Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 38(15): e95874, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31267558

RESUMEN

MAPK inhibitors (MAPKi) show outstanding clinical response rates in melanoma patients harbouring BRAF mutations, but resistance is common. The ability of melanoma cells to switch from melanocytic to mesenchymal phenotypes appears to be associated with therapeutic resistance. High-throughput, subcellular proteome analyses and RNAseq on two panels of primary melanoma cells that were either sensitive or resistant to MAPKi revealed that only 15 proteins were sufficient to distinguish between these phenotypes. The two proteins with the highest discriminatory power were PTRF and IGFBP7, which were both highly upregulated in the mesenchymal-resistant cells. Proteomic analysis of CRISPR/Cas-derived PTRF knockouts revealed targets involved in lysosomal activation, endocytosis, pH regulation, EMT, TGFß signalling and cell migration and adhesion, as well as a significantly reduced invasive index and ability to form spheres in 3D culture. Overexpression of PTRF led to MAPKi resistance, increased cell adhesion and sphere formation. In addition, immunohistochemistry of patient samples showed that PTRF expression levels were a significant biomarker of poor progression-free survival, and IGFBP7 levels in patient sera were shown to be higher after relapse.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Melanoma/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteómica/métodos , Proteínas de Unión al ARN/metabolismo , Adulto , Anciano , Carbamatos/farmacología , Adhesión Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Masculino , Melanoma/tratamiento farmacológico , Melanoma/genética , Persona de Mediana Edad , Mapas de Interacción de Proteínas , Análisis de Secuencia de ARN , Sulfonamidas/farmacología , Análisis de Supervivencia , Regulación hacia Arriba , Vemurafenib/farmacología
2.
Exp Dermatol ; 25(6): 447-52, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26844814

RESUMEN

The link between solar radiation and melanoma is still elusive. Although infrared radiation (IR) accounts for over 50% of terrestrial solar energy, its influence on human skin is not well explored. There is increasing evidence that IR influences the expression patterns of several molecules independently of heat. A previous in vivo study revealed that pretreatment with IR might promote the development of UVR-induced non-epithelial skin cancer and possibly of melanoma in mice. To expand on this, the aim of the present study was to evaluate the impact of IR on UVR-induced apoptosis and DNA repair in normal human epidermal melanocytes. The balance between these two effects is a key factor of malignant transformation. Human melanocytes were exposed to physiologic doses of IR and UVR. Compared to cells irradiated with UVR only, simultaneous exposure to IR significantly reduced the apoptotic rate. However, IR did not influence the repair of UVR-induced DNA damage. IR partly reversed the pro-apoptotic effects of UVR via modification of the expression and activity of proteins mainly of the extrinsic apoptotic pathway. In conclusion, IR enhances the survival of melanocytes carrying UVR-induced DNA damage and thereby might contribute to melanomagenesis.


Asunto(s)
Apoptosis/efectos de la radiación , Rayos Infrarrojos , Melanocitos/efectos de la radiación , Melanoma/etiología , Rayos Ultravioleta/efectos adversos , Daño del ADN/efectos de la radiación , Humanos , Recién Nacido , Cultivo Primario de Células
3.
Acta Derm Venereol ; 96(2): 207-12, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26271044

RESUMEN

This retrospective multicentre analysis from the Psoriasis Registry Austria (PsoRA) was conducted to determine drug effectiveness and survival of anti-tumour necrosis factor alpha (anti-TNF-α) agents in patients with moderate-to-severe chronic plaque psoriasis over a 9-year period. Data on 1,019 treatment cycles with adalimumab (n = 460), etanercept (n = 501), and/or infliximab (n = 58) administered to 827 patients (272 women, 555 men) were available for analysis. Compared with etanercept, adalimumab and infliximab showed superior short-term effectiveness. Intention-to-treat-calculated median drug survivals for adalimumab (1,264 days) and etanercept (1,438 days) were similar to each other (p = 0.74), but significantly superior to that of infliximab (477 days) (p = 7.0e-07 vs. adalimumab and p=2.2e-07 vs. etanercept, respectively). Their drug survival rates at 36 months were 51.6%, 56.0%, and 22.6%, respectively. Survival rates correlated significantly with effectiveness for adalimumab and etanercept, but not for infliximab.


Asunto(s)
Actividades Cotidianas , Productos Biológicos/uso terapéutico , Inmunosupresores/uso terapéutico , Psoriasis/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Austria , Productos Biológicos/efectos adversos , Femenino , Humanos , Inmunosupresores/efectos adversos , Análisis de Intención de Tratar , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Psoriasis/diagnóstico , Psoriasis/inmunología , Sistema de Registros , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Factores de Tiempo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/inmunología , Adulto Joven
4.
Electrophoresis ; 36(4): 564-76, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25395074

RESUMEN

To shed light on the multistep process of squamous cell carcinoma development and the underlying pathologic mechanisms, we performed comparative proteome analysis of keratinocytes, keratinocytes stimulated with Il-1beta, and A431 epidermoid carcinoma cells. Fractionation of the cells into supernatant, nucleus, and cytoplasm was followed by protein separation, proteolytic digest, and nano-LC separation, and fragmentation using an ion trap mass spectrometer. Specific bioinformatics tools were used to generate a list of keratinocyte-specific proteins. Ninety percent of these proteins were found to be upregulated in keratinocytes versus the A431 cells. Classification of the identified proteins by biologic function and gene set enrichment analysis revealed that keratinocytes produced more proteins involved in cell differentiation, cell adhesion, cell junction, calcium ion, calmodulin binding, cytoskeleton organization, and cytokinesis, whereas A431 produced more proteins involved in cell cycle checkpoint, cell cycle process, RNA processing and transport, DNA damage and repair, RNA and DNA binding, and chromatin remodeling. The protein signatures of A431 and normal keratinocytes treated with IL-1beta showed marked similarity, confirming that inflammation is an important step in malignant transformation in nonmelanoma skin cancer. Thus, proteome profiling and bioinformatic processing may support the understanding of the underlying mechanisms, with the potential to facilitate development of early biomarkers and patient-tailored therapy.


Asunto(s)
Queratinocitos/metabolismo , Queratinocitos/patología , Proteoma/análisis , Proteómica/métodos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica , Células Cultivadas , Dermatitis/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Interleucina-1beta/farmacología , Queratinocitos/efectos de los fármacos , Proteoma/metabolismo , Valores de Referencia , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas
5.
Am J Ther ; 22(1): 54-60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24176884

RESUMEN

Metastatic melanoma has a poor prognosis; the median survival for patients with stage IV melanoma ranges from 8 to 18 months after diagnosis. Interferon-α provides significant improvement in disease-free survival at the cost of poor tolerability. Identifying patients who benefit the most may improve the cost:benefit ratio. In addition, no data exist for the role of adjuvant therapy in noncutaneous melanoma. Molecular profiles may help to identify patients who benefit the most from adjuvant interferon therapy. In this review, the American Joint Commission on Cancer 2009 staging criteria and emerging biomarker data to guide adjuvant treatment decisions will be discussed. Several criteria to guide selection of patients are discussed in detail. These include Breslow thickness, number of positive lymph nodes, whether or not the primary lesion has ulcerated, immunologic markers, and cytokine profiles. Substantial progress has been made in deciding which patients benefit from interferon-α adjuvant therapy. Interferon-α is the only agent currently approved for the adjuvant treatment of this deadly disease, despite its side effect profile. More effective drugs with better tolerability are needed.


Asunto(s)
Antineoplásicos/uso terapéutico , Interferón-alfa/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Antineoplásicos/efectos adversos , Antineoplásicos/economía , Quimioterapia Adyuvante/economía , Quimioterapia Adyuvante/métodos , Análisis Costo-Beneficio , Supervivencia sin Enfermedad , Humanos , Interferón-alfa/efectos adversos , Interferón-alfa/economía , Melanoma/economía , Melanoma/patología , Estadificación de Neoplasias , Selección de Paciente , Pronóstico , Neoplasias Cutáneas/economía , Neoplasias Cutáneas/patología , Tasa de Supervivencia
6.
Photodermatol Photoimmunol Photomed ; 31(4): 175-83, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25495608

RESUMEN

BACKGROUND: Extracorporeal photopheresis (ECP) improves skin sclerosis in systemic sclerosis (SSc) patients. SSc is associated with an increased risk of lung cancer. As ECP is supposed to exert immunomodulatory effects, a possible impact of ECP on the incidence of lung cancer in SSc patients was evaluated. METHODS: Seventy-one SSc patients treated with ECP at the Photopheresis Unit of the Department of Dermatology at the Medical University of Vienna between 1991 and 2013 were analyzed retrospectively. RESULTS: We calculated a standardized incidence ratio (SIR) for lung cancer in ECP-treated SSc patients of 2.34 [95% confidence interval (CI) 1.63-2.49]. This is in accordance with recent meta-analyses demonstrating a significantly enhanced risk of lung carcinoma in SSc patients. Comparison of the lung cancer risks of these patients with our ECP-treated patients revealed that ECP has no influence. Each patient with lung carcinoma had previously been diagnosed with lung involvement of the non-specific interstitial pneumonitis (NSIP) type. CONCLUSION: We confirm that SSc patients are at significantly increased risk for lung cancer. However, ECP does not influence this risk. NSIP may be a risk factor for lung cancer in SSc patients.


Asunto(s)
Neoplasias Pulmonares/epidemiología , Fotoféresis/efectos adversos , Esclerodermia Sistémica/epidemiología , Esclerodermia Sistémica/terapia , Femenino , Humanos , Incidencia , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Factores de Riesgo , Esclerodermia Sistémica/patología
9.
J Proteome Res ; 12(7): 3264-76, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23713901

RESUMEN

Drug resistance is a major obstacle in melanoma treatment. Recognition of specific resistance patterns, the understanding of the patho-physiology of drug resistance, and identification of remaining options for individual melanoma treatment would greatly improve therapeutic success. We performed mass spectrometry-based proteome profiling of A375 melanoma cells and HeLa cells characterized as sensitive to cisplatin in comparison to cisplatin resistant M24met and TMFI melanoma cells. Cells were fractionated into cytoplasm, nuclei and secretome and the proteome profiles classified according to Gene Ontology. The cisplatin resistant cells displayed increased expression of lysosomal as well as Ca²âº ion binding and cell adherence proteins. These findings were confirmed using Lysotracker Red staining and cell adhesion assays with a panel of extracellular matrix proteins. To discriminate specific survival proteins, we selected constitutively expressed proteins of resistant M24met cells which were found expressed upon challenging the sensitive A375 cells. Using the CPL/MUW proteome database, the selected lysosomal, cell adherence and survival proteins apparently specifying resistant cells were narrowed down to 47 proteins representing a potential resistance signature. These were tested against our proteomics database comprising more than 200 different cell types/cell states for its predictive power. We provide evidence that this signature enables the automated assignment of resistance features as readout from proteome profiles of any human cell type. Proteome profiling and bioinformatic processing may thus support the understanding of drug resistance mechanism, eventually guiding patient tailored therapy.


Asunto(s)
Resistencia a Antineoplásicos/genética , Melanoma/genética , Proteínas de Neoplasias/clasificación , Proteómica/métodos , Línea Celular Tumoral , Cisplatino/uso terapéutico , Biología Computacional , Células HeLa , Humanos , Espectrometría de Masas , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas de Neoplasias/aislamiento & purificación , Transcriptoma/efectos de los fármacos
10.
J Dtsch Dermatol Ges ; 11(3): 251-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23078407

RESUMEN

BACKGROUND AND OBJECTIVES: The aim of this study is to show data regarding trends in the diagnosis of melanoma over the last ten years by looking at the University Clinic of Dermatology in Vienna as an example. PATIENTS AND METHODS: All excised melanocytic lesions from 1998 to 2008 were included. RESULTS: The ratio of excisions of benign to malignant lesions fell from 7:1 (1998) to 4:1 (2008). The mean percentage of in situ melanomas was 39% in 1998 and did not change significantly over time and no change was seen in tumor thickness of invasive melanomas. The median invasion depth was 0.7 mm in 1998 and 0.65 mm in 2008. The absolute number of diagnosed melanomas did not change significantly over time. CONCLUSION: The proportion of in situ melanomas was consistently high. Tumor thickness stayed at a low level, whereas the number of excised benign melanocytic lesions decreased significantly.


Asunto(s)
Centros Médicos Académicos/estadística & datos numéricos , Melanoma/diagnóstico , Melanoma/epidemiología , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/epidemiología , Adulto , Distribución por Edad , Austria/epidemiología , Femenino , Humanos , Incidencia , Estudios Longitudinales , Masculino , Melanoma/cirugía , Medición de Riesgo , Factores de Riesgo , Distribución por Sexo , Neoplasias Cutáneas/cirugía , Resultado del Tratamiento
11.
Genes Chromosomes Cancer ; 50(9): 680-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21584902

RESUMEN

It is commonly accepted that cancer cell progression is accompanied by accumulation of genetic changes. Here we searched for copy number variations in melanoma and asked whether homozygous losses always cumulate during tumor cell progression. Therefore we investigated either melanoma cell lines or tissue derived from the primary lesion and from the lymph node metastasis of the same individual patient. In vitro studies of melanoma cell lines revealed high migratory and anchorage independent growth of metastasis-derived cells. Surprisingly, whole genome DNA analysis of a primum-derived cell line revealed a total of 10 homozygous losses, whereas the matched metastasis-derived cell line only shared five of those losses. We further tested these cells in a mouse model for intradermal melanoma growth and detected fast growth of the metastasis-derived cell line and no growth of primum-derived cells. Additionally, we screened matched pairs of patient-derived melanoma primum and metastasis samples and we could also identify a case with homozygous deletions exclusively present in the primary lesion. Therefore, we suggest that tumor cell progression at the metastatic niche can occur parallel and independently from the primary tumor. We propose that for mutation-targeted therapy genotyping should be performed not only from primary, but also from metastatic melanoma.


Asunto(s)
Deleción Cromosómica , Melanoma/patología , Neoplasias Cutáneas/patología , Adulto , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Forma de la Célula , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 10/genética , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 4/genética , Variaciones en el Número de Copia de ADN , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Metástasis Linfática , Masculino , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Cromosoma X/genética
13.
Mol Cancer ; 9: 200, 2010 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-20667089

RESUMEN

BACKGROUND: Heme Oxygenase-1 (HO-1) is expressed in many cancers and promotes growth and survival of neoplastic cells. Recently, HO-1 has been implicated in tumor cell invasion and metastasis. However, the molecular mechanisms underlying these biologic effects of HO-1 remain largely unknown. To identify a common mechanism of action of HO-1 in cancer, we determined the global effect of HO-1 on the transcriptome of multiple tumor entities and identified a universal HO-1-associated gene expression signature. RESULTS: Genome-wide expression profiling of Heme Oxygenase-1 expressing versus HO-1 silenced BeWo choriocarcinoma cells as well as a comparative meta-profiling of the preexisting expression database of 190 human tumors of 14 independent cancer types led to the identification of 14 genes, the expression of which correlated strongly and universally with that of HO-1 (P = 0.00002). These genes included regulators of cell plasticity and extracellular matrix (ECM) remodeling (MMP2, ADAM8, TGFB1, BGN, COL21A1, PXDN), signaling (CRIP2, MICB), amino acid transport and glycosylation (SLC7A1 and ST3GAL2), estrogen and phospholipid biosynthesis (AGPAT2 and HSD17B1), protein stabilization (IFI30), and phosphorylation (ALPPL2). We selected PXDN, an adhesion molecule involved in ECM formation, for further analysis and functional characterization. Immunofluorescence and Western blotting confirmed the positive correlation of expression of PXDN and HO-1 in BeWo cancer cells as well as co-localization of these two proteins in invasive extravillous trophoblast cells. Modulation of HO-1 expression in both loss-of and gain-of function cell models (BeWo and 607B melanoma cells, respectively) demonstrated a direct relationship of HO-1 expression with cell adhesion to Fibronectin and Laminin coated wells. The adhesion-promoting effects of HO-1 were dependent on PXDN expression, as loss of PXDN in HO-1 expressing BeWo and 607B cells led to reduced cell attachment to Laminin and Fibronectin coated wells. CONCLUSIONS: Collectively, our results show that HO-1 expression determines a distinct 'molecular signature' in cancer cells, which is enriched in genes associated with tumorigenesis. The protein network downstream of HO-1 modulates adhesion, signaling, transport, and other critical cellular functions of neoplastic cells and thus promotes tumor cell growth and dissemination.


Asunto(s)
Adhesión Celular , Perfilación de la Expresión Génica , Hemo Oxigenasa (Desciclizante)/metabolismo , Neoplasias/genética , Secuencia de Bases , Cartilla de ADN , Técnicas de Silenciamiento del Gen , Hemo Oxigenasa (Desciclizante)/genética , Humanos , Neoplasias/enzimología , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
14.
Clin Cancer Res ; 15(10): 3495-502, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19447871

RESUMEN

PURPOSE: Melanoma is one of the most aggressive types of cancer with currently no chance of cure once the disease has spread to distant sites. Therapeutic options for advanced stage III and IV are very limited, mainly palliative, and show response in only approximately 20% of all cases. The presented preclinical study was done to investigate the influence of a combined treatment of the epidermal growth factor receptor inhibitor erlotinib and the vascular endothelial growth factor monoclonal antibody bevacizumab in melanoma. EXPERIMENTAL DESIGN AND RESULTS: The epidermal growth factor receptor was expressed in all cell lines tested, and treatment with erlotinib did inhibit the activation of the MEK/extracellular signal-regulated kinase and AKT signaling pathways. Whereas in vitro no influence on tumor cell proliferation was seen with erlotinib or bevacizumab monotherapy, a decreased invasive potential on erlotinib treatment in a three-dimensional Matrigel assay was observed. Furthermore, both drugs inhibited proliferation and sprouting of endothelial cells. In vivo, in a severe combined immunodeficient mouse xenotransplantation model, reduction in tumor volume under combined treatment with erlotinib and bevacizumab was superior to the additive effect of both single agents. This was associated with reduced cell proliferation, increased apoptosis, and a reduction in tumor angiogenesis compared with control or single treatment groups. Likewise, the reduction in the extent of lymph node and lung metastasis was most pronounced in animals treated with both drugs. CONCLUSION: The presented data strongly support the use of a combination of erlotinib and bevacizumab as a novel treatment regimen for metastatic melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bevacizumab , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Antígeno Ki-67/análisis , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones SCID , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
16.
Clin Cancer Res ; 14(24): 8178-83, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19088033

RESUMEN

PURPOSE: The high molecular weight melanoma-associated antigen (HMW-MAA) is an attractive target for immunotherapy of malignant melanoma. We have recently generated a vaccine based on the HMW-MAA mimotope 225D9.2+ that was able to induce anti-HMW-MAA antibodies with antitumor activity in vitro. Here, we investigated the antitumor activity of these antibodies in a human melanoma xenotransplant severe combined immunodeficient (SCID) mouse model. EXPERIMENTAL DESIGN: Tumors were established by injecting the human melanoma 518A2 cells into C.B.17 SCID/SCID mice. In tumor prevention experiments, 200 microg purified total IgG antibodies were injected intravenously the same day or on day 5 in therapeutic experiments. Antibody administration was repeated every fourth day and tumor volumes were measured. Antibody specificity and tumor infiltration by macrophages were investigated by immunohistochemistry. RESULTS: Within 35 days after cell inoculation, antibody treatment reduced tumor growth up to 40% in the therapeutic and up to 62% in the tumor prevention experiments compared with the control mice. In tumors of all groups, a similar distribution of the HMW-MAA and no differences in infiltration of macrophages were detected by immunohistochemistry. CONCLUSIONS: Here, we showed that antibodies induced by the 225D9.2+ mimotope effectively inhibited melanoma tumor growth. Additional mechanisms besides antibody-dependent cell cytotoxicity like disruption of interactions of melanoma cells mediated by extracellular matrix components seem to be involved in tumor growth inhibition. Based on our findings, we suggest that active immunization with this mimotope might be a promising strategy for treatment of melanoma.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma Experimental/terapia , Animales , Antígenos de Neoplasias/análisis , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Macrófagos/fisiología , Melanoma Experimental/inmunología , Ratones , Ratones SCID , Peso Molecular , Trasplante de Neoplasias , Trasplante Heterólogo
17.
Cancer Res ; 67(7): 3406-11, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409451

RESUMEN

A role of IgE antibodies in cancer surveillance has been implicated for a long time. Studies dealing with IgE antibodies directly targeted to tumor antigens have shown marked anticancer effects mediated by this antibody class. Thus, the basic function of IgE antibodies may be to control tumor growth. Thus far, cancer-specific IgE has only been applied passively. Consequently, the aim of this study was to establish an active vaccination protocol to induce tumor antigen-specific IgE antibodies, and to evaluate functional properties. We previously generated epitope mimics, so-called mimotopes, for the epitope recognized by the anti-HER-2 antibody trastuzumab. Upon i.p. immunizations, IgG antibodies with trastuzumab-like properties could be elicited. In the present study, we immunized BALB/c mice via the oral route with these trastuzumab mimotopes, under simultaneous neutralization and suppression of gastric acid. As shown in preceding experiments, this feeding regimen effectively induces Th2 immune responses. Oral immunizations with trastuzumab mimotopes under hypoacidic conditions indeed resulted in the formation of IgE antibodies towards the HER-2 antigen. Moreover, anti-HER-2 IgE-sensitized effector cells mediated SK-BR-3 target cell lysis in an antibody-dependent cytotoxicity assay. We conclude that directed and epitope-specific induction of IgE against tumor antigens is feasible with an oral mimotope vaccination regimen, and that these antibodies mediate anticancer effects.


Asunto(s)
Anticuerpos Antineoplásicos/biosíntesis , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/inmunología , Inmunoglobulina E/biosíntesis , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Anticuerpos Antineoplásicos/inmunología , Especificidad de Anticuerpos , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Epítopos/inmunología , Humanos , Inmunoglobulina E/inmunología , Ratas , Receptor ErbB-2/inmunología , Trastuzumab
18.
Eur J Dermatol ; 18(6): 651-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18955209

RESUMEN

Actinic keratoses (AKs) are lesions caused by chronic UV radiations that have the potential to progress to invasive SCCs. The prevalence of AK is increasing worldwide, and although there are a variety of treatment modalities, along with a number of published guidelines, there is a lack of advice on treatment recommendations for AK in clinical practice. There are also considerable variations in the care of patients with AK in Europe, and so the Skin Academy (a group of leading experts in the field of dermatology and skin cancer) formed to discuss best practice in the treatment of AK. The result of these discussions is an algorithm designed to assist physicians with their treatment decisions, giving recommendations for both field directed and lesion directed treatment. The new AK Treatment Algorithm described in this paper comprises 5 key, decision making steps, which are simple and allow the flexibility to reflect cultural differences between and within countries. The algorithm also considers factors such as patient profile, medical history, and personal preference (e.g. cosmesis, and pain).This is a novel algorithm that has the capacity to grow and expand as more clinical evidence becomes available. It offers, for the first time, a summary of European best practice recommendations for the treatment of AK.


Asunto(s)
Queratosis Actínica/terapia , Algoritmos , Humanos
19.
Cancer Res ; 66(24): 11888-96, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17178886

RESUMEN

Dimethylfumarate (DMF) inhibits signals transmitted by Rel proteins and is used for the treatment of inflammatory skin diseases such as psoriasis, but potential effects of DMF on tumor progression have yet not been analyzed. We show that DMF reduced melanoma growth and metastasis in severe combined immunodeficient mouse models. To identify targets of DMF action, we analyzed mRNA expression in DMF-treated melanomas by gene chip arrays. Using BiblioSphere software for data analysis, significantly regulated genes were mapped to Gene Ontology terms cell death, cell growth, and cell cycle. Indeed, we found that DMF inhibited proliferation of human melanoma cells A375 and M24met in vitro. The cell cycle was arrested at the G(2)-M boundary. Moreover, DMF was proapoptotic, as shown by cell cycle analysis and by Annexin V and Apo2.7 staining. These results were confirmed in vivo. DMF reduced proliferation rates of tumor cells as assessed by Ki-67 immunostaining and increased apoptosis as assessed by terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling staining. In conclusion, DMF is antiproliferative and proapoptotic and reduces melanoma growth and metastasis in animal models.


Asunto(s)
Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Fumaratos/uso terapéutico , Melanoma/patología , Metástasis de la Neoplasia/prevención & control , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Línea Celular Tumoral , Dimetilfumarato , Femenino , Humanos , Metástasis Linfática/patología , Metástasis Linfática/prevención & control , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Trasplante Heterólogo
20.
Cancer Res ; 66(3): 1658-63, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452225

RESUMEN

We previously described the expression of melanoma-associated endogenous retrovirus (MERV) proteins and viral particles in human melanomas and metastases. The objective of the present study was to determine whether a humoral immune response to MERV proteins occurs in melanoma. Candidate B-cell epitopes on MERV proteins were predicted using bioinformatic screening. The reactivity of MERV peptides corresponding to the predicted epitopes with antibodies prevalent in sera of melanoma patients was analyzed. An immunodominant peptide located in the env protein of MERV was identified. Subsequent analyzes using 81 samples from stage I to stage IV melanoma patients and 95 sera from healthy subjects revealed statistically significant differences in seroprevalence of antibodies in melanoma sera samples when compared with reference samples from healthy subjects. The prevalence of anti-MERV antibodies in melanoma patient sera was confirmed by immunofluorescence on env-transfected cells. These data indicate the potential of this candidate peptide as target for diagnosis and immunotherapy.


Asunto(s)
Biomarcadores de Tumor/inmunología , Retrovirus Endógenos/inmunología , Melanoma/virología , Proteínas del Envoltorio Viral/inmunología , Anticuerpos Antineoplásicos/sangre , Anticuerpos Antineoplásicos/inmunología , Reacciones Cruzadas , Epítopos de Linfocito B/inmunología , Células HeLa , Humanos , Epítopos Inmunodominantes/inmunología , Melanoma/sangre , Melanoma/inmunología , Melanoma/patología , Estadificación de Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA