Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Mol Cancer ; 16(1): 85, 2017 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-28454577

RESUMEN

BACKGROUND: The BRAF protein kinase is widely studied as a cancer driver and therapeutic target. However, the regulation of its expression is not completely understood. RESULTS: Taking advantage of the RNA-seq data of more than 4800 patients belonging to 9 different cancer types, we show that BRAF mRNA exists as a pool of 3 isoforms (reference BRAF, BRAF-X1, and BRAF-X2) that differ in the last part of their coding sequences, as well as in the length (BRAF-ref: 76 nt; BRAF-X1 and BRAF-X2: up to 7 kb) and in the sequence of their 3'UTRs. The expression levels of BRAF-ref and BRAF-X1/X2 are inversely correlated, while the most prevalent among the three isoforms varies from cancer type to cancer type. In melanoma cells, the X1 isoform is expressed at the highest level in both therapy-naïve cells and cells with acquired resistance to vemurafenib driven by BRAF gene amplification or expression of the Δ[3-10] splicing variant. In addition to the BRAF-ref protein, the BRAF-X1 protein (the full length as well as the Δ[3-10] variant) is also translated. The expression levels of the BRAF-ref and BRAF-X1 proteins are similar, and together they account for BRAF functional activities. In contrast, the endogenous BRAF-X2 protein is hard to detect because the C-terminal domain is selectively recognized by the ubiquitin-proteasome pathway and targeted for degradation. CONCLUSIONS: By shedding light on the repertoire of BRAF mRNA and protein variants, and on the complex regulation of their expression, our work paves the way to a deeper understanding of a crucially important player in human cancer and to a more informed development of new therapeutic strategies.


Asunto(s)
Melanoma/genética , Neoplasias/genética , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Empalme Alternativo/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Exones/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Indoles/administración & dosificación , Melanoma/tratamiento farmacológico , Melanoma/patología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , ARN Mensajero/genética , Sulfonamidas/administración & dosificación , Vemurafenib
2.
Pigment Cell Melanoma Res ; 36(6): 576-582, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37759408

RESUMEN

To commemorate the 20th Anniversary of the Society of Melanoma Research and the first International Melanoma Research Congress held in June of 2003, we have described in brief, how the Society for Melanoma Research (SMR) began, the purpose, goals, and governance of the SMR, and how the society has evolved to support new melanoma researchers. In celebration of the immense progress in treating melanoma patients over the last 20 years and the impact of the SMR on these advances, we have highlighted memories and insight from early SMR members and founders.


Asunto(s)
Amigos , Melanoma , Humanos , Melanoma/terapia , Sociedades Médicas
3.
Pigment Cell Melanoma Res ; 36(6): 594-601, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37819777

RESUMEN

Upon the 20th Anniversary of the Society for Melanoma Research, we highlight the perspectives of patients aiming to help improve future experiences, outcomes, and their quality of life over the next 20 years. Five melanoma patients generously shared their inspiring and enlightening stories of diagnosis, treatment, and outcomes. Many patients had excellent medical teams that synergistically worked together to provide an accurate diagnosis, effective treatment options, and supportive care. However, it is clear that health inequities persist in communities where people of color are predominant, affecting early detection, patient experience, and outcomes. These stories shed light on the unique challenges faced by patients and how the lack of melanoma awareness and adequate resources, especially in communities of color or low socioeconomic status, can contribute to disparate outcomes in melanoma care. We expect that these stories will raise awareness about the progress in melanoma treatment but also the existent disparities in melanoma diagnosis and treatment and the importance of early detection and prevention.


Asunto(s)
Melanoma , Calidad de Vida , Humanos , Melanoma/diagnóstico , Melanoma/terapia
4.
Pigment Cell Melanoma Res ; 36(5): 441-447, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37093838

RESUMEN

The inaugural Diversity and Inclusion in Science Session was held during the 2021 Society for Melanoma Research (SMR) congress. The goal of the session was to discuss diversity, equity, and inclusion in the melanoma research community and strategies to promote the advancement of underrepresented melanoma researchers. An international survey was conducted to assess the diversity, equity, and inclusion (DEI) climate among researchers and clinicians within the Society for Melanoma Research (SMR). The findings suggest there are feelings and experiences of inequity, bias, and harassment within the melanoma community that correlate with one's gender, ethnic/racial group, and/or geographic location. Notably, significant reports of inequity in opportunity, discrimination, and sexual harassment demonstrate there is much work remaining to ensure all scientists in our community experience an academic workplace culture built on mutual respect, fair access, inclusion, and equitable opportunity.


Asunto(s)
Diversidad, Equidad e Inclusión , Melanoma , Humanos
5.
Pigment Cell Melanoma Res ; 36(6): 588-593, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37819763

RESUMEN

The Society for Melanoma Research (SMR) was created 20 years ago and has unequivocally contributed to the vast progress of the field, particularly for the treatment of melanoma patients with metastatic disease by facilitating synergistic collaborations between clinicians, researchers at the bench, and industry. In commemoration of the 20th anniversary of the first SMR International Congress (held in 2003 in Philadelphia), we look to the future by highlighting the perspectives of the next generation of rising stars, medical, and graduate students across six continents.


Asunto(s)
Melanoma , Humanos , Melanoma/terapia , Melanoma/patología
6.
bioRxiv ; 2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36711814

RESUMEN

Resistance to combination BRAF/MEK inhibitor (BRAFi/MEKi) therapy arises in nearly every patient with BRAFV600E/K melanoma, despite promising initial responses. Achieving cures in this expanding BRAFi/MEKi-resistant cohort represents one of the greatest challenges to the field; few experience additional durable benefit from immunotherapy and no alternative therapies exist. To better personalize therapy in cancer patients to address therapy relapse, umbrella trials have been initiated whereby genomic sequencing of a panel of potentially actionable targets guide therapy selection for patients; however, the superior efficacy of such approaches remains to be seen. We here test the robustness of the umbrella trial rationale by analyzing relationships between genomic status of a gene and the downstream consequences at the protein level of related pathway, which find poor relationships between mutations, copy number amplification, and protein level. To profile candidate therapeutic strategies that may offer clinical benefit in the context of acquired BRAFi/MEKi resistance, we established a repository of patient-derived xenograft models from heavily pretreated patients with resistance to BRAFi/MEKi and/or immunotherapy (R-PDX). With these R-PDXs, we executed in vivo compound repurposing screens using 11 FDA-approved agents from an NCI-portfolio with pan-RTK, non-RTK and/or PI3K-mTOR specificity. We identify dasatinib as capable of restoring BRAFi/MEKi antitumor efficacy in ~70% of R-PDX tested. A systems-biology analysis indicates elevated baseline protein expression of canonical drivers of therapy resistance (e.g., AXL, YAP, HSP70, phospho-AKT) as predictive of MAPKi/dasatinib sensitivity. We therefore propose that dasatinib-based MAPKi therapy may restore antitumor efficacy in patients that have relapsed to standard-of-care therapy by broadly targeting proteins critical in melanoma therapy escape. Further, we submit that this experimental PDX paradigm could potentially improve preclinical evaluation of therapeutic modalities and augment our ability to identify biomarker-defined patient subsets that may respond to a given clinical trial.

7.
Int J Cancer ; 131(9): 2165-74, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22323315

RESUMEN

Melanoma is an increasingly common and potentially fatal malignancy of the skin and some mucous membranes. As no cure exists for metastatic disease, there is an urgent need for novel drugs. 2'-Deoxy-5-fluorouridylyl-(3'-5')-3'-C-ethynylcytidine [5-FdU(3'-5')ECyd] and 3'-C-ethynylcytidinylyl-(5' → 1-O)-2-O-octadecyl-sn-glycerylyl-(3-O → 5')-2'-deoxy-5-fluorouridine [ECyd-lipid-5-FdU] represent cytostatic active duplex drugs, which can be metabolized into various active antimetabolites. We evaluated the cytotoxicity of these heterodinucleoside phosphate analogs, their corresponding monomers ECyd and 5-FdU and combinations thereof on six metastatic melanoma cell lines and six ex vivo patient-derived melanoma cells in comparison to current standard cytostatic agents and the BRAF V600E inhibitor Vemurafenib. In vitro (real-time)-proliferation assays demonstrated that 5-FdU(3'-5')ECyd and ECyd-lipid-5-FdU had a high cytotoxic efficacy causing 75% melanoma cell death at concentrations in the nanomolar and micromolar range. Cytotoxicity was conducted by induction of DNA cleavage indicating apoptotic cells. Chicken embryotoxicity demonstrated that the duplex drugs were less toxic than 5-FdU at 0.01 µM. In vivo the duplex drug 5-FdU(3'-5')ECyd was efficacious in the murine LOX IMVI melanoma xenograph model on administration of 11.2 mg/kg/injection every fourth day. Both duplex drugs are promising novel cytostatic agents for the treatment of malignant melanoma meriting clinical evaluation.


Asunto(s)
Antineoplásicos/farmacología , Citidina Monofosfato/análogos & derivados , Citidina/análogos & derivados , Floxuridina/farmacología , Fluorodesoxiuridilato/análogos & derivados , Melanoma/tratamiento farmacológico , Melanoma/patología , Oligonucleótidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Embrión de Pollo , Citidina/farmacología , Citidina/uso terapéutico , Citidina Monofosfato/farmacología , Citidina Monofosfato/uso terapéutico , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Floxuridina/uso terapéutico , Fluorodesoxiuridilato/farmacología , Fluorodesoxiuridilato/uso terapéutico , Humanos , Indoles/farmacología , Ratones , Ratones Desnudos , Oligonucleótidos/uso terapéutico , Distribución Aleatoria , Sulfonamidas/farmacología , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncogene ; 40(37): 5590-5599, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34304249

RESUMEN

Targeting MAPK pathway using a combination of BRAF and MEK inhibitors is an efficient strategy to treat melanoma harboring BRAF-mutation. The development of acquired resistance is inevitable due to the signaling pathway rewiring. Combining western blotting, immunohistochemistry, and reverse phase protein array (RPPA), we aim to understanding the role of the mTORC1 signaling pathway, a center node of intracellular signaling network, in mediating drug resistance of BRAF-mutant melanoma to the combination of BRAF inhibitor (BRAFi) and MEK inhibitor (MEKi) therapy. The mTORC1 signaling pathway is initially suppressed by BRAFi and MEKi combination in melanoma but rebounds overtime after tumors acquire resistance to the combination therapy (CR) as assayed in cultured cells and PDX models. In vitro experiments showed that a subset of CR melanoma cells was sensitive to mTORC1 inhibition. The mTOR inhibitors, rapamycin and NVP-BEZ235, induced cell cycle arrest and apoptosis in CR cell lines. As a proof-of-principle, we demonstrated that rapamycin and NVP-BEZ235 treatment reduced tumor growth in CR xenograft models. Mechanistically, AKT or ERK contributes to the activation of mTORC1 in CR cells, depending on PTEN status of these cells. Our study reveals that mTOR activation is essential for drug resistance of melanoma to MAPK inhibitors, and provides insight into the rewiring of the signaling networks in CR melanoma.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf , Serina-Treonina Quinasas TOR , Humanos
9.
Cancer Cell ; 39(5): 610-631, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33545064

RESUMEN

There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Microambiente Tumoral/inmunología , Animales , Humanos , Inmunidad/inmunología , Inmunoterapia/métodos , Melanoma/patología , Neoplasias Cutáneas/patología
10.
Mol Biol Cell ; 18(4): 1457-63, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17314399

RESUMEN

The ERK subfamily of MAP kinases is a critical regulator of S phase entry. ERK activity regulates the induction of cyclin D1, and a sustained ERK signal is thought to be required for this effect, at least in fibroblasts. We now show that early G1 phase ERK activity is dispensable for the induction of cyclin D1 and that the critical ERK signaling period is restricted to 3-6 h after mitogenic stimulation of quiescent fibroblasts. Similarly, early G1 phase ERK activity is dispensable for entry into S phase. Moreover, if cyclin D1 is expressed ectopically, ERK activity becomes dispensable throughout the G1 phase. In addition to its effect on cyclin D1, ERK activity is thought to contribute to the down-regulation of p27kip1. We found that this effect is restricted to late G1/S phase. Mechanistic analysis showed that the ERK effect on p27kip1 is mediated by Skp2 and is secondary to its effect on cyclin D1. Our results emphasize the importance of mid-G1 phase ERK activity and resolve primary versus secondary ERK targets within the G1 phase cyclin-dependent kinases.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fase G1/fisiología , Animales , Butadienos/farmacología , Células Cultivadas , Ciclina D1/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Nitrilos/farmacología , Proteínas Quinasas Asociadas a Fase-S/efectos de los fármacos , Proteínas Quinasas Asociadas a Fase-S/metabolismo
11.
Oncogene ; 39(36): 5811-5824, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32733068

RESUMEN

Telomere maintenance via telomerase reactivation is a nearly universal hallmark of cancer cells which enables replicative immortality. In contrast, telomerase activity is silenced in most adult somatic cells. Thus, telomerase represents an attractive target for highly selective cancer therapeutics. However, development of telomerase inhibitors has been challenging and thus far there are no clinically approved strategies exploiting this cancer target. The discovery of prevalent mutations in the TERT promoter region in many cancers and recent advances in telomerase biology has led to a renewed interest in targeting this enzyme. Here we discuss recent efforts targeting telomerase, including immunotherapies and direct telomerase inhibitors, as well as emerging approaches such as targeting TERT gene expression driven by TERT promoter mutations. We also address some of the challenges to telomerase-directed therapies including potential therapeutic resistance and considerations for future therapeutic applications and translation into the clinical setting. Although much work remains to be done, effective strategies targeting telomerase will have a transformative impact for cancer therapy and the prospect of clinically effective drugs is boosted by recent advances in structural models of human telomerase.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Telomerasa/antagonistas & inhibidores , Animales , Biomarcadores de Tumor , Estudios Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/diagnóstico , Neoplasias/etiología , Telómero/genética , Telómero/metabolismo , Homeostasis del Telómero , Resultado del Tratamiento
12.
Pigment Cell Melanoma Res ; 33(2): 372-377, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31696640

RESUMEN

Bromodomain and extra-terminal inhibitors (BETi) delay tumor growth, in part, through tumor cell intrinsic alterations and initiation of anti-tumor CD8+ T-cell responses. By contrast, BETi effects on pro-tumoral immune responses remain unclear. Here, we show that the next-generation BETi, PLX51107, delayed tumor growth to differing degrees in Braf V600E melanoma syngeneic mouse models. These differential responses were associated with the influx of tumor-associated macrophages during BETi treatment. Tumors that were poorly responsive to PLX51107 showed increased influx of colony-stimulating factor-1 receptor (CSF-1R)-positive tumor-associated macrophages. We depleted CSF-1R+ tumor-associated macrophages with the CSF-1R inhibitor, PLX3397, in combination with PLX51107. Treatment with PLX3397 enhanced the efficacy of PLX51107 in poorly responsive Braf V600E syngeneic melanomas in vivo. These findings suggest that tumor-associated macrophage accumulation limits BETi efficacy and that co-treatment with PLX3397 can improve response to PLX51107, offering a potential novel combination therapy for metastatic melanoma patients.


Asunto(s)
Aminopiridinas/farmacología , Macrófagos/patología , Melanoma/patología , Proteínas/antagonistas & inhibidores , Pirroles/farmacología , Neoplasias Cutáneas/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Proteínas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Resultado del Tratamiento
13.
Pigment Cell Melanoma Res ; 32(5): 687-696, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31063649

RESUMEN

Epigenetic agents such as bromodomain and extra-terminal region inhibitors (BETi) slow tumor growth via tumor intrinsic alterations; however, their effects on antitumor immunity remain unclear. A recent advance is the development of next-generation BETi that are potent and display a favorable half-life. Here, we tested the BETi, PLX51107, for immune-based effects on tumor growth in BRAF V600E melanoma syngeneic models. PLX51107 delayed melanoma tumor growth and increased activated, proliferating, and functional CD8+ T cells in tumors leading to CD8+ T-cell-mediated tumor growth delay. PLX51107 decreased Cox2 expression, increased dendritic cells, and lowered PD-L1, FasL, and IDO-1 expression in the tumor microenvironment. Importantly, PLX51107 delayed the growth of tumors that progressed on anti-PD-1 therapy; a response associated with decreased Cox2 levels, decreased PD-L1 expression on non-immune cells, and increased intratumoral CD8+ T cells. Thus, next-generation BETi represent a potential first-line and secondary treatment strategy for metastatic melanoma by eliciting effects, at least in part, on antitumor CD8+ T cells.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Oxazoles/farmacología , Proteínas/antagonistas & inhibidores , Piridinas/farmacología , Pirroles/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Humanos , Masculino , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Oxazoles/uso terapéutico , Piridinas/uso terapéutico , Pirroles/uso terapéutico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Curr Oncol Rep ; 10(5): 439-46, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18706274

RESUMEN

Melanoma arises through the accrual of mutations in genes critical for proliferation and survival. Although melanoma had been traditionally conceptualized as a cell-autonomous event, increasing evidence supports the notion that these tumors are not isolated entities but rather depend, interact with, and react to the adjacent microenvironment. Melanoma is composed of not only the malignant cells but also the supporting stroma, which includes fibroblasts, endothelial cells, immune cells, soluble molecules, and the extracellular matrix (ECM). Tumor cells actively interact with the microenvironment in a bidirectional manner through molecular signals that modulate the malignant phenotype. This article briefly reviews the molecular basis of melanomagenesis as well as the interplay of melanoma with other cells of the tumor microenvironment and components of the ECM. It also discusses the influence of the microenvironment on therapeutic targeting of melanoma, highlighting recent studies that propose novel strategies to target tumor-microenvironment interactions.


Asunto(s)
Melanoma/patología , Melanoma/terapia , Mutación , Proliferación Celular , Supervivencia Celular , Matriz Extracelular/metabolismo , Humanos , Metaloproteinasas de la Matriz/metabolismo , Modelos Biológicos , Modelos Genéticos , Invasividad Neoplásica , Neovascularización Patológica , Fenotipo , Resultado del Tratamiento
17.
Cancer Res ; 78(19): 5694-5705, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30115697

RESUMEN

The tumor suppressor TP53 is the most frequently mutated gene in human cancer and serves to restrict tumor initiation and progression. Single-nucleotide polymorphisms (SNP) in TP53 and p53 pathway genes can have a marked impact on p53 tumor suppressor function, and some have been associated with increased cancer risk and impaired response to therapy. Approximately 6% of Africans and 1% of African Americans express a p53 allele with a serine instead of proline at position 47 (Pro47Ser). This SNP impairs p53-mediated apoptosis in response to radiation and genotoxic agents and is associated with increased cancer risk in humans and in a mouse model. In this study, we compared the ability of wild-type (WT) and S47 p53 to suppress tumor development and respond to therapy. Our goal was to find therapeutic compounds that are more, not less, efficacious in S47 tumors. We identified the superior efficacy of two agents, cisplatin and BET inhibitors, on S47 tumors compared with WT. Cisplatin caused dramatic decreases in the progression of S47 tumors by activating the p53/PIN1 axis to drive the mitochondrial cell death program. These findings serve as important proof of principle that chemotherapy can be tailored to p53 genotype.Significance: A rare African-derived radioresistant p53 SNP provides proof of principle that chemotherapy can be tailored to TP53 genotype. Cancer Res; 78(19); 5694-705. ©2018 AACR.


Asunto(s)
Polimorfismo de Nucleótido Simple , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , África , Negro o Afroamericano/genética , Alelos , Animales , Apoptosis , Población Negra/genética , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica , Cisplatino/farmacología , Progresión de la Enfermedad , Fibroblastos/metabolismo , Genotipo , Humanos , Ratones , Mitocondrias/metabolismo , Mutación/efectos de los fármacos , Trasplante de Neoplasias , Farmacogenética , Medicina de Precisión , Riesgo
18.
Oncogene ; 37(30): 4058-4072, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29695835

RESUMEN

Targeting RAS is one of the greatest challenges in cancer therapy. Oncogenic mutations in NRAS are present in over 25% of melanomas and patients whose tumors harbor NRAS mutations have limited therapeutic options and poor prognosis. Thus far, there are no clinical agents available to effectively target NRAS or any other RAS oncogene. An alternative approach is to identify and target critical tumor vulnerabilities or non-oncogene addictions that are essential for tumor survival. We investigated the consequences of NRAS blockade in NRAS-mutant melanoma and show that decreased expression of the telomerase catalytic subunit, TERT, is a major consequence. TERT silencing or treatment of NRAS-mutant melanoma with the telomerase-dependent telomere uncapping agent, 6-thio-2'-deoxyguanosine (6-thio-dG), led to rapid cell death, along with evidence of both telomeric and non-telomeric DNA damage, increased ROS levels, and upregulation of a mitochondrial antioxidant adaptive response. Combining 6-thio-dG with the mitochondrial inhibitor Gamitrinib attenuated this adaptive response and more effectively suppressed NRAS-mutant melanoma. Our study uncovers a robust dependency of NRAS-mutant melanoma on TERT, and provides proof-of-principle for a new combination strategy to combat this class of tumors, which could be expanded to other tumor types.


Asunto(s)
GTP Fosfohidrolasas/genética , Melanoma/genética , Proteínas de la Membrana/genética , Mutación/genética , Telomerasa/genética , Animales , Antioxidantes/metabolismo , Línea Celular , Línea Celular Tumoral , Daño del ADN/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mitocondrias/genética , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/genética
19.
EMBO Mol Med ; 10(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29650805

RESUMEN

Despite novel therapies for melanoma, drug resistance remains a significant hurdle to achieving optimal responses. NRAS-mutant melanoma is an archetype of therapeutic challenges in the field, which we used to test drug combinations to avert drug resistance. We show that BET proteins are overexpressed in NRAS-mutant melanoma and that high levels of the BET family member BRD4 are associated with poor patient survival. Combining BET and MEK inhibitors synergistically curbed the growth of NRAS-mutant melanoma and prolonged the survival of mice bearing tumors refractory to MAPK inhibitors and immunotherapy. Transcriptomic and proteomic analysis revealed that combining BET and MEK inhibitors mitigates a MAPK and checkpoint inhibitor resistance transcriptional signature, downregulates the transcription factor TCF19, and induces apoptosis. Our studies demonstrate that co-targeting MEK and BET can offset therapy resistance, offering a salvage strategy for melanomas with no other therapeutic options, and possibly other treatment-resistant tumor types.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Proteínas/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Acetanilidas/farmacología , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/genética , Melanoma/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/metabolismo , Proteómica/métodos , Terapia Recuperativa/métodos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA