Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Fetal Pediatr Pathol ; 42(2): 241-252, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36062956

RESUMEN

Background: Ewing sarcoma (ES) can be confirmed by identifying the EWSR1-FLI1 fusion transcript. This study is to investigate whether immunostaining (IHC) of PRKCB-a protein directly regulated by EWSR1-FLI1 is a surrogate maker for diagnosing ES in routine practice. Methods: Microarray gene expression analyses were conducted. RKCB IHC was applied to 69 ES confirmed by morphology and molecular methods, and 41 non-Ewing small round cell tumors. EWSR1 rearrangement, EWSR1-FLI1 fusion or t(11;22)(q24;q12) were identified by fluorescence in situ hybridization, reverse transcriptase polymerase chain reaction, or cytogenetic analysis, respectively. Results: Gene array analyses showed significant overexpression of the PRKCB in ES. PRKCB IHC was positive in 19 cases of ES with EWSR1-FLI1 fusion, 3 cases with cytogenetic 11:22 translocation and 59 cases with EWSR1 rearrangement while negative in only one EWSR1 rearranged case. PRKCB IHC is sensitive (98%) and specific (96%) in detecting EWSR1 rearranged ES. Conclusions: PRKCB is a reliable antibody for diagnosing ES in routine practice.


Asunto(s)
Sarcoma de Ewing , Sarcoma , Humanos , Sarcoma de Ewing/diagnóstico , Sarcoma de Ewing/genética , Inmunohistoquímica , Hibridación Fluorescente in Situ , Proteína EWS de Unión a ARN/genética , Biomarcadores , Proteínas de Fusión Oncogénica/genética , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo
2.
Cancer Immunol Immunother ; 67(12): 1853-1862, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30209589

RESUMEN

The GM.CD40L vaccine, which recruits and activates dendritic cells, migrates to lymph nodes, activating T cells and leading to systemic tumor cell killing. When combined with the CCL21 chemokine, which recruits T cells and enhances T-cell responses, additive effects have been demonstrated in non-small cell lung cancer mouse models. Here, we compared GM.CD40L versus GM.CD40L plus CCL21 (GM.CD40L.CCL21) in lung adenocarcinoma patients with ≥ 1 line of treatment. In this phase I/II randomized trial (NCT01433172), patients received intradermal vaccines every 14 days (3 doses) and then monthly (3 doses). A two-stage minimax design was used. During phase I, no dose-limiting toxicities were shown in three patients who received GM.CD40L.CCL21. During phase II, of evaluable patients, 5/33 patients (15.2%) randomized for GM.DCD40L (p = .023) and 3/32 patients (9.4%) randomized for GM.DCD40L.CCL21 (p = .20) showed 6-month progression-free survival. Median overall survival was 9.3 versus 9.5 months with GM.DCD40L versus GM.DCD40L.CCL21 (95% CI 0.70-2.25; p = .44). For GM.CD40L versus GM.CD40L.CCL21, the most common treatment-related adverse events (TRAEs) were grade 1/2 injection site reaction (51.4% versus 61.1%) and grade 1/2 fatigue (35.1% versus 47.2%). Grade 1 immune-mediated TRAEs were isolated to skin. No patients showed evidence of pseudo-progression or immune-related TRAEs of grade 1 or greater of pneumonitis, endocrinopathy, or colitis, and none discontinued treatment due to toxicity. Although we found no significant associations between vaccine immunogenicity and outcomes, in limited biopsies, one patient treated with GMCD40L.CCL21 displayed abundant tumor-infiltrating lymphocytes. This possible effectiveness warrants further investigation of GM.CD40L in combination approaches.


Asunto(s)
Adenocarcinoma/terapia , Ligando de CD40/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/terapia , Quimiocina CCL21/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Inmunoterapia , Adenocarcinoma/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
3.
PLoS One ; 12(1): e0169797, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28085924

RESUMEN

The tumor microenvironment is composed of cellular and stromal components such as tumor cells, mesenchymal cells, immune cells, cancer associated fibroblasts and the supporting extracellular matrix. The tumor microenvironment provides crucial support for growth and progression of tumor cells and affects tumor response to therapeutic interventions. To better understand tumor biology and to develop effective cancer therapeutic agents it is important to develop preclinical platforms that can faithfully recapitulate the tumor microenvironment and the complex interaction between the tumor and its surrounding stromal elements. Drug studies performed in vitro with conventional two-dimensional cancer cell line models do not optimally represent clinical drug response as they lack true tumor heterogeneity and are often performed in static culture conditions lacking stromal tumor components that significantly influence the metabolic activity and proliferation of cells. Recent microfluidic approaches aim to overcome such obstacles with the use of cell lines derived in artificial three-dimensional supportive gels or micro-chambers. However, absence of a true tumor microenvironment and full interstitial flow, leads to less than optimal evaluation of tumor response to drug treatment. Here we report a continuous perfusion microfluidic device coupled with microscopy and image analysis for the assessment of drug effects on intact fresh tumor tissue. We have demonstrated that fine needle aspirate biopsies obtained from patient-derived xenograft models of adenocarcinoma of the lung can successfully be analyzed for their response to ex vivo drug treatment within this biopsy trapping microfluidic device, wherein a protein kinase C inhibitor, staurosporine, was used to assess tumor cell death as a proof of principle. This approach has the potential to study tumor tissue within its intact microenvironment to better understand tumor response to drug treatments and eventually to choose the most effective drug and drug combination for individual patients in a cost effective and timely manner.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Doxorrubicina/farmacología , Técnicas Analíticas Microfluídicas/instrumentación , Neoplasias/patología , Estaurosporina/farmacología , Microambiente Tumoral/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Biopsia , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Ratones , Ratones Pelados , Ratones SCID , Neoplasias/tratamiento farmacológico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Sci Rep ; 5: 16991, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26601688

RESUMEN

Systemic therapy has improved osteosarcoma event-free and overall survival, but 30-50% of patients originally diagnosed will have progressive or recurrent disease, which is difficult to cure. Osteosarcoma has a complex karyotype, with loss of p53 in the vast majority of cases and an absence of recurrent, targetable pathways. In this study, we explored 54 agents that are clinically approved for other oncologic indications, agents in active clinical development, and others with promising preclinical data in osteosarcoma at clinically achievable concentrations in 5 osteosarcoma cell lines. We found significant single-agent activity of multiple agents and tested 10 drugs in all permutations of two-drug combinations to define synergistic combinations by Chou and Talalay analysis. We then evaluated order of addition to choose the combinations that may be best to translate to the clinic. We conclude that the repurposing of chemotherapeutics in osteosarcoma by using an in vitro system may define novel drug combinations with significant in vivo activity. In particular, combinations of proteasome inhibitors with histone deacetylase inhibitors and ixabepilone and MK1775 demonstrated excellent activity in our assays.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Osteosarcoma/tratamiento farmacológico , Inhibidores de Proteasoma/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/patología , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Epotilonas/uso terapéutico , Epotilonas/toxicidad , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Persona de Mediana Edad , Osteosarcoma/patología , Inhibidores de Proteasoma/farmacología , Pirazoles/uso terapéutico , Pirazoles/toxicidad , Pirimidinas/uso terapéutico , Pirimidinas/toxicidad , Pirimidinonas , Trasplante Heterólogo , Células Tumorales Cultivadas
5.
Cancer Control ; 22(2): 193-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26068764

RESUMEN

BACKGROUND: Worldwide, lung cancer is the most common cause of mortality. Toxins from tobacco smoke are known to increase the risk of lung cancer; however, up to 15% of lung cancer-related deaths in men and up to 50% of lung cancer-related deaths in women occur in people who do not smoke. Despite the fact that chemotherapy generally provides a survival benefit for non-small-cell lung cancer, not every patient will respond to therapy and many experience therapy-related adverse events. Thus, predictive markers are used to determine which patients are more likely to respond to a given regimen. METHODS: We reviewed the current medical literature in English relating to predictive markers that may be positive, such as the presence of an activating EGFR mutation. RESULTS: The advances in using EGFR as a molecular predictive marker were summarized. This biomarker influences therapeutic response in patients with lung adenocarcinoma. Clinical evidence supporting its value is also reviewed. CONCLUSIONS: The use of EGFR as a predictive factor in lung adenocarcinoma may help target therapy to individual tumors to achieve the best likelihood for long-term survival and to avoid adverse events from medications unlikely to be effective.


Asunto(s)
Adenocarcinoma/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Sustitución de Aminoácidos , Biomarcadores de Tumor , Carcinoma de Pulmón de Células no Pequeñas/patología , Deleción Cromosómica , Resistencia a Antineoplásicos/genética , Exones/genética , Humanos , Neoplasias Pulmonares/patología , Mutación , Pronóstico
6.
Cancer Control ; 22(2): 226-31, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26068769

RESUMEN

BACKGROUND: Predictive assays for cancer treatment are not new technology, but they have failed to meet the criteria necessary for standardized use in clinical decision-making. METHODS: The authors summarize the use of predictive assays and the challenges and values associated with these assays in the clinical setting. RESULTS: Predictive assays commercially available in the clinical setting are not standardized, have significant obstacles to overcome, and cannot be relied upon by health care professionals due to the limited value these assays provide to the decision-making process for the treatment of patients. CONCLUSIONS: A method that more closely recapitulates the human tumor microenvironment and accurately predicts response with high reproducibility would be beneficial to patient outcomes and quality of life.


Asunto(s)
Neoplasias/patología , Medicina de Precisión/métodos , Biomarcadores de Tumor , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Inmunohistoquímica , Reproducibilidad de los Resultados , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Sci Signal ; 8(359): ra4, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25587191

RESUMEN

Strategies to measure functional signaling-associated protein complexes have the potential to augment current molecular biomarker assays, such as genotyping and expression profiling, used to annotate diseases. Aberrant activation of epidermal growth factor receptor (EGFR) signaling contributes to diverse cancers. We used a proximity ligation assay (PLA) to detect EGFR in a complex with growth factor receptor-bound protein 2 (GRB2), the major signaling adaptor for EGFR. We used multiple lung cancer cell lines to develop and characterize EGFR:GRB2 PLA and correlated this assay with established biochemical measures of EGFR signaling. In a panel of patient-derived xenografts in mice, the intensity of EGFR:GRB2 PLA correlated with the reduction in tumor size in response to the EGFR inhibitor cetuximab. In tumor biopsies from three cohorts of lung cancer patients, positive EGFR:GRB2 PLA was observed in patients with and without EGFR mutations, and the intensity of EGFR:GRB2 PLA was predictive of overall survival in an EGFR inhibitor-treated cohort. Thus, we established the feasibility of using PLA to measure EGFR signaling-associated protein complexes in patient-based materials, suggesting the potential for similar assays for a broader array of receptor tyrosine kinases and other key signaling molecules.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Receptores ErbB/metabolismo , Inmunoensayo/métodos , Neoplasias Pulmonares/diagnóstico , Complejos Multiproteicos/metabolismo , Mapeo de Interacción de Proteínas/métodos , Transducción de Señal/fisiología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Línea Celular Tumoral , Cetuximab , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Proteína Adaptadora GRB2/metabolismo , Xenoinjertos/efectos de los fármacos , Xenoinjertos/patología , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Complejos Multiproteicos/fisiología
8.
J Thorac Oncol ; 9(7): 974-982, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24922005

RESUMEN

INTRODUCTION: Interleukin-6 (IL-6) can activate downstream signaling pathways in lung cancer cells, such as the STAT3 pathway, and is reported to be produced by tumor cells with activating EGFR mutations. We examined IL-6/STAT3 in lung cancer tumor tissues and the effects of siltuximab, a neutralizing antibody to human IL-6, in mouse models of lung cancer. METHODS: IL-6 and STAT3 activation levels were compared with tumor histology and presence of KRAS mutations in snap-frozen, non-small-cell lung cancer tumors. The effects of siltuximab alone or in combination with erlotinib were examined in mouse xenograft models constructed using three cell line xenograft models and one primary explant mouse model. We examined the influence of cancer-associated fibroblasts (CAFs) on tumor growth and siltuximab effects. RESULTS: IL-6 levels were higher in tumors of squamous cell versus adenocarcinoma histology and were not associated with presence of KRAS mutations. Tyrosine phosphorylation status of STAT3 did not correlate with tumor IL-6 levels. Serine phosphorylation of STAT3 was correlated with KRAS mutation status. Both tumor and stromal cells contributed to total IL-6 within tumors. Siltuximab had minimal effect as a single agent in xenografts with tumor cells alone; however, in models coadministered with CAFs, siltuximab had more potent effects on tumor inhibition. We observed no effects of combined erlotinib and siltuximab. CONCLUSIONS: IL-6 is elevated in subsets of human NSCLCs, especially with squamous cell histology. Tumors supported by stromal production of IL-6 seem to be the most vulnerable to tumor growth inhibition by siltuximab.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Adenocarcinoma/química , Adenocarcinoma/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/química , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Clorhidrato de Erlotinib , Femenino , Fibroblastos , Humanos , Interleucina-6/análisis , Interleucina-6/inmunología , Neoplasias Pulmonares/química , Neoplasias Pulmonares/genética , Ratones , Fosforilación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras) , Quinazolinas/administración & dosificación , Factor de Transcripción STAT3/metabolismo , Serina/metabolismo , Tirosina/metabolismo , Proteínas ras/genética
10.
Clin Cancer Res ; 20(6): 1644-55, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24429877

RESUMEN

PURPOSE: Panobinostat, a histone deacetylase (HDAC) inhibitor, enhances antiproliferative activity in non-small cell lung cancer (NSCLC) cell lines when combined with erlotinib. We evaluated this combination in patients with advanced NSCLC and head and neck cancer. EXPERIMENTAL DESIGN: Eligible patients were enrolled in a 3+3 dose-escalation design to determine the maximum tolerated dose (MTD) of twice weekly panobinostat plus daily erlotinib at four planned dose levels (DL). Pharmacokinetics, blood, fat pad biopsies (FPB) for histone acetylation, and paired pre and posttherapy tumor biopsies for checkpoint kinase 1 (CHK1) expression were assessed. RESULTS: Of 42 enrolled patients, 33 were evaluable for efficacy. Dose-limiting toxicities were prolonged-QTc and nausea at DL3. Adverse events included fatigue and nausea (grades 1-3), and rash and anorexia (grades 1-2). Disease control rates were 54% for NSCLC (n = 26) and 43% for head and neck cancer (n = 7). Of 7 patients with NSCLC with EGF receptor (EGFR) mutations, 3 had partial response, 3 had stable disease, and 1 progressed. For EGFR-mutant versus EGFR wild-type patients, progression-free survival (PFS) was 4.7 versus 1.9 months (P = 0.43) and overall survival was 41 (estimated) versus 5.2 months (P = 0.39). Erlotinib pharmacokinetics was not significantly affected. Correlative studies confirmed panobinostat's pharmacodynamic effect in blood, FPB, and tumor samples. Low CHK1 expression levels correlated with PFS (P = 0.006) and response (P = 0.02). CONCLUSIONS: We determined MTD at 30 mg (panobinostat) and 100 mg (erlotinib). Further studies are needed to further explore the benefits of HDAC inhibitors in patients with EGFR-mutant NSCLC, investigate FPB as a potential surrogate source for biomarker investigations, and validate CHK1's predictive role.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Ácidos Hidroxámicos/administración & dosificación , Indoles/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/administración & dosificación , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Supervivencia sin Enfermedad , Clorhidrato de Erlotinib , Femenino , Neoplasias de Cabeza y Cuello/mortalidad , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/efectos adversos , Humanos , Ácidos Hidroxámicos/efectos adversos , Ácidos Hidroxámicos/farmacocinética , Indoles/efectos adversos , Indoles/farmacocinética , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Panobinostat , Quinazolinas/efectos adversos , Quinazolinas/farmacocinética , Resultado del Tratamiento
11.
PLoS One ; 8(12): e82875, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24358232

RESUMEN

Combinations of targeted drugs have been employed to treat sarcomas, however, response rates have not improved notably, therefore emphasizing the need for novel treatments. In addition, imaging approaches to assess therapeutic response is lacking, as currently measurable indices, such as volume and/or diameter, do not accurately correlate with changes in tumor biology. In this study, quantitative and profound analyses of magnetic resonance imaging (MRI) were developed to evaluate these as imaging biomarkers for MK1775 and Gem in an osteosarcoma xenotransplant model at early time-points following treatment. Notably, we showed that Gem and Gem+MK1775 groups had significantly inhibited tumor growth by day 4, which was presaged by elevations in mean ADC by 24 hours post treatment. Significant differences were also observed at later time points for the Gem+MK1775 combination and MK1775 therapy. ADC distribution and entropy (randomness of ADC values) were also elevated by 24 hours following therapy. Immunohistochemistry demonstrated that these treatment-related increases in ADC correlated with apoptosis and observed cell condensations (dense- and exploded bodies). These findings underline the role of ADC as a quantitative imaging biomarker for therapy-induced response and show promising clinical relevance in the sarcoma patient population.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Imagen de Difusión por Resonancia Magnética , Osteosarcoma/diagnóstico , Osteosarcoma/tratamiento farmacológico , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Animales , Neoplasias Óseas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Femenino , Humanos , Ratones , Ratones SCID , Persona de Mediana Edad , Terapia Molecular Dirigida , Osteosarcoma/patología , Pronóstico , Pirimidinonas , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
12.
J Proteome Res ; 12(9): 4005-17, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23901793

RESUMEN

While targeted therapy based on the idea of attenuating the activity of a preselected, therapeutically relevant protein has become one of the major trends in modern cancer therapy, no truly specific targeted drug has been developed and most clinical agents have displayed a degree of polypharmacology. Therefore, the specificity of anticancer therapeutics has emerged as a highly important but severely underestimated issue. Chemical proteomics is a powerful technique combining postgenomic drug-affinity chromatography with high-end mass spectrometry analysis and bioinformatic data processing to assemble a target profile of a desired therapeutic molecule. Due to high demands on the starting material, however, chemical proteomic studies have been mostly limited to cancer cell lines. Herein, we report a down-scaling of the technique to enable the analysis of very low abundance samples, as those obtained from needle biopsies. By a systematic investigation of several important parameters in pull-downs with the multikinase inhibitor bosutinib, the standard experimental protocol was optimized to 100 µg protein input. At this level, more than 30 well-known targets were detected per single pull-down replicate with high reproducibility. Moreover, as presented by the comprehensive target profile obtained from miniaturized pull-downs with another clinical drug, dasatinib, the optimized protocol seems to be extendable to other drugs of interest. Sixty distinct human and murine targets were finally identified for bosutinib and dasatinib in chemical proteomic experiments utilizing core needle biopsy samples from xenotransplants derived from patient tumor tissue. Altogether, the developed methodology proves robust and generic and holds many promises for the field of personalized health care.


Asunto(s)
Compuestos de Anilina/química , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Neoplasias Pulmonares/enzimología , Nitrilos/química , Proteínas Tirosina Quinasas/aislamiento & purificación , Pirimidinas/química , Quinolinas/química , Tiazoles/química , Animales , Biopsia , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Cromatografía de Afinidad , Dasatinib , Humanos , Células K562 , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Anotación de Secuencia Molecular , Terapia Molecular Dirigida , Trasplante de Neoplasias , Mapas de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/química , Proteómica
13.
PLoS One ; 8(3): e57523, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23520471

RESUMEN

Sarcomas are rare and heterogeneous mesenchymal tumors affecting both pediatric and adult populations with more than 70 recognized histologies. Doxorubicin and ifosfamide have been the main course of therapy for treatment of sarcomas; however, the response rate to these therapies is about 10-20% in metastatic setting. Toxicity with the drug combination is high, response rates remain low, and improvement in overall survival, especially in the metastatic disease, remains negligible and new agents are needed. Wee1 is a critical component of the G2/M cell cycle checkpoint control and mediates cell cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by MK1775 has been reported to enhance the cytotoxic effect of DNA damaging agents in different types of carcinomas. In this study we investigated the therapeutic efficacy of MK1775 in various sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo both alone and in combination with gemcitabine, which is frequently used in the treatment of sarcomas. Our data demonstrate that MK1775 treatment as a single agent at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. Additionally, MK1775 significantly enhances the cytotoxic effect of gemcitabine in sarcoma cells lines with different p53 mutational status. In patient-derived bone and soft tissue sarcoma samples we showed that MK1775 alone and in combination with gemcitabine causes significant apoptotic cell death. Magnetic resonance imaging (MRI) and histopathologic studies showed that MK1775 induces significant cell death and terminal differentiation in a patient-derived xenograft mouse model of osteosarcoma in vivo. Our results together with the high safety profile of MK1775 strongly suggest that this drug can be used as a potential therapeutic agent in the treatment of both adult as well as pediatric sarcoma patients.


Asunto(s)
Antimetabolitos Antineoplásicos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Neoplasias Femorales/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Osteosarcoma/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles , Pirimidinas , Adolescente , Adulto , Animales , Antimetabolitos Antineoplásicos/agonistas , Antimetabolitos Antineoplásicos/farmacología , Muerte Celular , Diferenciación Celular , Línea Celular Tumoral , Niño , Preescolar , Desoxicitidina/agonistas , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Neoplasias Femorales/patología , Humanos , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Osteosarcoma/patología , Pirazoles/agonistas , Pirazoles/farmacología , Pirimidinas/agonistas , Pirimidinas/farmacología , Pirimidinonas , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
14.
Diagn Cytopathol ; 41(2): 159-63, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23335454

RESUMEN

Squamous cell carcinoma with rhabdoid features (SCCRF) is a very rare and unusual cutaneous tumor. Here, we report an extraordinary case diagnosed by fine needle aspiration biopsy, in a 66-year-old man, status post multiple organ transplantation. The patient presented with a large ulcerating fungating mass in his forearm that had all the light microscopic and immunohistochemical features of a SCCRF. Previously six cases of SCCRF phenotype diagnosed by surgical pathology have been reported. This is the first case diagnosed cytologically. A review of the literature with emphasis on the differential diagnoses of such unusual rhabdoid-like tumors in fine-needle aspiration biopsy and the potential molecular mechanism for rhabdoid phenotype in transplant patients are discussed.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Cutáneas/patología , Neoplasias de los Tejidos Blandos/patología , Anciano , Biopsia con Aguja Fina , Carcinoma de Células Escamosas/diagnóstico , Núcleo Celular/patología , Antebrazo , Humanos , Masculino , Trasplante de Órganos , Fenotipo , Tumor Rabdoide/patología , Neoplasias Cutáneas/diagnóstico , Neoplasias de los Tejidos Blandos/diagnóstico
15.
Cancer ; 119(5): 1023-32, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23065656

RESUMEN

BACKGROUND: Chemoprevention for lung cancer with nutraceutical or anti-inflammatory agents has had mixed clinical benefit. Novel targeted agents hold the promise of greater efficacy and selectivity. The authors of this report evaluated enzastaurin, a selective protein kinase C-ß (PKC-ß) inhibitor with antiproliferative and proapoptotic properties, in former smokers. METHODS: The primary objective of this study was to compare the average fraction of Ki-67-stained cells (the Ki-67 labeling index [LI]) in bronchial biopsy specimens that were collected before and after treatment. Participants were randomized (2:1) to receive either 6 months of daily oral enzastaurin (500 mg) or placebo. Stratification was based on morphology, history of lung cancer, and airway obstruction. RESULTS: In pretrial investigations, the rationale for PKC-ß inhibition and pathway interrogation was established in premalignant lesions and early stage lung cancer. In an intent-to-treat analysis, of 40 randomized participants, there was no significant difference in the pretreatment/post-treatment change in the Ki-67 LI between the enzastaurin group and the placebo group (P = .53). Six participants discontinued enzastaurin, including 4 participants who had adverse events, including abdominal distension, deep vein thrombosis, hyponatremia, and rash, and 2 participants who decided to discontinue. One participant in the placebo group was discontinued on the study because of noncompliance. Two participants had ≥1 serious adverse event (bradycardia, deep vein thrombosis, and hypotension). CONCLUSIONS: To the authors' knowledge, this represents the first chemoprevention trial with a non-US Food and Drug Administration-approved, oral, small-molecule-targeted agent. Although the primary endpoint was not met, enzastaurin was tolerable for 6 months by 75% of participants, and there was a suggestion of response in a subset analysis that was restricted to those who had metaplastic or dysplastic lesions.


Asunto(s)
Indoles/farmacología , Neoplasias Pulmonares/prevención & control , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Fumar/efectos adversos , Anciano , Femenino , Humanos , Indoles/efectos adversos , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/tratamiento farmacológico , Proteína Quinasa C beta , Riesgo
16.
Int J Clin Exp Pathol ; 5(9): 982-90, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23119117

RESUMEN

Mixed germ cell tumors are rare neoplasms that are known to occur in the anterior mediastinum. Characterized by two or more types of germ cell components, these tumors comprise upwards of 25% of mediastinal germ cell tumors. Even rarer are those harboring somatic-type malignancies such as carcinoma, sarcoma, and hematopoietic malignancies. To date, however, there are no known cases of melanoma arising in a malignant mixed germ cell tumor of the anterior mediastinum. We describe the first case of malignant melanoma with spindle and epithelioid components arising from respiratory epithelium in a mediastinal malignant mixed germ cell tumor of a 32-year-old male. In addition, we also provide evidence supporting the theory of neuroendocrine cells as the origin of melanoma arising in the respiratory epithelium. This case emphasizes the need to carefully evaluate all germ cell tumors, not only for a myriad of benign embryological components, but also for malignancies arising in these components, as they might change the prognosis and patient's course of treatment. This microscopic approach should bring to light the diversity of mixed germ cell tumors in addition to somatic malignancies with corresponding biologic potentials.


Asunto(s)
Neoplasias del Mediastino/patología , Melanoma/patología , Neoplasias Complejas y Mixtas/patología , Mucosa Respiratoria/patología , Teratoma/patología , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/análisis , Biopsia , Quimioterapia Adyuvante , Humanos , Inmunohistoquímica , Masculino , Neoplasias del Mediastino/química , Neoplasias del Mediastino/terapia , Melanoma/química , Melanoma/terapia , Terapia Neoadyuvante , Neoplasias Complejas y Mixtas/química , Neoplasias Complejas y Mixtas/terapia , Mucosa Respiratoria/química , Teratoma/química , Teratoma/terapia , Tomografía Computarizada por Rayos X
17.
Cancer Res ; 72(21): 5483-93, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942258

RESUMEN

Autophagy attenuates the efficacy of conventional chemotherapy but its effects on immunotherapy have been little studied. Here, we report that chemotherapy renders tumor cells more susceptible to lysis by CTL in vivo. Moreover, bystander tumor cells that did not express antigen were killed by CTL. This effect was mediated by transient but dramatic upregulation of the mannose-6-phosphate receptor (MPR) on the tumor cell surface. Antitumor effects of combined treatment related to the kinetics of MPR upregulation and abrogation of this event abolished the combined effect of immunotherapy and chemotherapy. MPR accumulation on the tumor cell surface during chemotherapy was observed in different mouse tumor models and in patients with multiple myeloma. Notably, this effect was the result of redistribution of the receptor caused by chemotherapy-inducible autophagy. Together, our findings reveal one molecular mechanism through which the antitumor effects of conventional cancer chemotherapy and immunotherapy are realized.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Citotoxicidad Inmunológica/fisiología , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Western Blotting , Terapia Combinada , Quimioterapia , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal , Neoplasias/inmunología , Neoplasias/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor IGF Tipo 2/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
18.
Mol Cancer ; 11: 73, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23009336

RESUMEN

BACKGROUND: Cancer stem cells are thought to be responsible for the initiation and progression of cancers. In non-small cell lung cancers (NSCLCs), Hoechst 33342 dye effluxing side population (SP) cells are shown to have stem cell like properties. The oncogenic capacity of cancer stem-like cells is in part due to their ability to self-renew; however the mechanistic correlation between oncogenic pathways and self-renewal of cancer stem-like cells has remained elusive. Here we characterized the SP cells at the molecular level and evaluated its ability to generate tumors at the orthotopic site in the lung microenvironment. Further, we investigated if the self-renewal of SP cells is dependent on EGFR mediated signaling. RESULTS: SP cells were detected and isolated from multiple NSCLC cell lines (H1650, H1975, A549), as well as primary human tumor explants grown in nude mice. SP cells demonstrated stem-like properties including ability to self-renew and grow as spheres; they were able to generate primary and metastatic tumors upon orthotopic implantation into the lung of SCID mice. In vitro study revealed elevated expression of stem cell associated markers like Oct4, Sox2 and Nanog as well as demonstrated intrinsic epithelial to mesenchymal transition features in SP cells. Further, we show that abrogation of EGFR, Src and Akt signaling through pharmacological or genetic inhibitors suppresses the self-renewal growth and expansion of SP-cells and resulted in specific downregulation of Sox2 protein expression. siRNA mediated depletion of Sox2 significantly blocked the SP phenotype as well as its self-renewal capacity; whereas other transcription factors like Oct4 and Nanog played a relatively lesser role in regulating self-renewal. Interestingly, Sox2 was elevated in metastatic foci of human NSCLC samples. CONCLUSIONS: Our findings suggest that Sox2 is a novel target of EGFR-Src-Akt signaling in NSCLCs that modulates self-renewal and expansion of stem-like cells from NSCLC. Therefore, the outcome of the EGFR-Src-Akt targeted therapy may rely upon the expression and function of Sox2 within the NSCLC-CSCs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Receptores ErbB/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción SOXB1/genética , Familia-src Quinasas/metabolismo , Animales , Biomarcadores/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Receptores ErbB/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Células de Población Lateral/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 72(10): 2501-11, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22461510

RESUMEN

Driver tyrosine kinase mutations are rare in sarcomas, and patterns of tyrosine phosphorylation are poorly understood. To better understand the signaling pathways active in sarcoma, we examined global tyrosine phosphorylation in sarcoma cell lines and human tumor samples. Anti-phosphotyrosine antibodies were used to purify tyrosine phosphorylated peptides, which were then identified by liquid chromatography and tandem mass spectrometry. The findings were validated with RNA interference, rescue, and small-molecule tyrosine kinase inhibitors. We identified 1,936 unique tyrosine phosphorylated peptides, corresponding to 844 unique phosphotyrosine proteins. In sarcoma cells alone, peptides corresponding to 39 tyrosine kinases were found. Four of 10 cell lines showed dependence on tyrosine kinases for growth and/or survival, including platelet-derived growth factor receptor (PDGFR)α, MET, insulin receptor/insulin-like growth factor receptor signaling, and SRC family kinase signaling. Rhabdomyosarcoma samples showed overexpression of PDGFRα in 13% of examined cases, and sarcomas showed abundant tyrosine phosphorylation and expression of a number of tyrosine phosphorylated tyrosine kinases, including DDR2, EphB4, TYR2, AXL, SRC, LYN, and FAK. Together, our findings suggest that integrating global phosphoproteomics with functional analyses with kinase inhibitors can identify drivers of sarcoma growth and survival.


Asunto(s)
Proteínas Tirosina Quinasas/metabolismo , Proteómica/métodos , Sarcoma/enzimología , Línea Celular Tumoral , Humanos , Fosforilación , Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
20.
Mol Cancer Ther ; 11(1): 174-82, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22084170

RESUMEN

Wee1 is a critical component of the G(2)-M cell-cycle checkpoint control and mediates cell-cycle arrest by regulating the phosphorylation of CDC2. Inhibition of Wee1 by a selective small molecule inhibitor MK1775 can abrogate G(2)-M checkpoint, resulting in premature mitotic entry and cell death. MK1775 has recently been tested in preclinical and clinical studies of human carcinoma to enhance the cytotoxic effect of DNA-damaging agents. However, its role in mesenchymal tumors, especially as a single agent, has not been explored. Here, we studied the cytotoxic effect of MK1775 in various sarcoma cell lines and patient-derived tumor explants ex vivo. Our data show that MK1775 treatment at clinically relevant concentrations leads to unscheduled entry into mitosis and initiation of apoptotic cell death in all sarcomas tested. In MK1775-treated cells, CDC2 activity was enhanced, as determined by decreased inhibitory phosphorylation of tyrosine-15 residue and increased expression of phosphorylated histone H3, a marker of mitotic entry. The cytotoxic effect of Wee1 inhibition on sarcoma cells seems to be independent of p53 status as all sarcoma cell lines with different p53 mutation were highly sensitive to MK1775 treatment. Finally, in patient-derived sarcoma samples, we showed that MK1775 as a single agent causes significant apoptotic cell death, suggesting that Wee1 inhibition may represent a novel approach in the treatment of sarcomas.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Mitosis/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Sarcoma/patología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Histonas/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Pirimidinonas , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...