Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Cancer Educ ; 36(6): 1295-1305, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-32683629

RESUMEN

The University of Toronto - Department of Radiation Oncology (UTDRO) has had a well-established Fellowship Program for over 20 years. An assessment of its graduates was conducted to evaluate training experience and perceived impact on professional development. Graduates of the UTDRO Fellowship Program between 1991 and 2015 were the focus of our review. Current employment status was collected using online tools. A study-specific web-based questionnaire was distributed to 263/293 graduates for whom active e-mails were identified; questions focused on training experience, and impact on career progression and academic productivity. As a surrogate measure for the impact of UTDRO Fellowship training, a comparison of current employment and scholarly activities of individuals who obtained their Fellow of the Royal College of Physicians of Canada (FRCPC) designation in Radiation Oncology between 2000 and 2012, with (n = 57) or without (n = 230) UTDRO Fellowship training, was conducted. Almost all UTDRO Fellowship graduates were employed as staff radiation oncologists (291/293), and most of those employed were associated with additional academic (130/293), research (53/293), or leadership (68/293) appointments. Thirty-eight percent (101/263) of alumni responded to the online survey. The top two reasons for completing the Fellowship were to gain specific clinical expertise and exposure to research opportunities. Respondents were very satisfied with their training experience, and the vast majority (99%) would recommend the program to others. Most (96%) felt that completing the Fellowship was beneficial to their career development. University of Toronto, Department of Radiation Oncology Fellowship alumni were more likely to hold university, research, and leadership appointments, and author significantly more publications than those with FRCPC designation without fellowship training from UTDRO. The UTDRO Fellowship Program has been successful since its inception, with the majority of graduates reporting positive training experiences, benefits to scholarly output, and professional development for their post-fellowship careers. Key features that would optimize the fellowship experience and its long-term impact on trainees were also identified.


Asunto(s)
Internado y Residencia , Oncología por Radiación , Selección de Profesión , Becas , Humanos , Liderazgo , Oncólogos de Radiación , Encuestas y Cuestionarios
2.
Int J Radiat Oncol Biol Phys ; 115(1): 39-47, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36309074

RESUMEN

PURPOSE: During the COVID-19 pandemic, many radiation oncology departments worldwide adopted the use of shorter and more intense hypofractionated regimens. Hospital foot traffic was reduced through virtual care. This study's primary objective was to assess the collective environmental effect of these strategic changes by identifying sources of carbon dioxide equivalents (CO2e). The rate of radiation-related adverse events from the increased use of hypofractionated treatments was assessed. METHODS AND MATERIALS: All patients treated with external beam radiation therapy from April 1, 2019, to March 31, 2021, at our single institution were identified (n = 10,175) along with their radiation therapy visits (176,423 fractions) and unplanned visits to the radiation nursing clinic or emergency department. Out-patient hospital and virtual visits (n = 75,853) during this same period were also analyzed. Environmental effect measures, including linear accelerator power usage, patient travel distances, and personal protection equipment consumption were all converted into CO2e. RESULTS: The use of curative hypofractionated regimens increased from 17% to 27% during the pandemic year. Carbon footprint was reduced by 39% during the pandemic year (1,332,388 kg CO2e) compared with the prepandemic year (2,024,823 kg CO2e). Comparing patients in the prepandemic versus pandemic year, there was a significant reduction in the proportion of hypofractionated patients who needed a visit to either the radiation nursing clinic (39% vs 25%; P < .001) or emergency department (6% vs 2%; P < .001) during and within 90 days of radiation therapy. CONCLUSIONS: This is the first study to demonstrate the environmental benefits of increased use of hypofractionated regimens and virtual care, while assuring that there was no added acute radiation-related adverse event. Our findings support their continued use as one of many long-term strategies to reduce the environmental footprint of health care delivery.


Asunto(s)
COVID-19 , Oncología por Radiación , Humanos , Pandemias/prevención & control , COVID-19/epidemiología , Servicio de Urgencia en Hospital , Hospitales
3.
BMC Cancer ; 11: 500, 2011 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-22128797

RESUMEN

Micro-RNAs (miRs) are important regulators of mRNA and protein expression; the ability of miR expression profilings to distinguish different cancer types and classify their sub-types has been well-described. They also represent a novel biological entity with potential value as tumour biomarkers, which can improve diagnosis, prognosis, and monitoring of treatment response for human cancers. This endeavour has been greatly facilitated by the stability of miRs in formalin-fixed paraffin-embedded (FFPE) tissues, and their detection in circulation. This review will summarize some of the key dysregulated miRs described to date in human epithelial malignancies, and their potential value as molecular bio-markers in FFPE tissues and blood samples. There remain many challenges in this domain, however, with the evolution of different platforms, the complexities of normalizing miR profiling data, and the importance of evaluating sufficiently-powered training and validation cohorts. Nonetheless, well-conducted miR profiling studies should contribute important insights into the molecular aberrations driving human cancer development and progression.


Asunto(s)
Biomarcadores de Tumor/análisis , MicroARNs/análisis , Neoplasias Glandulares y Epiteliales/metabolismo , Humanos , MicroARNs/sangre , Análisis por Micromatrices , Adhesión en Parafina
4.
Int J Radiat Oncol Biol Phys ; 109(3): 656-660, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33516435

RESUMEN

PURPOSE: The Accelerated Education Program (AEP) at the Princess Margaret Cancer Centre (PM) has been offering continuing medical education courses since 2006. The purpose of this study was to assess learner experiences, perspectives, and outcomes using Kirkpatrick's Four Level Training Evaluation Model (ie, reaction, learning, behavior, results) to ascertain whether it was meeting stated goals. METHODS AND MATERIALS: Past course participants (2010-2018) were invited to participate in a semistructured interview. Interviews were transcribed verbatim; thematic analysis was conducted by a 4-person research team. RESULTS: Seventeen participants including 2 medical physicists, 6 radiation oncologists, and 9 radiation therapists from 6 countries on 4 continents participated in the study. Interviews lasted an average of 25 minutes. Consistently positive outcomes were reported at each level of Kirkpatrick's model. At the reaction level, participants liked the small, interactive case-based design, exposure to renowned faculty and practices from PM and other major centers, and the interprofessional practice (IPP) approach. Suggestions for improvements include enhancing practical content. At the learning level, participants reported gaining new knowledge or skills and new awareness or attitudes. Behavior changes described included sharing learnings with colleagues, implementing changes in practice or techniques, departmental structure, and IPP. Participants described the effects on clinical practice (results) in quality of care, access to care, and academic contribution. Identified barriers to change related to the restricted internal capacity for change and the need for wider staff training. CONCLUSIONS: AEP courses were found to have a positive effect on local practices ranging from confirmation of current practice through to increased access to and quality of advanced radiotherapeutic techniques and care. Our findings confirm that AEP is achieving its goal of "putting innovation to work" and suggest curricular improvements that can enhance these effects.


Asunto(s)
Educación Médica Continua , Internacionalidad , Oncología por Radiación/educación , Actitud del Personal de Salud , Educación Médica Continua/métodos , Humanos , Aprendizaje , Ontario , Investigación Cualitativa , Oncólogos de Radiación/educación , Oncólogos de Radiación/estadística & datos numéricos
5.
Int J Cancer ; 126(9): 2036-48, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19739117

RESUMEN

Polo-like kinase 1 (Plk1) is a critical regulator of many stages of mitosis; increasing evidence indicates that Plk1 overexpression correlates with poor clinical outcome, yet its mechanism of regulation remains unknown. Hence, a detailed evaluation was undertaken of Plk1 expression in human nasopharyngeal cancer (NPC), the cellular effects of targeting Plk1 using siRNA in combination with ionizing radiation (RT) and potential upstream microRNAs (miRs) that might regulate Plk1 expression. Using immunohistochemistry, Plk1 was observed to be overexpressed in 28 of 40 (70%) primary NPC biopsies, which in turn was associated with a higher likelihood of recurrence (p = 0.018). SiPlk1 significantly inhibited Plk1 mRNA and protein expression, and decreased Cdc25c levels in NPC cell lines. This depletion resulted in cytotoxicity of C666-1 cells, enhanced by the addition of RT, mediated by G2/M arrest, increased DNA double-strand breaks, apoptosis, and caspase activation. Immunofluorescence demonstrated that the G2/M arrest was associated with aberrant spindle formation, leading to mitotic arrest. In vivo, transfection of C666-1 cells and systemic delivery of siPlk1 decreased tumour growth. MicroRNA-100 (miR-100) was predicted to target Plk1 mRNA, which was indeed underexpressed in C666-1 cells, inversely correlating with Plk1 expression. Using luciferase constructs containing the 3'-UTR of Plk1 sequence, we document that miR-100 can directly target Plk1. Hence, our data demonstrate for the first time that underexpressed miR-100 leads to Plk1 overexpression, which in turn contributes to NPC progression. Targeting Plk1 will cause mitotic catastrophe, with significant cytotoxicity both in vitro and in vivo, underscoring the important therapeutic opportunity of Plk1 in NPC.


Asunto(s)
Proteínas de Ciclo Celular/genética , MicroARNs/fisiología , Neoplasias Nasofaríngeas/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Animales , Apoptosis , Proteínas de Ciclo Celular/antagonistas & inhibidores , Supervivencia Celular/efectos de la radiación , Femenino , Regulación de la Expresión Génica , Histonas/análisis , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Nasofaríngeas/enzimología , Neoplasias Nasofaríngeas/terapia , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Mensajero/análisis , ARN Interferente Pequeño/genética , Quinasa Tipo Polo 1
6.
Clin Cancer Res ; 15(11): 3716-24, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19470731

RESUMEN

PURPOSE: Seliciclib is a small-molecule cyclin-dependent kinase inhibitor, which has been reported to induce apoptosis and cell cycle arrest in EBV-negative nasopharyngeal carcinoma cell lines. Because most nasopharyngeal carcinoma patients harbor EBV, we proceeded to evaluate the cytotoxic effects of seliciclib in EBV-positive nasopharyngeal carcinoma models. EXPERIMENTAL DESIGN: Cytotoxicity of seliciclib was investigated in the EBV-positive cell line C666-1 and the C666-1 and C15 xenograft models. Caspase activities and cell cycle analyses were measured by flow cytometry. Efficacy of combined treatment of seliciclib with radiation therapy was also evaluated. RESULTS: Seliciclib caused significant cytotoxicity in the C666-1 cells in a time- and dose-dependent manner, with accumulation of cells in both sub-G(1) and G(2)-M phases, indicative of apoptosis and cell cycle arrest, respectively. Caspase-2, -3, -8, and -9 activities were all increased, with caspase-3 being the most significantly activated at 48 h after treatment. These cells also showed a reduction of Mcl-1 mRNA and protein levels. Combined treatment of seliciclib with radiation therapy showed a synergistic interaction with enhanced cytotoxicity in C666-1 cells and delayed repair of double-strand DNA breaks. For in vivo models, significant delays in tumor growth were observed for both C666-1 and C15 tumors, which were associated with enhanced apoptosis as determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and immunohistochemistry analyses. CONCLUSIONS: Seliciclib enhanced the antitumor efficacy of radiation therapy in EBV-positive nasopharyngeal carcinoma, characterized by G(2)-M arrest, and apoptosis, associated with an induction in caspase activity. This process is mediated by reduction in Mcl-1 expression and by attenuation of double-strand DNA break repair.


Asunto(s)
Neoplasias Nasofaríngeas/terapia , Purinas/uso terapéutico , Radiación Ionizante , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Western Blotting , Caspasa 2/metabolismo , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Terapia Combinada , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Roscovitina , Factores de Tiempo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Pharmacol ; 76(5): 969-83, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19654225

RESUMEN

A potential therapeutic agent for human head and neck cancer (HNC), cetrimonium bromide (CTAB), was identified through a cell-based phenotype-driven high-throughput screen (HTS) of 2000 biologically active or clinically used compounds, followed by in vitro and in vivo characterization of its antitumor efficacy. The preliminary and secondary screens were performed on FaDu (hypopharyngeal squamous cancer) and GM05757 (primary normal fibroblasts), respectively. Potential hit compounds were further evaluated for their anticancer specificity and efficacy in combination with standard therapeutics on a panel of normal and cancer cell lines. Mechanism of action, in vivo antitumor efficacy, and potential lead compound optimizations were also investigated. In vitro, CTAB interacted additively with gamma radiation and cisplatin, two standard HNC therapeutic agents. CTAB exhibited anticancer cytotoxicity against several HNC cell lines, with minimal effects on normal fibroblasts; a selectivity that exploits cancer-specific metabolic aberrations. The central mode of cytotoxicity was mitochondria-mediated apoptosis via inhibition of H(+)-ATP synthase activity and mitochondrial membrane potential depolarization, which in turn was associated with reduced intracellular ATP levels, caspase activation, elevated sub-G(1) cell population, and chromatin condensation. In vivo, CTAB ablated tumor-forming capacity of FaDu cells and delayed growth of established tumors. Thus, using an HTS approach, CTAB was identified as a potential apoptogenic quaternary ammonium compound possessing in vitro and in vivo efficacy against HNC models.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Compuestos de Cetrimonio/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/fisiología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cetrimonio , Compuestos de Cetrimonio/farmacología , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Biotechniques ; 44(2): ix-xiv, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18422490

RESUMEN

The colony formation assay (CFA) is the gold standard for measuring the effects of cytotoxic agents on cancer cells in vitro; however, in its traditional 6-well format, it is a time-consuming assay, particularly when evaluating combination therapies. In the interest of increased efficiency, the 6-well CFA was converted to a 96-well format using an automated colony counting algorithm. The 96-well CFA was validated using ionizing radiation therapy on the FaDu (human hypopharyngeal squamous cell) and A549 (human lung) cancer cell lines. Its ability to evaluate combination therapies was investigated by the generation of dose-response curves for the combination of cisplatin and radiation therapy on FaDu and A549 cells. The 96-well CFA was then transferred to a robotic platform for evaluating its potential as a high-throughput screening (HTS) readout. The LOPAC1280 library was screened against FaDu cells, and eight putative hits were identified. Using the 96-well CFA to validate the eight putative chemicals, six of the eight were confirmed, resulting in a positive hit rate of 75%. These data indicate that the 96-well CFA can be adopted as an efficient alternative assay to the 6-well CFA in evaluating single and combination therapies in vitro, providing a possible readout that could be used on a HTS platform.


Asunto(s)
Ensayo de Tumor de Célula Madre/instrumentación , Ensayo de Tumor de Célula Madre/métodos , Línea Celular Tumoral , Cisplatino/farmacología , Terapia Combinada , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas/análisis , Ensayo de Tumor de Célula Madre/normas
9.
Mol Cancer Ther ; 5(9): 2234-40, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16985057

RESUMEN

Despite advances in surgery, radiation, and chemotherapy, novel therapeutics are needed for head and neck cancer treatment. The objective of this current study was to evaluate alexidine dihydrochloride as a novel compound lead for head and neck cancers. Using a tetrazolium-based assay, the dose required to reduce cell viability by 50% (ED50) was found to be approximately 1.8 micromol/L in FaDu (human hypopharyngeal squamous cancer) and approximately 2.6 micromol/L in C666-1 (human undifferentiated nasopharyngeal cancer) cells. In contrast, the ED50 values were much higher in untransformed cells, specifically at approximately 8.8 micromol/L in GM05757 (primary normal human fibroblast), approximately 8.9 micromol/L in HNEpC (primary normal human nasal epithelial), and approximately 19.6 micromol/L in NIH/3T3 (mouse embryonic fibroblast) cells. Alexidine dihydrochloride did not interfere with the activities of cisplatin, 5-fluorouracil, or radiation, and interacted in a less-than-additive manner. DNA content analyses and Hoechst 33342 staining revealed that this compound induced apoptosis. Alexidine dihydrochloride-induced mitochondrial damage was visualized using transmission electron microscopy. Mitochondrial membrane potential (DeltaPsiM) depolarization was detectable after only 3 hours of treatment, and was followed by cytosolic Ca2+ increase along with loss of membrane integrity/cell death. Caspase-2 and caspase-9 activities were detectable at 12 hours, caspase-8 at 24 hours, and caspase-3 at 48 hours. FaDu cell clonogenic survival was reduced to < 5% with 1 micromol/L alexidine dihydrochloride, and, correspondingly, this compound decreased the in vivo tumor-forming potential of FaDu cells. Thus, we have identified alexidine dihydrochloride as the first bisbiguanide compound with anticancer specificity.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Biguanidas/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias Hipofaríngeas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biguanidas/administración & dosificación , Carcinoma de Células Escamosas/patología , Caspasa 2/metabolismo , Caspasa 9/metabolismo , Cisplatino/administración & dosificación , Activación Enzimática/efectos de los fármacos , Femenino , Fluorouracilo/administración & dosificación , Humanos , Neoplasias Hipofaríngeas/patología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/fisiología , Células 3T3 NIH , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Clin Oncol ; 33(29): 3346-55, 2015 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-26351340

RESUMEN

Nasopharyngeal carcinoma (NPC) is a unique epithelial malignancy arising from the superior aspect of the pharyngeal mucosal space, associated with latent Epstein-Barr virus infection in most cases. The capacity to characterize cancer genomes in unprecedented detail is now providing insights into the genesis and molecular underpinnings of this disease. Herein, we provide an overview of the molecular aberrations that likely drive nasopharyngeal tumor development and progression. The contributions of major Epstein-Barr virus-encoded factors, including proteins, small RNAs, and microRNAs, along with their interactions with pathways regulating cell proliferation and survival are highlighted. We review recent analyses that clearly define the role of genetic and epigenetic variations affecting the human genome in NPC. These findings point to the impact of DNA methylation and histone modifications on gene expression programs that promote this malignancy. The molecular interactions that allow NPC cells to evade immune recognition and elimination, which is crucial for the survival of cells expressing potentially immunogenic viral proteins, are also described. Finally, the potential utility of detecting host and viral factors for the diagnosis and prognosis of NPC is discussed. Altogether, the studies summarized herein have greatly expanded our knowledge of the molecular biology of NPC, yet much remains to be uncovered. Emerging techniques for using and analyzing well-annotated biospecimens from patients with NPC will ultimately lead to a greater level of understanding, and enable improvements in precision therapies and clinical outcomes.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virología , Proliferación Celular , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , MicroARNs/genética , ARN Viral/genética , Factores de Riesgo
12.
PLoS One ; 9(5): e98557, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24853121

RESUMEN

Epidermal growth factor receptor (EGFR) is over-expressed in nearly all cases of squamous cell carcinoma of the head and neck (SCCHN), and is an important driver of disease progression. EGFR targeted therapies have demonstrated clinical benefit for SCCHN treatment. In this report, we investigated the pre-clinical efficacy of Dacomitinib (PF-00299804), an irreversible pan-ErbB inhibitor, both alone and in combination with ionizing radiation (IR), a primary curative modality for SCCHN. One normal oral epithelial (NOE) and three SCCHN (FaDu, UT-SCC-8, UT-SCC-42a) cell lines were used to conduct cell viability, clonogenic survival, cell cycle, and immunoblotting assays in vitro, using increasing doses of Dacomitinib (10-500 nM), both with and without IR (2-4 Gy). The FaDu xenograft model was utilized for tumor growth delay assays in vivo, and immunohistochemical analyses were conducted on extracted tumors. A dose-dependent reduction in cell viability and clonogenic survival after Dacomitinib treatment was observed in all three SCCHN models. Treatment led to a significant reduction in EGFR signalling, with a subsequent decrease in phosphorylation of downstream targets such as ERK, AKT, and mTOR. In vivo, Dacomitinib treatment delayed tumor growth, while decreasing phospho-EGFR and Ki-67 immunoexpression. These effects were further enhanced when combined with IR, both in vitro and in vivo. The preclinical data support the further evaluations of Dacomitinib combined with IR for the future management of patients with SCCHN.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Quinazolinonas/uso terapéutico , Animales , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Evaluación Preclínica de Medicamentos , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación , Quinazolinonas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello
13.
PLoS One ; 8(8): e74075, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23991213

RESUMEN

INTRODUCTION: Despite improvements in treatment strategies for head and neck squamous cell carcinoma (HNSCC), outcomes have not significantly improved; highlighting the importance of identifying novel therapeutic approaches to target this disease. To address this challenge, we proceeded to evaluate the role of iron in HNSCC. EXPERIMENTAL DESIGN: Expression levels of iron-related genes were evaluated in HNSCC cell lines using quantitative RT-PCR. Cellular phenotypic effects were assessed using viability (MTS), clonogenic survival, BrdU, and tumor formation assays. The prognostic significance of iron-related proteins was determined using immunohistochemistry. RESULTS: In a panel of HNSCC cell lines, hemochromatosis (HFE) was one of the most overexpressed genes involved in iron regulation. In vitro knockdown of HFE in HNSCC cell lines significantly decreased hepcidin (HAMP) expression and intracellular iron level. This in turn, resulted in a significant decrease in HNSCC cell viability, clonogenicity, DNA synthesis, and Wnt signalling. These cellular changes were reversed by re-introducing iron back into HNSCC cells after HFE knockdown, indicating that iron was mediating this phenotype. Concordantly, treating HNSCC cells with an iron chelator, ciclopirox olamine (CPX), significantly reduced viability and clonogenic survival. Finally, patients with high HFE expression experienced a reduced survival compared to patients with low HFE expression. CONCLUSIONS: Our data identify HFE as potentially novel prognostic marker in HNSCC that promotes tumour progression via HAMP and elevated intracellular iron levels, leading to increased cellular proliferation and tumour formation. Hence, these findings suggest that iron chelators might have a therapeutic role in HNSCC management.


Asunto(s)
Progresión de la Enfermedad , Neoplasias de Cabeza y Cuello/patología , Hemocromatosis/patología , Hierro/metabolismo , Regulación hacia Arriba , Neoplasias de Cabeza y Cuello/metabolismo , Hemocromatosis/metabolismo , Humanos
14.
Int J Radiat Oncol Biol Phys ; 83(5): e563-70, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22520480

RESUMEN

PURPOSE: To describe and assess an interdisciplinary research training program for graduate students, postdoctoral fellows, and clinical fellows focused on radiation medicine; funded by the Canadian Institutes for Health Research since 2003, the program entitled "Excellence in Radiation Research for the 21st Century" (EIRR21) aims to train the next generation of interdisciplinary radiation medicine researchers. METHODS AND MATERIALS: Online surveys evaluating EIRR21 were sent to trainees (n=56), mentors (n=36), and seminar speakers (n=72). Face-to-face interviews were also conducted for trainee liaisons (n=4) and participants in the international exchange program (n=2). RESULTS: Overall response rates ranged from 53% (mentors) to 91% (trainees). EIRR21 was well received by trainees, with the acquisition of several important skills related to their research endeavors. An innovative seminar series, entitled Brainstorm sessions, imparting "extracurricular" knowledge in intellectual property protection, commercialization strategies, and effective communication, was considered to be the most valuable component of the program. Networking with researchers in other disciplines was also facilitated owing to program participation. CONCLUSIONS: EIRR21 is an innovative training program that positively impacts the biomedical community and imparts valuable skill sets to foster success for the future generation of radiation medicine researchers.


Asunto(s)
Oncología por Radiación/educación , Investigación/educación , Comercio/educación , Comunicación , Organización de la Financiación , Humanos , Propiedad Intelectual , Comunicación Interdisciplinaria , Mentores , Ontario , Evaluación de Programas y Proyectos de Salud , Oncología por Radiación/normas , Investigación/normas , Escritura
15.
Cancer Res ; 71(6): 2381-91, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21385904

RESUMEN

Nasopharayngeal carcinoma (NPC) is an Epstein-Barr virus-associated malignancy most common in East Asia and Africa. Here we report frequent downregulation of the microRNA miR-218 in primary NPC tissues and cell lines where it plays a critical role in NPC progression. Suppression of miR-218 was associated with epigenetic silencing of SLIT2 and SLIT3, ligands of ROBO receptors that have been previously implicated in tumor angiogenesis. Exogenous expression of miR-218 caused significant toxicity in NPC cells in vitro and delayed tumor growth in vivo. We used an integrated trimodality approach to identify targets of miR-218 in NPC, cervical, and breast cell lines. Direct interaction between miR-218 and the 3'-untranslated regions (UTR) of mRNAs encoding ROBO1, survivin (BIRC5), and connexin43 (GJA1) was validated in a luciferase-based transcription reporter assay. Mechanistic investigations revealed a negative feedback loop wherein miR-218 regulates NPC cell migration via the SLIT-ROBO pathway. Pleotropic effects of miR-218 on NPC survival and migration were rescued by enforced expression of miR-218-resistant, engineered isoforms of survivin and ROBO1, respectively. In clinical specimens of NPC (n=71), ROBO1 overexpression was significantly associated with worse overall (P=0.04, HR=2.4) and nodal relapse-free survival (P=0.008, HR=6.0). Our findings define an integrative tumor suppressor function for miR-218 in NPC and further suggest that restoring miR-218 expression in NPC might be useful for its clinical management.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/genética , Péptidos y Proteínas de Señalización Intercelular/genética , MicroARNs/genética , Neoplasias Nasofaríngeas/genética , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/genética , Regiones no Traducidas 3'/genética , Adulto , Anciano , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Línea Celular , Línea Celular Tumoral , Conexina 43/genética , Conexina 43/metabolismo , Metilación de ADN/efectos de los fármacos , Decitabina , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Survivin , Proteínas Roundabout
16.
Sci Transl Med ; 3(67): 67ra7, 2011 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-21270338

RESUMEN

Head and neck cancer (HNC) is the eighth most common malignancy worldwide, comprising a diverse group of cancers affecting the head and neck region. Despite advances in therapeutic options over the last few decades, treatment toxicities and overall clinical outcomes have remained disappointing, thereby underscoring a need to develop novel therapeutic approaches in HNC treatment. Uroporphyrinogen decarboxylase (UROD), a key regulator of heme biosynthesis, was identified from an RNA interference-based high-throughput screen as a tumor-selective radiosensitizing target for HNC. UROD knockdown plus radiation induced caspase-mediated apoptosis and cell cycle arrest in HNC cells in vitro and suppressed the in vivo tumor-forming capacity of HNC cells, as well as delayed the growth of established tumor xenografts in mice. This radiosensitization appeared to be mediated by alterations in iron homeostasis and increased production of reactive oxygen species, resulting in enhanced tumor oxidative stress. Moreover, UROD was significantly overexpressed in HNC patient biopsies. Lower preradiation UROD mRNA expression correlated with improved disease-free survival, suggesting that UROD could potentially be used to predict radiation response. UROD down-regulation also radiosensitized several different models of human cancer, as well as sensitized tumors to chemotherapeutic agents, including 5-fluorouracil, cisplatin, and paclitaxel. Thus, our study has revealed UROD as a potent tumor-selective sensitizer for both radiation and chemotherapy, with potential relevance to many human malignancies.


Asunto(s)
Técnicas de Silenciamiento del Gen , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/radioterapia , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Uroporfirinógeno Descarboxilasa/genética , Uroporfirinógeno Descarboxilasa/metabolismo , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Homeostasis , Humanos , Hierro/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Trasplante de Neoplasias , Estrés Oxidativo , Interferencia de ARN
17.
Eur J Cancer ; 46(11): 1967-78, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20451372

RESUMEN

Nasopharyngeal carcinoma (NPC) differs from other head and neck cancers in its aetiology, epidemiology and potential therapeutic options. Despite cure for the majority of the patients, challenges still exist in the prevention of disease relapse, treatment of patients with refractory or metastatic NPC and the management of long-term toxicities. This article discusses the specific challenges in pushing the boundaries of NPC treatments further, with an emphasis on prognostic/predictive markers, molecularly targeted therapies, immunotherapies and the areas of interest with regard to long-term toxicities arising from therapeutic interventions.


Asunto(s)
Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/radioterapia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores , Humanos , Inmunoterapia/métodos , Pronóstico
18.
Int J Radiat Oncol Biol Phys ; 77(1): 253-60, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20394857

RESUMEN

PURPOSE: To investigate the efficacy of targeting polo-like kinase 1 (Plk1) combined with ionizing radiotherapy (RT) for head-and-neck squamous cell carcinoma (HNSCC). METHODS AND MATERIALS: Polo-like kinase 1 messenger ribonucleic acid (mRNA) was targeted by small interfering RNA (siRNA) transfection into the FaDu HNSCC cell line; reduction was confirmed using quantitative real-time polymerase chain reaction. The cellular effects were assessed using [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium], clonogenic, flow cytometric, and caspase assays. In vivo efficacy of siPlk1 was evaluated using mouse xenograft models. RESULTS: Small interfering Plk1 significantly decreased Plk1 mRNA expression, while also increasing cyclin B1 and p21(Waf1/CIP1) mRNA levels after 24 h. This depletion resulted in a time-dependent increase in FaDu cytotoxicity, which was enhanced by the addition of RT. Flow cytometric and caspase assays demonstrated progressive apoptosis, DNA double-strand breaks (gamma-H2AX), G2/M arrest, and activation of caspases 3 and 7. Implantation of siPlk1-treated FaDu cells in severe combined immunodeficient mice delayed tumor formation, and systemic administration of siPlk1 inhibited tumor growth enhanced by RT. CONCLUSIONS: These data demonstrate the suitability of Plk1 as a potential therapeutic target for HNSCC, because Plk1 depletion resulted in significant cytotoxicity in vitro and abrogated tumor-forming potential in vivo. The effects of Plk1 depletion were enhanced with the addition of RT, indicating that Plk1 represents an important potential radiation sensitizer for HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/radioterapia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Mensajero/antagonistas & inhibidores , ARN Interferente Pequeño/uso terapéutico , Animales , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Ciclina B1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Histonas/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/metabolismo , Sales de Tetrazolio , Tiazoles , Transfección , Ensayo de Tumor de Célula Madre/métodos , Quinasa Tipo Polo 1
19.
Clin Cancer Res ; 16(3): 898-911, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20103674

RESUMEN

PURPOSE: Rapidly metabolizing tumor cells have elevated levels of nicotinamide phosphoribosyltransferase, an enzyme involved in NAD(+) biosynthesis, which serves as an important substrate for proteins involved in DNA repair. GMX1777, which inhibits nicotinamide phosphoribosyltransferase, was evaluated in two human head and neck cancer models in combination with radiotherapy. EXPERIMENTAL DESIGN: Effects of GMX1777-mediated radiosensitization were examined via metabolic and cytotoxicity assays in vitro; mechanism of action, in vivo antitumor efficacy, and radiosensitization were also investigated. RESULTS: IC(50) values of GMX1777 for FaDu and C666-1 cells were 10 and 5 nmol/L, respectively, which interacted synergistically with radiotherapy. GMX1777 induced a rapid decline in intracellular NAD(+) followed by ATP reduction associated with significant cytotoxicity. These metabolic changes were slightly increased with the addition of radiotherapy, although poly(ADP-ribose) polymerase activity was significantly reduced when GMX1777 was combined with radiotherapy, thereby accounting for the synergistic cytotoxicity of these two modalities. Systemic GMX1777 administration with local tumor radiotherapy caused complete disappearance of FaDu and C666-1 tumors for 50 and 20 days, respectively. There was also significant reduction in tumor vascularity, particularly for the more sensitive FaDu model. [(18)F]FDG-positron emission tomography/computed tomography images showed reduction in [(18)F]FDG uptake after GMX1777 administration, showing decreased glucose metabolism in vivo. CONCLUSIONS: Our data represent the first report showing that GMX1777 plus radiotherapy is an effective therapeutic strategy for head and neck cancer, mediated via pleiotropic effects of inhibition of DNA repair and tumor angiogenesis, while sparing normal tissues. Therefore, GMX1777 combined with radiotherapy definitely warrants clinical evaluation in human head and neck cancer patients.


Asunto(s)
Guanidinas/administración & dosificación , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Animales , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Terapia Combinada , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Guanidinas/farmacología , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Humanos , Ratones , Ratones SCID , NAD/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Radiat Oncol Biol Phys ; 73(4): 988-96, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19251086

RESUMEN

Radiation therapy is a highly effective cancer treatment modality, and extensive investigations have been undertaken over the years to augment its efficacy in the clinic. This review summarizes the current understanding of the biologic bases underpinning many of the clinically used radiosensitizers. In addition, this review illustrates how the advent of innovative, high-throughput technologies with integration of different disciplines could be harnessed for an expeditious discovery process for novel radiosensitizers, providing an exciting future for such pursuits in radiation biology and oncology.


Asunto(s)
Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Hipoxia de la Célula/fisiología , Terapia Combinada/métodos , Daño del ADN/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Perfilación de la Expresión Génica/métodos , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Neoplasias/tratamiento farmacológico , Tolerancia a Radiación/fisiología , Fármacos Sensibilizantes a Radiaciones/clasificación , Fármacos Sensibilizantes a Radiaciones/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA