Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38612892

RESUMEN

Glioblastoma (GBM) is a fatal brain tumor with limited treatment options. O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status is the central molecular biomarker linked to both the response to temozolomide, the standard chemotherapy drug employed for GBM, and to patient survival. However, MGMT status is captured on tumor tissue which, given the difficulty in acquisition, limits the use of this molecular feature for treatment monitoring. MGMT protein expression levels may offer additional insights into the mechanistic understanding of MGMT but, currently, they correlate poorly to promoter methylation. The difficulty of acquiring tumor tissue for MGMT testing drives the need for non-invasive methods to predict MGMT status. Feature selection aims to identify the most informative features to build accurate and interpretable prediction models. This study explores the new application of a combined feature selection (i.e., LASSO and mRMR) and the rank-based weighting method (i.e., MGMT ProFWise) to non-invasively link MGMT promoter methylation status and serum protein expression in patients with GBM. Our method provides promising results, reducing dimensionality (by more than 95%) when employed on two large-scale proteomic datasets (7k SomaScan® panel and CPTAC) for all our analyses. The computational results indicate that the proposed approach provides 14 shared serum biomarkers that may be helpful for diagnostic, prognostic, and/or predictive operations for GBM-related processes, given further validation.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/genética , Proteómica , Temozolomida/uso terapéutico , Proteínas Sanguíneas , Neoplasias Encefálicas/genética , O(6)-Metilguanina-ADN Metiltransferasa , Metilasas de Modificación del ADN/genética , Proteínas Supresoras de Tumor/genética , Enzimas Reparadoras del ADN/genética
3.
Biomolecules ; 13(10)2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37892181

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common brain tumor with an overall survival (OS) of less than 30% at two years. Valproic acid (VPA) demonstrated survival benefits documented in retrospective and prospective trials, when used in combination with chemo-radiotherapy (CRT). PURPOSE: The primary goal of this study was to examine if the differential alteration in proteomic expression pre vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA as compared to standard-of-care CRT. The second goal was to explore the associations between the proteomic alterations in response to VPA/RT/TMZ correlated to patient outcomes. The third goal was to use the proteomic profile to determine the mechanism of action of VPA in this setting. MATERIALS AND METHODS: Serum obtained pre- and post-CRT was analyzed using an aptamer-based SOMAScan® proteomic assay. Twenty-nine patients received CRT plus VPA, and 53 patients received CRT alone. Clinical data were obtained via a database and chart review. Tests for differences in protein expression changes between radiation therapy (RT) with or without VPA were conducted for individual proteins using two-sided t-tests, considering p-values of <0.05 as significant. Adjustment for age, sex, and other clinical covariates and hierarchical clustering of significant differentially expressed proteins was carried out, and Gene Set Enrichment analyses were performed using the Hallmark gene sets. Univariate Cox proportional hazards models were used to test the individual protein expression changes for an association with survival. The lasso Cox regression method and 10-fold cross-validation were employed to test the combinations of expression changes of proteins that could predict survival. Predictiveness curves were plotted for significant proteins for VPA response (p-value < 0.005) to show the survival probability vs. the protein expression percentiles. RESULTS: A total of 124 proteins were identified pre- vs. post-CRT that were differentially expressed between the cohorts who received CRT plus VPA and those who received CRT alone. Clinical factors did not confound the results, and distinct proteomic clustering in the VPA-treated population was identified. Time-dependent ROC curves for OS and PFS for landmark times of 20 months and 6 months, respectively, revealed AUC of 0.531, 0.756, 0.774 for OS and 0.535, 0.723, 0.806 for PFS for protein expression, clinical factors, and the combination of protein expression and clinical factors, respectively, indicating that the proteome can provide additional survival risk discrimination to that already provided by the standard clinical factors with a greater impact on PFS. Several proteins of interest were identified. Alterations in GALNT14 (increased) and CCL17 (decreased) (p = 0.003 and 0.003, respectively, FDR 0.198 for both) were associated with an improvement in both OS and PFS. The pre-CRT protein expression revealed 480 proteins predictive for OS and 212 for PFS (p < 0.05), of which 112 overlapped between OS and PFS. However, FDR-adjusted p values were high, with OS (the smallest p value of 0.586) and PFS (the smallest p value of 0.998). The protein PLCD3 had the lowest p-value (p = 0.002 and 0.0004 for OS and PFS, respectively), and its elevation prior to CRT predicted superior OS and PFS with VPA administration. Cancer hallmark genesets associated with proteomic alteration observed with the administration of VPA aligned with known signal transduction pathways of this agent in malignancy and non-malignancy settings, and GBM signaling, and included epithelial-mesenchymal transition, hedgehog signaling, Il6/JAK/STAT3, coagulation, NOTCH, apical junction, xenobiotic metabolism, and complement signaling. CONCLUSIONS: Differential alteration in proteomic expression pre- vs. post-completion of concurrent chemoirradiation (CRT) is present with the addition of VPA. Using pre- vs. post-data, prognostic proteins emerged in the analysis. Using pre-CRT data, potentially predictive proteins were identified. The protein signals and hallmark gene sets associated with the alteration in the proteome identified between patients who received VPA and those who did not, align with known biological mechanisms of action of VPA and may allow for the identification of novel biomarkers associated with outcomes that can help advance the study of VPA in future prospective trials.


Asunto(s)
Glioblastoma , Humanos , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Estudios Retrospectivos , Proteoma , Proteómica , Antineoplásicos Alquilantes , Proteínas Hedgehog
4.
Front Oncol ; 13: 1127645, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37637066

RESUMEN

Background: Glioblastomas (GBM) are rapidly progressive, nearly uniformly fatal brain tumors. Proteomic analysis represents an opportunity for noninvasive GBM classification and biological understanding of treatment response. Purpose: We analyzed differential proteomic expression pre vs. post completion of concurrent chemoirradiation (CRT) in patient serum samples to explore proteomic alterations and classify GBM by integrating clinical and proteomic parameters. Materials and methods: 82 patients with GBM were clinically annotated and serum samples obtained pre- and post-CRT. Serum samples were then screened using the aptamer-based SOMAScan® proteomic assay. Significant traits from uni- and multivariate Cox models for overall survival (OS) were designated independent prognostic factors and principal component analysis (PCA) was carried out. Differential expression of protein signals was calculated using paired t-tests, with KOBAS used to identify associated KEGG pathways. GSEA pre-ranked analysis was employed on the overall list of differentially expressed proteins (DEPs) against the MSigDB Hallmark, GO Biological Process, and Reactome databases with weighted gene correlation network analysis (WGCNA) and Enrichr used to validate pathway hits internally. Results: 3 clinical clusters of patients with differential survival were identified. 389 significantly DEPs pre vs. post-treatment were identified, including 284 upregulated and 105 downregulated, representing several pathways relevant to cancer metabolism and progression. The lowest survival group (median OS 13.2 months) was associated with DEPs affiliated with proliferative pathways and exhibiting distinct oppositional response including with respect to radiation therapy related pathways, as compared to better-performing groups (intermediate, median OS 22.4 months; highest, median OS 28.7 months). Opposite signaling patterns across multiple analyses in several pathways (notably fatty acid metabolism, NOTCH, TNFα via NF-κB, Myc target V1 signaling, UV response, unfolded protein response, peroxisome, and interferon response) were distinct between clinical survival groups and supported by WGCNA. 23 proteins were statistically signficant for OS with 5 (NETO2, CST7, SEMA6D, CBLN4, NPS) supported by KM. Conclusion: Distinct proteomic alterations with hallmarks of cancer, including progression, resistance, stemness, and invasion, were identified in serum samples obtained from GBM patients pre vs. post CRT and corresponded with clinical survival. The proteome can potentially be employed for glioma classification and biological interrogation of cancer pathways.

5.
Cancers (Basel) ; 15(10)2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37345009

RESUMEN

Glioblastomas (GBM) are rapidly growing, aggressive, nearly uniformly fatal, and the most common primary type of brain cancer. They exhibit significant heterogeneity and resistance to treatment, limiting the ability to analyze dynamic biological behavior that drives response and resistance, which are central to advancing outcomes in glioblastoma. Analysis of the proteome aimed at signal change over time provides a potential opportunity for non-invasive classification and examination of the response to treatment by identifying protein biomarkers associated with interventions. However, data acquired using large proteomic panels must be more intuitively interpretable, requiring computational analysis to identify trends. Machine learning is increasingly employed, however, it requires feature selection which has a critical and considerable effect on machine learning problems when applied to large-scale data to reduce the number of parameters, improve generalization, and find essential predictors. In this study, using 7k proteomic data generated from the analysis of serum obtained from 82 patients with GBM pre- and post-completion of concurrent chemoirradiation (CRT), we aimed to select the most discriminative proteomic features that define proteomic alteration that is the result of administering CRT. Thus, we present a novel rank-based feature weighting method (RadWise) to identify relevant proteomic parameters using two popular feature selection methods, least absolute shrinkage and selection operator (LASSO) and the minimum redundancy maximum relevance (mRMR). The computational results show that the proposed method yields outstanding results with very few selected proteomic features, with higher accuracy rate performance than methods that do not employ a feature selection process. While the computational method identified several proteomic signals identical to the clinical intuitive (heuristic approach), several heuristically identified proteomic signals were not selected while other novel proteomic biomarkers not selected with the heuristic approach that carry biological prognostic relevance in GBM only emerged with the novel method. The computational results show that the proposed method yields promising results, reducing 7k proteomic data to 7 selected proteomic features with a performance value of 93.921%, comparing favorably with techniques that do not employ feature selection.

7.
Neurooncol Pract ; 7(3): 268-276, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32537176

RESUMEN

Glioblastoma (GBM) is a challenging diagnosis with almost universally poor prognosis. Though the survival advantage of postoperative radiation (RT) is well established, around 90% of patients will fail in the RT field. The high likelihood of local failure suggests the efficacy of RT needs to be improved to improve clinical outcomes. Radiosensitizers are an established method of enhancing RT cell killing through the addition of a pharmaceutical agent. Though the majority of trials using radiosensitizers have historically been unsuccessful, there continues to be interest with a variety of approaches having been employed. Epidermal growth factor receptor inhibitors, histone deacetylase inhibitors, antiangiogenic agents, and a number of other molecularly targeted agents have all been investigated as potential methods of radiosensitization in the temozolomide era. Outcomes have varied both in terms of toxicity and survival, but some agents such as valproic acid and bortezomib have demonstrated promising results. However, reporting of results in phase 2 trials in newly diagnosed GBM have been inconsistent, with no standard in reporting progression-free survival and toxicity. There is a pressing need for investigation of new agents; however, nearly all phase 3 trials of GBM patients of the past 25 years have demonstrated no improvement in outcomes. One proposed explanation for this is the selection of agents lacking sufficient preclinical data and/or based on poorly designed phase 2 trials. Radiosensitization may represent a viable strategy for improving GBM outcomes in newly diagnosed patients, and further investigation using agents with promising phase 2 data is warranted.

8.
J Biochem Anal Stud ; 4(1)2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33884377

RESUMEN

PURPOSE: Glioblastoma (GBM) is the most common form of brain tumor and has a uniformly poor prognosis. Development of prognostic biomarkers in easily accessible serum samples have the potential to improve the outcomes of patients with GBM through personalized therapy planning. MATERIAL/METHODS: In this study pre-treatment serum samples from 30 patients newly diagnosed with GBM were evaluated using a 40-protein multiplex ELISA platform. Analysis of potentially relevant gene targets using The Cancer Genome Atlas database was done using the Glioblastoma Bio Discovery Portal (GBM-BioDP). A ten-biomarker subgroup of clinically relevant molecules was selected using a functional grouping analysis of the 40 plex genes with two genes selected from each group on the basis of degree of variance, lack of co-linearity with other biomarkers and clinical interest. A Multivariate Cox proportional hazard approach was used to analyze the relationship between overall survival (OS), gene expression, and resection status as covariates. RESULTS: Thirty of 40 of the MSD molecules mapped to known genes within TCGA and separated the patient cohort into two main clusters centered predominantly around a grouping of classical and proneural versus the mesenchymal subtype as classified by Verhaak. Using the values for the 30 proteins in a prognostic index (PI) demonstrated that patients in the entire cohort with a PI below the median lived longer than those patients with a PI above the median (HR 1.8, p=0.001) even when stratified by both age and MGMT status. This finding was also consistent within each Verhaak subclass and highly significant (range p=0.0001-0.011). Additionally, a subset of ten proteins including, CRP, SAA, VCAM1, VEGF, MDC, TNFA, IL7, IL8, IL10, IL16 were found to have prognostic value within the TCGA database and a positive correlation with overall survival in GBM patients who had received gross tumor resection followed by conventional radiation therapy and temozolomide treatment concurrent with the addition of valproic acid. CONCLUSION: These findings demonstrate that proteomic approaches to the development of prognostic assays for treatment of GBM may hold potential clinical value.

9.
Neurooncol Pract ; 5(4): 246-250, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30402263

RESUMEN

BACKGROUND: Valproic acid (VPA) is an antiepileptic agent with histone deacetylase inhibitor activity shown to enhance overall survival and progression free survival in patients with newly diagnosed glioblastoma (GBM). This reports on the late toxicity of the VPA/radiotherapy (RT)/temozolomide (TMZ) combination in the long-term survivors of a phase 2 study evaluating this regimen. METHODS: 37 patients with newly diagnosed GBM were initially enrolled on this trial and received combination therapy. VPA/RT/TMZ related late toxicities were evaluated in the 6 patients that lived greater than 3 years using the Cancer Therapy and Evaluation Program Common Toxicity Criteria (CTC) Version 4.0 for toxicity and adverse event reporting as well as the RTOG/EORTC Radiation Morbidity Scoring Scheme. RESULTS: The median duration of follow-up for these 6 patients was 69.5m. In this cohort, the median OS was 73.8m (60.8-103.8m) and median PFS was 53.1m (37.3 - 103.8m). The most common late toxicity of VPA in conjunction with RT/TMZ were the CTC classifications of neurological, pain, and blood/ bone marrow toxicity and most were grade 1/2. There were only two grade 3/4 toxicities. CONCLUSIONS: The addition of VPA to concurrent RT/TMZ in patients with newly diagnosed GBM was well tolerated with little late toxicity. Additionally, VPA may result in improved outcomes as compared to historical data and merits further study.

10.
J Neurooncol ; 139(1): 145-152, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29767308

RESUMEN

INTRODUCTION: Pseudoprogression (PsP) is a diagnostic dilemma in glioblastoma (GBM) after chemoradiotherapy (CRT). Magnetic resonance imaging (MRI) features may fail to distinguish PsP from early true progression (eTP), however clinical findings may aid in their distinction. METHODS: Sixty-seven patients received CRT for GBM between 2003 and 2016, and had pre- and post-treatment imaging suitable for retrospective evaluation using RANO criteria. Patients with signs of progression within the first 12-weeks post-radiation (P-12) were selected. Lesions that improved or stabilized were defined as PsP, and lesions that progressed were defined as eTP. RESULTS: The median follow up for all patients was 17.6 months. Signs of progression developed in 35/67 (52.2%) patients within P-12. Of these, 20/35 (57.1%) were subsequently defined as eTP and 15/35 (42.9%) as PsP. MRI demonstrated increased contrast enhancement in 84.2% of eTP and 100% of PsP, and elevated CBV in 73.7% for eTP and 93.3% for PsP. A decrease in FLAIR was not seen in eTP patients, but was seen in 26.7% PsP patients. Patients with eTP were significantly more likely to require increased steroid doses or suffer clinical decline than PsP patients (OR 4.89, 95% CI 1.003-19.27; p = 0.046). KPS declined in 25% with eTP and none of the PsP patients. CONCLUSIONS: MRI imaging did not differentiate eTP from PsP, however, KPS decline or need for increased steroids was significantly more common in eTP versus PsP. Investigation and standardization of clinical assessments in response criteria may help address the diagnostic dilemma of pseudoprogression after frontline treatment for GBM.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/terapia , Glioblastoma/diagnóstico por imagen , Glioblastoma/terapia , Imagen por Resonancia Magnética , Encéfalo/diagnóstico por imagen , Quimioradioterapia , Medios de Contraste , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Esteroides/uso terapéutico , Resultado del Tratamiento
11.
Radiat Oncol ; 12(1): 191, 2017 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-29187219

RESUMEN

PURPOSE/OBJECTIVES: Despite mounting evidence for the use of re-irradiation (re-RT) in recurrent high grade glioma, optimal patient selection criteria for re-RT remain unknown. We present a novel scoring system based on radiobiology principles including target independent factors, the likelihood of target control, and the anticipated organ at risk (OAR) toxicity to allow for proper patient selection in the setting of recurrent glioma. MATERIALS/METHODS: Thirty one patients with recurrent glioma who received re-RT (2008-2016) at NCI - NIH were included in the analysis. A novel scoring system for overall survival (OS) and progression free survival (PFS) was designed to include:1) target independent factors (age, KPS (Karnofsky Performance Status), histology, presence of symptoms), 2) target control, and 3) OAR toxicity risk. Normal tissue complication probability (NTCP) calculations were performed using the Lyman model. Kaplan-Meier analysis was performed for overall survival (OS) and progression free survival (PFS) for comparison amongst variables. RESULTS: No patient, including those who received dose to OAR above the published tolerance dose, experienced any treatment related grade 3-5 toxicity with a median PFS and OS from re-RT of 4 months (0.5-103) and 6 months (0.7-103) respectively. Based on cumulative maximum doses the average NTCP was 25% (0-99%) for the chiasm, 21% (0-99%) for the right optic nerve, 6% (0-92%) for the left optic nerve, and 59% (0-100%) for the brainstem. The independent factor and target control scores were each statistically significant for OS and the combination of independent factors plus target control was also significant for both OS (p = 0.02) and PFS (p = 0.006). The anticipated toxicity risk score was not statistically significant. CONCLUSION: Our scoring system may represent a novel approach to patient selection for re-RT in recurrent high grade glioma. Further validation in larger patient cohorts including compilation of doses to tumor and OAR may help refine this further for inclusion into clinical trials and general practice.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Recurrencia Local de Neoplasia/patología , Órganos en Riesgo/efectos de la radiación , Reirradiación/mortalidad , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/radioterapia , Femenino , Glioma/radioterapia , Humanos , Estado de Ejecución de Karnofsky , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/radioterapia , Pronóstico , Dosificación Radioterapéutica , Radioterapia de Intensidad Modulada/métodos , Tasa de Supervivencia , Adulto Joven
12.
Proc Natl Acad Sci U S A ; 114(15): E3110-E3118, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28356516

RESUMEN

Gold nanorods (AuNRs)-assisted plasmonic photothermal therapy (AuNRs-PPTT) is a promising strategy for combating cancer in which AuNRs absorb near-infrared light and convert it into heat, causing cell death mainly by apoptosis and/or necrosis. Developing a valid PPTT that induces cancer cell apoptosis and avoids necrosis in vivo and exploring its molecular mechanism of action is of great importance. Furthermore, assessment of the long-term fate of the AuNRs after treatment is critical for clinical use. We first optimized the size, surface modification [rifampicin (RF) conjugation], and concentration (2.5 nM) of AuNRs and the PPTT laser power (2 W/cm2) to achieve maximal induction of apoptosis. Second, we studied the potential mechanism of action of AuNRs-PPTT using quantitative proteomic analysis in mouse tumor tissues. Several death pathways were identified, mainly involving apoptosis and cell death by releasing neutrophil extracellular traps (NETs) (NETosis), which were more obvious upon PPTT using RF-conjugated AuNRs (AuNRs@RF) than with polyethylene glycol thiol-conjugated AuNRs. Cytochrome c and p53-related apoptosis mechanisms were identified as contributing to the enhanced effect of PPTT with AuNRs@RF. Furthermore, Pin1 and IL18-related signaling contributed to the observed perturbation of the NETosis pathway by PPTT with AuNRs@RF. Third, we report a 15-month toxicity study that showed no long-term toxicity of AuNRs in vivo. Together, these data demonstrate that our AuNRs-PPTT platform is effective and safe for cancer therapy in mouse models. These findings provide a strong framework for the translation of PPTT to the clinic.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Oro/farmacología , Neoplasias de Cabeza y Cuello/terapia , Hipertermia Inducida , Rayos Láser , Nanotubos/química , Fototerapia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Femenino , Oro/química , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteómica , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Bioconjug Chem ; 25(2): 207-215, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24409808

RESUMEN

Nanoparticles as potential drug delivery vectors are drawing more attention every day. Here, we used gold nanopspheres (AuNSs) to selectively target the Wnt signaling pathway in human oral squamous cell carcinoma (HSC-3) cells. In a previously conducted study, XAV939, a small inhibiter, was found to strongly regulate the Wnt pathway by inhibiting the tankyrase enzyme and subsequent stabilization of cytoplasmic axin levels. In the present study, conjugating XAV939 molecules to AuNSs is found to enhance its potency by at least 100 times over its free form in killing HSC-3 cancer cells. Additionally, XAV 939 uptake studies have demonstrated an enhanced XAV939 bioconjugate delivery to the targeted cells compared to the passive cellular diffusion of the free drug at the same concentration. Furthermore, our study revealed that drug delivery and cytotoxicity are directly related to the size of the functionalized nanoparticles.


Asunto(s)
Oro/química , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Carcinoma de Células Escamosas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Nanopartículas del Metal/química , Microscopía Electrónica de Transmisión , Neoplasias de la Boca/patología
14.
J Phys Chem B ; 118(5): 1319-26, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24433049

RESUMEN

The development of new and improved photothermal contrast agents for the successful treatment of cancer (or other diseases) via plasmonic photothermal therapy (PPTT) is a crucial part of the application of nanotechnology in medicine. Gold nanorods (AuNRs) have been found to be the most effective photothermal contrast agents, both in vitro and in vivo. Therefore, determining the optimum AuNR size needed for applications in PPTT is of great interest. In the present work, we utilized theoretical calculations as well as experimental techniques in vitro to determine this optimum AuNR size by comparing plasmonic properties and the efficacy as photothermal contrast agents of three different sizes of AuNRs. Our theoretical calculations showed that the contribution of absorbance to the total extinction, the electric field, and the distance at which this field extends away from the nanoparticle surface all govern the effectiveness of the amount of heat these particles generate upon NIR laser irradiation. Comparing between three different AuNRs (38 × 11, 28 × 8, and 17 × 5 nm), we determined that the 28 × 8 nm AuNR is the most effective in plasmonic photothermal heat generation. These results encouraged us to carry out in vitro experiments to compare the PPTT efficacy of the different sized AuNRs. The 28 × 8 nm AuNR was found to be the most effective photothermal contrast agent for PPTT of human oral squamous cell carcinoma. This size AuNR has the best compromise between the total amount of light absorbed and the fraction of which is converted to heat. In addition, the distance at which the electric field extends from the particle surface is most ideal for this size AuNR, as it is sufficient to allow for coupling between the fields of adjacent particles in solution (i.e., particle aggregates), resulting in effective heating in solution.


Asunto(s)
Oro/química , Nanotubos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Medios de Contraste/química , Medios de Contraste/uso terapéutico , Medios de Contraste/toxicidad , Humanos , Rayos Infrarrojos , Rayos Láser , Modelos Teóricos , Neoplasias de la Boca/terapia , Nanotubos/toxicidad , Tamaño de la Partícula , Fototerapia , Polietilenglicoles/química
15.
Photochem Photobiol ; 90(2): 306-12, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24329577

RESUMEN

We have previously shown that plasmonic nanoparticles conjugated with nuclear-targeting and cytoplasm-targeting peptides (NLS and RGD, respectively) are capable of altering the cell cycle of human oral squamous carcinoma cells (HSC-3). In the present work, we show that this regulation of the cell cycle can be exploited to enhance the efficacy of a common chemotherapeutic agent, 5-Fluorouracil, by pretreating cells with gold nanoparticles. Utilizing flow cytometry cell cycle analysis, we were able to quantify the 5-Fluorouracil efficacy as an accumulation of cells in the S phase with a depletion of cells in the G2/M phase. Two gold nanoparticle sizes were tested in this work; 30 nm with a surface plasmon resonance at 530 nm and 15 nm with a surface plasmon resonance at 520 nm. The 30 nm nuclear-targeted gold nanoparticles (NLS-AuNPs) showed the greatest 5-Fluorouracil efficacy enhancement when 5-Fluorouracil treatment (500 µm, 48 h) is preceded by a 24-h treatment with nanoparticles. In conclusion, we show that nuclear-targeted 30 nm gold nanoparticles enhance 5-Fluorouracil drug efficacy in HSC-3 cells via regulation of the cell cycle, a chemosensitization technique that could potentially be expanded to different cell lines and different chemotherapies.


Asunto(s)
Ciclo Celular , Oro/química , Nanopartículas del Metal , Antimetabolitos Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Citometría de Flujo , Fluorouracilo/administración & dosificación , Humanos , Microscopía Electrónica de Transmisión , Espectrofotometría Ultravioleta
16.
ACS Nano ; 7(9): 7524-33, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-23952174

RESUMEN

We have successfully demonstrated the potential of surface-enhanced Raman spectroscopy (SERS) in monitoring the real time damage to genomic DNA. To reveal the capabilities of this technique, we exposed DNA to reactive oxygen species (ROS), an agent that has been implicated in causing DNA double-strand breaks, and the various stages of free radical-induced DNA damage have been monitored by using SERS. Besides this, we showed that prompt DNA aggregation followed by DNA double-strand scission and residual damage to the DNA bases caused by the ROS could be substantially reduced by the protective effect of Pt nanocages and nearly cubical Pt nanopartcles. The antioxidant activity of Pt nanoparticles was further confirmed by the cell viability studies. On the basis of SERS results, we identified various stages involved in the mechanism of action of ROS toward DNA damage, which involves the DNA double-strand scission and its aggregation followed by the oxidation of DNA bases. We found that Pt nanoparticles inhibit the DNA double-strand scission to a significant extent by the degradation of ROS. Our method illustrates the capability of SERS technique in giving vital information about the DNA degradation reactions at molecular level, which may provide insight into the effectiveness and mechanism of action of many drugs in cancer therapy.


Asunto(s)
Daño del ADN/efectos de los fármacos , ADN/química , Nanopartículas del Metal/química , Platino (Metal)/química , Especies Reactivas de Oxígeno/química , Espectrometría Raman/métodos , Sistemas de Computación , ADN/genética , Daño del ADN/genética , Interacciones Farmacológicas , Humanos , Nanopartículas del Metal/ultraestructura , Tamaño de la Partícula , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
17.
Bioconjug Chem ; 24(6): 897-906, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23777334

RESUMEN

Recently, we have shown that targeting the cancer cell nucleus with solid gold nanospheres, using a cancer cell penetrating/pro-apoptotic peptide (RGD) and a nuclear localization sequence peptide (NLS), inhibits cell division, thus leading to apoptosis. In the present work, flow cytometric analysis revealed an increase in cell death, via apoptosis and necrosis, in HSC cells upon treatment with peptide-conjugated hollow gold nanocages, compared to those treated with the peptide-conjugated solid gold nanospheres. This is consistent with a G0/G1 phase accumulation, S phase depletion, and G2/M phase depletion, as well as reduced ATP levels. Here, we investigate the possible causes for the observed enhanced cell death with the use of confocal microscopy. The fluorescence images of HSC cells treated with gold nanocages indicate the presence of reactive oxygen species, known to cause apoptosis. The formation of reactive oxygen species observed is consistent with a mechanism involving the oxidation of metallic silver on the inner cavity of the nanocage (inherent to the synthesis of the gold nanocages) to silver oxide. This oxidation is confirmed by an observed redshift in the surface plasmon resonance of the gold nanocages in cell culture medium. The silver oxide, a semiconductor known to photochemically generate hydroxyl radicals, a form of reactive oxygen species, is proposed as a mechanism for the enhanced cell death caused by gold nanocages. Thus, the enhanced cell death, via apoptosis and necrosis, observed with peptide-conjugated hollow gold nanocage-treated cells is considered to be a result of the metallic composition (silver remaining on the inner cavity) of the nanocage.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Oro/química , Nanopartículas del Metal/química , Neoplasias de la Boca/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Péptidos/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Carcinoma de Células Escamosas/patología , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias de la Boca/patología , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Tamaño de la Partícula , Porosidad , Relación Estructura-Actividad , Propiedades de Superficie
18.
J Am Chem Soc ; 135(12): 4815-21, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23470053

RESUMEN

Conformation-induced formation of a series of unique Raman marker bands in cancer cell DNA, upon dehydration, have been probed for the first time with the use of surface enhanced Raman spectroscopy (SERS). These bands are capable of distinguishing cancer cell DNA from healthy cell DNA. For this simple and label-free DNA detection approach, we used conventional spherical silver nanoparticles, at a high concentration, without any aggregating agents, which gave highly reproducible SERS spectra of DNA separated from various human cells irrespective of their highly complex compositions and sequences. The observed phenomenon is attributed to the change in the chemical environment due to the presence of nucleobase lesions in cancer cell DNA and subsequent variation in the nearby electronic cloud during the dehydration-driven conformational changes. Detailed analysis of the SERS spectra gave important insight about the lesion-induced structural modifications upon dehydration in the cancer cell DNA. These results have widespread implications in cancer diagnostics, where SERS provides vital information about the DNA modifications in the cancer cells.


Asunto(s)
Carcinoma de Células Escamosas/patología , ADN/química , Deshidratación/patología , Neoplasias de la Boca/patología , Espectrometría Raman/métodos , Línea Celular , Línea Celular Tumoral , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Agua/química
19.
Ther Deliv ; 3(4): 457-78, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22834077

RESUMEN

Cancer is the current leading cause of death worldwide, responsible for approximately one quarter of all deaths in the USA and UK. Nanotechnologies provide tremendous opportunities for multimodal, site-specific drug delivery to these disease sites and Au nanoparticles further offer a particularly unique set of physical, chemical and photonic properties with which to do so. This review will highlight some recent advances, by our laboratory and others, in the use of Au nanoparticles for systemic drug delivery to these malignancies and will also provide insights into their rational design, synthesis, physiological properties and clinical/preclinical applications, as well as strategies and challenges toward the clinical implementation of these constructs moving forward.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos , Oro/química , Nanopartículas del Metal , Nanomedicina , Neoplasias/tratamiento farmacológico , Tecnología Farmacéutica/métodos , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Transporte Biológico , Química Farmacéutica , Composición de Medicamentos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Tamaño de la Partícula , Distribución Tisular
20.
Chem Soc Rev ; 40(7): 3391-404, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21629885

RESUMEN

Gold nanoparticles possess a unique combination of properties which allow them to act as highly multifunctional anti-cancer agents (X. H. Huang, P. K. Jain, I. H. El-Sayed and M. A. El-Sayed, Nanomedicine, 2007, 2, 681-693; P. Ghosh, G. Han, M. De, C. K. Kim and V. M. Rotello, Adv. Drug Delivery Rev., 2008, 60, 1307-1315; S. Lal, S. E. Clare and N. J. Halas, Acc. Chem. Res., 2008, 41, 1842-1851; D. A. Giljohann, D. S. Seferos, W. L. Daniel, M. D. Massich, P. C. Patel and C. A. Mirkin, Angew. Chem., Int. Ed., 2010, 49, 3280-3294). Not only can they be used as targeted contrast agents for photothermal cancer therapy, they can serve as scaffolds for increasingly potent cancer drug delivery, as transfection agents for selective gene therapy, and as intrinsic antineoplastic agents. This tutorial review will highlight some of the many forms and recent applications of these gold nanoparticle conjugates by our lab and others, as well as their rational design and physiologic interactions.


Asunto(s)
Antineoplásicos/administración & dosificación , Oro/química , Nanopartículas del Metal/química , Neoplasias/terapia , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Diseño de Fármacos , Oro/metabolismo , Oro/farmacocinética , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA