Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Biol Toxicol ; 37(3): 461-478, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32979173

RESUMO

High-risk neuroblastoma (HR-NB) is branded with hematogenous metastasis, relapses, and dismal long-term survival. Intensification of consolidation therapy with tandem/triple autologous stem cell (SC) rescue (with bone marrow [BM]/peripheral blood [PB] CD34+ selection) after myeloablative chemotherapy has improved long-term survival. However, the benefit is limited by the indication of NB cells in CD34+ PBSCs, CD34 expression in NB cells, and the risk of reinfusing NB cancer stem cells (NB CSCs) that could lead to post-transplant relapse. We investigated the association of CD34 surface expression (92 patients) with NB evolution/clinical outcomes. CD34 gene-level status in NB was assessed through RNA-Seq data mining (18 cohorts, n, 3324). Genetic landscape of CD34-expressing NB CSCs (CD133+CD34+) was compared with CD34- CSCs (CD133+CD34-). RNA-seq data revealed equivocal association patterns of CD34 expression with patient survival. Our immunohistochemistry data revealed definite, but rare (mean, 0.73%; range 0.00-7.87%; median, 0.20%) CD34 positivity in NB. CD34+ significantly associated with MYCN amplification (p, 0.003), advanced disease stage (p, 0.016), and progressive disease (PD, p < 0.0009) after clinical therapy. A general high-is-worse tendency was observed in patients with relapsed disease. High CD34+ correlated with poor survival in patients with N-MYC-amplified HR-NB. Gene expression analysis of CD34+-NB CSCs identified significant up (4631) and downmodulation (4678) of genes compared with NB CSCs that lack CD34. IPA recognized the modulation of crucial signaling elements (EMT, stemness maintenance, differentiation, inflammation, clonal expansion, drug resistance, metastasis) that orchestrate NB disease evolution in CD34+ CSCs compared with CD34- CSCs. While the function of CD34 in NB evolution requires further in-depth investigation, careful consideration should be exercised for autologous stem cell rescue with CD34+ selection in NB patients. Graphical abstract.


Assuntos
Antígeno AC133/genética , Antígenos CD34/genética , Antígenos de Superfície/genética , Proteína Proto-Oncogênica N-Myc/genética , Neuroblastoma/genética , Pré-Escolar , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Lactente , Masculino , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neuroblastoma/epidemiologia , Neuroblastoma/patologia , Pediatria , Prognóstico , RNA-Seq
2.
BMC Cancer ; 19(1): 106, 2019 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-30691436

RESUMO

BACKGROUND: MYCN amplification directly correlates with the clinical course of neuroblastoma and poor patient survival, and serves as the most critical negative prognostic marker. Although fluorescence in situ hybridization (FISH) remains the gold standard for clinical diagnosis of MYCN status in neuroblastoma, its limitations warrant the identification of rapid, reliable, less technically challenging, and inexpensive alternate approaches. METHODS: In the present study, we examined the concordance of droplet digital PCR (ddPCR, in combination with immunohistochemistry, IHC) with FISH for MYCN detection in a panel of formalin-fixed paraffin-embedded (FFPE) human neuroblastoma samples. RESULTS: In 112 neuroblastoma cases, ddPCR analysis demonstrated a 96-100% concordance with FISH. Consistently, IHC grading revealed 92-100% concordance with FISH. Comparing ddPCR with IHC, we observed a concordance of 95-98%. CONCLUSIONS: The results demonstrate that MYCN amplification status in NB cases can be assessed with ddPCR, and suggest that ddPCR could be a technically less challenging method of detecting MYCN status in FFPE specimens. More importantly, these findings illustrate the concordance between FISH and ddPCR in the detection of MYCN status. Together, the results suggest that rapid, less technically demanding, and inexpensive ddPCR in conjunction with IHC could serve as an alternate approach to detect MYCN status in NB cases, with near-identical sensitivity to that of FISH.


Assuntos
Proteína Proto-Oncogênica N-Myc/genética , Neuroblastoma/diagnóstico , Reação em Cadeia da Polimerase , Biomarcadores Tumorais/genética , Formaldeído , Amplificação de Genes , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Neuroblastoma/genética , Neuroblastoma/patologia , Inclusão em Parafina , Sensibilidade e Especificidade
3.
Mol Cell Biochem ; 460(1-2): 175-193, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31367889

RESUMO

The upsurge of marine-derived therapeutics for cancer treatment is evident, with many drugs in clinical use and in clinical trials. Seaweeds harbor large amounts of polyphenols and their anti-cancer benefit is linear to their anti-oxidant activity. Our studies identified three superlative anti-cancer seaweed polyphenol drug candidates (SW-PD). We investigated the acquisition of oncogenic burden in radiation-resilient pancreatic cancer (PC) that could drive tumor relapse, and elucidated the efficacy of SW-PD candidates as adjuvants in genetically diverse in vitro systems and a mouse model of radiation-residual disease. QPCR profiling of 88 oncogenes in therapy-resilient PC cells identified a 'shared' activation of 40 oncogenes. SW-PD pretreatment inflicted a significant mitigation of acquired (shared) oncogenic burden, in addition to drug- and cell-line-specific repression signatures. Tissue microarray with IHC of radiation-residual tumors in mice signified acquired cellular localization of key oncoproteins and other critical architects. Conversely, SW-PD treatment inhibited the acquisition of these critical drivers of tumor genesis, dissemination, and evolution. Heightened death of resilient PC cells with SW-PD treatment validated the translation aspects. The results defined the acquisition of oncogenic burden in resilient PC and demonstrated that the marine polyphenols effectively target the acquired oncogenic burden and could serve as adjuvant(s) for PC treatment.


Assuntos
Organismos Aquáticos/química , Carcinogênese/patologia , Neoplasias Pancreáticas/patologia , Polifenóis/farmacologia , Acetatos/química , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Progressão da Doença , Humanos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/radioterapia , Polifenóis/uso terapêutico , Alga Marinha/química
4.
BMC Genomics ; 16: 501, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26148557

RESUMO

BACKGROUND: MetastamiRs have momentous clinical relevance and have been correlated with disease progression in many tumors. In this study, we identified neuroblastoma metastamiRs exploiting unique mouse models of favorable and high-risk metastatic human neuroblastoma. Further, we related their deregulation to the modulation of target proteins and established their association with clinical outcomes. RESULTS: Whole genome miRNA microarray analysis identified 74 metastamiRs across the manifold of metastatic tumors. RT-qPCR on select miRNAs validated profile expression. Results from bio-informatics across the ingenuity pathway, miRCancer, and literature data-mining endorsed the expression of these miRNAs in multiple tumor systems and showed their role in metastasis, identifying them as metastamiRs. Immunoblotting and TMA-IHC analyses revealed alterations in the expression/phosphorylation of metastamiRs' targets, including ADAMTS-1, AKT1/2/3, ASK1, AURKß, Birc1, Birc2, Bric5, ß-CATENIN, CASP8, CD54, CDK4, CREB, CTGF, CXCR4, CYCLIN-D1, EGFR, ELK1, ESR1, CFOS, FOSB, FRA, GRB10, GSK3ß, IL1α, JUND, kRAS, KRTAP1, MCP1, MEGF10, MMP2, MMP3, MMP9, MMP10, MTA2, MYB, cMYC, NF2, NOS3, P21, pP38, PTPN3, CLEAVED PARP, PKC, SDF-1ß, SEMA3D, SELE, STAT3, TLR3, TNFα, TNFR1, and VEGF in aggressive cells ex vivo and in a manifold of metastatic tumors in vivo. miRNA mimic (hsa-miR-125b, hsa-miR-27b, hsa-miR-93, hsa-miR-20a) and inhibitor (hsa-miR-1224-3p, hsa-miR-1260) approach for select miRNAs revealed the direct influence of the altered metastamiRs in the regulation of identified protein targets. Clinical outcome association analysis with the validated metastamiRs' targets corresponded strongly with poor overall and relapse-free survival. CONCLUSIONS: For the first time, these results identified a comprehensive list of neuroblastoma metastamiRs, related their deregulation to altered expression of protein targets, and established their association with poor clinical outcomes. The identified set of distinctive neuroblastoma metastamiRs could serve as potential candidates for diagnostic markers for the switch from favorable to high-risk metastatic disease.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , Metástase Neoplásica/genética , Neuroblastoma/genética , Animais , Linhagem Celular Tumoral , Biologia Computacional , Progressão da Doença , Intervalo Livre de Doença , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos , Camundongos Nus , Análise em Microsséries/métodos , Recidiva Local de Neoplasia/genética , Neuroblastoma/patologia , Prognóstico
5.
BMC Cancer ; 15: 514, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26159519

RESUMO

BACKGROUND: Determining the driving factors and molecular flow-through that define the switch from favorable to aggressive high-risk disease is critical to the betterment of neuroblastoma cure. METHODS: In this study, we examined the cytogenetic and tumorigenic physiognomies of distinct population of metastatic site- derived aggressive cells (MSDACs) from high-risk tumors, and showed the influence of acquired genetic rearrangements on poor patient outcomes. RESULTS: Karyotyping in SH-SY5Y and MSDACs revealed trisomy of 1q, with additional non-random chromosomal rearrangements on 1q32, 8p23, 9q34, 15q24, 22q13 (additions), and 7q32 (deletion). Array CGH analysis of individual clones of MSDACs revealed genetic alterations in chromosomes 1, 7, 8, and 22, corresponding to a gain in the copy numbers of LOC100288142, CD1C, CFHR3, FOXP2, MDFIC, RALYL, CSMD3, SAMD12-AS1, and MAL2, and a loss in ADAM5, LOC400927, APOBEC3B, RPL3, MGAT3, SLC25A17, EP300, L3MBTL2, SERHL, POLDIP3, A4GALT, and TTLL1. QPCR analysis and immunoblotting showed a definite association between DNA-copy number changes and matching transcriptional/translational expression in clones of MSDACs. Further, MSDACs exert a stem-like phenotype. Under serum-free conditions, MSDACs demonstrated profound tumorosphere formation ex vivo. Moreover, MSDACs exhibited high tumorigenic capacity in vivo and prompted aggressive metastatic disease. Tissue microarray analysis coupled with automated IHC revealed significant association of RALYL to the tumor grade in a cohort of 25 neuroblastoma patients. Clinical outcome association analysis showed a strong correlation between the expression of CFHR3, CSMD3, MDFIC, FOXP2, RALYL, POLDIP3, SLC25A17, SERHL, MGAT3, TTLL1, or LOC400927 and overall and relapse-free survival in patients with neuroblastoma. CONCLUSION: Together, these data highlight the ongoing acquired genetic rearrangements in undifferentiated tumor-forming neural crest cells, and suggest that these alterations could switch favorable neuroblastoma to high-risk aggressive disease, promoting poor clinical outcomes.


Assuntos
Estudos de Associação Genética , Neuroblastoma/genética , Neuroblastoma/mortalidade , Animais , Linhagem Celular Tumoral , Aberrações Cromossômicas , Bandeamento Cromossômico , Hibridização Genômica Comparativa , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Progressão da Doença , Xenoenxertos , Humanos , Metástase Neoplásica , Estadiamento de Neoplasias , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neuroblastoma/patologia , Prognóstico , Proteína Ribossômica L3
6.
J Biol Chem ; 286(24): 21588-600, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21527635

RESUMO

Induced radioresistance in the surviving cancer cells after radiotherapy could be associated with clonal selection leading to tumor regrowth at the treatment site. Previously we reported that post-translational modification of IκBα activates NFκB in response to ionizing radiation (IR) and plays a key role in regulating apoptotic signaling. Herein, we investigated the orchestration of NFκB after IR in human neuroblastoma. Both in vitro (SH-SY5Y, SK-N-MC, and IMR-32) and in vivo (xenograft) studies showed that IR persistently induced NFκB DNA binding activity and NFκB-dependent TNFα transactivation and secretion. Approaches including silencing NFκB transcription, blocking post-translational NFκB nuclear import, muting TNF receptor, overexpression, and physiological induction of either NFκB or TNFα precisely demonstrated the initiation and occurrence of NFκB → TNFα → NFκB positive feedback cycle after IR that leads to and sustains NFκB activation. Selective TNF-dependent NFκB regulation was confirmed with futile inhibition of AP-1 and SP-1 in TNF receptor muted cells. Moreover, IR increased both transactivation and translation of Birc1, Birc2, and Birc5 and induced metabolic activity and clonal expansion. This pathway was further defined to show that IR-induced functional p65 transcription (not NFκB1, NFκB2, or c-Rel) is necessary for activation of these survival molecules and associated survival advantage. Together, these results demonstrate for the first time the functional orchestration of NFκB in response to IR and further imply that p65-dependent survival advantage and initiation of clonal expansion may correlate with an unfavorable prognosis of human neuroblastoma.


Assuntos
Regulação Neoplásica da Expressão Gênica , NF-kappa B/metabolismo , Neuroblastoma/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Prognóstico , Radiação Ionizante , Fatores de Tempo
7.
Med Dosim ; 47(3): 273-279, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35644778

RESUMO

The aim of our study is to document our cases of choroidal melanoma treated with low dose rate (LDR) brachytherapy and to correlate the dosimetry and radiobiology with clinical effects and oncologic outcomes. Data from 157 patients treated from 2014 to 2018 with LDR brachytherapy were used for this investigation. Treatments used a collaborative ocular melanoma study eye plaque and Iodine-125 radioactive seeds. The seeds activities were chosen to deliver 85 Gy to the tumor apex or to a prescription point (if the apex < 5 mm). The plaque sizes used were 10, 12, 14, 16, 18, 20, and 22 mm including notched or deep notched. The plaques were modeled in Varian BrachyVision version 11.6 (Varian Medical Systems) with seed coordinates from the AAPM Task Group 129. The Task Group 43 from AAPM was used for brachytherapy dose planning. Dose data were extracted for the apex, prescription point, sclera, retina opposite to the implant, lens, macula, and optic disc. The radiobiological dosimetry were calculated using appropriate α/ß ratios found in the literature and then correlated to clinical side effects. Average biologically effective dose for associated organs at risk were calculated in cases where toxicity occurred. These included: radiation cataract (70.66 Gy), disc atrophy (475.49 Gy), foveal atrophy (263.07 Gy), radiation papillopathy (373.45 Gy), radiation maculopathy (213.62 Gy), vitreous hemorrhage (1437.68 Gy), vascular occlusion (1080.93 Gy), nonproliferative retinopathy (1066.89 Gy), proliferative retinopathy (1590.71 Gy), exudative retinal detachment (1364.32 Gy), and rhegmatogenous retinal detachment (2265.54 Gy). Average biologically effective dose was higher in patients who developed radiation induced long term side effects than in the whole patient population except for radiation maculopathy. In spite of the small patient population and short-term follow-up, it is of interest to correlate the radiation induced effects and create a guideline for the improvement of the treatment of patients treated with LDR brachytherapy.


Assuntos
Braquiterapia , Neoplasias Oculares , Degeneração Macular , Melanoma , Lesões por Radiação , Descolamento Retiniano , Doenças Retinianas , Atrofia/etiologia , Braquiterapia/efeitos adversos , Neoplasias Oculares/radioterapia , Humanos , Radioisótopos do Iodo , Degeneração Macular/etiologia , Melanoma/radioterapia , Lesões por Radiação/etiologia , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Descolamento Retiniano/etiologia , Doenças Retinianas/etiologia , Estudos Retrospectivos
8.
Am J Physiol Heart Circ Physiol ; 300(3): H736-44, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21186274

RESUMO

Whole brain radiation therapy (WBRT) leads to cognitive impairment in 40-50% of brain tumor survivors following treatment. Although the etiology of cognitive deficits post-WBRT remains unclear, vascular rarefaction appears to be an important component of these impairments. In this study, we assessed the effects of WBRT on the cerebrovasculature and the effects of systemic hypoxia as a potential mechanism to reverse the microvascular rarefaction. Transgenic mice expressing green fluorescent protein driven by the Acta2 (smooth muscle actin) promoter for blood vessel visualization were randomly assigned to control or radiated groups. Animals received a clinical series of 4.5 Gy WBRT two times weekly for 4 wk followed by 1 mo of recovery. Subsequently, mice were subjected to 11% (hypoxia) or 21% (normoxia) oxygen for 1 mo. Capillary density in subregions of the hippocampus revealed profound vascular rarefaction that persisted despite local tissue hypoxia. Nevertheless, systemic hypoxia was capable of completely restoring cerebrovascular density. Thus hippocampal microvascular rarefaction post-WBRT is not capable of stimulating angiogenesis and can be reversed by chronic systemic hypoxia. Our results indicate a potential shift in sensitivity to angiogenic stimuli and/or the existence of an independent pathway of regulating cerebral microvasculature.


Assuntos
Transtornos Cognitivos/fisiopatologia , Transtornos Cognitivos/terapia , Cognição/efeitos da radiação , Hipocampo/irrigação sanguínea , Hipocampo/efeitos da radiação , Oxigênio/fisiologia , Actinas/genética , Actinas/metabolismo , Anaerobiose , Animais , Cognição/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Microvasos/fisiopatologia , Microvasos/efeitos da radiação , Regiões Promotoras Genéticas
9.
Mutat Res ; 718(1-2): 44-55, 2011 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-21056117

RESUMO

Radiation-induced amplification of reactive oxygen species (ROS) may be a sensing mechanism for activation of signaling cascades that influence cell fate. However, the regulated intrinsic mechanisms and targets of low-dose ionizing radiation (LDIR) are still unclear. Accordingly, we investigated the effects of LDIR on NFκB signal transduction and manganese superoxide dismutase (SOD2) activity in mice brain and gut. LDIR resulted in both dose-dependent and persistent NFκB activation in gut and brain. QPCR displayed a dose- and tissue-dependent differential modulation of 88 signaling molecules. With stringent criteria, a total of 15 (2cGy), 43 (10cGy) and 19 (50cGy) genes were found to be commonly upregulated between brain and gut. SOD2 immunostaining showed a LDIR-dose dependent increase. Consistent with the NFκB results, we observed a persistent increase in SOD2 activity after LDIR. Moreover, muting of LDIR-induced NFκB attenuated SOD2 transactivation and cellular localization. These results imply that exposure of healthy tissues to LDIR results in induced NFκB and SOD2 activity and transcriptional activation of NFκB-signal transduction/target molecules. More importantly, the results suggest that NFκB initiates a feedback response through transcriptional activation of SOD2 that may play a key role in the LDIR-induced oxidative stress response and may control the switch that directs cell fate.


Assuntos
Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Sistema Digestório/efeitos dos fármacos , Sistema Digestório/metabolismo , Raios gama/efeitos adversos , NF-kappa B/metabolismo , Superóxido Dismutase/metabolismo , Animais , Sequência de Bases , Primers do DNA/genética , Relação Dose-Resposta à Radiação , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , NF-kappa B/genética , Estresse Oxidativo/efeitos da radiação , Reação em Cadeia da Polimerase , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Superóxido Dismutase/genética , Ativação Transcricional/efeitos da radiação
10.
Nutr Cancer ; 62(1): 93-104, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20043264

RESUMO

Black raspberry extracts (RSE) have been shown to inhibit cancer cell growth and stimulate apoptosis. Also, studies have demonstrated that RSE inhibits transcriptional regulators including NFkappa B. Accordingly, we investigated the effect of RSE in inhibiting radiation (IR) induced NFkappa B mediated radioprotection in breast adenocarcinoma cells. MCF-7 cells were exposed to IR (2Gy), treated with RSE (0.5, 1.0, 2.0 micro g/ml) or treated with RSE (1.0 micro g/ml) followed by IR exposure, and harvested after 1, 3, 6, 24, 48, and 72 h. NFkappa B DNA-binding activity was measured by EMSA and phosphorylated Ikappa Balpha by immunoblotting. Expression of IAP1, IAP2, XIAP and survivin were measured by QPCR and immunoblotting. Cell survival was measured using MTT assay and cell death using Caspase-3/7 activity. Effect of RSE on IR induced MnSOD, TNFalpha, IL-1alpha and MnSOD activity was also determined. RSE inhibited NFkappa B activity in a dose-dependent manner. Also, RSE inhibited IR-induced sustained activation of NFkappa B, and NFkappa B regulated IAP1, IAP2, XIAP, and survivin. In addition, RSE inhibited IR-induced TNFalpha, IL-1alpha, and MnSOD levels and MnSOD activity. RSE suppressed cell survival and enhanced cell death. These results suggest that RSE may act as a potent radiosensitizer by overcoming the effects of NFkappa B mediated radioprotection in human breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Frutas/química , NF-kappa B/antagonistas & inibidores , Extratos Vegetais/farmacologia , Protetores contra Radiação/farmacologia , Rosaceae/química , Apoptose/efeitos dos fármacos , Proteína 3 com Repetições IAP de Baculovírus , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , DNA/metabolismo , Raios gama , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Apoptose/genética , Interleucina-1alfa/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , NF-kappa B/metabolismo , NF-kappa B/farmacologia , Fenóis/análise , Extratos Vegetais/química , Radiossensibilizantes/farmacologia , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase/metabolismo , Survivina , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Ubiquitina-Proteína Ligases , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
11.
J Appl Clin Med Phys ; 11(1): 3133, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-20160698

RESUMO

The purpose of this study was to assess the impact of respiratory gating on tumor and normal tissue dosimetry in patients treated with SBRT for early stage non-small cell lung cancer (NSCLC). Twenty patients with stage I NSCLC were studied. Treatment planning was performed using four-dimensional computed tomography (4D CT) with free breathing (Plan I), near-end inhalation (Plan II), and near-end exhalation (Plan III). The prescription dose was 60 Gy in three fractions. The tumor displacement was most pronounced for lower peripheral lesions (average 7.0 mm, range 4.1-14.3 mm) when compared to upper peripheral (average 2.4mm, range 1.0-5.1 mm) or central lesions (average 2.9 mm, range 1.0-4.1 mm). In this study, the pencil beam convolution (PBC) algorithm with modified Batho power law for tissue heterogeneity was used for dose calculation. There were no significant differences in tumor and normal tissue dosimetry among the three gated plans. Tumor location however, significantly influenced tumor doses because of the necessity of respecting normal tissue constraints of centrally located structures. For plans I, II and III, average doses to central lesions were lower as compared with peripheral lesions by 4.88 Gy, 8.24 Gy and 6.93 Gy for minimum PTV and 0.98, 1.65 and 0.87 Gy for mean PTV dose, respectively. As a result, the mean single fraction equivalent dose (SFED) values were also lower for central compared to peripheral lesions. In addition, central lesions resulted in higher mean doses for lung, esophagus, and ipsilateral bronchus by 1.24, 1.93 and 7.75 Gy, respectively. These results indicate that the tumor location is the most important determinant of dosimetric optimization of SBRT plans. Respiratory gating proved unhelpful in the planning of these patients with severe COPD.


Assuntos
Neoplasias Pulmonares/radioterapia , Planejamento da Radioterapia Assistida por Computador/normas , Técnicas de Imagem de Sincronização Respiratória/normas , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Feminino , Tomografia Computadorizada Quadridimensional , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Tolerância a Radiação/fisiologia , Tolerância a Radiação/efeitos da radiação , Radiobiologia/normas , Dosagem Radioterapêutica , Mecânica Respiratória , Tecnologia Radiológica/normas
12.
Med Dosim ; 45(1): 21-27, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31101569

RESUMO

This work evaluated the difference in dosimetry of high dose rate (HDR) brachytherapy treatments between plans using advanced multichannel applicators and simplified base versions. Eighteen HDR patients treated using Interstitial Ring CT/MR Applicator Set (Elekta Brachytherapy, Netherlands) (TRN) (21 plans), CapriTM Applicator Set (Varian Medical Systems, Inc., Palo Alto, CA) (CC) (19 plans), Rotte Endometrial Applicator Set (Elekta Brachytherapy, Netherlands) (RDT) (18 plans), and the Advanced Gynecological Applicator Venezia (Vz) (Elekta Brachytherapy, Netherlands) (6 plans) were retrospectively reviewed. For each plan, "advanced" channels including any interstitial channels, the 12 noncentral channels in the CC, and the lateral extending aspects of the RDT were removed and a new plan with the original inverse planning settings was optimized using only the remaining "simplified" applicator and compared to the original. The new plans were renormalized to match the original percent dose to 90% of the high-risk clinical target volume (HR-CTV). Critical structures included bladder, rectum, sigmoid colon, and small bowel. Comparisons were made utilizing dose volume histograms of HR-CTVs, conformation number (CN), and the equivalent total dose in 2 Gy fractions (EQD2) to 2 cm3 of the normal structures. Comparing simplified to advanced plans, the average percent differences in EQD2 to 2 cm3 for Vz, with 95% confidence interval, were 101.7 ± 85.9%, 147.8 ± 76.7%, 95.3 ± 61.6%, and 44.0 ± 12.4% for Rectum, Bladder, Sigmoid, and Bowel, respectively. For TRN: 36.9 ± 18.5%, 38.2 ± 14.5%, 20.3 ± 8.8%, and 15.3 ± 8.2%. For CC: 18.9 ± 3.7%, 12.3 ± 5.3%, 27.8 ± 7.1%, and 17.1 ± 3.6%. For RDT: 1.5 ± 6.8%, 7.4 ± 6.7%, 11.1 ± 4.4%, and 8.0 ± 8.7%. The CN was better in advanced applications by 0.024 for RDT, 0.104 for TRN, 0.043 for CC, and 0.251 for Vz (all p < 0.05). Advanced multichannel treatments allow better target dose conformation and normal tissue dose manipulation. The biggest factors influencing the brachytherapy dose distributions are the number of available channels and their separation from each other within the target.


Assuntos
Braquiterapia/instrumentação , Neoplasias dos Genitais Femininos/radioterapia , Planejamento da Radioterapia Assistida por Computador/métodos , Adulto , Idoso , Braquiterapia/efeitos adversos , Braquiterapia/métodos , Feminino , Humanos , Pessoa de Meia-Idade , Órgãos em Risco , Dosagem Radioterapêutica , Estudos Retrospectivos , Neoplasias do Colo do Útero/radioterapia , Neoplasias Uterinas/radioterapia , Neoplasias Vaginais/radioterapia
13.
J Cell Biochem ; 106(6): 999-1009, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19204936

RESUMO

Ascertaining the upstream regulatory mechanisms of hyperthermia-induced apoptosis is important to understand the role of hyperthermia in combined modality cancer therapy. Accordingly, we investigated whether (i) hyperthermia-induced apoptosis is mediated through the nitric oxide (NO) signaling pathway and (ii) inhibition of post-translational modification of IkappaBalpha and down regulation of NFkappaB-DNA binding activity is an intermediate step in NO-dependent apoptosis in MCF-7 breast cancer cells. For hyperthermia treatment, the cells were exposed to 43 degrees C. Intracellular NO levels measured by the fluorescent intensity of DAF-2A and iNOS expression by immunobloting revealed an increased level of iNOS dependent NO production after 43 degrees C. Apoptosis measured by Annexin V expression and cell survival by clonogenic assay showed a 20% increase in apoptosis after 43 degrees C treatments. EMSA analysis showed a dose-dependent inhibition of NFkappaB-DNA binding activity. The hyperthermia-mediated inhibition of NFkappaB was persistent even after 48 h. Inhibition of NO by L-NAME rescued the NFkappaB-DNA binding activity and inhibits heat-induced apoptosis. Similarly, over-expression of NFkappaB by transient transfection inhibits heat-induced apoptosis. These results demonstrate that apoptosis upon hyperthermia exposure of MCF-7 cells is regulated by NO-mediated suppression of NFkappaB.


Assuntos
Apoptose/fisiologia , Neoplasias da Mama/terapia , Hipertermia Induzida , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Animais , Linhagem Celular Tumoral , Terapia Combinada , DNA/metabolismo , Inibidores Enzimáticos/metabolismo , Feminino , Humanos , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Ligação Proteica
14.
J Radiat Res ; 50(4): 311-24, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19436149

RESUMO

Radiotherapy has been used as an adjunctive local-control modality for high-risk neuroblastoma. However, relapse due to radioresistance affects the success of radiotherapy. Ascertaining the fractionated radiation (FIR) modulated molecular targets is imperative in targeted molecular therapy. Accordingly, we investigated the (i) expression of genes representing six functional pathways; (ii) NFkappaB DNA-binding activity and (iii) expression of radioresponsive molecules after single dose (10 Gy) radiation (SDR) and FIR (2 Gy x 5) in human neuroblastoma cells. Alterations in gene expression were analyzed using QPCR-profiling, NFkappaB activity using electrophoretic mobility shift assay (EMSA) and pIkappaBalpha using immunoblotting. Modulations in TNFalpha, IL-1alpha, pAKT, IAP1, IAP2, XIAP, survivin, MnSOD, BID, Bak, MyD88 and Vegfc were determined using quantitative real-time PCR (Q-PCR) and immunoblotting. Compared to SDR, FIR significantly induced the expression of 25 genes and completely suppressed another 30 genes. Furthermore, FIR induced NFkappaB-DNA-binding activity and IkappaBalpha phosphorylation. Similarly, we observed an induced expression of IAP1, IAP2, XIAP, Survivin, IL-1alpha, MnSOD, Bid, Bak, MyD88, TNFalpha and pAKT in cells exposed to FIR. The results of the study clearly show distinct differences in the molecular response of cells between SDR and FIR. We identified several potential targets confining to NFkappaB signaling cascade that may affect radio-resistance after FIR.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , NF-kappa B/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Transdução de Sinais/efeitos da radiação , Linhagem Celular Tumoral , Fracionamento da Dose de Radiação , Relação Dose-Resposta à Radiação , Humanos , Doses de Radiação
15.
Cancer Drug Resist ; 2: 948-967, 2019 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-31867574

RESUMO

Neuroblastoma (NB) is the most common cancer of infancy and accounts for nearly one tenth of pediatric cancer deaths. This mortality rate has been attributed to the > 50% frequency of relapse despite intensive, multimodal clinical therapy in patients with progressive NB. Given the disease's heterogeneity and developed resistance, attaining a cure after relapse of progressive NB is highly challenging. A rapid decrease in the timeline between successive recurrences is likely due to the ongoing acquisition of genetic rearrangements in undifferentiated NB-cancer stem cells (CSCs). In this review, we present the current understanding of NB-CSCs, their intrinsic role in tumorigenesis, their function in disease progression, and their influence on acquired therapy resistance and tumor evolution. In particular, this review focus on the intrinsic involvement of stem cells and signaling in the genesis of NB, the function of pre-existing CSCs in NB progression and therapy response, the formation and influence of induced CSCs (iCSCs) in drug resistance and tumor evolution, and the development of a CSC-targeted therapeutic approach.

16.
Cancer Drug Resist ; 2: 1086-1105, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31867575

RESUMO

Neuroblastoma (NB) deriving from neural crest cells is the most common extra-cranial solid cancer at infancy. NB originates within the peripheral sympathetic ganglia in adrenal medulla and along the midline of the body. Clinically, NB exhibits significant heterogeneity stretching from spontaneous regression to rapid progression to therapy resistance. MicroRNAs (miRNAs, miRs) are small (19-22 nt in length) non-coding RNAs that regulate human gene expression at the post-transcriptional level and are known to regulate cellular signaling, growth, differentiation, death, stemness, and maintenance. Consequently, the function of miRs in tumorigenesis, progression and resistance is of utmost importance for the understanding of dysfunctional cellular pathways that lead to disease evolution, therapy resistance, and poor clinical outcomes. Over the last two decades, much attention has been devoted to understanding the functional roles of miRs in NB biology. This review focuses on highlighting the important implications of miRs within the context of NB disease progression, particularly miRs' influences on NB disease evolution and therapy resistance. In this review, we discuss the functions of both the "oncomiRs" and "tumor suppressor miRs" in NB progression/therapy resistance. These are the critical components to be considered during the development of novel miR-based therapeutic strategies to counter therapy resistance.

17.
Med Dosim ; 44(3): 245-250, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30366619

RESUMO

The primary occurrence of mucoepidermoid carcinoma (MEC) of the conjunctiva is extremely rare, aggressive, and easily mistaken for squamous cell carcinoma. With fewer than 50 cases reported in the literature, there is no consensus on the most effective treatment. Radiation is an alternative to enucleation or orbital exenteration with the potential of eye preservation. We investigated several radiation approaches for a case with postresection positive margins at Tenon's fascia, and reported the patient's clinical course during the treatment and for a short time thereafter. An otherwise healthy 64-year-old male presented with MEC extending to the deep margin at the Tenon's fascia. Plans for 4 different radiation therapy treatment modalities (Intensity Modulated Radiation Therapy [IMRT], Volumetric Modulated Radiation Therapy [VMAT], double scattering (DS) proton, and reverse eye plaque low dose rate [LDR] ) were created and compared based on tumor coverage and normal tissue doses. The planning target volume (PTV) was too large and nonuniform for an eye plaque application. Using the largest plaque available (22 mm), the calculated minimum dose to the PTV was 57% while the dose to the skin was 1000% of the prescription. The proton plan completely spared the contralateral ocular structures and reduced the max doses to the ipsilateral macula and optic nerve, but was not clinically available at the time of treatment. The IMRT and VMAT plans produced similar dose distributions to each other, but VMAT further minimized dose to the ipsilateral eye. Due to the uniqueness of this case, a thorough study of the available radiation treatment options was deemed necessary. All of the external beam treatment techniques produced acceptable plans with VMAT producing the best available plan in this case. The patient was treated with the VMAT plan with a prescription of 6600 cGy in 30 fractions. At 5 months post-treatment, the patient is recovering from expected acute responses to radiation with follow ups scheduled.


Assuntos
Carcinoma Mucoepidermoide/radioterapia , Neoplasias da Túnica Conjuntiva/radioterapia , Braquiterapia , Carcinoma Mucoepidermoide/cirurgia , Neoplasias da Túnica Conjuntiva/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Radioterapia de Intensidade Modulada
18.
Oncotarget ; 8(4): 5717-5734, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-27974694

RESUMO

Therapy-resistant pancreatic cancer (PC) cells play a crucial role in tumor relapse, recurrence, and metastasis. Recently, we showed the anti-PC potential of an array of seaweed polyphenols and identified efficient drug deliverables. Herein, we investigated the benefit of one such deliverable, Hormophysa triquerta polyphenol (HT-EA), in regulating the dissemination physiognomy of therapy-resistant PC cells in vitro,and residual PC in vivo. Human PC cells exposed to ionizing radiation (IR), with/without HT-EA pre-treatment were examined for the alterations in the tumor invasion/metastasis (TIM) transcriptome (93 genes, QPCR-profiling). Utilizing a mouse model of residual PC, we investigated the benefit of HT-EA in the translation regulation of crucial TIM targets (TMA-IHC). Radiation activated 30, 50, 15, and 38 TIM molecules in surviving Panc-1, Panc-3.27, BxPC3, and MiaPaCa-2 cells. Of these, 15, 44, 12, and 26 molecules were suppressed with HT-EA pre-treatment. CXCR4 and COX2 exhibited cell-line-independent increases after IR, and was completely suppressed with HT-EA, across all PC cells. HT-EA treatment resulted in translational repression of IR-induced CXCR4, COX2, ß-catenin, MMP9, Ki-67, BAPX, PhPT-1, MEGF10, and GRB10 in residual PC. Muting CXCR4 or COX2 regulated the migration/invasion potential of IR-surviving cells, while forced expression of CXCR4 or COX2 significantly increased migration/invasion capabilities of PC cells. Further, treatment with HT-EA significantly inhibited IR-induced and CXCR4/COX2 forced expression-induced PC cell migration/invasion. This study (i) documents the TIM blueprint in therapy-resistant PC cells, (ii) defines the role of CXCR4 and COX2 in induced metastatic potential, and (iii) recognizes the potential of HT-EA in deterring the CXCR4/COX2-dependent dissemination destiny of therapy-resistant residual PC cells.


Assuntos
Ciclo-Oxigenase 2/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/radioterapia , Phaeophyceae/química , Polifenóis/administração & dosagem , Receptores CXCR4/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Invasividade Neoplásica , Neoplasia Residual , Neoplasias Pancreáticas/genética , Extratos Vegetais/química , Polifenóis/farmacologia , Alga Marinha/química , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Oncol Rep ; 37(4): 2382-2390, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28260023

RESUMO

Overexpression of BMI1 in human cancer cells, a member of the polycomb group of repressive complexes, correlates with advanced stage of disease, aggressive clinico-pathological behavior, poor prognosis, and resistance to radiation and chemotherapy. Studies have shown that experimental reduction of BMI1 protein level in tumor cells results in inhibition of cell proliferation, induction of apoptosis and/or senescence, and increased susceptibility to cytotoxic agents and radiation therapy. Although a role for BMI1 in cancer progression and its importance as a molecular target for cancer therapy has been established, information on the impact of silencing BMI1 in triple-negative breast cancer (TNBC) and its consequence on radiotherapy have not been well studied. Therefore, in the present study we investigated the potential therapeutic benefit of radiation therapy in BMI1-silenced breast cancer cells and studied the mechanism(s) of radiosensitization. Human MDA-MB-231 and SUM159PT breast cancer cells that were either stably transfected with a lentiviral vector expressing BMI1 shRNA (shBMI1) or control shRNA (shControl) or transient transfection with a BMI1-specific siRNA were used. Silencing of BMI1 resulted in marked reduction in BMI1 both at the mRNA and protein level that was accompanied by a significant reduction in cell migration compared to control cells. Further, BMI1 knockdown produced a marked enhancement of DNA damage as evidenced by Comet Assay and γH2AX foci, resulting in a dose-dependent radiosensitization effect. Molecular studies revealed modulation of protein expression that is associated with the DNA damage response (DDR) and autophagy pathways. Our results demonstrate that BMI1 is an important therapeutic target in breast cancer and suppression of BMI1 produces radiation sensitivity. Further, combining BMI1-targeted therapeutics with radiation might benefit patients diagnosed with TNBC.


Assuntos
Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , RNA Interferente Pequeno/farmacologia , Radiossensibilizantes/farmacologia , Neoplasias de Mama Triplo Negativas/genética , Autofagia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Complexo Repressor Polycomb 1/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/radioterapia
20.
Sci Rep ; 7(1): 13154, 2017 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-29030614

RESUMO

The 195-amino-acid-long human Retinal Degeneration Protein 3 (RD3) is critical in the regulation of guanylate cyclase (GC) signaling and photoreceptor cell survival. Recently, we identified significant loss of RD3 in high-risk neuroblastoma and the influential role of RD3 in tumor progression. However, the functional characterization of RD3 in tumor systems has been hampered by the dearth of information on its localization in normal tissue and by the lack of antibodies suitable for staining FFPE tissue, primarily due to the inaccessibility of the epitopes. In this study, we validated a custom-synthesized RD3 antibody and investigated the expression/localization of RD3 in assorted human tissues. We observed stratified expression of RD3 in different cell types and subcellular location of retina. We demonstrated extensive positive RD3 immunoreactivity in various normal tissues and particularly strong dot-like perinuclear staining in the lining epithelial cells, suggesting that RD3 may play an important role in the normal functioning of epithelial cells. RD3 expression is limited in the CNS. While neuroblastoma is often RD3-positive, the adrenal medulla, where many neuroblastomas originate, is RD3-negative. Meta-analysis of RD3 transcriptional expression across normal tissues confirmed tissue-specific RD3 mRNA levels. Our results revealed the tissue-specific expression/localization profile of RD3 for the first time.


Assuntos
Proteínas do Olho/metabolismo , Glândulas Suprarrenais/metabolismo , Sistema Nervoso Central/metabolismo , Células Epiteliais/metabolismo , Proteínas do Olho/genética , Humanos , Técnicas In Vitro , Neuroblastoma/metabolismo , Retina/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa