurlstone, A., additional, and Wellbrock, C., additional
- Published
- 2016
- Full Text
- View/download PDF
10. Adhesion signalling in melanoma biology
- Author
-
Miskolczi, Z., primary, Smith, M., additional, and Wellbrock, C., additional
- Published
- 2016
- Full Text
- View/download PDF
11. Proffered Paper: Utilising non-mutational drug-tolerance prolongs and improves targeted melanoma therapy
- Author
-
Smith, M.P., primary, Brunton, H., additional, Rowling, E.J., additional, Girotti, M.R., additional, Marais, R., additional, Dummer, R., additional, Flaherty, K.T., additional, Cooper, Z.A., additional, Wargo, J.A., additional, and Wellbrock, C., additional
- Published
- 2016
- Full Text
- View/download PDF
12. A relay of IL-1R and CXCR2 signals in the tumour microenvironment confer tolerance to MAPK antagonism in melanoma
- Author
-
Young, H., primary, Rowling, E., additional, Smith, M., additional, Bugatti, M., additional, Vermi, W., additional, Luheshi, N., additional, Wellbrock, C., additional, and Hurlstone, A., additional
- Published
- 2016
- Full Text
- View/download PDF
13. 457 - The role of tumour heterogeneity in melanoma progression
- Author
-
Rowling, E., Chapman, A., Telfer, B., Hurlstone, A., and Wellbrock, C.
- Published
- 2016
- Full Text
- View/download PDF
14. 449 - Adhesion signalling in melanoma biology
- Author
-
Miskolczi, Z., Smith, M., and Wellbrock, C.
- Published
- 2016
- Full Text
- View/download PDF
15. STAT5 contributes to interferon resistance of melanoma cells
- Author
-
Wellbrock, C., Weisser, C., Hassel, J. C., Fischer, P., Becker, J., Vetter, C. S., Behrmann, Iris, Kortylewski, M., Heinrich, P. C., Schartl, M., Wellbrock, C., Weisser, C., Hassel, J. C., Fischer, P., Becker, J., Vetter, C. S., Behrmann, Iris, Kortylewski, M., Heinrich, P. C., and Schartl, M.
- Abstract
BACKGROUND: Malignant melanoma is a highly aggressive neoplastic disease whose incidence is increasing rapidly. In recent years, the use of interferon alpha (IFNalpha) has become the most established adjuvant immunotherapy for melanoma of advanced stage. IFNalpha is a potent inhibitor of melanoma cell proliferation, and the signal transducer and activator of transcription STAT1 is crucial for its antiproliferative action. Although advanced melanomas clinically resistant to IFNalpha are frequently characterized by inefficient STAT1 signaling, the mechanisms underlying advanced-stage interferon resistance are poorly understood. RESULTS: Here, we demonstrate that IFNalpha activates STAT5 in melanoma cells and that in IFNalpha-resistant cells STAT5 is overexpressed. Significantly, the knockdown of STAT5 in interferon-resistant melanoma cells restored the growth-inhibitory response to IFNalpha. When STAT5 was overexpressed in IFNalpha-sensitive cells, it counteracted interferon-induced growth inhibition. The overexpressed STAT5 diminished IFNalpha-triggered STAT1 activation, most evidently through upregulation of the inhibitor of cytokine-signaling CIS. CONCLUSIONS: Our data demonstrate that overexpression and activation of STAT5 enable melanoma cells to overcome cytokine-mediated antiproliferative signaling. Thus, overexpression of STAT5 can counteract IFNalpha signaling in melanoma cells, and this finally can result in cytokine-resistant and progressively growing tumor cells. These findings have significant implications for the clinical failure of IFNalpha therapy of advanced melanoma because they demonstrate that IFNalpha induces the activation of STAT5 in melanoma cells, and in STAT5-overexpressing cells, this contributes to IFNalpha resistance.
- Published
- 2005
16. 290 Modifying TGF-beta Signalling and MEK Inhibition in Melanoma
- Author
-
Smith, M.P., primary, Ferguson, J.E., additional, Chapman, A., additional, Hurlstone, A., additional, Arozarena, I., additional, and Wellbrock, C., additional
- Published
- 2012
- Full Text
- View/download PDF
17. 653 Activating TGF-beta Signalling Enhances the Efficacy of MAP-kinase Pathway Inhibitors in Melanoma
- Author
-
Wellbrock, C., primary, Smith, M., additional, Ferguson, J., additional, Arozarena, I., additional, Hayward, R., additional, Marais, R., additional, Chapmann, A., additional, and Hurlstone, A., additional
- Published
- 2012
- Full Text
- View/download PDF
18. 461 MEK and SRC Inhibitors as a Combinatorial Approach to Melanoma Therapy
- Author
-
Ferguson, J., primary, Arozarena, I., additional, Ehrhardt, M., additional, and Wellbrock, C., additional
- Published
- 2012
- Full Text
- View/download PDF
19. Combination of MEK and SRC inhibition suppresses melanoma cell growth and invasion
- Author
-
Ferguson, J, primary, Arozarena, I, additional, Ehrhardt, M, additional, and Wellbrock, C, additional
- Published
- 2012
- Full Text
- View/download PDF
20. 20 B-RAF is an oncogene and therapeutic target in melanoma
- Author
-
Marais, R., primary, Barford, D., additional, Garnett, M., additional, Hedley, D., additional, Jones, M., additional, Karasarides, M., additional, Martin, J., additional, Niculescu-Duvaz, D., additional, Lee, S., additional, Ogilvie, L., additional, Springer, C., additional, Wan, P., additional, and Wellbrock, C., additional
- Published
- 2004
- Full Text
- View/download PDF
21. IL-16 B-Raf in melanoma
- Author
-
Wellbrock, C., primary, Garnett, M., additional, and Marais, R., additional
- Published
- 2003
- Full Text
- View/download PDF
22. Report from the II Melanoma Translational Meeting of the Spanish Melanoma Group (GEM)
- Author
-
Gonzalez-Cao M, Berrocal A, Puig S, Karachaliou N, De Matos-Arruda L, Seoane J, Escors D, Alvarez C, Jp, Vaque, Prat A, Wellbrock C, Arozarena I, and Martin-Algarra S
23. In melanoma, beta-catenin is a suppressor of invasion
- Author
-
Claudia Wellbrock, Benedetta Belloni, Imanol Arozarena, Daniel Gilby, Helen Bischof, Reinhard Dummer, University of Zurich, and Wellbrock, C
- Subjects
Cancer Research ,Beta-catenin ,Colorectal cancer ,610 Medicine & health ,Cell morphology ,Molecular oncology ,03 medical and health sciences ,0302 clinical medicine ,1311 Genetics ,1312 Molecular Biology ,Genetics ,medicine ,1306 Cancer Research ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,Melanoma ,10177 Dermatology Clinic ,Cancer ,Cell cycle ,medicine.disease ,Microphthalmia-associated transcription factor ,3. Good health ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cancer research - Abstract
Cell-type-specific signalling determines cell fate under physiological conditions, but it is increasingly apparent that also in cancer development the impact of any given oncogenic pathway on the individual cancer pathology is dependent on cell-lineage-specific molecular traits. For instance in colon and liver cancer canonical Wnt signalling produces increased cytoplasmic and nuclear localised beta-catenin, which correlates with invasion and poor prognosis. In contrast, in melanoma increased cytoplasmic and nuclear beta-catenin is currently emerging as a marker for good prognosis, and thus seems to have a different function compared with other cancer types; however, this function is unknown. We discovered that in contrast to its function in other cancers, in melanoma, beta-catenin blocks invasion. We demonstrate that this opposing role of nuclear beta-catenin in melanoma is mediated through MITF, a melanoma-specific protein that defines the lineage background of this cancer type. Downstream of beta-catenin MITF not only suppresses the Rho-GTPase-regulated cell morphology of invading melanoma cells, but also interferes with beta-catenin-induced expression of the essential collagenase MT1-MMP, thus affecting all aspects of an invasive phenotype. Importantly, overexpression of MITF in invasive colon cancer cells modifies beta-catenin-directed signalling and induces a 'melanoma phenotype'. In summary, the cell-type-specific presence of MITF in melanoma affects beta-catenin's pro-invasive properties otherwise active in colon or liver cancer. Thus our study reveals the general importance of considering cell-type-specific signalling for the accurate interpretation of tumour markers and ultimately for the design of rational therapies.
- Published
- 2011
- Full Text
- View/download PDF
24. 5 - Proffered Paper: Utilising non-mutational drug-tolerance prolongs and improves targeted melanoma therapy.
- Author
-
Smith, M.P., Brunton, H., Rowling, E.J., Girotti, M.R., Marais, R., Dummer, R., Flaherty, K.T., Cooper, Z.A., Wargo, J.A., and Wellbrock, C.
- Published
- 2016
- Full Text
- View/download PDF
25. 337 - A relay of IL-1R and CXCR2 signals in the tumour microenvironment confer tolerance to MAPK antagonism in melanoma.
- Author
-
Young, H., Rowling, E., Smith, M., Bugatti, M., Vermi, W., Luheshi, N., Wellbrock, C., and Hurlstone, A.
- Published
- 2016
- Full Text
- View/download PDF
26. Drug-Induced Reorganisation of Lipid Metabolism Limits the Therapeutic Efficacy of Ponatinib in Glioma Stem Cells.
- Author
-
Aldaz P, Olias-Arjona A, Lasheras-Otero I, Ausin K, Redondo-Muñoz M, Wellbrock C, Santamaria E, Fernandez-Irigoyen J, and Arozarena I
- Abstract
The standard of care for glioblastoma (GBM) involves surgery followed by adjuvant radio- and chemotherapy, but often within months, patients relapse, and this has been linked to glioma stem cells (GSCs), self-renewing cells with increased therapy resistance. The identification of the epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR) as key players in gliomagenesis inspired the development of inhibitors targeting these tyrosine kinases (TKIs). However, results from clinical trials testing TKIs have been disappointing, and while the role of GSCs in conventional therapy resistance has been extensively studied, less is known about resistance of GSCs to TKIs. In this study, we have used compartmentalised proteomics to analyse the adaptive response of GSCs to ponatinib, a TKI with activity against PDGFR. The analysis of differentially expressed proteins revealed that GSCs respond to ponatinib by broadly rewiring lipid metabolism, involving fatty acid beta-oxidation, cholesterol synthesis, and sphingolipid degradation. Inhibiting each of these metabolic pathways overcame ponatinib adaptation of GSCs, but interrogation of patient data revealed sphingolipid degradation as the most relevant pathway in GBM. Our data highlight that targeting lipid metabolism, and particularly sphingolipid degradation in combinatorial therapies, could improve the outcome of TKI therapies using ponatinib in GBM.
- Published
- 2024
- Full Text
- View/download PDF
27. Metabolic rewiring induced by ranolazine improves melanoma responses to targeted therapy and immunotherapy.
- Author
-
Redondo-Muñoz M, Rodriguez-Baena FJ, Aldaz P, Caballé-Mestres A, Moncho-Amor V, Otaegi-Ugartemendia M, Carrasco-Garcia E, Olias-Arjona A, Lasheras-Otero I, Santamaria E, Bocanegra A, Chocarro L, Grier A, Dzieciatkowska M M, Bigas C, Martin J, Urdiroz-Urricelqui U, Marzo F, Santamaria E, Kochan G, Escors D, Larrayoz IM, Heyn H, D'Alessandro A, Attolini CS, Matheu A, Wellbrock C, Benitah SA, Sanchez-Laorden B, and Arozarena I
- Subjects
- United States, Animals, Mice, Ranolazine pharmacology, Ranolazine therapeutic use, Immunotherapy, Protein Kinase Inhibitors pharmacology, Methionine, Melanoma drug therapy, Melanoma metabolism
- Abstract
Resistance of melanoma to targeted therapy and immunotherapy is linked to metabolic rewiring. Here, we show that increased fatty acid oxidation (FAO) during prolonged BRAF inhibitor (BRAFi) treatment contributes to acquired therapy resistance in mice. Targeting FAO using the US Food and Drug Administration-approved and European Medicines Agency-approved anti-anginal drug ranolazine (RANO) delays tumour recurrence with acquired BRAFi resistance. Single-cell RNA-sequencing analysis reveals that RANO diminishes the abundance of the therapy-resistant NGFR
hi neural crest stem cell subpopulation. Moreover, by rewiring the methionine salvage pathway, RANO enhances melanoma immunogenicity through increased antigen presentation and interferon signalling. Combination of RANO with anti-PD-L1 antibodies strongly improves survival by increasing antitumour immune responses. Altogether, we show that RANO increases the efficacy of targeted melanoma therapy through its effects on FAO and the methionine salvage pathway. Importantly, our study suggests that RANO could sensitize BRAFi-resistant tumours to immunotherapy. Since RANO has very mild side-effects, it might constitute a therapeutic option to improve the two main strategies currently used to treat metastatic melanoma., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
28. The Regulators of Peroxisomal Acyl-Carnitine Shuttle CROT and CRAT Promote Metastasis in Melanoma.
- Author
-
Lasheras-Otero I, Feliu I, Maillo A, Moreno H, Redondo-Muñoz M, Aldaz P, Bocanegra A, Olias-Arjona A, Lecanda F, Fernandez-Irigoyen J, Santamaria E, Larrayoz IM, Gomez-Cabrero D, Wellbrock C, Vicent S, and Arozarena I
- Subjects
- Mice, Animals, Carnitine O-Acetyltransferase genetics, Carnitine O-Acetyltransferase metabolism, Carnitine Acyltransferases genetics, Carnitine Acyltransferases metabolism, Ranolazine, Oxidation-Reduction, Fatty Acids metabolism, Carnitine metabolism, Neoplastic Cells, Circulating, Melanoma drug therapy
- Abstract
Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
29. Oxidative stress from DGAT1 oncoprotein inhibition in melanoma suppresses tumor growth when ROS defenses are also breached.
- Author
-
Wilcock DJ, Badrock AP, Wong CW, Owen R, Guerin M, Southam AD, Johnston H, Telfer BA, Fullwood P, Watson J, Ferguson H, Ferguson J, Lloyd GR, Jankevics A, Dunn WB, Wellbrock C, Lorigan P, Ceol C, Francavilla C, Smith MP, and Hurlstone AFL
- Subjects
- Animals, Oncogene Proteins metabolism, Oxidative Stress, Reactive Oxygen Species, Triglycerides, Zebrafish metabolism, Diacylglycerol O-Acyltransferase genetics, Diacylglycerol O-Acyltransferase metabolism, Melanoma
- Abstract
Dysregulated cellular metabolism is a cancer hallmark for which few druggable oncoprotein targets have been identified. Increased fatty acid (FA) acquisition allows cancer cells to meet their heightened membrane biogenesis, bioenergy, and signaling needs. Excess FAs are toxic to non-transformed cells but surprisingly not to cancer cells. Molecules underlying this cancer adaptation may provide alternative drug targets. Here, we demonstrate that diacylglycerol O-acyltransferase 1 (DGAT1), an enzyme integral to triacylglyceride synthesis and lipid droplet formation, is frequently up-regulated in melanoma, allowing melanoma cells to tolerate excess FA. DGAT1 over-expression alone transforms p53-mutant zebrafish melanocytes and co-operates with oncogenic BRAF or NRAS for more rapid melanoma formation. Antagonism of DGAT1 induces oxidative stress in melanoma cells, which adapt by up-regulating cellular reactive oxygen species defenses. We show that inhibiting both DGAT1 and superoxide dismutase 1 profoundly suppress tumor growth through eliciting intolerable oxidative stress., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Human Melanoma-Associated Mast Cells Display a Distinct Transcriptional Signature Characterized by an Upregulation of the Complement Component 3 That Correlates With Poor Prognosis.
- Author
-
Bahri R, Kiss O, Prise I, Garcia-Rodriguez KM, Atmoko H, Martínez-Gómez JM, Levesque MP, Dummer R, Smith MP, Wellbrock C, and Bulfone-Paus S
- Subjects
- Complement C3 metabolism, Humans, Mast Cells, Peptide Hydrolases metabolism, Transforming Growth Factor beta metabolism, Tumor Microenvironment genetics, Up-Regulation, Melanoma, Cutaneous Malignant, Melanoma pathology, Skin Neoplasms pathology
- Abstract
Cutaneous melanoma is one of the most aggressive human malignancies and shows increasing incidence. Mast cells (MCs), long-lived tissue-resident cells that are particularly abundant in human skin where they regulate both innate and adaptive immunity, are associated with melanoma stroma (MAMCs). Thus, MAMCs could impact melanoma development, progression, and metastasis by secreting proteases, pro-angiogenic factors, and both pro-inflammatory and immuno-inhibitory mediators. To interrogate the as-yet poorly characterized role of human MAMCs, we have purified MCs from melanoma skin biopsies and performed RNA-seq analysis. Here, we demonstrate that MAMCs display a unique transcriptome signature defined by the downregulation of the FcεRI signaling pathway, a distinct expression pattern of proteases and pro-angiogenic factors, and a profound upregulation of complement component C3. Furthermore, in melanoma tissue, we observe a significantly increased number of C3
+ MCs in stage IV melanoma. Moreover, in patients, C3 expression significantly correlates with the MC-specific marker TPSAB1 , and the high expression of both markers is linked with poorer melanoma survival. In vitro , we show that melanoma cell supernatants and tumor microenvironment (TME) mediators such as TGF-β, IL-33, and IL-1β induce some of the changes found in MAMCs and significantly modulate C3 expression and activity in MCs. Taken together, these data suggest that melanoma-secreted cytokines such as TGF-β and IL-1β contribute to the melanoma microenvironment by upregulating C3 expression in MAMCs, thus inducing an MC phenotype switch that negatively impacts melanoma prognosis., Competing Interests: RD has intermittent, project-focused consulting and/or advisory relationships with Novartis, Merck Sharp & Dhome (MSD), Bristol-Myers Squibb (BMS), Roche, Amgen, Takeda, Pierre Fabre, Sun Pharma, Sanofi, Catalym, Second Genome, Regeneron, Alligator, MaxiVAX SA, and touchIME outside the submitted work. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Bahri, Kiss, Prise, Garcia-Rodriguez, Atmoko, Martínez-Gómez, Levesque, Dummer, Smith, Wellbrock and Bulfone-Paus.)- Published
- 2022
- Full Text
- View/download PDF
31. Identification of a Dexamethasone Mediated Radioprotection Mechanism Reveals New Therapeutic Vulnerabilities in Glioblastoma.
- Author
-
Aldaz P, Auzmendi-Iriarte J, Durántez M, Lasheras-Otero I, Carrasco-Garcia E, Zelaya MV, Bragado L, Olías-Arjona A, Egaña L, Samprón N, Morilla I, Redondo-Muñoz M, Rico M, Squatrito M, Maria-Alonso M, Fernández-Irigoyen J, Santamaria E, Larráyoz IM, Wellbrock C, Matheu A, and Arozarena I
- Abstract
(1) Background: Despite the indisputable effectiveness of dexamethasone (DEXA) to reduce inflammation in glioblastoma (GBM) patients, its influence on tumour progression and radiotherapy response remains controversial. (2) Methods: We analysed patient data and used expression and cell biological analyses to assess effects of DEXA on GBM cells. We tested the efficacy of tyrosine kinase inhibitors in vitro and in vivo. (3) Results: We confirm in our patient cohort that administration of DEXA correlates with worse overall survival and shorter time to relapse. In GBM cells and glioma stem-like cells (GSCs) DEXA down-regulates genes controlling G2/M and mitotic-spindle checkpoints, and it enables cells to override the spindle assembly checkpoint (SAC). Concurrently, DEXA up-regulates Platelet Derived Growth Factor Receptor (PDGFR) signalling, which stimulates expression of anti-apoptotic regulators BCL2L1 and MCL1, required for survival during extended mitosis. Importantly, the protective potential of DEXA is dependent on intact tyrosine kinase signalling and ponatinib, sunitinib and dasatinib, all effectively overcome the radio-protective and pro-proliferative activity of DEXA. Moreover, we discovered that DEXA-induced signalling creates a therapeutic vulnerability for sunitinib in GSCs and GBM cells in vitro and in vivo. (4) Conclusions: Our results reveal a novel DEXA-induced mechanism in GBM cells and provide a rationale for revisiting the use of tyrosine kinase inhibitors for the treatment of GBM.
- Published
- 2021
- Full Text
- View/download PDF
32. Cooperative behaviour and phenotype plasticity evolve during melanoma progression.
- Author
-
Rowling EJ, Miskolczi Z, Nagaraju R, Wilcock DJ, Wang P, Telfer B, Li Y, Lasheras-Otero I, Redondo-Muñoz M, Sharrocks AD, Arozarena I, and Wellbrock C
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Fibronectins metabolism, Humans, Melanocytes pathology, Mesoderm pathology, Mice, Neoplasm Metastasis, Neoplastic Cells, Circulating pathology, Phenotype, Adaptation, Physiological, Cell Communication, Disease Progression, Melanoma pathology
- Abstract
A major challenge for managing melanoma is its tumour heterogeneity based on individual co-existing melanoma cell phenotypes. These phenotypes display variable responses to standard therapies, and they drive individual steps of melanoma progression; hence, understanding their behaviour is imperative. Melanoma phenotypes are defined by distinct transcriptional states, which relate to different melanocyte lineage development phases, ranging from a mesenchymal, neural crest-like to a proliferative, melanocytic phenotype. It is thought that adaptive phenotype plasticity based on transcriptional reprogramming drives melanoma progression, but at which stage individual phenotypes dominate and moreover, how they interact is poorly understood. We monitored melanocytic and mesenchymal phenotypes throughout melanoma progression and detected transcriptional reprogramming at different stages, with a gain in mesenchymal traits in circulating melanoma cells (CTCs) and proliferative features in metastatic tumours. Intriguingly, we found that distinct phenotype populations interact in a cooperative manner, which generates tumours of greater "fitness," supports CTCs and expands organotropic cues in metastases. Fibronectin, expressed in mesenchymal cells, acts as key player in cooperativity and promotes survival of melanocytic cells. Our data reveal an important role for inter-phenotype communications at various stages of disease progression, suggesting these communications could act as therapeutic target., (© 2020 The Authors. Pigment Cell & Melanoma Research published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
33. Osteoblasts contribute to a protective niche that supports melanoma cell proliferation and survival.
- Author
-
Ferguson J, Wilcock DJ, McEntegart S, Badrock AP, Levesque M, Dummer R, Wellbrock C, and Smith MP
- Subjects
- Cell Differentiation, Cell Line, Tumor, Cell Proliferation, Cell Survival, Gene Expression Regulation, Neoplastic, Humans, Melanoma genetics, Microphthalmia-Associated Transcription Factor genetics, Microphthalmia-Associated Transcription Factor metabolism, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf metabolism, RANK Ligand metabolism, Signal Transduction, Melanoma pathology, Osteoblasts pathology
- Abstract
Melanoma is the deadliest form of skin cancer; a primary driver of this high level of morbidity is the propensity of melanoma cells to metastasize. When malignant tumours develop distant metastatic lesions the new local tissue niche is known to impact on the biology of the cancer cells. However, little is known about how different metastatic tissue sites impact on frontline targeted therapies. Intriguingly, melanoma bone lesions have significantly lower response to BRAF or MEK inhibitor therapies. Here, we have investigated how the cellular niche of the bone can support melanoma cells by stimulating growth and survival via paracrine signalling between osteoblasts and cancer cells. Melanoma cells can enhance the differentiation of osteoblasts leading to increased production of secreted ligands, including RANKL. Differentiated osteoblasts in turn can support melanoma cell proliferation and survival via the secretion of RANKL that elevates the levels of the transcription factor MITF, even in the presence of BRAF inhibitor. By blocking RANKL signalling, either via neutralizing antibodies, genetic alterations or the RANKL receptor inhibitor SPD304, the survival advantage provided by osteoblasts could be overcome., (© 2019 The Authors. Pigment Cell & Melanoma Research published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
34. Phenotype plasticity as enabler of melanoma progression and therapy resistance.
- Author
-
Arozarena I and Wellbrock C
- Subjects
- Disease Progression, Drug Resistance, Neoplasm, Epithelial-Mesenchymal Transition, Humans, Immunotherapy, Melanoma therapy, Phenotype, Adaptation, Physiological physiology, Melanoma genetics, Melanoma pathology
- Abstract
Malignant melanoma is notorious for its inter- and intratumour heterogeneity, based on transcriptionally distinct melanoma cell phenotypes. It is thought that these distinct phenotypes are plastic in nature and that their transcriptional reprogramming enables heterogeneous tumours both to undergo different stages of melanoma progression and to adjust to drug exposure during treatment. Recent advances in genomic technologies and the rapidly expanding availability of large gene expression datasets have allowed for a refined definition of the gene signatures that characterize these phenotypes and have revealed that phenotype plasticity plays a major role in the resistance to both targeted therapy and immunotherapy. In this Review we discuss the definition of melanoma phenotypes through particular transcriptional states and reveal the prognostic relevance of the related gene expression signatures. We review how the establishment of phenotypes is controlled and which roles phenotype plasticity plays in melanoma development and therapy. Because phenotype plasticity in melanoma bears a great resemblance to epithelial-mesenchymal transition, the lessons learned from melanoma will also benefit our understanding of other cancer types.
- Published
- 2019
- Full Text
- View/download PDF
35. A PAX3/BRN2 rheostat controls the dynamics of BRAF mediated MITF regulation in MITF high /AXL low melanoma.
- Author
-
Smith MP, Rana S, Ferguson J, Rowling EJ, Flaherty KT, Wargo JA, Marais R, and Wellbrock C
- Subjects
- Base Sequence, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Melanoma pathology, Microphthalmia-Associated Transcription Factor metabolism, Phenotype, Protein Kinase Inhibitors pharmacology, Axl Receptor Tyrosine Kinase, Homeodomain Proteins metabolism, Melanoma genetics, Microphthalmia-Associated Transcription Factor genetics, PAX3 Transcription Factor metabolism, POU Domain Factors metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins B-raf metabolism, Receptor Protein-Tyrosine Kinases genetics
- Abstract
The BRAF kinase and the MAPK pathway are targets of current melanoma therapies. However, MAPK pathway inhibition results in dynamic changes of downstream targets that can counteract inhibitor-action not only in during treatment, but also in acquired resistant tumours. One such dynamic change involves the expression of the transcription factor MITF, a crucial regulator of cell survival and proliferation in untreated as well as drug-addicted acquired resistant melanoma. Tight control over MITF expression levels is required for optimal melanoma growth, and while it is well established that the MAPK pathway regulates MITF expression, the actual mechanism is insufficiently understood. We reveal here, how BRAF through action on the transcription factors BRN2 and PAX3 executes control over the regulation of MITF expression in a manner that allows for considerable plasticity. This plasticity provides robustness to the BRAF mediated MITF regulation and explains the dynamics in MITF expression that are observed in patients in response to MAPK inhibitor therapy., (© 2018 The Authors. Pigment Cell & Melanoma Research Published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
36. Collagen abundance controls melanoma phenotypes through lineage-specific microenvironment sensing.
- Author
-
Miskolczi Z, Smith MP, Rowling EJ, Ferguson J, Barriuso J, and Wellbrock C
- Subjects
- Acyltransferases, Adaptor Proteins, Signal Transducing genetics, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Adhesion, Cell Differentiation, Cell Line, Tumor, Cell Lineage, Cell Nucleus metabolism, Cell Proliferation, Extracellular Matrix metabolism, Extracellular Matrix pathology, Fibroblasts metabolism, Fibroblasts pathology, Gene Expression Regulation, Neoplastic, Humans, Melanoma genetics, Melanoma metabolism, Microphthalmia-Associated Transcription Factor genetics, Microphthalmia-Associated Transcription Factor metabolism, Phosphoproteins genetics, Transcription Factors genetics, Tumor Microenvironment, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Collagen metabolism, Melanoma pathology, Phosphoproteins metabolism, Transcription Factors metabolism
- Abstract
Despite the general focus on an invasive and de-differentiated phenotype as main driver of cancer metastasis, in melanoma patients many metastatic lesions display a high degree of pigmentation, indicative for a differentiated phenotype. Indeed, studies in mice and fish show that melanoma cells switch to a differentiated phenotype at secondary sites, possibly because in melanoma differentiation is closely linked to proliferation through the lineage-specific transcriptional master regulator MITF. Importantly, while a lot of effort has gone into identifying factors that induce the de-differentiated/invasive phenotype, it is not well understood how the switch to the differentiated/proliferative phenotype is controlled. We identify collagen as a contributor to this switch. We demonstrate that collagen stiffness induces melanoma differentiation through a YAP/PAX3/MITF axis and show that in melanoma patients increased collagen abundance correlates with nuclear YAP localization. However, the interrogation of large patient datasets revealed that in the context of the tumour microenvironment, YAP function is more complex. In the absence of fibroblasts, YAP/PAX3-mediated transcription prevails, but in the presence of fibroblasts tumour growth factor-β suppresses YAP/PAX3-mediated MITF expression and induces YAP/TEAD/SMAD-driven transcription and a de-differentiated phenotype. Intriguingly, while high collagen expression is correlated with poorer patient survival, the worst prognosis is seen in patients with high collagen expression, who also express MITF target genes such as the differentiation markers TRPM1, TYR and TYRP1, as well as CDK4. In summary, we reveal a distinct lineage-specific route of YAP signalling that contributes to the regulation of melanoma pigmentation and uncovers a set of potential biomarkers predictive for poor survival.
- Published
- 2018
- Full Text
- View/download PDF
37. Overcoming resistance to BRAF inhibitors.
- Author
-
Arozarena I and Wellbrock C
- Abstract
The discovery of activating mutations in the serine/threonine (S/T) kinase BRAF followed by a wave of follow-up research manifested that the MAPK-pathway plays a critical role in melanoma initiation and progression. BRAF and MEK inhibitors produce an unparalleled response rate in melanoma, but it is now clear that most responses are transient, and while some patients show long lasting responses the majority progress within 1 year. In accordance with the key role played by the MAPK-pathway in BRAF mutant melanomas, disease progression is mostly due to the appearance of drug-resistance mechanisms leading to restoration of MAPK-pathway activity. In the present article we will review the development, application and clinical effects of BRAF and MEK inhibitors both, as single agent and in combination in the context of targeted therapy in melanoma. We will then describe the most prominent mechanisms of resistance found in patients progressed on these targeted therapies. Finally we will discuss strategies for further optimizing the use of MAPK inhibitors and will describe the potential of alternative combination therapies to either delay the onset of resistance to MAPK inhibitors or directly target specific mechanisms of resistance to BRAF/MEK inhibitors., Competing Interests: Conflicts of Interest: The authors have no conflicts of interest to declare.
- Published
- 2017
- Full Text
- View/download PDF
38. PDL1 Signals through Conserved Sequence Motifs to Overcome Interferon-Mediated Cytotoxicity.
- Author
-
Gato-Cañas M, Zuazo M, Arasanz H, Ibañez-Vea M, Lorenzo L, Fernandez-Hinojal G, Vera R, Smerdou C, Martisova E, Arozarena I, Wellbrock C, Llopiz D, Ruiz M, Sarobe P, Breckpot K, Kochan G, and Escors D
- Subjects
- Animals, Cell Line, Tumor, Female, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Signal Transduction, B7-H1 Antigen immunology, Interferons immunology, Neoplasms immunology
- Abstract
PDL1 blockade produces remarkable clinical responses, thought to occur by T cell reactivation through prevention of PDL1-PD1 T cell inhibitory interactions. Here, we find that PDL1 cell-intrinsic signaling protects cancer cells from interferon (IFN) cytotoxicity and accelerates tumor progression. PDL1 inhibited IFN signal transduction through a conserved class of sequence motifs that mediate crosstalk with IFN signaling. Abrogation of PDL1 expression or antibody-mediated PDL1 blockade strongly sensitized cancer cells to IFN cytotoxicity through a STAT3/caspase-7-dependent pathway. Moreover, somatic mutations found in human carcinomas within these PDL1 sequence motifs disrupted motif regulation, resulting in PDL1 molecules with enhanced protective activities from type I and type II IFN cytotoxicity. Overall, our results reveal a mode of action of PDL1 in cancer cells as a first line of defense against IFN cytotoxicity., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
39. Biomarker Accessible and Chemically Addressable Mechanistic Subtypes of BRAF Melanoma.
- Author
-
Eskiocak B, McMillan EA, Mendiratta S, Kollipara RK, Zhang H, Humphries CG, Wang C, Garcia-Rodriguez J, Ding M, Zaman A, Rosales TI, Eskiocak U, Smith MP, Sudderth J, Komurov K, Deberardinis RJ, Wellbrock C, Davies MA, Wargo JA, Yu Y, De Brabander JK, Williams NS, Chin L, Rizos H, Long GV, Kittler R, and White MA
- Subjects
- Animals, Carcinogenesis drug effects, Drug Resistance, Neoplasm genetics, Humans, I-kappa B Kinase antagonists & inhibitors, I-kappa B Kinase genetics, MAP Kinase Signaling System drug effects, Melanoma classification, Melanoma pathology, Mice, Mutation, Protein Kinase Inhibitors administration & dosage, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Xenograft Model Antitumor Assays, Biomarkers, Tumor genetics, Melanoma drug therapy, Melanoma genetics, Proto-Oncogene Proteins B-raf genetics
- Abstract
Genomic diversity among melanoma tumors limits durable control with conventional and targeted therapies. Nevertheless, pathologic activation of the ERK1/2 pathway is a linchpin tumorigenic mechanism associated with the majority of primary and recurrent disease. Therefore, we sought to identify therapeutic targets that are selectively required for tumorigenicity in the presence of pathologic ERK1/2 signaling. By integration of multigenome chemical and genetic screens, recurrent architectural variants in melanoma tumor genomes, and patient outcome data, we identified two mechanistic subtypes of BRAF
V600 melanoma that inform new cancer cell biology and offer new therapeutic opportunities. Subtype membership defines sensitivity to clinical MEK inhibitors versus TBK1/IKBKε inhibitors. Importantly, subtype membership can be predicted using a robust quantitative five-feature genetic biomarker. This biomarker, and the mechanistic relationships linked to it, can identify a cohort of best responders to clinical MEK inhibitors and identify a cohort of TBK1/IKBKε inhibitor-sensitive disease among nonresponders to current targeted therapy. Significance: This study identified two mechanistic subtypes of melanoma: (1) the best responders to clinical BRAF/MEK inhibitors (25%) and (2) nonresponders due to primary resistance mechanisms (9.9%). We identified robust biomarkers that can detect these subtypes in patient samples and predict clinical outcome. TBK1/IKBKε inhibitors were selectively toxic to drug-resistant melanoma. Cancer Discov; 7(8); 832-51. ©2017 AACR. See related commentary by Jenkins and Barbie, p. 799 This article is highlighted in the In This Issue feature, p. 783 ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
40. Targeting endothelin receptor signalling overcomes heterogeneity driven therapy failure.
- Author
-
Smith MP, Rowling EJ, Miskolczi Z, Ferguson J, Spoerri L, Haass NK, Sloss O, McEntegart S, Arozarena I, von Kriegsheim A, Rodriguez J, Brunton H, Kmarashev J, Levesque MP, Dummer R, Frederick DT, Andrews MC, Cooper ZA, Flaherty KT, Wargo JA, and Wellbrock C
- Subjects
- Animals, Bosentan, Cell Line, Tumor, Disease Models, Animal, Heterografts, Humans, Mice, Nude, Neoplasm Transplantation, Treatment Outcome, Zebrafish, Antineoplastic Agents therapeutic use, Endothelin Receptor Antagonists administration & dosage, Melanoma drug therapy, Melanoma pathology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Sulfonamides administration & dosage
- Abstract
Approaches to prolong responses to BRAF targeting drugs in melanoma patients are challenged by phenotype heterogeneity. Melanomas of a "MITF-high" phenotype usually respond well to BRAF inhibitor therapy, but these melanomas also contain subpopulations of the de novo resistance "AXL-high" phenotype. > 50% of melanomas progress with enriched "AXL-high" populations, and because AXL is linked to de-differentiation and invasiveness avoiding an "AXL-high relapse" is desirable. We discovered that phenotype heterogeneity is supported during the response phase of BRAF inhibitor therapy due to MITF-induced expression of endothelin 1 (EDN1). EDN1 expression is enhanced in tumours of patients on treatment and confers drug resistance through ERK re-activation in a paracrine manner. Most importantly, EDN1 not only supports MITF-high populations through the endothelin receptor B (EDNRB), but also AXL-high populations through EDNRA, making it a master regulator of phenotype heterogeneity. Endothelin receptor antagonists suppress AXL-high-expressing cells and sensitize to BRAF inhibition, suggesting that targeting EDN1 signalling could improve BRAF inhibitor responses without selecting for AXL-high cells., (© 2017 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2017
- Full Text
- View/download PDF
41. Targeting invasive properties of melanoma cells.
- Author
-
Arozarena I and Wellbrock C
- Subjects
- Animals, Cell Differentiation, Gene Expression Regulation, Neoplastic drug effects, Humans, Melanoma metabolism, Neoplasm Invasiveness, Neoplasm Metastasis, Phenotype, Signal Transduction drug effects, Skin Neoplasms metabolism, Antineoplastic Agents therapeutic use, Melanoma drug therapy, Melanoma pathology, Molecular Targeted Therapy, Neoplasm Proteins antagonists & inhibitors, Skin Neoplasms drug therapy, Skin Neoplasms pathology
- Abstract
Melanoma is a skin cancer notorious for its metastatic potential. As an initial step of the metastatic cascade, melanoma cells part from the primary tumour and invade the surrounding tissue, which is crucial for their dissemination and the formation of distant secondary tumours. Over the last two decades, our understanding of both, general and melanoma specific mechanisms of invasion has significantly improved, but to date no efficient therapeutic strategy tackling the invasive properties of melanoma cells has reached the clinic. In this review, we assess the major contributions towards the understanding of the molecular biology of melanoma cell invasion with a focus on melanoma specific traits. These traits are based on the neural crest origin of melanoma cells and explain their intrinsic invasive nature. A particular emphasis is given not only to lineage specific signalling mediated by TGFβ, and noncanonical and canonical WNT signalling, but also to the role of PDE5A and RHO-GTPases in modulating modes of melanoma cell invasion. We discuss existing caveats in the current understanding of the metastatic properties of melanoma cells, as well as the relevance of the 'phenotype switch' model and 'co-operativity' between different phenotypes in heterogeneous tumours. At the centre of these phenotypes is the lineage commitment factor microphthalmia-associated transcription factor, one of the most crucial regulators of the balance between de-differentiation (neural crest specific gene expression) and differentiation (melanocyte specific gene expression) that defines invasive and noninvasive melanoma cell phenotypes. Finally, we provide insight into the current evidence linking resistance to targeted therapies to invasive properties of melanoma cells., (© 2017 Federation of European Biochemical Societies.)
- Published
- 2017
- Full Text
- View/download PDF
42. An adaptive signaling network in melanoma inflammatory niches confers tolerance to MAPK signaling inhibition.
- Author
-
Young HL, Rowling EJ, Bugatti M, Giurisato E, Luheshi N, Arozarena I, Acosta JC, Kamarashev J, Frederick DT, Cooper ZA, Reuben A, Gil J, Flaherty KT, Wargo JA, Vermi W, Smith MP, Wellbrock C, and Hurlstone A
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Chemokine CXCL1 metabolism, Fibroblasts drug effects, Fibroblasts metabolism, Humans, Interleukin-1 metabolism, Interleukin-1beta metabolism, Interleukin-8 metabolism, Ligands, Macrophages drug effects, Macrophages metabolism, Mice, Knockout, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, NF-kappa B metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Receptors, Interleukin-1 metabolism, Receptors, Interleukin-8B metabolism, Stromal Cells metabolism, Stromal Cells pathology, Inflammation enzymology, Inflammation pathology, MAP Kinase Signaling System drug effects, Melanoma enzymology, Melanoma pathology, Skin Neoplasms enzymology, Skin Neoplasms pathology
- Abstract
Mitogen-activated protein kinase (MAPK) pathway antagonists induce profound clinical responses in advanced cutaneous melanoma, but complete remissions are frustrated by the development of acquired resistance. Before resistance emerges, adaptive responses establish a mutation-independent drug tolerance. Antagonizing these adaptive responses could improve drug effects, thereby thwarting the emergence of acquired resistance. In this study, we reveal that inflammatory niches consisting of tumor-associated macrophages and fibroblasts contribute to treatment tolerance through a cytokine-signaling network that involves macrophage-derived IL-1β and fibroblast-derived CXCR2 ligands. Fibroblasts require IL-1β to produce CXCR2 ligands, and loss of host IL-1R signaling in vivo reduces melanoma growth. In tumors from patients on treatment, signaling from inflammatory niches is amplified in the presence of MAPK inhibitors. Signaling from inflammatory niches counteracts combined BRAF/MEK (MAPK/extracellular signal-regulated kinase kinase) inhibitor treatment, and consequently, inhibiting IL-1R or CXCR2 signaling in vivo enhanced the efficacy of MAPK inhibitors. We conclude that melanoma inflammatory niches adapt to and confer drug tolerance toward BRAF and MEK inhibitors early during treatment., (© 2017 Young et al.)
- Published
- 2017
- Full Text
- View/download PDF
43. Glucose availability controls ATF4-mediated MITF suppression to drive melanoma cell growth.
- Author
-
Ferguson J, Smith M, Zudaire I, Wellbrock C, and Arozarena I
- Subjects
- Cell Line, Tumor, Cell Proliferation, Cell Survival, Humans, Microphthalmia-Associated Transcription Factor genetics, Multiple Myeloma genetics, Promoter Regions, Genetic, Reactive Oxygen Species metabolism, Signal Transduction, Activating Transcription Factor 4 metabolism, Glucose metabolism, Microphthalmia-Associated Transcription Factor metabolism, Multiple Myeloma metabolism
- Abstract
It is well know that cancer cells have adopted an altered metabolism and that glucose is a major source of energy for these cells. In melanoma, enhanced glucose usage is favoured through the hyper-activated MAPK pathway, which suppresses OXPHOS and stimulates glycolysis. However, it has not been addressed how glucose availability impacts on melanoma specific signaling pathways that drive melanoma cell proliferation. Here we show that melanoma cells are dependent on high glucose levels for efficient growth. Thereby, glucose metabolism controls the expression of the melanoma fate transcription factor MITF, a master regulator of melanoma cell survival and proliferation, invasion and therapy resistance. Restriction of glucose availability to physiological concentrations induces the production of reactive oxygen species (ROS). Increased ROS levels lead to the up-regulation of AFT4, which in turn suppresses MITF expression by competing with CREB, an otherwise potent inducer of the MITF promoter. Our data give new insight into the complex regulation of MITF, a key regulator of melanoma biology, and support previous findings that link metabolic disorders such as hyperglycemia and diabetes with increased melanoma risk.
- Published
- 2017
- Full Text
- View/download PDF
44. Molecular Pathways: Maintaining MAPK Inhibitor Sensitivity by Targeting Nonmutational Tolerance.
- Author
-
Smith MP and Wellbrock C
- Subjects
- Gene Expression drug effects, Humans, Neoplasms metabolism, Protein Kinase Inhibitors, Tumor Microenvironment drug effects, Antineoplastic Agents therapeutic use, Mitogen-Activated Protein Kinases antagonists & inhibitors, Neoplasms drug therapy, Signal Transduction drug effects
- Abstract
Targeting hyperactive MAPK signaling has proven to be an effective treatment for a variety of different cancers. Responses to the BRAF inhibitors vemurafenib and dabrafenib and the MEK inhibitors trametinib and cobimetinib are, however, transient, and complete remission is rarely observed; rather, outgrowth of resistant clones within progressed tumors appears inevitable. These resistant tumors display great heterogeneity, which poses a major challenge to any salvage therapy. Recent focus has, therefore, been on the early dynamics of inhibitor response during tumor regression. During this time, cells can persist in an adapted tolerant state, which results in a phase of nonmutational drug tolerance. In this article, we discuss how inhibition of the MAPK pathway leads to an adaptive rewiring that evolves from the relief of immediate negative feedback loops to short-term gene expression changes and adaptation of intracellular signaling. Tolerance can also be mediated by external signaling from the tumor microenvironment, which itself adapts upon treatment and the selection for cells with an innate drug-tolerant phenotype. In preclinical models, combination treatment with receptor tyrosine kinase (RTK) inhibitors (lapatinib and dasatinib), histone deacetylase (HDAC) inhibitors (vorinostat and entinostat), or drugs targeting cancer-specific mechanisms (nelfinavir in melanoma) can overcome this early tolerance. A better understanding of how nonmutational tolerance is created and supported may hold the key to better combinational strategies that maintain drug sensitivity. Clin Cancer Res; 22(24); 5966-70. ©2016 AACR., Competing Interests: The authors declare no conflict of interest, (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
45. Targeting MITF in the tolerance-phase.
- Author
-
Arozarena I, Smith MP, and Wellbrock C
- Subjects
- Drug Tolerance, Humans, Proto-Oncogene Proteins B-raf, Melanoma
- Published
- 2016
- Full Text
- View/download PDF
46. Melanoma and the Microenvironment--Age Matters.
- Author
-
Wellbrock C
- Subjects
- Animals, Humans, Male, Aging metabolism, Drug Resistance, Neoplasm, Melanoma drug therapy, Melanoma pathology, Membrane Proteins metabolism, Neoplasm Metastasis, Tumor Microenvironment
- Published
- 2016
- Full Text
- View/download PDF
47. The Complexity of the ERK/MAP-Kinase Pathway and the Treatment of Melanoma Skin Cancer.
- Author
-
Wellbrock C and Arozarena I
- Abstract
The central role played by the ERK/MAPK pathway downstream of RAS in human neoplasias is best exemplified in the context of melanoma skin cancer. Signaling through the MAPK pathway is crucial for the proliferation of melanocytes, the healthy pigment cells that give rise to melanoma. However, hyper-activation of the MAPK-pathway is found in over 90% of melanomas with approximately 50% of all patients displaying mutations in the kinase BRAF, and approximately 28% of all patients harboring mutations in the MAPK-pathway up-stream regulator NRAS. This finding has led to the development of BRAF and MEK inhibitors whose application in the clinic has shown unprecedented survival responses. Unfortunately the responses to MAPK pathway inhibitors are transient with most patients progressing within a year and a median progression free survival of 7-10 months. The disease progression is due to the development of drug-resistance based on various mechanisms, many of them involving a rewiring of the MAPK pathway. In this article we will review the complexity of MAPK signaling in melanocytic cells as well as the mechanisms of action of different MAPK-pathway inhibitors and their correlation with clinical response. We will reflect on mechanisms of innate and acquired resistance that limit patient's response, with a focus on the MAPK signaling network. Because of the resurgence of antibody-based immune-therapies there is a growing feeling of failure in the targeted therapy camp. However, recent studies have revealed new windows of therapeutic opportunity for melanoma sufferers treated with drugs targeting the MAPK pathway, and these opportunities will be discussed.
- Published
- 2016
- Full Text
- View/download PDF
48. Inhibiting Drivers of Non-mutational Drug Tolerance Is a Salvage Strategy for Targeted Melanoma Therapy.
- Author
-
Smith MP, Brunton H, Rowling EJ, Ferguson J, Arozarena I, Miskolczi Z, Lee JL, Girotti MR, Marais R, Levesque MP, Dummer R, Frederick DT, Flaherty KT, Cooper ZA, Wargo JA, and Wellbrock C
- Subjects
- Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, GTP Phosphohydrolases genetics, HIV Protease Inhibitors pharmacology, Humans, Membrane Proteins genetics, Mitogen-Activated Protein Kinases metabolism, Nelfinavir pharmacology, PAX3 Transcription Factor, Paired Box Transcription Factors genetics, Proto-Oncogene Proteins B-raf genetics, Up-Regulation drug effects, Up-Regulation genetics, Drug Tolerance genetics, Melanoma drug therapy, Melanoma genetics, Mutation drug effects, Mutation genetics, Protein Kinase Inhibitors pharmacology
- Abstract
Once melanomas have progressed with acquired resistance to mitogen-activated protein kinase (MAPK)-targeted therapy, mutational heterogeneity presents a major challenge. We therefore examined the therapy phase before acquired resistance had developed and discovered the melanoma survival oncogene MITF as a driver of an early non-mutational and reversible drug-tolerance state, which is induced by PAX3-mediated upregulation of MITF. A drug-repositioning screen identified the HIV1-protease inhibitor nelfinavir as potent suppressor of PAX3 and MITF expression. Nelfinavir profoundly sensitizes BRAF and NRAS mutant melanoma cells to MAPK-pathway inhibitors. Moreover, nelfinavir is effective in BRAF and NRAS mutant melanoma cells isolated from patients progressed on MAPK inhibitor (MAPKi) therapy and in BRAF/NRAS/PTEN mutant tumors. We demonstrate that inhibiting a driver of MAPKi-induced drug tolerance could improve current approaches of targeted melanoma therapy., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
49. Spatial intra-tumour heterogeneity in acquired resistance to targeted therapy complicates the use of PDX models for co-clinical cancer studies.
- Author
-
Wellbrock C
- Subjects
- Humans, Vemurafenib, Antineoplastic Agents therapeutic use, Drug Resistance, Neoplasm genetics, Indoles therapeutic use, Melanoma drug therapy, Mutation, Neoplasm Metastasis drug therapy, Skin Neoplasms drug therapy, Sulfonamides therapeutic use
- Published
- 2015
- Full Text
- View/download PDF
50. Microphthalmia-associated transcription factor in melanoma development and MAP-kinase pathway targeted therapy.
- Author
-
Wellbrock C and Arozarena I
- Subjects
- Animals, Carcinogenesis metabolism, Carcinogenesis pathology, Cell Cycle, Humans, Melanoma genetics, Melanoma pathology, Microphthalmia-Associated Transcription Factor genetics, Skin Neoplasms, Melanoma, Cutaneous Malignant, MAP Kinase Signaling System, Melanoma enzymology, Melanoma therapy, Microphthalmia-Associated Transcription Factor metabolism, Molecular Targeted Therapy
- Abstract
Malignant melanoma is a neoplasm of melanocytes, and the microphthalmia-associated transcription factor (MITF) is essential for the existence of melanocytes. MITF's relevance for this cell lineage is maintained in melanoma, where it is an important regulator of survival and balances melanoma cell proliferation with terminal differentiation (pigmentation). The MITF gene is amplified in ~20% of melanomas and MITF mutation can predispose to melanoma development. Furthermore, the regulation of MITF expression and function is strongly linked to the BRAF/MEK/ERK/MAP-kinase (MAPK) pathway, which is deregulated in >90% of melanomas and central target of current therapies. MITF expression in melanoma is heterogeneous, and recent findings highlight the relevance of this heterogeneity for the response of melanoma to MAPK pathway targeting drugs, as well as for MITF's role in melanoma progression. This review aims to provide an updated overview on the regulation of MITF function and plasticity in melanoma with a focus on its link to MAPK signaling., (© 2015 The Authors. Pigment Cell & Melanoma Research Published by John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.