Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 29(3): 1199-1213, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33212301

RESUMEN

T cell receptor (TCR)-engineered T cell therapy is a promising cancer treatment approach. Human telomerase reverse transcriptase (hTERT) is overexpressed in the majority of tumors and a potential target for adoptive cell therapy. We isolated a novel hTERT-specific TCR sequence, named Radium-4, from a clinically responding pancreatic cancer patient vaccinated with a long hTERT peptide. Radium-4 TCR-redirected primary CD4+ and CD8+ T cells demonstrated in vitro efficacy, producing inflammatory cytokines and killing hTERT+ melanoma cells in both 2D and 3D settings, as well as malignant, patient-derived ascites cells. Importantly, T cells expressing Radium-4 TCR displayed no toxicity against bone marrow stem cells or mature hematopoietic cells. Notably, Radium-4 TCR+ T cells also significantly reduced tumor growth and improved survival in a xenograft mouse model. Since hTERT is a universal cancer antigen, and the very frequently expressed HLA class II molecules presenting the hTERT peptide to this TCR provide a very high (>75%) population coverage, this TCR represents an attractive candidate for immunotherapy of solid tumors.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Inmunoterapia/métodos , Melanoma/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Telomerasa/antagonistas & inhibidores , Animales , Apoptosis , Proliferación Celular , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Gynecol Oncol ; 152(2): 408-415, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30448261

RESUMEN

OBJECTIVE: The objective of this study was to analyze the expression level and clinical role of soluble AXL (sAXL) in cancers affecting the serosal surfaces, with focus on ovarian carcinoma. METHODS: sAXL protein expression by ELISA was analyzed in 572 effusion supernatants, including 424 peritoneal, 147 pleural and 1 pericardial specimens. RESULTS: sAXL was overexpressed in peritoneal effusions compared to pleural and pericardial specimens (p < 0.001). sAXL levels were additionally significantly higher in effusions from patients with ovarian carcinoma, malignant mesothelioma and breast carcinoma compared to specimens from patients with other cancers (predominantly carcinomas of lung, gastrointestinal or uterine corpus/cervix origin) or benign reactive effusions (p < 0.001). sAXL was further overexpressed in high-grade serous carcinoma (HGSC; n = 373) compared to low-grade serous carcinoma (LGSC; n = 32; p = 0.036). In HGSC, sAXL levels were significantly lower in post-chemotherapy effusions compared to primary diagnosis pre-chemotherapy specimens (p = 0.002). sAXL levels in HGSC were unrelated to chemoresponse at diagnosis, progression-free survival or overall survival. Levels were similarly unrelated to survival in LGSC and breast carcinoma. CONCLUSIONS: sAXL is widely expressed in malignant effusions, particularly in ovarian and breast carcinoma and in malignant mesothelioma. sAXL is overexpressed in HGSC compared to LGSC and its levels are lower following exposure to chemotherapy. However, sAXL levels are not informative of chemoresponse or survival.


Asunto(s)
Líquido Ascítico/enzimología , Neoplasias de la Mama/enzimología , Cistadenocarcinoma Seroso/enzimología , Neoplasias Pulmonares/enzimología , Mesotelioma/enzimología , Neoplasias Ováricas/enzimología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Líquido Ascítico/efectos de los fármacos , Líquido Ascítico/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Mesotelioma/tratamiento farmacológico , Mesotelioma/patología , Mesotelioma Maligno , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Adulto Joven , Tirosina Quinasa del Receptor Axl
3.
Gynecol Oncol ; 150(1): 136-142, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29804637

RESUMEN

OBJECTIVE: To analyze the expression and clinical role of CHK1 and CHK2 in metastatic high-grade serous carcinoma (HGSC). METHODS: HGSC effusions (n = 335; 280 peritoneal, 55 pleural) were analyzed for protein expression of total CHK1 and its phosphorylated forms p-ser317 and p-ser296, as well as total CHK2 and its phosphorylated form p-thr68 using immunohistochemistry. Expression was analyzed for association with clinicopathologic parameters, including chemotherapy response, and survival. RESULTS: Carcinoma cells stained positive, predominantly at the nuclei, in the majority of cases (range 83-100% for the five antibodies), while expression in reactive mesothelial cells and tumor-associated macrophages was more variable. Total CHK1 (p = 0.037), p-CHK1ser317 (p = 0.001), p-CHK1ser296 (p = 0.002) and p-CHK2thr68 (p < 0.001) expression was significantly higher in post-chemotherapy disease recurrence compared to pre-chemotherapy effusions obtained at diagnosis. CHK1, p-CHK1ser296, p-CHK2thr68 and p-CHK1ser317 nuclear expression was positively related to expression of the checkpoint regulator WEE1, previously studied in this cohort (p = 0.003, p = 0.013, p = 0.001 and p = 0.01, respectively). Higher total CHK1 (p = 0.007), p-CHK1ser317 (p = 0.004), CHK2 (p = 0.01) and p-CHK2thr68 (p = 0.048) expression was significantly related to shorter overall survival in univariate analysis, and CHK1ser317 was an independent prognostic marker in multivariate analysis (p = 0.025). Higher p-CHK1ser317 (p = 0.03) and CHK2 (p = 0.034) expression was additionally associated with poor progression-free survival. CONCLUSIONS: CHK1 and CHK2 and their activated forms are frequently expressed in HGSC effusions, with higher expression following exposure to chemotherapy, and their expression is related to survival.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quinasa de Punto de Control 2/biosíntesis , Quinasa de Punto de Control 2/metabolismo , Cistadenocarcinoma Seroso/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/biosíntesis , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa de Punto de Control 2/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Activación Enzimática , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Análisis de Supervivencia , Adulto Joven
4.
Tumour Biol ; 37(9): 12133-12140, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27220319

RESUMEN

Wee1 is a nuclear kinase regulating cell cycle progression, and has emerged as a promising therapeutic target in cancer. Expression of Wee1 has been associated with poor outcome in certain tumor types, but the prognostic impact and clinical significance in colorectal cancer is unknown. The expression of Wee1 was examined by immunohistochemistry in primary colorectal carcinomas from a prospectively collected patient cohort, and associations with clinicopathological parameters and outcome were investigated. Cell culture experiments were performed using the cell lines RKO and SW620, and the relationship with the metastasis-promoting protein S100A4 was investigated. Nuclear expression was detected in 229 of the 258 tumors analyzed (89 %). Wee1 staining was associated with low pT stage, but no other significant associations with demographic or histopathological variables were found. Moderate Wee1 staining intensity was a predictor of favorable metastasis-free and overall survival compared to strong intensity and no or weak staining. The fraction of positive cells was not a prognostic factor in the present cohort. Inhibition of Wee1 expression using siRNA or treatment with the Wee1 inhibitor MK-1775 reduced expression of the metastasis-promoting protein S100A4, but no relationship between Wee1 and S100A4 was found in the patient samples. In conclusion, Wee1 is highly expressed in primary colorectal carcinomas, but few relevant associations with clinicopathological parameters or outcome were found. The lack of clinical significance of Wee1 expression could indicate that other tumor types might be better suited for further development of Wee1 inhibitors.


Asunto(s)
Proteínas de Ciclo Celular/análisis , Neoplasias Colorrectales/química , Proteínas Nucleares/análisis , Proteínas Tirosina Quinasas/análisis , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Humanos , Inmunohistoquímica , Estadificación de Neoplasias , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Pirimidinonas , Proteína de Unión al Calcio S100A4/análisis
5.
BMC Cancer ; 15: 462, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26054341

RESUMEN

BACKGROUND: Malignant melanoma has an increasing incidence rate and the metastatic disease is notoriously resistant to standard chemotherapy. Loss of cell cycle checkpoints is frequently found in many cancer types and makes the cells reliant on compensatory mechanisms to control progression. This feature may be exploited in therapy, and kinases involved in checkpoint regulation, such as Wee1 and Chk1/2, have thus become attractive therapeutic targets. METHODS: In the present study we combined a Wee1 inhibitor (MK1775) with Chk1/2 inhibitor (AZD7762) in malignant melanoma cell lines grown in vitro (2D and 3D cultures) and in xenografts models. RESULTS: Our in vitro studies showed that combined inhibition of Wee1 and Chk1/2 synergistically decreased viability and increased apoptosis (cleavage of caspase 3 and PARP), which may be explained by accumulation of DNA-damage (increased expression of γ-H2A.X)--and premature mitosis of S-phase cells. Compared to either inhibitor used as single agents, combined treatment reduced spheroid growth and led to greater tumour growth inhibition in melanoma xenografts. CONCLUSIONS: These data provide a rationale for further evaluation of the combination of Wee1 and Chk1/2 inhibitors in malignant melanoma.


Asunto(s)
Proteínas de Ciclo Celular/genética , Quinasa de Punto de Control 2/genética , Sinergismo Farmacológico , Melanoma/tratamiento farmacológico , Proteínas Nucleares/genética , Proteínas Tirosina Quinasas/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa de Punto de Control 2/antagonistas & inhibidores , Humanos , Melanoma/genética , Melanoma/patología , Ratones , Proteínas Nucleares/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Pirimidinonas , Neoplasias Cutáneas , Tiofenos/administración & dosificación , Urea/administración & dosificación , Urea/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto , Melanoma Cutáneo Maligno
6.
Gynecol Oncol ; 135(1): 118-24, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25093290

RESUMEN

OBJECTIVE: Wee1-like kinase (Wee1) is a tyrosine kinase which negatively regulates entry into mitosis at the G2 to M-phase transition and has a role in inhibition of unscheduled DNA replication in S-phase. The present study investigated the clinical role of Wee1 in advanced-stage (FIGO III-IV) ovarian serous carcinoma. METHODS: Wee1 protein expression was analyzed in 287 effusions using immunohistochemistry. Expression was analyzed for association with clinicopathologic parameters, including survival. Forty-five effusions were additionally studied using Western blotting. Wee1 was further silenced in SKOV3 and OVCAR8 cells by siRNA knockdown and proliferation was assessed. RESULTS: Nuclear expression of Wee1 in tumor cells was observed in 265/287 (92%) and 45/45 (100%) effusions by immunohistochemistry and Western blotting, respectively. Wee1 expression by immunohistochemistry was significantly higher in post-chemotherapy disease recurrence compared to pre-chemotherapy effusions obtained at diagnosis (p=0.002). Wee1 silencing in SKOV3 and OVCAR8 cells reduced proliferation. In univariate survival analysis of the entire cohort, a trend was observed between high (>25% of cells) Wee1 expression and poor overall survival (p=0.083). Survival analysis for 109 patients with post-chemotherapy effusions showed significant association between Wee1 expression and poor overall survival (p=0.004), a finding which retained its independent prognostic role in Cox multivariate analysis (p=0.003). CONCLUSIONS: Wee1 is frequently expressed in ovarian serous carcinoma effusions, and its expression is significantly higher following exposure to chemotherapy. The present study is the first to report that Wee1 is an independent prognostic marker in serous ovarian carcinoma.


Asunto(s)
Líquido Ascítico/metabolismo , Biomarcadores de Tumor/biosíntesis , Proteínas de Ciclo Celular/biosíntesis , Proteínas Nucleares/biosíntesis , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Derrame Pleural Maligno/metabolismo , Proteínas Tirosina Quinasas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Líquido Ascítico/patología , Células Cultivadas , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Derrame Pleural Maligno/patología , Pronóstico , Tasa de Supervivencia , Adulto Joven
7.
Int J Cancer ; 133(6): 1497-506, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23494569

RESUMEN

Cytoreductive surgery and intraperitoneal (i.p.) chemotherapy constitute a curative treatment option in mucinous peritoneal surface malignancies of intestinal origin, but treatment outcome is highly variable and the search for novel therapies is warranted. Immunotoxins are attractive candidates for targeted therapy in the peritoneal cavity because of direct cytotoxicity, distinct mechanisms of action and tumor cell selectivity. The MOC31PE immunotoxin targets the tumor-associated adhesion protein EpCAM (Epithelial Cell Adhesion Molecule), and has been administered safely in early clinical trials. In our work, the efficacy of i.p. administration of MOC31PE alone and together with mitomycin C (MMC) was investigated in unique animal models of human mucinous peritoneal surface malignancies. In initial model validation experiments, clear differences in efficacy were demonstrated between MMC and oxaliplatin, favoring MMC in five investigated tumor models. Subsequently, MOC31PE and MMC were given as single i.p. injections alone and in combination. In the PMCA-2 model, moderate growth inhibition was obtained with both drugs, while the combination resulted in at least additive effects; whereas the PMP-2 model was highly sensitive to both drugs separately and in combination and intermediate sensitivity was found for the PMCA-3 model. Furthermore, results from ex vivo experiments on freshly obtained mucinous tumor tissue from animals and patients suggested that classic mechanisms of immunotoxin activity were involved, i.e., inhibition of protein synthesis and induction of apoptosis. The present results suggest that adding MOC31PE to MMC-based i.p. chemotherapy should be further explored for EpCAM-expressing peritoneal surface malignancies, and a phase I trial is in preparation.


Asunto(s)
ADP Ribosa Transferasas/uso terapéutico , Antígenos de Neoplasias/inmunología , Toxinas Bacterianas/uso terapéutico , Moléculas de Adhesión Celular/inmunología , Exotoxinas/uso terapéutico , Inmunotoxinas/uso terapéutico , Neoplasias Peritoneales/tratamiento farmacológico , Factores de Virulencia/uso terapéutico , Animales , Modelos Animales de Enfermedad , Molécula de Adhesión Celular Epitelial , Femenino , Ratones , Ratones Endogámicos BALB C , Mitomicina/uso terapéutico , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Neoplasias Peritoneales/patología , Exotoxina A de Pseudomonas aeruginosa
8.
BMC Cancer ; 13: 288, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23767999

RESUMEN

BACKGROUND: Vulvar squamous cell carcinoma is a cancer form with increasing incidence rate and few treatment options. Wee1 is a central regulator of the G2/M DNA-damage checkpoint, and has in previous studies been described as a prognostic biomarker and a potential target for therapy in other cancer forms. METHODS: In the present study we analyzed the expression of Wee1 in a panel of 297 vulvar tumors by immunohistochemistry. Furthermore, siRNA transfections were carried out in two vulvar cancer cell lines (SW-954 and CAL-39) in order to study the effect on cell cycle distribution (flow cytometry) and proteins (western blot) involved in DNA damage response and apoptosis. RESULTS: Wee1 kinase is increased in vulvar squamous cell carcinomas, as compared to expression in normal epithelium, and a high Wee1 expression is associated with markers of malignancy, such as lymph node metastasis and poor differentiation. Our in vitro results showed that siRNA mediated Wee1 silencing only led to a modest reduction in viability, when examined in vulvar cancer cell lines. Nonetheless, a marked increase in DNA damages, as assessed by augmented levels of γ-H2AX, was observed in both cell lines in the absence of Wee1. CONCLUSIONS: Our results suggest that Wee1 may be involved in the progression of vulvar carcinomas. Based on our in vitro results, Wee1 is unlikely to function as a target for mono-treatment of these patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/biosíntesis , Proteínas Nucleares/biosíntesis , Proteínas Tirosina Quinasas/biosíntesis , Neoplasias de la Vulva/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/análisis , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Proteínas Nucleares/análisis , Pronóstico , Proteínas Tirosina Quinasas/análisis , ARN Interferente Pequeño , Transfección , Neoplasias de la Vulva/patología
9.
Biochem Biophys Res Commun ; 420(3): 516-22, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22446330

RESUMEN

The novel synthetic retinoid, CD437, shows potent anti-tumor activity in a range of different cancer cell lines and now serves as a prototype for development of new retinoid related molecules (RRMs). The purpose of this study was to examine the effect and cellular targets of CD437 in the human metastatic melanoma cell lines FEMX-1 and WM239. We showed that treatment with CD437 led to cell cycle arrest and induced apoptosis through both the extrinsic- and intrinsic pathways (caspase 8, -9 and PARP cleavage) in both cell lines. Interestingly, apoptosis was induced independently of DNA-fragmentation in FEMX-1 cells, and appeared partially caspase-independent in the WM239 cells. Additionally, up-regulation of CHOP mRNA and cathepsin D protein expression, following retinoid treatment, suggests involvement of the endoplasmatic reticulum (ER) and lysosomes, respectively. Combination of suboptimal concentrations of CD437 and lexatumumab, a TRAIL death receptor-2 agonist, resulted in synergistic reduction of viable cells, along with increased PARP cleavage. These results indicate that CD437 has a strong anti-neoplastic effect alone and in combination with lexatumumab in melanoma cell lines.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Melanoma/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Retinoides/farmacología , Neoplasias Cutáneas/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Catepsina D/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Mensajero/biosíntesis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Factor de Transcripción CHOP/biosíntesis , Regulación hacia Arriba , Receptor fas/biosíntesis
10.
BMC Cancer ; 12: 73, 2012 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-22356677

RESUMEN

BACKGROUND/AIMS: Breast cancer metastasis suppressor 1 (BRMS1) blocks metastasis in melanoma xenografts; however, its usefulness as a biomarker in human melanomas has not been widely studied. The goal was to measure BRMS1 expression in benign nevi, primary and metastatic melanomas and evaluate its impact on disease progression and prognosis. METHODS: Paraffin-embedded tissue from 155 primary melanomas, 69 metastases and 15 nevi was examined for BRMS1 expression using immunohistochemistry. siRNA mediated BRMS1 down-regulation was used to study impact on invasion and migration in melanoma cell lines. RESULTS: A significantly higher percentage of nevi (87%), compared to primary melanomas (20%) and metastases (48%), expressed BRMS1 in the nucelus (p < 0.0001). Strong nuclear staining intensity was observed in 67% of nevi, and in 9% and 24% of the primary and metastatic melanomas, respectively (p < 0.0001). Comparable cytoplasmic expression was observed (nevi; 87%, primaries; 86%, metastases; 72%). However, a decline in cytoplasmic staining intensity was observed in metastases compared to nevi and primary tumors (26%, 47%, and 58%, respectively, p < 0.0001). Score index (percentage immunopositive celles multiplied with staining intensity) revealed that high cytoplasmic score index (≥ 4) was associated with thinner tumors (p = 0.04), lack of ulceration (p = 0.02) and increased disease-free survival (p = 0.036). When intensity and percentage BRMS1 positive cells were analyzed separately, intensity remained associated with tumor thickness (p = 0.024) and ulceration (p = 0.004) but was inversely associated with expression of proliferation markers (cyclin D3 (p = 0.008), cyclin A (p = 0.007), and p21Waf1/Cip1 (p = 0.009)). Cytoplasmic score index was inversely associated with nuclear p-Akt (p = 0.013) and positively associated with cytoplasmic p-ERK1/2 expression (p = 0.033). Nuclear BRMS1 expression in ≥ 10% of primary melanoma cells was associated with thicker tumors (p = 0.016) and decreased relapse-free period (p = 0.043). Nuclear BRMS1 was associated with expression of fatty acid binding protein 7 (FABP7; p = 0.011), a marker of invasion in melanomas. In line with this, repression of BRMS1 expression reduced the ability of melanoma cells to migrate and invade in vitro. CONCLUSION: Our data suggest that BRMS1 is localized in cytoplasm and nucleus of melanocytic cells and that cellular localization determines its in vivo effect. We hypothesize that cytoplasmic BRMS1 restricts melanoma progression while nuclear BRMS1 possibly promotes melanoma cell invasion.Please see related article: http://www.biomedcentral.com/1741-7015/10/19.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Citoplasma/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Nevo/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Movimiento Celular , Núcleo Celular/metabolismo , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Melanoma/mortalidad , Melanoma/patología , Invasividad Neoplásica/patología , Nevo/mortalidad , Nevo/patología , Pronóstico , Proteínas Represoras , Células Tumorales Cultivadas
11.
Gynecol Oncol ; 126(3): 460-5, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22652154

RESUMEN

OBJECTIVE: To investigate the expression of MGST1 in primary tumors, solid metastases and metastatic effusions in advanced-stage serous ovarian carcinoma (OC) and analyze the association with clinicopathologic parameters, including chemotherapy resistance and survival. METHODS: MGST1 mRNA expression was investigated in 178 tumors (88 effusions, 38 primary carcinomas, 52 solid metastases) from 144 patients using real-time quantitative PCR (qRT-PCR). Forty-two of the 88 effusions were additionally analyzed for MGST1 protein expression by Western blotting. RESULTS: mRNA expression of MGST1 was higher in primary carcinomas and solid metastases compared to effusions (p=0.008 and p=0.012, respectively). In patient-matched samples, mRNA expression of MGST1 was higher in solid metastases compared to effusions (p=0.023), and a trend for higher MGST1 levels in solid metastases compared to primary tumors was observed (p=0.06). Biopsies from primary carcinomas obtained from patients with >200 ml ascites at diagnosis had higher mRNA expression of MGST1 compared to samples from patients with <200 ml ascites (p=0.037). MGST1 mRNA expression was not associated with age, histological grade, tumor stage, residual disease volume, response to chemotherapy, chemotherapy resistance or survival. Western blot analysis of patient-matched effusions showed high concordance between MGST1 protein and mRNA levels measured by qRT-PCR (p<0.001). CONCLUSIONS: The present study documents frequent MGST1 mRNA and protein expression in OC. The data suggest increased activity of oxidative response pathways, reflected by higher mRNA expression, in solid OC tumors compared to metastatic effusions. Additionally, a tumor microenvironment consisting of ascites may induce antioxidant activity.


Asunto(s)
Líquido Ascítico/enzimología , Carcinoma/enzimología , Glutatión Transferasa/metabolismo , Neoplasias Ováricas/enzimología , Derrame Pleural Maligno/enzimología , Ascitis/enzimología , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Estimación de Kaplan-Meier , Metástasis de la Neoplasia , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , ARN Mensajero/metabolismo , Estadísticas no Paramétricas
12.
Sci Rep ; 12(1): 5076, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35332208

RESUMEN

More than half of metastatic melanoma patients receiving standard therapy fail to achieve a long-term survival due to primary and/or acquired resistance. Tumor cell ability to switch from epithelial to a more aggressive mesenchymal phenotype, attributed with AXLhigh molecular profile in melanoma, has been recently linked to such event, limiting treatment efficacy. In the current study, we investigated the therapeutic potential of the AXL inhibitor (AXLi) BGB324 alone or in combination with the clinically relevant BRAF inhibitor (BRAFi) vemurafenib. Firstly, AXL was shown to be expressed in majority of melanoma lymph node metastases. When treated ex vivo, the largest reduction in cell viability was observed when the two drugs were combined. In addition, a therapeutic benefit of adding AXLi to the BRAF-targeted therapy was observed in pre-clinical AXLhigh melanoma models in vitro and in vivo. When searching for mechanistic insights, AXLi was found to potentiate BRAFi-induced apoptosis, stimulate ferroptosis and inhibit autophagy. Altogether, our findings propose AXLi as a promising treatment in combination with standard therapy to improve therapeutic outcome in metastatic melanoma.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Melanoma/patología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/patología , Vemurafenib/farmacología
13.
Int J Cancer ; 126(2): 350-61, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19588500

RESUMEN

The insulin like growth factor (IGF) signaling pathway has been shown to contribute to melanoma progression, but little is known about the role of the IGF binding protein 3 (IGFBP-3) in melanoma biology. The aim of the present study was to characterize expression, function and regulation of IGFBP-3 in malignant melanomas and study its potential as a biomarker. The expression of IGFBP-3 varied between different human melanoma cell lines and reintroduction of the protein in non-expressing cells led to induction of apoptosis. Interestingly, in cell lines expressing endogenous IGFBP-3, siRNA silencing of the protein led to a cell line-dependent decrease in proliferation, but had no effect on apoptosis and invasion. Examination of patient material showed that IGFBP-3 is unexpressed in benign nevi while a slight increase in protein expression was seen in primary and metastatic melanoma. However, expression of the protein was low and no correlation was found with circulating levels of IGFBP-3 in serum, suggesting that IGFBP-3 has limited potential as a predictive marker in malignant melanoma. We showed that promoter methylation of IGFBP-3 occurred in both melanoma cell lines and patient material, implicating epigenetic silencing as a regulation mechanism. Furthermore, expression of the protein was shown to be regulated by the PI3-kinase/AKT and MAPK/ERK1/2 pathways. In summary, our findings suggest that IGFBP-3 can exert dual functional effects influencing both apoptosis and proliferation. Development of resistance to the antiproliferative effects of IGFBP-3 may be an important step in progression of malignant melanomas.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Melanoma/genética , Apoptosis , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Melanoma/metabolismo , Melanoma/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
14.
BMC Cancer ; 10: 233, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20500813

RESUMEN

BACKGROUND: CDC25 phosphatases are important regulators of the cell cycle. Their abnormal expression detected in a number of tumors implies that their dysregulation is involved in malignant transformation. However, the role of CDC25s in vulvar cancer is still unknown. To shed light on their roles in the pathogenesis and to clarify their prognostic values, expression of CDC25A, CDC25B and CDC25C in a large series of vulvar squamous cell carcinomas were examined. METHODS: Expression of CDC25A, CDC25B, CDC25C and phosphorylated (phospho)-CDC25C (Ser216) were examined in 300 vulvar carcinomas using immunohistochemistry. Western blot analysis was utilized to demonstrate CDC25s expression in vulvar cancer cell lines. Kinase and phosphatase assays were performed to exclude cross reactivity among CDC25s isoform antibodies. RESULTS: High nuclear CDC25A and CDC25B expression were observed in 51% and 16% of the vulvar carcinomas, respectively, whereas high cytoplasmic CDC25C expression was seen in 63% of the cases. In cytoplasm, nucleus and cytoplasm/nucleus high phospho-CDC25C (Ser216) expression was identified in 50%, 70% and 77% of the carcinomas, respectively. High expression of CDC25s correlated significantly with malignant features, including poor differentiation and infiltration of vessel for CDC25B, high FIGO stage, presence of lymph node metastases, large tumor diameter, poor differentiation for CDC25C and high FIGO stage, large tumor diameter, deep invasion and poor differentiation for phospho-CDC25C (Ser216). In univariate analysis, high expression of phospho-CDC25C (Ser216) was correlated with poor disease-specific survival (p = 0.04). However, such an association was annulled in multivariate analysis. CONCLUSIONS: Our results suggest that CDC25C and phospho-CDC25C (Ser216) play a crucial role and CDC25B a minor role in the pathogenesis and/or progression of vulvar carcinomas. CDC25B, CDC25C and phospho-CDC25C (Ser216) were associated with malignant features and aggressive cancer phenotypes. However, the CDC25s isoforms were not independently correlated to prognosis.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Neoplasias de la Vulva/enzimología , Fosfatasas cdc25/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Especificidad de Anticuerpos , Western Blotting , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Diferenciación Celular , Línea Celular Tumoral , Núcleo Celular/enzimología , Distribución de Chi-Cuadrado , Citoplasma/enzimología , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Metástasis Linfática , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Fenotipo , Fosforilación , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Serina , Factores de Tiempo , Regulación hacia Arriba , Neoplasias de la Vulva/mortalidad , Neoplasias de la Vulva/patología
15.
Virchows Arch ; 477(6): 857-864, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32533344

RESUMEN

The objective of this study was to analyze the expression and clinical role of mitosis regulators α-thalassemia/mental retardation syndrome X-linked (ATRX) and death-domain-associated protein (DAXX) in metastatic high-grade serous carcinoma (HGSC). ATRX and DAXX protein expression by immunohistochemistry was analyzed in 400 HGSC effusions. DAXX expression was additionally studied in 15 cancer cell lines, including 4 ovarian carcinoma lines, and in 81 of the 400 HGSC effusions using Western blotting. ATRX and DAXX were expressed in HGSC cells in 386/400 (96%) and 348/400 (87%) effusions, respectively. Western blotting showed DAXX expression in all 15 cell lines and in 70/81 (86%) HGSC effusions. DAXX expression by immunohistochemistry was higher in pleural compared to peritoneal effusions (p = 0.006) and in post-chemotherapy compared to pre-chemotherapy effusions (p = 0.004), and its expression was significantly associated with poor overall survival in univariate of the entire cohort (p = 0.014), as well as analysis limited to chemo-naïve effusions tapped at diagnosis (p = 0.038). The former association retained its prognostic role in Cox multivariate survival analysis (p = 0.011). ATRX expression was unrelated to clinicopathologic parameters or survival. In conclusion, DAXX is associated with disease progression and could be a prognostic marker in metastatic HGSC. Silencing this molecule may have therapeutic relevance in this cancer.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas Co-Represoras/biosíntesis , Cistadenocarcinoma Seroso/patología , Chaperonas Moleculares/biosíntesis , Neoplasias Ováricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Líquido Ascítico/metabolismo , Líquido Ascítico/patología , Biomarcadores de Tumor/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/mortalidad , Femenino , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Derrame Pleural Maligno/metabolismo , Derrame Pleural Maligno/mortalidad , Derrame Pleural Maligno/patología , Pronóstico , Proteína Nuclear Ligada al Cromosoma X/biosíntesis , Adulto Joven
16.
PLoS One ; 15(1): e0227187, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31917795

RESUMEN

Receptor tyrosine kinase AXL is a one-pass transmembrane protein upregulated in cancers and associated with lower survival and therapy resistance. AXL can be cleaved by the A Disintegrin and Metalloproteinases (ADAM)10 and ADAM17, yielding a soluble version of the protein. Elevated soluble AXL (sAXL) has been reported to be associated with disease progression in hepatocellular carcinoma, renal cancer, neurofibromatosis type 1 and inflammatory diseases. In the present work, we analyzed sAXL levels in blood from melanoma patients and showed that sAXL increases with disease progression. Additionally, increased sAXL levels were found correlated with shorter two-year survival in stage IV patients treated with ipilimumab. Furthermore, we showed that sAXL levels were related to the percentage of cells expressing AXL in resected melanoma lymph node metastases. This finding was verified in vitro, where sAXL levels in the cell media corresponded to AXL expression in the cells. AXL inhibition using the small-molecular inhibitor BGB324 reduced sAXL levels, while the cellular expression was elevated through increased protein stability. Our findings signify that quantification of sAXL blood levels is a simple and easily assessable method to determine cellular AXL levels and should be further evaluated for its use as a biomarker of disease progression and treatment response.


Asunto(s)
Progresión de la Enfermedad , Melanoma/sangre , Melanoma/mortalidad , Proteínas Proto-Oncogénicas/sangre , Proteínas Tirosina Quinasas Receptoras/sangre , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/uso terapéutico , Benzocicloheptenos/farmacología , Biomarcadores de Tumor/sangre , Línea Celular Tumoral , Femenino , Humanos , Ipilimumab/efectos adversos , Ipilimumab/uso terapéutico , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/química , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Solubilidad , Tasa de Supervivencia , Triazoles/farmacología , Tirosina Quinasa del Receptor Axl
17.
Pigment Cell Melanoma Res ; 33(3): 446-457, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31660681

RESUMEN

MX2 protein is a dynamin-like GTPase2 that has recently been identified as an interferon-induced restriction factor of HIV-1 and other primate lentiviruses. A single nucleotide polymorphism (SNP), rs45430, in an intron of the MX2 gene, was previously reported as a novel melanoma susceptibility locus in genome-wide association studies. Functionally, however, it is still unclear whether and how MX2 contributes to melanoma susceptibility and tumorigenesis. Here, we show that MX2 is differentially expressed in melanoma tumors and cell lines, with most metastatic cell lines showing lower MX2 expression than primary melanoma cell lines and melanocytes. Furthermore, high expression of MX2 RNA in primary melanoma tumors is associated with better patient survival. Overexpression of MX2 reduces in vivo proliferation partially through inhibition of AKT activation, suggesting that it can act as a tumor suppressor in melanoma. However, we have also identified a subset of melanoma cell lines with high endogenous MX2 expression where downregulation of MX2 leads to reduced proliferation. In these cells, MX2 downregulation interfered with DNA replication and cell cycle processes. Collectively, our data for the first time show that MX2 is functionally involved in the regulation of melanoma proliferation but that its function is context-dependent.


Asunto(s)
Ciclo Celular , Melanoma/patología , Proteínas de Resistencia a Mixovirus/metabolismo , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Melanoma/genética , Ratones Desnudos , Proteínas de Resistencia a Mixovirus/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
18.
Mol Cancer Ther ; 19(3): 895-905, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31871265

RESUMEN

Receptor tyrosine kinase AXL is found upregulated in various types of cancer, including melanoma, and correlates with an aggressive cancer phenotype, inducing cell proliferation and epithelial-to-mesenchymal transition. In addition, AXL has recently been linked to chemotherapy resistance, and inhibition of AXL is found to increase DNA damage and reduce expression of DNA repair proteins. In light of this, we aimed to investigate whether targeting AXL together with DNA damage response proteins would be therapeutically beneficial. Using melanoma cell lines, we observed that combined reduction of AXL and CHK1/CHK2 signaling decreased proliferation, deregulated cell-cycle progression, increased apoptosis, and reduced expression of DNA damage response proteins. Enhanced therapeutic effect of combined treatment, as compared with mono-treatment, was further observed in a patient-derived xenograft model and, of particular interest, when applying a three-dimensional ex vivo spheroid drug sensitivity assay on tumor cells harvested directly from 27 patients with melanoma lymph node metastases. Together, these results indicate that targeting AXL together with the DNA damage response pathway could be a promising treatment strategy in melanoma, and that further investigations in patient groups lacking treatment alternatives should be pursued.


Asunto(s)
Daño del ADN , Reparación del ADN , Resistencia a Antineoplásicos , Melanoma/patología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Tiofenos/farmacología , Urea/análogos & derivados , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Puntos de Control del Ciclo Celular , Proliferación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa de Punto de Control 2/antagonistas & inhibidores , Quimioterapia Combinada , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Melanoma/tratamiento farmacológico , Melanoma/genética , Ratones , Ratones Desnudos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/genética , Células Tumorales Cultivadas , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
19.
Tumour Biol ; 30(1): 15-25, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19194111

RESUMEN

The present study analyzed the site-specific expression and clinical role of S100A4 in advanced-stage ovarian carcinoma (OC). S100A4 expression was analyzed in 161 effusions, 67 primary carcinomas and 127 solid metastases using immunohistochemistry. S100A4 levels were additionally measured in 20 effusion supernatants using the immunofluorometric assay IFMA. Cytoplasmic and nuclear S100A4 was detected in cancer cells at all anatomic sites, while myofibroblasts in solid tumors expressed only cytoplasmic S100A4. Nuclear expression was higher in solid tumors than in effusions (p < 0.001), while stromal cell expression was higher in metastases than primary tumors (p = 0.001). IFMA analysis detected S100A4 in all 20 supernatants. Nuclear S100A4 expression in primary carcinomas (p = 0.049), FIGO stage IV (p = 0.019) and poor response to chemotherapy at diagnosis (p < 0.001) were associated with worse overall survival. All three parameters were independent prognosticators in Cox analysis (S100A4: p = 0.048, FIGO stage: p = 0.015, response to chemotherapy: p < 0.001). S100A4 is frequently expressed in tumor and host cells in OC, with upregulated stromal expression in solid metastases. Tumor cell nuclear S100A4 expression is higher in solid tumors than in effusions, and is associated with more aggressive clinical disease in primary carcinoma, suggesting that the tumor-promoting role of this molecule is predominantly exerted at this anatomic site.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Neoplasias Quísticas, Mucinosas y Serosas/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas S100/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/inmunología , Líquido Ascítico/metabolismo , Líquido Ascítico/patología , Núcleo Celular/patología , Citoplasma/patología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Estimación de Kaplan-Meier , Persona de Mediana Edad , Análisis Multivariante , Neoplasias Quísticas, Mucinosas y Serosas/patología , Neoplasias Quísticas, Mucinosas y Serosas/secundario , Neoplasias Ováricas/patología , Neoplasias Ováricas/secundario , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Derrame Pleural/metabolismo , Derrame Pleural/patología , Pronóstico , Estudios Retrospectivos , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Proteínas S100/inmunología
20.
Transl Oncol ; 12(7): 951-958, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31096111

RESUMEN

Although clinical management of melanoma has changed considerably in recent years, intrinsic treatment resistance remains a severe problem and strategies to design personal treatment regimens are highly warranted. We have applied a three-dimensional (3D) ex vivo drug efficacy assay, exposing disaggregated cells from 38 freshly harvested melanoma lymph node metastases and 21 patient derived xenografts (PDXs) to clinical relevant drugs for 7 days, and examined its potential to evaluate therapy response. A strong association between Vemurafenib response and BRAF mutation status was achieved (P < .0001), while enhanced viability was seen in some NRAS mutated tumors. BRAF and NRAS mutated tumors responded comparably to the MEK inhibitor Cobimetinib. Based on the ex vivo results, two tumors diagnosed as BRAF wild-type by routine pathology examinations had to be re-evaluated; one was subsequently found to have a complex V600E mutation, the other a double BRAF mutation (V600E/K601 N). No BRAF inhibitor resistance mechanisms were identified, but PIK3CA and NF1 mutations were identified in two highly responsive tumors. Concordance between ex vivo drug responses using tissue from PDXs and corresponding patient tumors demonstrate that PDX models represent an indefinite source of tumor material that may allow ex vivo evaluation of numerous drugs and combinations, as well as studies of underlying molecular mechanisms. In conclusion, we have established a rapid and low cost ex vivo drug efficacy assay applicable on tumor tissue from patient biopsies. The 3D/spheroid format, limiting the influence from normal adjacent cells and allowing assessment of drug sensitivity to numerous drugs in one week, confirms its potential as a supplement to guide clinical decision, in particular in identifying non-responding patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA