Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Br J Cancer ; 122(7): 1023-1036, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32063604

RESUMO

BACKGROUND: Drug resistance remains as one of the major challenges in melanoma therapy. It is well known that tumour cells undergo phenotypic switching during melanoma progression, increasing melanoma plasticity and resistance to mitogen-activated protein kinase inhibitors (MAPKi). METHODS: We investigated the melanoma phenotype switching using a partial reprogramming model to de-differentiate murine melanoma cells and target melanoma therapy adaptation against MAPKi. RESULTS: Here, we show that partially reprogrammed cells are a less proliferative and more de-differentiated cell population, expressing a gene signature for stemness and suppressing melanocyte-specific markers. To investigate adaptation to MAPKi, cells were exposed to B-Raf Proto-Oncogene (BRAF) and mitogen-activated protein kinase kinase (MEK) inhibitors. De-differentiated cells became less sensitive to MAPKi, showed increased cell viability and decreased apoptosis. Furthermore, T-type calcium channels expression increased in adaptive murine cells and in human adaptive melanoma cells. Treatment with the calcium channel blocker mibefradil induced cell death, differentiation and susceptibility to MAPKi in vitro and in vivo. CONCLUSION: In summary, we show that partial reprogramming of melanoma cells induces de-differentiation and adaptation to MAPKi. Moreover, we postulated a calcium channel blocker such as mibefradil, as a potential candidate to restore sensitivity to MAPKi in adaptive melanoma cells.


Assuntos
Canais de Cálcio Tipo T/genética , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Humanos , Melanoma/patologia , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Proto-Oncogene Mas
2.
Int J Cancer ; 145(12): 3462-3477, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31131878

RESUMO

Alterations in histone modifications play a crucial role in the progression of various types of cancer. The histone methyltransferase SETDB1 catalyzes the addition of methyl groups to histone H3 at lysine 9. Here, we describe how overexpression of SETDB1 contributes to melanoma tumorigenesis. SETDB1 is highly amplified in melanoma cells and in the patient tumors. Increased expression of SETDB1, which correlates with SETDB1 amplification, is associated with a more aggressive phenotype in in vitro and in vivo studies. Mechanistically, SETDB1 implements its effects via regulation of thrombospondin 1, and the SET-domain of SETDB1 is essential for the maintenance of its tumorigenic activity. Inhibition of SETDB1 reduces cell growth in melanomas resistant to targeted treatments. Our results indicate that SETDB1 is a major driver of melanoma development and may serve as a potential future target for the treatment of this disease.


Assuntos
Carcinogênese/genética , Histona-Lisina N-Metiltransferase/genética , Melanoma/genética , Melanoma/patologia , Animais , Carcinogênese/patologia , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica/genética , Histonas/genética , Humanos , Lisina/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
3.
Cancer Immunol Immunother ; 68(2): 297-303, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30478475

RESUMO

BACKGROUND: An association between immune-related adverse events (irAEs) caused by immunotherapeutic agents and the clinical benefit of immunotherapy has been suggested. We retrospectively evaluated by means of 18F-FDG PET/CT lymphoid tissue changes in the mediastinal/hilar lymph nodes and the spleen in response to ipilimumab administration in metastatic melanoma. METHODS: A total of 41 patients with unresectable metastatic melanoma underwent 18F-FDG PET/CT before the start of ipilimumab (baseline PET/CT), after two cycles (interim PET/CT) and at the end of treatment (late PET/CT). Data analysis was focused on the mediastinal/hilar lymph nodes and the spleen. The patients' best clinical response (BCR) was used as reference. RESULTS: According to the BCR reference, 31 patients showed disease control (DC) and 10 patients showed progressive disease (PD). Mediastinal/hilar lymph node evaluation revealed that in total 4 patients in the interim or late PET/CT (10%) demonstrated a 'sarcoid-like lymphadenopathy' as response to treatment (LN-positive). All LN-positive patients responded to ipilimumab with DC. On the other hand, no significant differences between the DC and PD groups regarding both semi-quantitative and quantitative 18F-FDG PET spleen-related parameters at baseline and as response to treatment were detected. CONCLUSION: Based on our findings, 10% patients in the interim or late PET/CT showed 'sarcoid-like lymphadenopathy' as response to treatment. All these patients showed disease control, implying a relation between the appearance of sarcoid-like lymphadenopathy and the clinical benefit of anti-CTLA-4 therapy. On the other hand, quantitative 18F-FDG PET analysis of the spleen showed a poor performance in predicting clinical benefit to ipilimumab.


Assuntos
Imunoterapia/métodos , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/diagnóstico por imagem , Melanoma/terapia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Antineoplásicos Imunológicos/uso terapêutico , Feminino , Fluordesoxiglucose F18/farmacocinética , Humanos , Ipilimumab/uso terapêutico , Masculino , Melanoma/imunologia , Melanoma/metabolismo , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Estudos Retrospectivos
4.
Int J Cancer ; 143(11): 2962-2972, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30110134

RESUMO

Recent studies suggest that malignant melanoma heterogeneity includes subpopulations of cells with features of multipotent neural crest (NC) cells. Zebrafish and mouse models have shown that reactivation of neural crest-specific pathways during transformation determines the invasiveness of melanoma cells. In our study, we show that the neural crest-associated transcription factor FOXD1 plays a key role in the invasion and the migration capacities of metastatic melanomas both in vivo and in vitro. Gene expression profiling analysis identified both an upregulation of FOXD1 in NC and melanoma cells, as well as a downregulation of several genes related to cell invasion in FOXD1 knockdown cells, including MMP9 and RAC1B. Furthermore, we demonstrate that knockdown of RAC1B a tumor-specific isoform of RAC1, significantly impaired melanoma cell migration and invasion and could abrogate enhanced invasiveness induced by FOXD1 overexpression. We conclude that FOXD1 may influence invasion and migration via indirect regulation of MMP9 and RAC1B alternative splicing in melanoma cells.


Assuntos
Movimento Celular/genética , Regulação para Baixo/genética , Fatores de Transcrição Forkhead/genética , Melanoma/genética , Invasividade Neoplásica/genética , Crista Neural/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Metaloproteinase 9 da Matriz/genética , Transdução de Sinais/genética
5.
Int J Cancer ; 143(12): 3131-3142, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29905375

RESUMO

Melanoma is often characterized by a constitutively active RAS-RAF-MEK-ERK pathway. For targeted therapy, BRAF inhibitors are available that are powerful in the beginning but resistance occurs rather fast. A better understanding of the mechanisms of resistance is urgently needed to increase the success of the treatment. Here, we observed that SOX2 and CD24 are upregulated upon BRAF inhibitor treatment. A similar upregulation was seen in targeted therapy-resistant, melanoma-derived induced pluripotent cancer cells (iPCCs). SOX2 and CD24 are known to promote an undifferentiated and cancer stem cell-like phenotype associated with resistance. We, therefore, elucidated the role of SOX2 and CD24 in targeted therapy resistance in more detail. We found that the upregulation of SOX2 and CD24 required activation of STAT3 and that SOX2 induced the expression of CD24 by binding to its promoter. We find that the overexpression of SOX2 or CD24 significantly increases the resistance toward BRAF inhibitors, while SOX2 knock-down rendered cells more sensitivity toward treatment. The overexpression of CD24 or SOX2 induced Src and STAT3 activity. Importantly, by either CD24 knock-down or Src/STAT3 inhibition in resistant SOX2-overexpressing cells, the sensitivity toward BRAF inhibitors was re-established. Hence, we suggest a novel mechanism of adaptive resistance whereby BRAF inhibition is circumvented via the activation of STAT3, SOX2 and CD24. Thus, to prevent adaptive resistance, it might be beneficial to combine Src/STAT3 inhibitors together with MAPK pathway inhibitors.


Assuntos
Antineoplásicos/uso terapêutico , Antígeno CD24/metabolismo , Melanoma/tratamento farmacológico , Terapia de Alvo Molecular , Fatores de Transcrição SOXB1/fisiologia , Neoplasias Cutâneas/tratamento farmacológico , Regulação para Cima/fisiologia , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Antígeno CD24/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/genética , Melanoma/metabolismo , Células-Tronco Neoplásicas , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/efeitos dos fármacos , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Fatores de Transcrição SOXB1/genética , Fator de Transcrição STAT3/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo
6.
Cancer Immunol Immunother ; 67(8): 1261-1270, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29872898

RESUMO

BACKGROUND: Immunotherapy has raised the issue of appropriate treatment response evaluation, due to the unique mechanism of action of the immunotherapeutic agents. Aim of this analysis is to evaluate the potential role of quantitative analysis of 2-deoxy-2-(18F)fluoro-D-glucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) data in monitoring of patients with metastatic melanoma undergoing ipilimumab therapy. METHODS: 25 patients with unresectable metastatic melanoma underwent dynamic PET/CT (dPET/CT) of the thorax and upper abdomen as well as static, whole body PET/CT with 18F-FDG before the start of ipilimumab treatment (baseline PET/CT), after two cycles of treatment (interim PET/CT) and at the end of treatment after four cycles (late PET/CT). The evaluation of dPET/CT studies was based on semi-quantitative (standardized uptake value, SUV) calculation as well as quantitative analysis, based on two-tissue compartment modeling and a fractal approach. Patients' best clinical response, assessed at a mean of 59 weeks, was used as reference. RESULTS: According to their best clinical response, patients were dichotomized in those demonstrating clinical benefit (CB, n = 16 patients) and those demonstrating no clinical benefit (no-CB, n = 9 patients). No statistically significant differences were observed between CB and no-CB regarding either semi-quantitative or quantitative parameters in all scans. On contrary, the application of the recently introduced PET response evaluation criteria for immunotherapy (PERCIMT) led to a correct classification rate of 84% (21/25 patients). CONCLUSION: Quantitative analysis of 18F-FDG PET data does not provide additional information in treatment response evaluation of metastatic melanoma patients receiving ipilimumab. PERCIMT criteria correlated better with clinical response.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Fluordesoxiglucose F18 , Ipilimumab/uso terapêutico , Melanoma/secundário , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Compostos Radiofarmacêuticos , Feminino , Humanos , Cinética , Estudos Longitudinais , Masculino , Melanoma/diagnóstico por imagem , Melanoma/tratamento farmacológico , Pessoa de Meia-Idade , Prognóstico
7.
Eur J Nucl Med Mol Imaging ; 45(8): 1289-1296, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29478079

RESUMO

PURPOSE: The aim of the present study was to assess the value of interim 18F-FDG PET/CT performed after the first two cycles of ipilimumab treatment in the prediction of the final clinical response to this type of immunotherapy. METHODS: The study group comprised 41 patients with unresectable metastatic melanoma scheduled for ipilimumab therapy. Whole-body 18F-FDG PET/CT was performed before the start of ipilimumab treatment (baseline PET/CT) and after the initial two cycles of ipilimumab treatment (interim PET/CT). Evaluation of patient response to treatment was based on the European Organization for Research and Treatment of Cancer (EORTC) 1999 criteria for PET as well as the recently proposed PET Response Evaluation Criteria for Immunotherapy (PERCIMT). The patients' best clinical response, assessed at a median of 21.4 months (range 6.3-41.9 months) was used as reference. RESULTS: According to their best clinical response, the patients were divided into two groups: those showing clinical benefit (CB) including stable disease, partial response and complete response (31 patients), and those showing no clinical benefit (no-CB including progressive disease (10 patients). According to the EORTC criteria, interim PET/CT demonstrated progressive metabolic disease (PMD) in 20 patients, stable metabolic disease (SMD) in 11 patients, partial metabolic response (PMR) in 8 patients, and complete metabolic response (CMR) in 2 patients. According to the PERCIMT, interim PET/CT demonstrated PMD in 9 patients, SMD in 24 patients, PMR in 6 patients and CMR in 2 patients. On the basis of the interim PET, the patients were divided in a similar manner to the division according to clinical response into those showing metabolic benefit (MB) including SMD, PMR and CMR, and those showing no metabolic benefit (no-MB) including PMD. According to this dichotomization, the EORTC criteria showed a sensitivity (correctly predicting CB) of 64.5%, a specificity (correctly predicting no-CB) of 90.0%, a positive predictive value (PPV) of 95.2%, a negative predictive value (NPV) of 45.0% and an accuracy of 70.7% in predicting best clinical response. The PERCIMT showed a sensitivity of 93.6%, a specificity of 70.0%, a PPV of 90.6%, a NPV of 77.8% and an accuracy of 87.8%. The McNemar test showed that the PERCIMT had a significantly higher sensitivity than EORTC criteria (p = 0.004), while there was no significant difference in specificity (p = 0.5). The agreement between the two sets of criteria was poor (McNemar test p = 0.001, and accordingly kappa = 0.46). CONCLUSION: The application of the recently proposed PERCIMT to interim 18F-FDG PET/CT provides a more sensitive predictor of final clinical response to immunotherapy than the application of the EORTC criteria in patients with metastatic melanoma.


Assuntos
Ipilimumab/uso terapêutico , Melanoma/tratamento farmacológico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Neoplasias Cutâneas/tratamento farmacológico , Feminino , Fluordesoxiglucose F18 , Humanos , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Neoplasias Cutâneas/patologia , Resultado do Tratamento
8.
Stem Cells ; 34(4): 832-46, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26753613

RESUMO

The combination of cancer-focused studies and research related to nuclear reprogramming has gained increasing importance since both processes-reprogramming towards pluripotency and malignant transformation-share essential features. Studies have revealed that incomplete reprogramming of somatic cells leads to malignant transformation indicating that epigenetic regulation associated with iPSC generation can drive cancer development [J Mol Cell Biol 2011;341-350; Cell 2012;151:1617-1632; Cell 2014;156:663-677]. However, so far it is unclear whether incomplete reprogramming also affects cancer cells and their function. In the context of melanoma, dedifferentiation correlates to therapy resistance in mouse studies and has been documented in melanoma patients [Nature 2012;490:412-416; Clin Cancer Res 2014;20:2498-2499]. Therefore, we sought to investigate directed dedifferentiation using incomplete reprogramming of melanoma cells. Using a murine model we investigated the effects of partial reprogramming on the cellular plasticity of melanoma cells. We demonstrate for the first time that induced partial reprogramming results in a reversible phenotype switch in melanoma cells. Partially reprogrammed cells at day 12 after transgene induction display elevated invasive potential in vitro and increased lung colonization in vivo. Additionally, using global gene expression analysis of partially reprogrammed cells, we identified SNAI3 as a novel invasion-related marker in human melanoma. SNAI3 expression correlates with tumor thickness in primary melanomas and thus, may be of prognostic value. In summary, we show that investigating intermediate states during the process of reprogramming melanoma cells can reveal novel insights into the pathogenesis of melanoma progression. We propose that deeper analysis of partially reprogrammed melanoma cells may contribute to identification of yet unknown signaling pathways that can drive melanoma progression.


Assuntos
Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/patologia , Melanoma/genética , Neoplasias Experimentais/genética , Fatores de Transcrição da Família Snail/biossíntese , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Desdiferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/patologia , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/biossíntese , Neoplasias Experimentais/patologia , Transdução de Sinais , Fatores de Transcrição da Família Snail/genética
9.
Int J Cancer ; 138(9): 2257-62, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26684394

RESUMO

Recent therapeutic advances have improved melanoma patients clinical outcome. Novel therapeutics targeting BRAF, NRAS and cKit mutant melanomas are widely used in clinical practice. However therapeutic options in NRAS(wild-type) /BRAF(wild-type) /cKit(wild-type) melanoma patients are limited. Our study shows that gene copy numbers of members of the MAPK signaling pathway vary in different melanoma subgroups. NRAS(wild-type) /BRAF(wild-type) melanoma metastases are characterized by significant gains of MAP2K1 (MEK1) and MAPK3 (ERK1) gene loci. These additional gene copies could lead to an activation of the MAPK signaling pathway via a gene-dosage effect. Our results suggest that downstream analyses of the pMEK and pERK expression status in NRAS(wild-type) /BRAF(wild-type) melanoma patients identify patients that could benefit from targeted therapies with MEK and ERK inhibitors.


Assuntos
Dosagem de Genes , Sistema de Sinalização das MAP Quinases/genética , Melanoma/genética , Melanoma/patologia , Neoplasias Cutâneas/genética , Idoso , Análise Mutacional de DNA , Feminino , GTP Fosfo-Hidrolases/genética , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/patologia
10.
Eur J Nucl Med Mol Imaging ; 42(3): 386-96, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25359635

RESUMO

PURPOSE: Ipilimumab is a newly approved immunotherapeutic agent that has been shown to provide a survival benefit in patients with metastatic melanoma. (18)F-FDG PET/CT has demonstrated very satisfying results in detecting melanoma metastases in general. Using (18)F-FDG PET/CT we monitored patients with metastatic melanoma undergoing ipilimumab therapy during the course of treatment. The aim of our study was to evaluate the role of (18)F-FDG PET/CT performed after two cycles of ipilimumab in predicting the final response to therapy. METHODS: In 22 patients suffering from unresectable metastatic melanoma, scheduled for ipilimumab treatment PET/CT scanning was performed before the start of treatment (baseline scan), after two cycles of treatment (early response) and at the end of treatment after four cycles (late response). Evaluation of the patient response to treatment on PET was based on the European Organization for Research and Treatment of Cancer 1999 criteria. Progression-free survival (PFS) and overall survival (OS) data are presented. RESULTS: After the end of treatment, 15 patients were characterized as having progressive metabolic disease (PMD) and five as having stable metabolic disease (SMD), and two patients showed a partial metabolic response (PMR). Early PET/CT performed after two ipilimumab cycles predicted treatment response in 13 of the 15 PMD patients, in five of the five SMD patients and in neither of the two PMR patients. Both patients with PMR showed pseudoprogression after the second cycle and were therefore wrongly classified. According to the patients' clinical outcome, patients with late PMD had a median PFS of 3.6 months (mean 5.6 months), while patients with late SMD had a median PFS of 9.8 months (mean 9.0 months). In comparison, patients with early PMD had a median PFS of 2.7 months (mean 5.5 months) and patients with early SMD had a median PFS of 6.3 months (mean 7.5 months). The difference in PFS between the two groups was statistically significant for both early and late responses (log-rank p < 0.001). Median OS among patients with late PMD was 9.1 months (mean 11.2 months) and among those with late SMD 9.8 months (mean 10.7 months). The difference in OS between the two groups was statistically significant (log-rank p = 0.013). The median OS among patients with early PMD was 8.8 months (mean 12.0 months) and among those with early SMD 9.8 months (mean 10.0 months). The difference in OS between the two groups was statistically significant (log-rank p < 0.001). CONCLUSION: (18)F-FDG PET/CT after two cycles of ipilimumab is highly predictive of the final treatment outcome in patients with PMD and SMD.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Fluordesoxiglucose F18 , Melanoma/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Adolescente , Adulto , Idoso , Feminino , Humanos , Ipilimumab , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Pessoa de Meia-Idade , Imagem Multimodal , Metástase Neoplásica , Valor Preditivo dos Testes , Tomografia Computadorizada por Raios X , Resultado do Tratamento
11.
Proc Natl Acad Sci U S A ; 108(36): 14861-6, 2011 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-21856949

RESUMO

Melanocytes are essential for skin homeostasis and protection, and their defects in humans lead to a wide array of diseases that are potentially extremely severe. To date, the analysis of molecular mechanisms and the function of human melanocytes have been limited because of the difficulties in accessing large numbers of cells with the specific phenotypes. This issue can now be addressed via a differentiation protocol that allows melanocytes to be obtained from pluripotent stem cell lines, either induced or of embryonic origin, based on the use of moderate concentrations of a single cytokine, bone morphogenic protein 4. Human melanocytes derived from pluripotent stem cells exhibit all the characteristic features of their adult counterparts. This includes the enzymatic machinery required for the production and functional delivery of melanin to keratinocytes. Melanocytes also integrate appropriately into organotypic epidermis reconstructed in vitro. The availability of human cells committed to the melanocytic lineage in vitro will enable the investigation of those mechanisms that guide the developmental processes and will facilitate analysis of the molecular mechanisms responsible for genetic diseases. Access to an unlimited resource may also prove a vital tool for the treatment of hypopigmentation disorders when donors with matching haplotypes become available in clinically relevant banks of pluripotent stem cell lines.


Assuntos
Células-Tronco Adultas/citologia , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Epidérmicas , Melanócitos/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Adultas/metabolismo , Linhagem Celular , Epiderme/metabolismo , Humanos , Hipopigmentação/metabolismo , Hipopigmentação/terapia , Melanócitos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transplante de Células-Tronco
12.
Cancers (Basel) ; 13(22)2021 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-34830853

RESUMO

The ability to detect minimal residual disease (MRD) after a curative-intent surgery or treatment is of paramount importance, because it offers the possibility to help guide the clinical decisions related adjuvant therapy. Thus, the earlier MRD is detected, the earlier potentially beneficial treatment can be proposed to patients who might need it. Liquid biopsies, and in particular the next-generation sequencing of circulating tumor DNA (ctDNA) in the blood, have been the focus of an increasing amount of research in the past years. The ctDNA detection at advanced cancer stages is practicable for several solid tumors, and complements molecular information on acquired therapy resistance. In the context of MRD, it is by definition more challenging to detect ctDNA, but it is technically achievable and provides information on treatment response and probability of relapse significantly earlier than standard imaging methods. The clinical benefit of implementing this new technique in the routine is being tested in interventional clinical trials at the moment. We propose here an update of the current use of ctDNA detection by NGS as a tool to assess the presence of MRD and improve adjuvant treatment of solid tumors. We also discuss the main limitations and medium-term perspectives of this process in the clinic.

13.
J Clin Med ; 10(15)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34362135

RESUMO

The precise role played by the tumor suppressor gene NF1 in melanocyte biology and during the transformation into melanoma is not completely understood. In particular, understanding the interaction during melanocyte development between NF1 and key signaling pathways, which are known to be reactivated in advanced melanoma, is still under investigation. Here, we used RNAseq datasets from either situation to better understand the transcriptomic regulation mediated by an NF1 partial loss of function. We found that NF1 mutations had a differential impact on pluripotency and on melanoblast differentiation. In addition, major signaling pathways such as VEGF, senescence/secretome, endothelin, and cAMP/PKA are likely to be upregulated upon NF1 loss of function in both melanoblasts and metastatic melanoma. In sum, these data bring new light on the transcriptome regulation of the NF1-mutated melanoma subgroup and will help improve the possibilities for specific treatment.

14.
Artigo em Inglês | MEDLINE | ID: mdl-34281000

RESUMO

To date, more than 160 million people have been infected with COVID-19 worldwide. In the present study, we investigated the history of SARS-CoV-2 infection among 3067 healthcare workers (HCW) in a German COVID-19 treatment center during the early phase of the pandemic (July 2020) based on the seroprevalence of SARS-CoV-2 antibodies and self-reported previous PCR results. The results demonstrate a low prevalence of SARS-CoV-2 infection (n = 107 [3.5%]) with no increased risk for employees with a high level of patient exposure in general or working in COVID-19-confined areas in particular. This suggests that the local hygiene standards implemented in our hospital during the first wave of COVID-19 pandemic were effective in preventing patient-to-HCW transmission. No evidence for highly mobile staff serving as a vector for SARS-CoV-2 transmission could be found. In addition, impairment of smell and/or taste was strongly associated with SARS-CoV-2 history.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Pessoal de Saúde , Humanos , Pandemias , Estudos Soroepidemiológicos
15.
J Cell Biol ; 170(1): 49-59, 2005 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-15983061

RESUMO

In melanocytes and melanoma cells alpha-melanocyte stimulating hormone (alpha-MSH), via the cAMP pathway, elicits a large array of biological responses that control melanocyte differentiation and influence melanoma development or susceptibility. In this work, we show that cAMP transcriptionally activates Hif1a gene in a melanocyte cell-specific manner and increases the expression of a functional hypoxia-inducible factor 1alpha (HIF1alpha) protein resulting in a stimulation of Vegf expression. Interestingly, we report that the melanocyte-specific transcription factor, microphthalmia-associated transcription factor (MITF), binds to the Hif1a promoter and strongly stimulates its transcriptional activity. Further, MITF "silencing" abrogates the cAMP effect on Hif1a expression, and overexpression of MITF in human melanoma cells is sufficient to stimulate HIF1A mRNA. Our data demonstrate that Hif1a is a new MITF target gene and that MITF mediates the cAMP stimulation of Hif1a in melanocytes and melanoma cells. Importantly, we provide results demonstrating that HIF1 plays a pro-survival role in this cell system. We therefore conclude that the alpha-MSH/cAMP pathway, using MITF as a signal transducer and HIF1alpha as a target, might contribute to melanoma progression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Melanócitos/metabolismo , Melanoma/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , AMP Cíclico/metabolismo , AMP Cíclico/farmacologia , Proteínas de Ligação a DNA/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/genética , Genes Reguladores/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Melanócitos/citologia , Melanócitos/efeitos dos fármacos , Melanoma/genética , Camundongos , Fator de Transcrição Associado à Microftalmia , Células NIH 3T3 , Regiões Promotoras Genéticas/fisiologia , Interferência de RNA/efeitos dos fármacos , Interferência de RNA/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/genética , alfa-MSH/metabolismo
16.
Cancers (Basel) ; 12(6)2020 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-32532044

RESUMO

Tumorigenesis is correlated with abnormal expression and activity of G protein-coupled receptors (GPCRs) and associated G proteins. Oncogenic mutations in both GPCRs and G proteins (GNAS, GNAQ or GNA11) encoding genes have been identified in a significant number of tumors. Interestingly, uveal melanoma driver mutations in GNAQ/GNA11 were identified for a decade, but their discovery did not lead to mutation-specific drug development, unlike it the case for BRAF mutations in cutaneous melanoma which saw enormous success. Moreover, new immunotherapies strategies such as immune checkpoint inhibitors have given underwhelming results. In this review, we summarize the current knowledge on cancer-associated alterations of GPCRs and G proteins and we focus on the case of uveal melanoma. Finally, we discuss the possibilities that this signaling might represent in regard to novel drug development for cancer prevention and treatment.

17.
Cancers (Basel) ; 12(1)2020 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-31906480

RESUMO

Melanoma patients carrying an oncogenic NRAS mutation represent 20% of all cases and present worse survival, relapse rate and therapy response than patients with wild type NRAS or with BRAF mutations. Nevertheless, no efficient targeted therapy has emerged so far for this group of patients in comparison with the classical combination of BRAF and MEK inhibitors for the patient group carrying a BRAF mutation. NRAS key downstream actors should therefore be identified for drug targeting, possibly in combination with MEK inhibitors. Here, we investigated the influence of different melanoma-associated NRAS mutations (codon 12, 13 or 61) on several parameters such as oncogene-induced senescence, cell proliferation, migration or colony formation in immortalized melanocytes and in melanoma cell lines. We identified AXL/STAT3 axis as a main regulator of NRASQ61-induced oncogene-induced senescence (OIS) and observed that NRASQ61 mutations are not only more tumorigenic than NRASG12/13 mutations but also associated to STAT3 activation. In conclusion, these data bring new evidence of the potential tumorigenic role of STAT3 in NRAS-mutant melanomas and will help improving current therapy strategies for this particular patient group.

18.
Mol Ther Oncolytics ; 18: 83-99, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32637583

RESUMO

Malignant melanoma is the most deadly skin cancer, associated with rising incidence and mortality rates. Most of the patients with melanoma, treated with current targeted therapies, develop a drug resistance, causing tumor relapse. The attainment of a better understanding of novel cancer-promoting molecular mechanisms driving melanoma progression is essential for the development of more effective targeted therapeutic approaches. Recent studies, including the research previously conducted in our laboratory, reported that the histone methyltransferase SETDB1 contributes to melanoma pathogenesis. In this follow-up study, we further elucidated the role of SETDB1 in melanoma, showing that SETDB1 modulated relevant transcriptomic effects in melanoma, in particular, as activator of cancer-related secreted (CRS) factors and as repressor of melanocyte-lineage differentiation (MLD) and metabolic enzymes. Next, we investigated the effects of SETDB1 inhibition via compounds belonging to the mithramycin family, mithramycin A and mithramycin analog (mithralog) EC-8042: melanoma cells showed strong sensitivity to these drugs, which effectively suppressed the expression of SETDB1 and induced changes at the transcriptomic, morphological, and functional level. Moreover, SETDB1 inhibitors enhanced the efficacy of mitogen-activated protein kinase (MAPK) inhibitor-based therapies against melanoma. Taken together, this work highlights the key regulatory role of SETDB1 in melanoma and supports the development of SETDB1-targeting therapeutic strategies for the treatment of melanoma patients.

19.
Transl Oncol ; 13(12): 100858, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32891903

RESUMO

Metastases's spreading is the main cause of mortality for advanced stage cancer patients, including melanoma. The formation of metastases is favored by enhanced migratory and invasive capacities of tumor cells. Tumor suppressor gene NF1 is a negative regulator of RAS and its deregulation plays an important role in several aspects of melanoma transformation and progression. However, very little is described about the role of NF1 in cellular migration and invasion. In this study, our results show on the one hand, that the loss of NF1 expression delays migration of human melanoblasts via a RAC1-dependent mechanism. On the other hand, our data indicate that NF1 loss in melanoma cells is enhancing migration, intravasation and metastases formation in vivo. Moreover, not only this phenotype is associated with an upregulation of PREX1 but also patient-derived melanoma samples with low NF1 expression present increased levels of PREX1. In sum, our study brings new elements on the mechanism controlling cellular migration in the context of NF1 loss. These data are of prime interest to improve treatment strategies against all NF1-mutated tumors, including this subtype of melanoma.

20.
Front Mol Neurosci ; 12: 111, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118886

RESUMO

During development, neural crest (NC) cells are early precursors of several lineages including melanocytes. Along their differentiation from multipotent cells to mature melanocytes, NC cells will go through successive steps which require either proliferative or motile capacities. For example, they will undergo Epithelial to Mesenchymal Transition (EMT) in order the separate from the neural tube and migrate to their final location in the epidermis (Larribere and Utikal, 2013; Skrypek et al., 2017). The differentiated melanocytes are the cells of origin of melanoma tumors which progress through several stages such as radial growth phase, vertical growth phase, metastasis formation, and often resistance to current therapies. Interestingly, depending on the stage of the disease, melanoma tumor cells share phenotypes with NC cells (proliferative, motile, EMT). These phenotypes are tightly controlled by specific signaling pathways and transcription factors (TFs) which tend to be reactivated during the onset of melanoma. In this review, we summarize first the main TFs which control these common phenotypes. Then, we focus on the existing strategies used to generate human NCs. Finally we discuss how identification and regulation of NC-associated genes provide an additional approach to improving current melanoma targeted therapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA