Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep Med ; 4(11): 101287, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37967556

RESUMEN

The efficacy of immune checkpoint inhibitors varies in clear-cell renal cell carcinoma (ccRCC), with notable primary resistance among patients. Here, we integrate epigenetic (DNA methylation) and transcriptome data to identify a ccRCC subtype characterized by cancer-specific promoter hypermethylation and epigenetic silencing of Polycomb targets. We develop and validate an index of methylation-based epigenetic silencing (iMES) that predicts primary resistance to immune checkpoint inhibition (ICI) in the BIONIKK trial. High iMES is associated with VEGF pathway silencing, endothelial cell depletion, immune activation/suppression, EZH2 activation, BAP1/SETD2 deficiency, and resistance to ICI. Combination therapy with hypomethylating agents or tyrosine kinase inhibitors may benefit patients with high iMES. Intriguingly, tumors with low iMES exhibit increased endothelial cells and improved ICI response, suggesting the importance of angiogenesis in ICI treatment. We also develop a transcriptome-based analogous system for extended applicability of iMES. Our study underscores the interplay between epigenetic alterations and tumor microenvironment in determining immunotherapy response.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Metilación de ADN/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Microambiente Tumoral/genética , Células Endoteliales/metabolismo , Inmunoterapia
2.
Nat Commun ; 14(1): 7884, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-38036539

RESUMEN

Wilms tumors are highly curable in up to 90% of cases with a combination of surgery and radio-chemotherapy, but treatment-resistant types such as diffuse anaplastic Wilms tumors pose significant therapeutic challenges. Our multi-omics profiling unveils a distinct desert-like diffuse anaplastic Wilms tumor subtype marked by immune/stromal cell depletion, TP53 alterations, and cGAS-STING pathway downregulation, accounting for one-third of all diffuse anaplastic cases. This subtype, also characterized by reduced CD8 and CD3 infiltration and active oncogenic pathways involving histone deacetylase and DNA repair, correlates with poor clinical outcomes. These oncogenic pathways are found to be conserved in anaplastic Wilms tumor cell models. We identify histone deacetylase and/or WEE1 inhibitors as potential therapeutic vulnerabilities in these tumors, which might also restore tumor immunogenicity and potentially enhance the effects of immunotherapy. These insights offer a foundation for predicting outcomes and personalizing treatment strategies for aggressive pediatric Wilms tumors, tailored to individual immunological landscapes.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Niño , Humanos , Neoplasias Renales/genética , Neoplasias Renales/terapia , Neoplasias Renales/metabolismo , Tumor de Wilms/genética , Tumor de Wilms/terapia , Histona Desacetilasas
3.
Clin Cancer Res ; 29(7): 1279-1291, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36374555

RESUMEN

PURPOSE: Immune checkpoint inhibitors (ICI) have revolutionized the treatment of patients with clear-cell renal cell carcinomas (ccRCC). Although analyses of transcriptome, genetic alterations, and the tumor microenvironment (TME) have shed light into mechanisms of response and resistance to these agents, the role of epigenetic alterations in this process remains fully unknown. EXPERIMENTAL DESIGN: We investigated the methylome of six ccRCC cohorts as well as one cell line dataset. Of note, we took advantage of the BIONIKK trial aiming to tailor treatments according to Paris Descartes 4-gene expression subgroups, and performed Illumina EPIC profiling for 46 samples related to patients treated with ipilimumab plus nivolumab, and 17 samples related to patients treated with sunitinib. RESULTS: A group of tumors associated with enhancer demethylation was discovered, namely TED. TED was associated with tumors with sarcomatoid differentiation and poor clinical outcome. TED harbored TET1 promoter demethylation, activated the gene expression signature of epithelial-mesenchymal transition and IL6/JAK/STAT3 pathways, and displayed a TME characterized by both immune activation and suppressive populations, fibroblast infiltration, and endothelial depletion. In addition, TED was a predictive factor of resistance to the combination of first-line ipilimumab-nivolumab in the BIONIKK clinical trial. Finally, TED was associated with activation of specific regulons, which we also found to be predictive of resistance to immunotherapy in an independent cohort. CONCLUSIONS: We report on the discovery of a novel epigenetic phenotype associated with resistance to ICIs that may pave the way to better personalizing patients' treatments. See related commentary by Zhou and Kim, p. 1170.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Nivolumab/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/patología , Ipilimumab/administración & dosificación , Metilación de ADN , Fenotipo , Microambiente Tumoral/genética , Oxigenasas de Función Mixta , Proteínas Proto-Oncogénicas/genética
4.
Genome Biol ; 22(1): 7, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397444

RESUMEN

BACKGROUND: Crosstalk between genetic, epigenetic, and immune alterations in upper tract urothelial carcinomas and their role in shaping muscle invasiveness and patient outcome are poorly understood. RESULTS: We perform an integrative genome- and methylome-wide profiling of diverse non-muscle-invasive and muscle-invasive upper tract urothelial carcinomas. In addition to mutations of FGFR3 and KDM6A, we identify ZFP36L1 as a novel, significantly mutated tumor suppressor gene. Overall, mutations of ZFP36 family genes (ZFP36, ZFP36L1, and ZFP36L2) are identified in 26.7% of cases, which display a high mutational load. Unsupervised DNA methylation subtype classification identifies two epi-clusters associated with distinct muscle-invasive status and patient outcome, namely, EpiC-low and EpiC-high. While the former is hypomethylated, immune-depleted, and enriched for FGFR3-mutated, the latter is hypermethylated, immune-infiltrated, and tightly associated with somatic mutations of SWI/SNF genes. CONCLUSIONS: Our study delineates for the first time the key role for convergence between genetic and epigenetic alterations in shaping clinicopathological and immune upper tract urothelial carcinoma features.


Asunto(s)
Carcinoma de Células Transicionales/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Vejiga Urinaria/genética , Factor 1 de Respuesta al Butirato/genética , Carcinoma de Células Transicionales/inmunología , Línea Celular Tumoral , Metilación de ADN , Epigenómica , Histona Demetilasas/genética , Humanos , Inmunidad , Mutación , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Factores de Transcripción/genética , Transcriptoma , Tristetraprolina/genética , Neoplasias de la Vejiga Urinaria/inmunología
5.
Sci Rep ; 10(1): 701, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959902

RESUMEN

Sarcomatoid clear-cell renal cell carcinomas (sRCC) are associated with dismal prognosis. Genomic alterations associated with sarcomatoid dedifferentiation are poorly characterized. We sought to define the genomic landscape of sRCC and uncover potentially actionable therapeutic targets. We assessed the genomic landscape of sRCC using targeted panel sequencing including patients with microdissected sarcomatoid and epithelial components. Along with common genomic alterations associated with clear-cell histology, we found that Hippo was one of the most frequently altered pathways in these tumours. Hippo alterations were differentially enriched in sRCC compared to non-sRCC. Functional analysis showed that Hippo members mutations were associated with higher nuclear accumulation of YAP/TAZ, core effectors of the Hippo pathway. In a NF2-mutant sRCC model, YAP1 knockdown and NF2 reconstitution suppressed cell proliferation, tumour growth and invasion, both in vitro and in vivo. Overall, we show that Hippo pathway alterations are a feature of sRCC, and enable the exploration of the Hippo pathway as a novel potential therapeutic target.


Asunto(s)
Carcinoma de Células Renales/genética , Redes Reguladoras de Genes , Neoplasias Renales/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Predisposición Genética a la Enfermedad , Vía de Señalización Hippo , Humanos , Captura por Microdisección con Láser , Masculino , Ratones , Trasplante de Neoplasias , Neurofibromina 2/genética , Proteínas Serina-Treonina Quinasas/genética , Análisis de Secuencia de ADN/métodos , Transducción de Señal , Transactivadores/genética , Factores de Transcripción/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Regulación hacia Arriba , Proteínas Señalizadoras YAP
6.
Cell Rep ; 25(3): 811-821.e5, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332658

RESUMEN

Esthesioneuroblastoma (ENB) is a rare cancer of the olfactory mucosa, with no established molecular stratification to date. We report similarities of ENB with tumors arising in the neural crest and perform integrative analysis of these tumors. We propose a molecular-based subtype classification of ENB as basal or neural, both of which have distinct pathological, transcriptomic, proteomic, and immune features. Among the basal subtype, we uncovered an IDH2 R172 mutant-enriched subgroup (∼35%) harboring a CpG island methylator phenotype reminiscent of IDH2 mutant gliomas. Compared with the basal ENB methylome, the neural ENB methylome shows genome-wide reprogramming with loss of DNA methylation at the enhancers of axonal guidance genes. Our study reveals insights into the molecular pathogenesis of ENB and provides classification information of potential therapeutic relevance.


Asunto(s)
Biomarcadores de Tumor/análisis , Linaje de la Célula/genética , Metilación de ADN , Estesioneuroblastoma Olfatorio/genética , Variación Genética , Cavidad Nasal/metabolismo , Neoplasias Nasales/genética , Biología Computacional , Islas de CpG , Epigénesis Genética , Estesioneuroblastoma Olfatorio/clasificación , Estesioneuroblastoma Olfatorio/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfocitos Infiltrantes de Tumor , Masculino , Persona de Mediana Edad , Cavidad Nasal/patología , Neoplasias Nasales/clasificación , Neoplasias Nasales/metabolismo , Pronóstico , Proteoma/análisis , Tasa de Supervivencia , Transcriptoma
7.
Sci Rep ; 7(1): 8540, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819235

RESUMEN

Prediction of recurrence is a challenge for the development of adjuvant treatments in clear-cell renal cell carcinoma (ccRCC). In these tumors, expression of long non-coding RNAs (lncRNAs) are deregulated and closely associated with prognosis. Thus, we aimed to predict ccRCC recurrence risk using lncRNA expression. We identified prognostic lncRNAs in a training set of 351 localized ccRCCs from The Cancer Genome Atlas and validated lncRNA-based recurrence classification in an independent cohort of 167 localized ccRCCs. We identified lncRNA MFI2-AS1 as best candidate in the training set. In the validation cohort, MFI2-AS1 expression was independently associated with shorter disease-free survival (Hazard Ratio (HR) for relapse 3.5, p = 0.0001). Combined with Leibovich classification, MFI2-AS1 status improved prediction of recurrence (C-index 0.70) compared to MFI2-AS1 alone (0.67) and Leibovich classification alone (0.66). In patients with aggressive tumors (Leibovich ≥5), MFI2-AS1 expression was associated with dramatically increased risk of relapse (HR 12.16, p < 0.0001) compared to patients with undetectable MFI2-AS1 who had favorable outcomes. Compared to normal samples, MFI2-AS1 was upregulated in tumor tissue, and higher expression was associated with metastatic dissemination. Overall, MFI2-AS1 status improves patient stratification in localized ccRCC, which supports further integration of lncRNAs in molecular cancer classifications.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales/genética , ARN Largo no Codificante/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Renales/patología , Estudios de Cohortes , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Pronóstico
8.
Cancer Res ; 77(18): 4835-4845, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28754676

RESUMEN

Extensive dysregulation of chromatin-modifying genes in clear cell renal cell carcinoma (ccRCC) has been uncovered through next-generation sequencing. However, a scientific understanding of the cross-talk between epigenetic and genomic aberrations remains limited. Here we identify three ccRCC epigenetic clusters, including a clear cell CpG island methylator phenotype (C-CIMP) subgroup associated with promoter methylation of VEGF genes (FLT4, FLT1, and KDR). C-CIMP was furthermore characterized by silencing of genes related to vasculature development. Through an integrative analysis, we discovered frequent silencing of the histone H3 K36 methyltransferase NSD1 as the sole chromatin-modifying gene silenced by DNA methylation in ccRCC. Notably, tumors harboring NSD1 methylation were of higher grade and stage in different ccRCC datasets. NSD1 promoter methylation correlated with SETD2 somatic mutations across and within spatially distinct regions of primary ccRCC tumors. ccRCC harboring epigenetic silencing of NSD1 displayed a specific genome-wide methylome signature consistent with the NSD1 mutation methylome signature observed in Sotos syndrome. Thus, we concluded that epigenetic silencing of genes involved in angiogenesis is a hallmark of the methylator phenotype in ccRCC, implying a convergence toward loss of function of epigenetic writers of the H3K36 histone mark as a root feature of aggressive ccRCC. Cancer Res; 77(18); 4835-45. ©2017 AACR.


Asunto(s)
Carcinoma de Células Renales/patología , Metilación de ADN , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Renales/patología , Mutación , Proteínas Nucleares/genética , Anciano , Apoptosis , Biomarcadores de Tumor , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Proliferación Celular , Epigénesis Genética , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Humanos , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , Tasa de Supervivencia , Células Tumorales Cultivadas
9.
Sci Rep ; 6: 30988, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27484008

RESUMEN

Collecting duct carcinoma (CDC) is a kidney cancer subtype that is thought to arise from principal cells in distal parts of the collecting ducts. Some studies suggested an overlap of CDC with upper tract urothelial carcinoma (UTUC), making the pathological diagnosis challenging. Herein, we performed for the first time transcriptome sequencing of CDC and compared them to UTUC and renal cell carcinoma subtypes. We discovered that CDC displays a unique transcriptomic signature among kidney cancer subtypes, with a putative cell of origin in the distal convoluted tubules. Hierarchical unsupervised clustering reveals that the CDC signature is closer to that of other RCC subtypes than to UTUC, which is similar to that of bladder carcinoma. CDC is characterized by a metabolic shift, with impairment of oxidoreductase activity, pyruvate metabolism and the tricarboxlyic acid cycle, as well as an immunogenic response consistent with increased tumor infiltrating lymphocytes, particularly within metastatic cases. In addition, pathways differentially altered between CDC and UTUC point to a basal-like phenotype of CDC in contrast to the luminal-like signature of UTUC. We conclude that CDC harbors a pathognomonic transcriptomic signature characterized by immunogenic and a metabolic aberrations, indicating that targeting these processes might provide therapeutic options for patients.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Neoplasias Urológicas/genética , Adolescente , Adulto , Anciano , Carcinoma de Células Renales/patología , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Transcriptoma , Neoplasias Urológicas/patología , Adulto Joven
10.
Clin Cancer Res ; 22(24): 6236-6246, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27256309

RESUMEN

PURPOSE: DNA methylation is a heritable covalent modification that is developmentally regulated and is critical in tissue-type definition. Although genotype-phenotype correlations have been described for different subtypes of renal cell carcinoma (RCC), it is unknown if DNA methylation profiles correlate with morphological or ontology based phenotypes. Here, we test the hypothesis that DNA methylation signatures can discriminate between putative precursor cells in the nephron. EXPERIMENTAL DESIGNS: We performed deep profiling of DNA methylation and transcriptome in diverse histopathological RCC subtypes and validated DNA methylation in an independent dataset as well as in The Cancer Genome Atlas Clear Cell and Chromophobe Renal Cell Carcinoma Datasets. RESULTS: Our data provide the first mapping of methylome epi-signature and indicate that RCC subtypes can be grouped into two major epi-clusters: C1, which encompasses clear-cell RCC, papillary RCC, mucinous and spindle cell carcinomas and translocation RCC; C2, which comprises oncocytoma and chromophobe RCC. Interestingly, C1 epi-cluster displayed 3-fold more hypermethylation as compared with C2 epi-cluster. Of note, differentially methylated regions between C1 and C2 epi-clusters occur in gene bodies and intergenic regions, instead of gene promoters. Transcriptome analysis of C1 epi-cluster suggests a functional convergence on Polycomb targets, whereas C2 epi-cluster displays DNA methylation defects. Furthermore, we find that our epigenetic ontogeny signature is associated with worse outcomes of patients with clear-cell RCC. CONCLUSIONS: Our data define the epi-clusters that can discriminate between distinct RCC subtypes and for the first time define the epigenetic basis for proximal versus distal tubule derived kidney tumors. Clin Cancer Res; 22(24); 6236-46. ©2016 AACR.


Asunto(s)
Carcinoma de Células Renales/genética , Metilación de ADN/genética , Neoplasias Renales/genética , Ontologías Biológicas , Carcinoma de Células Renales/patología , Epigénesis Genética/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Estudios de Asociación Genética , Humanos , Neoplasias Renales/patología , Masculino , Nefronas/patología , Regiones Promotoras Genéticas/genética , Transcriptoma/genética
11.
Oncotarget ; 7(33): 54082-54093, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27340923

RESUMEN

Inter-tumor heterogeneity might explain divergent clinical evolution of cancers bearing similar pathological features. In the last decade, genomic has highly improved tumor subtypes classification through the identification of oncogenic or tumor suppressor drivers. In addition, epigenetics and long non-coding RNAs (lncRNAs) are emerging as new fields for investigation, which might also account for tumor heterogeneity. There is growing evidence that modifications of lncRNA expression profiles are involved in cancer progression through epigenetic regulation, activation of pro-oncogenic pathways and crosstalks with other RNA subtypes. Consequently, the study of lncRNA expression profile will be a key factor in the future for charting cancer subtype classifications as well as defining prognostic and progression biomarkers. Herein we discuss the interest of lncRNA as potent prognostic and predictive biomarkers, and provide a glimpse on the impact of emerging cancer subtypes classification based on lncRNAs.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias/clasificación , Neoplasias/genética , ARN Largo no Codificante/biosíntesis , Animales , Perfilación de la Expresión Génica , Humanos , ARN Largo no Codificante/análisis
12.
Epigenetics ; 10(9): 872-81, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26224146

RESUMEN

With the goal of studying epigenetic alterations in fibrolamellar hepatocellular carcinoma (FLC) and establish an associated DNA methylation signature, we analyzed LINE-1 methylation in a cohort of FLC and performed next-generation sequencing of DNA methylation in a training set of pure-FLCs and non-cirrhotic hepatocellular carcinomas (nc-HCC). DNA methylation was correlated with gene expression. Furthermore, we established and validated an epigenetic signature differentiating pure-FLC from other HCCs. LINE-1 methylation correlated with shorter recurrence-free survival and overall survival in resected pure-FLC patients. Unsupervised clustering using CG sites located in islands distinguished pure-FLC from nc-HCC. Major DNA methylation changes occurred outside promoters, mainly in gene bodies and intergenic regions located in the vicinity of liver developmental genes (i.e., SMARCA4 and RXRA). Partially methylated domains were more prone to DNA methylation changes. Furthermore, we identified several putative tumor suppressor genes (e.g., DLEU7) and oncogenes (e.g., DUSP4). While ∼ 70% of identified gene promoters gaining methylation were marked by bivalent histone marks (H3K4me3/H3K27me3) in embryonic stem cells, ∼ 70% of those losing methylation were marked by H3K4me3. Finally, we established a pure FLC DNA methylation signature and validated it in an independent dataset. Our analysis reveals a distinct epigenetic signature of pure FLC as compared to nc-HCC, with DNA methylation changes occurring in the vicinity of liver developmental genes. These data suggest new options for targeting FLC based on cancer epigenome aberrations.


Asunto(s)
Carcinoma Hepatocelular/genética , Metilación de ADN , Análisis de Secuencia de ADN/métodos , Adolescente , Carcinoma Hepatocelular/patología , Niño , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Elementos de Nucleótido Esparcido Largo , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas , Análisis de Supervivencia , Adulto Joven
16.
Arch Dermatol ; 146(6): 615-23, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20566924

RESUMEN

OBJECTIVES: To investigate skin manifestations of the polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes (POEMS) syndrome and their correlation with serum vascular endothelial growth factor (s-VEGF-A) levels and to describe the impact of autologous peripheral blood stem cell transplantation (aPBSCT) on these manifestations and the correlation with s-VEGF-A levels. DESIGN: Case series from January 1993 through June 2007. SETTING: Hospitalized care in Assistance Publique-Hôpitaux de Paris in Pitié-Salpêtrière and Tenon hospitals. PATIENTS: Twenty-three patients with POEMS syndrome, 10 of whom were clinically followed up after aPBSCT. MAIN OUTCOME MEASURES: Description and distribution of clinical lesions at POEMS syndrome diagnosis, skin evaluation after aPBSCT, and s-VEGF-A levels measured at POEMS syndrome diagnosis and after aPBSCT. RESULTS: In 21 patients with skin manifestations at POEMS syndrome diagnosis, the most common skin manifestations were hemangiomas (18 patients [86%]), hyperpigmentation (16 [76%]), skin thickening (12 [57%]), acrocyanosis (12 [57%]), hypertrichosis (11 [52%]), acquired facial lipoatrophy (11 [52%]), and white nails (8 [38%]). The median s-VEGF-A level was not different between patients with and without skin manifestations except in those with hypertrichosis (P = .04). After aPBSCT, no significant correlation was observed between s-VEGF-A level decreases and response of skin manifestations, again except for hypertrichosis (P = .007). CONCLUSIONS: Acquired facial lipoatrophy and livedo should be added to the skin manifestations of POEMS syndrome. Despite a role of s-VEGF-A in various skin manifestations, the impact of s-VEGF-A level decreases on skin outcomes is weak after aPBSCT, mostly resulting in clinical stabilization.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Síndrome POEMS/patología , Piel/patología , Factor A de Crecimiento Endotelial Vascular/sangre , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Síndrome POEMS/sangre , Síndrome POEMS/cirugía , Pronóstico , Estudios Retrospectivos , Piel/metabolismo , Trasplante Autólogo
17.
Melanoma Res ; 20(2): 67-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20093989

RESUMEN

Biomarkers play an important role in the diagnosis and prognostic classification of various cancers and can be useful in monitoring the patient's clinical course of disease and response to therapy. Generally, biomarkers are proteins and their expressions are associated with malignant disease. In the majority of cases, the marker molecules are expressed by the tumour cells themselves or by the tumour microenvironment cells. Thus, most biomarkers can primarily be found in malignant tissues, but after active secretion or passive release at tumour destruction, they become detectable in body fluids such as blood. Besides morphological and histopathological biomarkers (anatomic site, type of the primary tumour, tumour size, invasion depth, vascular invasion and ulceration), an increasing variety of serological markers have been identified, providing the possibility of a more detailed diagnostic and prognostic subgrouping of tumour entities, up to and even changing existing classification systems. The goal of this review is to provide an overview of old and more recent serological biomarkers in malignant melanoma. We will first focus on confirmed and nonconfirmed serum tumour markers, followed by proteomic profiling, an innovative approach to identify new and better serological biomarkers in melanoma, and ending with the predictive factors for treatments in this pathology.


Asunto(s)
Biomarcadores de Tumor/sangre , Melanoma/sangre , Humanos , Melanoma/clasificación , Melanoma/diagnóstico , Pronóstico
18.
Crit Rev Oncol Hematol ; 74(1): 27-39, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19781957

RESUMEN

The number of melanoma cases worldwide is increasing faster than any other cancer and remains one of the most treatment-refractory malignancies. Despite decades of clinical trials testing chemotherapy and immunotherapy, a standard first-line treatment for metastatic melanoma has not yet been established; tough single agent dacarbazine represents the most common option. This review will focus on metastatic malignant melanoma treatment from single agent until new therapies. An overview of established chemotherapies and immunotherapies, followed by a summary of trials testing the potential combination and new agent are explored.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Productos Biológicos/uso terapéutico , Drogas en Investigación/uso terapéutico , Inmunoterapia , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Humanos , Inmunoterapia/métodos , Interferón-alfa/uso terapéutico , Interleucina-2/uso terapéutico , Melanoma/inmunología , Melanoma/secundario , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Resultado del Tratamiento
19.
Eur J Cancer ; 45(8): 1407-14, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19157860

RESUMEN

PURPOSE: The presence of metastases in regional lymph nodes is a strong indicator of poor patient survival in many types of cancer. It has recently been shown that vascular endothelial growth factor-C (VEGF-C), and its receptor VEGFR-3, may play a pivotal role in the promotion of metastasis to regional lymph nodes. This study was designed to detect and evaluate whether the expression of VEGFR-3 or its soluble form plays a role in metastatic malignant melanoma and to determine the relationship with clinicopathological parameters and patients outcome. EXPERIMENTAL DESIGN: VEGFR-3 expression on melanoma tumour was evaluated by immunohistochemical study. Using a sensitive enzyme-linked immunosorbent assay, sVEGFR-3 was measured in sera of 60 metastatic melanoma patients in comparison with 30 healthy controls. RESULTS: Immunohistochemical study demonstrated a high expression of VEGFR-3 in melanoma cells. Median level of pre-treatment sVEGFR-3 was significantly higher (p=0.00001) in melanoma patients as compared to healthy donors. No association was noted between VEGFR-3 in situ or in sera and gender, age or LDH level. Median serum VEGFR-3 levels were significantly higher in patients with high tumour burden as compared to those with low tumour burden (p=0.013) as well as in non-responding patients (n=33) as compared to responding ones (n=27). Finally, low level of VEGFR-3 was also related positively to disease free survival (X(2)=3.85, p=0.022). CONCLUSION: These results suggest that the expression and high pre-treatment sVEGFR-3 level are significantly correlated to poorer prognosis, and may be promising targets for new therapeutic strategies in melanoma disease.


Asunto(s)
Biomarcadores de Tumor/análisis , Melanoma/química , Neoplasias Cutáneas/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis , Adulto , Anciano , Biomarcadores de Tumor/sangre , Estudios de Casos y Controles , Supervivencia sin Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Masculino , Melanoma/sangre , Melanoma/mortalidad , Persona de Mediana Edad , Neovascularización Fisiológica , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/mortalidad , Tasa de Supervivencia , Receptor 3 de Factores de Crecimiento Endotelial Vascular/sangre , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA