Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS One ; 15(9): e0239551, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32946518

RESUMEN

Pathway analysis is an informative method for comparing and contrasting drug-induced gene expression in cellular systems. Here, we define the effects of the marine natural product fucoxanthin, separately and in combination with the prototypic phosphatidylinositol 3-kinase (PI3K) inhibitor LY-294002, on gene expression in a well-established human glioblastoma cell system, U87MG. Under conditions which inhibit cell proliferation, LY-294002 and fucoxanthin modulate many pathways in common, including the retinoblastoma, DNA damage, DNA replication and cell cycle pathways. In sharp contrast, we see profound differences in the expression of genes characteristic of pathways such as apoptosis and lipid metabolism, contributing to the development of a differentiated and distinctive drug-induced gene expression signature for each compound. Furthermore, in combination, fucoxanthin synergizes with LY-294002 in inhibiting the growth of U87MG cells, suggesting complementarity in their molecular modes of action and pointing to further treatment combinations. The synergy we observe between the dietary nutraceutical fucoxanthin and the synthetic chemical LY-294002 in producing growth arrest in glioblastoma, illustrates the potential of nutri-pharmaceutical combinations in targeting this challenging disease.


Asunto(s)
Cromonas/administración & dosificación , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Morfolinas/administración & dosificación , Xantófilas/administración & dosificación , Apoptosis/efectos de los fármacos , Productos Biológicos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Suplementos Dietéticos , Sinergismo Farmacológico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Glioblastoma/dietoterapia , Humanos , Inhibidores de las Quinasa Fosfoinosítidos-3/administración & dosificación
2.
J Neurooncol ; 147(1): 213-227, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32036576

RESUMEN

PURPOSE: We conducted a feasibility study to investigate the use of ketogenic diets (KDs) as an adjuvant therapy for patients with glioblastoma (GBM), investigating (i) trial feasibility; (ii) potential impacts of the trial on patients' quality of life and health; (iii) patients' perspectives of their decision-making when invited to participate in the trial and (iv) recommending improvements to optimize future phase III trials. METHODS: A single-center, prospective, randomized, pilot study (KEATING), with an embedded qualitative design. Twelve newly diagnosed patients with GBM were randomized 1:1 to modified ketogenic diet (MKD) or medium chain triglyceride ketogenic diet (MCTKD). Primary outcome was retention at three months. Semi-structured interviews were conducted with a purposive sample of patients and caregivers (n = 15). Descriptive statistics were used for quantitative outcomes and qualitative data were analyzed thematically aided by NVivo. RESULTS: KEATING achieved recruitment targets, but the recruitment rate was low (28.6%). Retention was poor; only four of 12 patients completed the three-month diet (MCTKD n = 3; MKD n = 1). Participants' decisions were intuitive and emotional; caregivers supported diet implementation and influenced the patients' decision to participate. Those who declined made a deliberative and considered decision factoring diet burden and quality of life. A three-month diet was undesirable to patients who declined and withdrew. CONCLUSION: Recruitment to a KD trial for patients with GBM is possible. A six-week intervention period is proposed for a phase III trial. The role of caregivers should not be underestimated. Future trials should optimize and adequately support the decision-making of patients.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Dieta Cetogénica , Glioblastoma/dietoterapia , Adulto , Anciano , Terapia Combinada , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Investigación Cualitativa , Calidad de Vida , Resultado del Tratamiento
3.
Int J Oncol ; 56(2): 606-617, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31894296

RESUMEN

Abnormal metabolism serves a critical role in the development and progression of different types of malignancies including glioblastoma (GBM), and may therefore serve as a promising target for treatment of cancer. Preclinical studies have indicated that a ketogenic diet (KD) may exhibit beneficial effects in patients with GBM; however, the underlying mechanisms remain incompletely understood. The aim of the present study was to evaluate the effects of a KD on glioma stem­like cells (GSCs), by culturing patient­derived primary GSCs as well as a GSC cell line in glucose­restricted, ß­hydroxybutyrate­containing medium (BHB­Glow) which was used to mimic clinical KD treatment. GSCs cultured in BHB­Glow medium exhibited reduced proliferation and increased apoptosis compared with cells grown in the control medium. Furthermore, decreased expression of stem cell markers, diminished self­renewal in vitro, and reduced tumorigenic capacity in vivo, providing evidence that the stemness of GSCs was compromised. Mechanistically, culturing in BHB­Glow medium reduced glucose uptake and inhibited glycolysis in GSCs. Furthermore, culturing in the BHB­Glow medium resulted in morphological and functional disturbances to the mitochondria of GSCs. These metabolic changes may have reduced ATP production, promoted lactic acid accumulation, and thus, increased the production of reactive oxygen species (ROS) in GSCs. The expression levels and activation of mammalian target of rapamycin, hypoxia­inducible factor 1 and B­cell lymphoma 2 were decreased, consistent with the reduced proliferation of GSCs in BHB­Glow medium. ROS scavenging reversed the inhibitory effects of a KD on GSCs. Taken together, the results demonstrate that treatment with KD inhibited proliferation of GSCs, increased apoptosis and attenuated the stemness in GSCs by increasing ROS production.


Asunto(s)
Ácido 3-Hidroxibutírico/farmacología , Neoplasias Encefálicas/dietoterapia , Dieta Cetogénica , Glioblastoma/dietoterapia , Células Madre Neoplásicas/patología , Adolescente , Adulto , Anciano , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Proliferación Celular/efectos de los fármacos , Medios de Cultivo/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/patología , Glioblastoma/cirugía , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/metabolismo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Adulto Joven
4.
Acta Neuropathol Commun ; 6(1): 81, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30129426

RESUMEN

Glioblastoma (GB) is the most frequent primary brain tumor in adults with a dismal prognosis despite aggressive treatment including surgical resection, radiotherapy and chemotherapy with the alkylating agent temozolomide. Thus far, the successful implementation of the concept of targeted therapy where a drug targets a selective alteration in cancer cells was mainly limited to model diseases with identified genetic drivers. One of the most commonly altered oncogenic drivers of GB and therefore plausible therapeutic target is the epidermal growth factor receptor (EGFR). Trials targeting this signaling cascade, however, have been negative, including the phase III OSAG 101-BSA-05 trial. This highlights the need for further patient selection to identify subgroups of GB with true EGFR-dependency. In this retrospective analysis of treatment-naïve samples of the OSAG 101-BSA-05 trial cohort, we identify the EGFR signaling activity markers phosphorylated PRAS40 and phosphorylated ribosomal protein S6 as predictive markers for treatment efficacy of the EGFR-blocking antibody nimotuzumab in MGMT promoter unmethylated GBs. Considering the total trial population irrespective of MGMT status, a clear trend towards a survival benefit from nimotuzumab was already detectable when tumors had above median levels of phosphorylated ribosomal protein S6. These results could constitute a basis for further investigations of nimotuzumab or other EGFR- and downstream signaling inhibitors in selected patient cohorts using the reported criteria as candidate predictive biomarkers.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/dietoterapia , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al Calcio , Estudios de Cohortes , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Receptores ErbB/inmunología , Femenino , Humanos , Masculino , Proteínas de Microfilamentos , Necrosis/etiología , Necrosis/metabolismo , Fosforilación/efectos de los fármacos , Quinazolinas , Proteína S6 Ribosómica/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Proteínas Supresoras de Tumor/metabolismo
5.
Cancer Res ; 78(15): 4360-4369, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29844123

RESUMEN

Glioblastoma (GBM) is the most common primary brain malignancy and carries an extremely poor prognosis. Recent molecular studies revealed the CDK4/6-Rb-E2F axis and receptor tyrosine kinase (RTK) signaling to be deregulated in most GBM, creating an opportunity to develop more effective therapies by targeting both pathways. Using a phospho-RTK protein array, we found that both c-Met and TrkA-B pathways were significantly activated upon CDK4/6 inhibition in GBM cells. We therefore investigated the efficacy of combined CDK4/6 and c-Met/TrkA-B inhibition against GBM. We show that both c-Met and TrkA-B pathways transactivate each other, and targeting both pathways simultaneously results in more efficient pathway suppression. Mechanistically, inhibition of CDK4/6 drove NF-κB-mediated upregulation of hepatocyte growth factor, brain-derived neurotrophic factor, and nerve growth factor that in turn activated both c-Met and TrkA-B pathways. Combining the CDK4/6 inhibitor abemaciclib with the c-Met/Trk inhibitor altiratinib or the corresponding siRNAs induced apoptosis, leading to significant synergy against GBM. Collectively, these findings demonstrate that the activation of c-Met/TrkA-B pathways is a novel mechanism involved in therapeutic resistance of GBM to CDK4/6 inhibition and that dual inhibition of c-Met/Trk with CDK4/6 should be considered in future clinical trials.Significance: CDK4/6 inhibition in glioblastoma activates the c-Met and TrkA-B pathways mediated by NF-κB and can be reversed by a dual c-Met/Trk inhibitor. Cancer Res; 78(15); 4360-9. ©2018 AACR.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Glioblastoma/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor trkA/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glioblastoma/dietoterapia , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos
6.
Nutr Cancer ; 70(4): 643-649, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29668317

RESUMEN

There is an increasing interest in the use of the ketogenic diet (KD) as an adjuvant therapy for glioma patients. We assessed the tolerability and feasibility of a modified ketogenic diet (MKD) in patients with glioma, along with willingness of patients to participate in future randomized controlled trials. The study was undertaken in two parts; a patient questionnaire and evaluation of the diet. One hundred and seventy-two questionnaires were completed; 69% (n = 119) of the population reported MKD should be offered to patients with glioma and 73% (n = 125) would be willing to try MKD for 3 months. Six male patients with high grade gliomas tried the diet; 4 completed the 3-month feasibility period. Ketosis was achieved in all patients. The only gastrointestinal side effect was constipation (n = 2). Minimal changes were observed in weight, body mass index, fat mass and cholesterol profiles. MKD was well tolerated, with few side effects and is deliverable within a financially viable, NHS service. There is a high level of interest in the diet within the glioma patient community to ensure adequate recruitment for a clinical trial. Further studies are required to demonstrate efficacy and patient benefit before implementing a service.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Dieta Cetogénica/métodos , Glioblastoma/dietoterapia , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/patología , Costos y Análisis de Costo , Dieta Cetogénica/economía , Estudios de Factibilidad , Femenino , Glioblastoma/patología , Humanos , Cetosis , Masculino , Persona de Mediana Edad , Participación del Paciente/psicología , Ensayos Clínicos Controlados Aleatorios como Asunto , Encuestas y Cuestionarios
7.
Future Oncol ; 14(8): 699-707, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29521523

RESUMEN

AIM: MGMT promoter methylation has been associated with improved survival in glioblastoma multiforme treated with temozolomide. However, there is no consensus on specific cut-off levels of methylation. The aims of the study were to explore the prognostic impact of MGMT methylation status and to analyze the role of specific cut-off values. MATERIALS & METHODS: We analyzed 108 glioblastoma multiforme patients treated between 2008 and 2013 stratified according to three pyrosequencing-based quantitative methylation in: unmethylated (methylation <9%), intermediate (9-29%) and highly methylated (>29%). RESULTS: The three-class stratification has a prognostic impact (median progression-free survival: 7.97, 11.6 and 15 months respectively; p = 0.004; median OS: 13.2, 15.8 and 19.5 months, respectively; p = 0.0002), especially in patients exposed to temozolomide. CONCLUSION: Our study confirmed that the independent prognostic role of MGMT methylation status. An average level of methylation between all investigated CpGs of 9% may help discriminating between methylated and unmethylated tumors.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Glioblastoma/dietoterapia , Pronóstico , Proteínas Supresoras de Tumor/genética , Anciano , Islas de CpG/genética , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Supervivencia sin Enfermedad , Femenino , Glioblastoma/genética , Glioblastoma/patología , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/efectos de los fármacos , Temozolomida
8.
Nutr Cancer ; 68(6): 943-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27340742

RESUMEN

BACKGROUND: The oral bioavailability of curcuminoids is low, but can be enhanced by incorporation into micelles. The major curcuminoid curcumin has antitumor effects on glioblastoma cells in vitro and in vivo. We therefore aimed to determine intratumoral concentrations and the clinical tolerance of highly bioavailable micellar curcuminoids in glioblastoma patients. METHODS: Thirteen glioblastoma patients ingested 70 mg micellar curcuminoids [57.4 mg curcumin, 11.2 mg demethoxycurcumin (DMC), and 1.4 mg bis-demethoxycurcumin (BDMC)] three times per day for 4 days (total amount of 689 mg curcumin, 134 mg DMC, and 17 mg BDMC) prior to planned resection of their respective brain tumors. Tumor and blood samples were taken during the surgery and analyzed for total curcuminoid concentrations. (31)P magnetic resonance spectroscopic imaging was performed before and after curcuminoid consumption. RESULTS: Ten patients completed the study. The mean intratumoral concentration of curcumin was 56 pg/mg of tissue (range 9-151), and the mean serum concentration was 253 ng/ml (range 129-364). Inorganic phosphate was significantly increased within the tumor (P = 0.034). The mean ratio of phosphocreatine to inorganic phosphate decreased, and the mean intratumoral pH increased (P = 0.08) after curcuminoid intervention. CONCLUSION: Oral treatment with micellar curcuminoids led to quantifiable concentrations of total curcuminoids in glioblastomas and may alter intratumoral energy metabolism.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Curcumina/análogos & derivados , Curcumina/administración & dosificación , Suplementos Dietéticos , Glioblastoma/metabolismo , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/metabolismo , Antineoplásicos Fitogénicos/uso terapéutico , Transporte Biológico , Terapia Combinada/efectos adversos , Curcumina/efectos adversos , Curcumina/metabolismo , Curcumina/uso terapéutico , Diarilheptanoides , Suplementos Dietéticos/efectos adversos , Metabolismo Energético , Femenino , Jugos de Frutas y Vegetales , Glioblastoma/diagnóstico por imagen , Glioblastoma/dietoterapia , Glioblastoma/cirugía , Humanos , Concentración de Iones de Hidrógeno , Absorción Intestinal , Imagen por Resonancia Magnética , Masculino , Micelas , Neuroimagen , Fosfatos/metabolismo , Fosfocreatina/metabolismo , Cuidados Preoperatorios , Pyrus
9.
BMC Cancer ; 16: 310, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27178315

RESUMEN

BACKGROUND: Glioblastoma multiforme is a highly aggressive brain tumor with a poor prognosis, and advances in treatment have led to only marginal increases in overall survival. We and others have shown previously that the therapeutic ketogenic diet (KD) prolongs survival in mouse models of glioma, explained by both direct tumor growth inhibition and suppression of pro-inflammatory microenvironment conditions. The aim of this study is to assess the effects of the KD on the glioma reactive immune response. METHODS: The GL261-Luc2 intracranial mouse model of glioma was used to investigate the effects of the KD on the tumor-specific immune response. Tumor-infiltrating CD8+ T cells, CD4+ T cells and natural killer (NK) cells were analyzed by flow cytometry. The expression of immune inhibitory receptors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD-1) on CD8+ T cells were also analyzed by flow cytometry. Analysis of intracellular cytokine production was used to determine production of IFN, IL-2 and IFN- in tumor-infiltrating CD8+ T and natural killer (NK) cells and IL-10 production by T regulatory cells. RESULTS: We demonstrate that mice fed the KD had increased tumor-reactive innate and adaptive immune responses, including increased cytokine production and cytolysis via tumor-reactive CD8+ T cells. Additionally, we saw that mice maintained on the KD had increased CD4 infiltration, while T regulatory cell numbers stayed consistent. Lastly, mice fed the KD had a significant reduction in immune inhibitory receptor expression as well as decreased inhibitory ligand expression on glioma cells. CONCLUSIONS: The KD may work in part as an immune adjuvant, boosting tumor-reactive immune responses in the microenvironment by alleviating immune suppression. This evidence suggests that the KD increases tumor-reactive immune responses, and may have implications in combinational treatment approaches.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Citocinas/metabolismo , Dieta Cetogénica/métodos , Glioblastoma/dietoterapia , Animales , Neoplasias Encefálicas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Glioblastoma/inmunología , Humanos , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Clin Cancer Res ; 22(10): 2482-95, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26631612

RESUMEN

PURPOSE: Dysregulated energetics coupled with uncontrolled proliferation has become a hallmark of cancer, leading to increased interest in metabolic therapies. Glioblastoma (GB) is highly malignant, very metabolically active, and typically resistant to current therapies. Dietary treatment options based on glucose deprivation have been explored using a restrictive ketogenic diet (KD), with positive anticancer reports. However, negative side effects and a lack of palatability make the KD difficult to implement in an adult population. Hence, we developed a less stringent, supplemented high-fat low-carbohydrate (sHFLC) diet that mimics the metabolic and antitumor effects of the KD, maintains a stable nutritional profile, and presents an alternative clinical option for diverse patient populations. EXPERIMENTAL DESIGN: The dietary paradigm was tested in vitro and in vivo, utilizing multiple patient-derived gliomasphere lines. Cellular proliferation, clonogenic frequency, and tumor stem cell population effects were determined in vitro using the neurosphere assay (NSA). Antitumor efficacy was tested in vivo in preclinical xenograft models and mechanistic regulation via the mTOR pathway was explored. RESULTS: Reducing glucose in vitro to physiologic levels, coupled with ketone supplementation, inhibits proliferation of GB cells and reduces tumor stem cell expansion. In vivo, while maintaining animal health, the sHFLC diet significantly reduces the growth of tumor cells in a subcutaneous model of tumor progression and increases survival in an orthotopic xenograft model. Dietary-mediated anticancer effects correlate with the reduction of mTOR effector expression. CONCLUSIONS: We demonstrate that the sHFLC diet is a viable treatment alternative to the KD, and should be considered for clinical testing. Clin Cancer Res; 22(10); 2482-95. ©2015 AACR.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Glioblastoma/dietoterapia , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Dieta Baja en Carbohidratos/métodos , Dieta Alta en Grasa/métodos , Dieta Cetogénica/métodos , Modelos Animales de Enfermedad , Glioblastoma/metabolismo , Glucosa/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
12.
Adv Biol Regul ; 57: 64-74, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25442674

RESUMEN

Mechanistic target of rapamycin (mTOR) is a serine-threonine kinase that functions via two multiprotein complexes, namely mTORC1 and mTORC2, each characterized by different binding partners that confer separate functions. mTORC1 function is tightly regulated by PI3-K/Akt and is sensitive to rapamycin. mTORC2 is sensitive to growth factors, not nutrients, and is associated with rapamycin-insensitivity. mTORC1 regulates protein synthesis and cell growth through downstream molecules: 4E-BP1 (also called EIF4E-BP1) and S6K. Also, mTORC2 is thought to modulate growth factor signaling by phosphorylating the C-terminal hydrophobic motif of some AGC kinases such as Akt and SGK. Recent evidence has suggested that mTORC2 may play an important role in maintenance of normal as well as cancer cells by virtue of its association with ribosomes, which may be involved in metabolic regulation of the cell. Rapamycin (sirolimus) and its analogs known as rapalogues, such as RAD001 (everolimus) and CCI-779 (temsirolimus), suppress mTOR activity through an allosteric mechanism that acts at a distance from the ATP-catalytic binding site, and are considered incomplete inhibitors. Moreover, these compounds suppress mTORC1-mediated S6K activation, thereby blocking a negative feedback loop, leading to activation of mitogenic pathways promoting cell survival and growth. Consequently, mTOR is a suitable target of therapy in cancer treatments. However, neither of these complexes is fully inhibited by the allosteric inhibitor rapamycin or its analogs. In recent years, new pharmacologic agents have been developed which can inhibit these complexes via ATP-binding mechanism, or dual inhibition of the canonical PI3-K/Akt/mTOR signaling pathway. These compounds include WYE-354, KU-003679, PI-103, Torin1, and Torin2, which can target both complexes or serve as a dual inhibitor for PI3-K/mTOR. This investigation describes the mechanism of action of pharmacological agents that effectively target mTORC1 and mTORC2 resulting in suppression of growth, proliferation, and migration of tumor and cancer stem cells.


Asunto(s)
Movimiento Celular , Glioblastoma/metabolismo , Mitosis , Complejos Multiproteicos/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Antibióticos Antineoplásicos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Everolimus , Glioblastoma/dietoterapia , Glioblastoma/genética , Glioblastoma/patología , Humanos , Inmunosupresores/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/análogos & derivados , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
13.
Int J Oncol ; 44(6): 1843-52, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24728273

RESUMEN

Limiting dietary carbohydrates inhibits glioma growth in preclinical models. Therefore, the ERGO trial (NCT00575146) examined feasibility of a ketogenic diet in 20 patients with recurrent glioblastoma. Patients were put on a low-carbohydrate, ketogenic diet containing plant oils. Feasibility was the primary endpoint, secondary endpoints included the percentage of patients reaching urinary ketosis, progression-free survival (PFS) and overall survival. The effects of a ketogenic diet alone or in combination with bevacizumab was also explored in an orthotopic U87MG glioblastoma model in nude mice. Three patients (15%) discontinued the diet for poor tolerability. No serious adverse events attributed to the diet were observed. Urine ketosis was achieved at least once in 12 of 13 (92%) evaluable patients. One patient achieved a minor response and two patients had stable disease after 6 weeks. Median PFS of all patients was 5 (range, 3-13) weeks, median survival from enrollment was 32 weeks. The trial allowed to continue the diet beyond progression. Six of 7 (86%) patients treated with bevacizumab and diet experienced an objective response, and median PFS on bevacizumab was 20.1 (range, 12-124) weeks, for a PFS at 6 months of 43%. In the mouse glioma model, ketogenic diet alone had no effect on median survival, but increased that of bevacizumab-treated mice from 52 to 58 days (p<0.05). In conclusion, a ketogenic diet is feasible and safe but probably has no significant clinical activity when used as single agent in recurrent glioma. Further clinical trials are necessary to clarify whether calorie restriction or the combination with other therapeutic modalities, such as radiotherapy or anti-angiogenic treatments, could enhance the efficacy of the ketogenic diet.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Dieta Cetogénica/efectos adversos , Glioblastoma/dietoterapia , Recurrencia Local de Neoplasia/terapia , Adulto , Anciano , Animales , Bevacizumab , Terapia Combinada , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Cetosis/orina , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Neoplasias Experimentales , Proyectos Piloto , Calidad de Vida , Análisis de Supervivencia , Resultado del Tratamiento
14.
J Neurooncol ; 117(1): 125-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24442482

RESUMEN

Retrospective data suggests that low serum glucose levels during the treatment of glioblastoma multiforme (GBM) may improve clinical outcomes. As such, many patients are implementing a ketogenic diet (KD) in order to decrease serum glucose flux while simultaneously elevating circulating ketones during radiation therapy and chemotherapy for the treatment of GBM. With IRB approval, a retrospective review of patients with high-grade glioma treated with concurrent chemoradiotherapy and adjuvant chemotherapy was carried out from August 2010 to April 2013. Serum glucose and ketone levels, dexamethasone dose, and toxicity of patients undergoing a KD during treatment were also assessed. Blood glucose levels were compared between patients on an unspecified/standard diet and a KD. Toxicity was assessed by Common Terminology Criteria for Adverse Events version 4. In total, 53 patients were analyzed. Six underwent a KD during treatment. The diet was well tolerated with no grade III toxicity and one episode of grade II fatigue. No episodes of symptomatic hypoglycemia were experienced. Four patients are alive at a median follow-up of 14 months. The mean blood glucose of patients on a standard diet was 122 versus 84 mg/dl for those on a KD. Based on this retrospective study, a KD appears safe and well tolerated during the standard treatment of GBM. Dietary restriction of carbohydrates through a KD reduces serum glucose levels significantly, even in conjunction with high dose steroids, which may affect the response to standard treatment and prognosis. Larger prospective trials to confirm this relationship are warranted.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Dieta Cetogénica , Glioblastoma/dietoterapia , Glioma/dietoterapia , Adulto , Antineoplásicos Hormonales/administración & dosificación , Antineoplásicos Hormonales/uso terapéutico , Glucemia/análisis , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Quimioradioterapia , Quimioterapia Adyuvante , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Dieta Cetogénica/efectos adversos , Estudios de Seguimiento , Glioblastoma/sangre , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Glioma/sangre , Glioma/tratamiento farmacológico , Glioma/radioterapia , Humanos , Cetonas/sangre , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento
15.
Mol Cancer Ther ; 12(11): 2529-40, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23960094

RESUMEN

The major cytotoxic DNA adduct induced by temozolomide and other methylating agents used in malignant glioma and metastasized melanoma therapy is O(6)-methylguanine (O(6)-MeG). This primary DNA damage is converted by mismatch repair into secondary lesions, which block replication and in turn induce DNA double-strand breaks that trigger the DNA damage response (DDR). Key upstream players in the DDR are the phosphoinositide 3-kinases ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3 related (ATR). Here, we addressed the question of the importance of ATM and ATR in the cell death response following temozolomide. We show that (i) ATM- and ATR-mutated cells are hypersensitive to temozolomide, (ii) O(6)-MeG triggers ATM and ATR activation, (iii) knockdown of ATM and ATR enhances cell kill in gliobalstoma and malignant melanoma cells with a stronger and significant effect in ATR knockdown cells, (iv) ATR, but not ATM, knockdown abolished phosphorylation of H2AX, CHK1, and CHK2 in glioma cells, and (v) temozolomide-induced cell death was more prominently enhanced by pharmacologic inhibition of CHK1 compared with CHK2. The data suggest that ATM and, even better, ATR inhibition is a useful strategy in sensitizing cancer cells to temozolomide and presumably also other anticancer drugs.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos , Glioblastoma/metabolismo , Melanoma/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Roturas del ADN de Doble Cadena/efectos de los fármacos , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/farmacología , Técnicas de Silenciamiento del Gen , Glioblastoma/dietoterapia , Glioblastoma/genética , Guanina/análogos & derivados , Guanina/farmacología , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Metilnitronitrosoguanidina/farmacología , Fosforilación , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Temozolomida , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
16.
J Child Neurol ; 28(8): 1002-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23670248

RESUMEN

Glioblastoma multiforme is the most common malignant primary brain tumor in adults and generally considered to be universally fatal. Glioblastoma multiforme accounts for 12% to 15% of all intracranial neoplasms and affects 2 to 3 adults per every 100,000 in the United States annually. In children glioblastoma multiforme accounts for only approximately 7% to 9% of central nervous system tumors. The mean survival rate in adults after diagnosis ranges from 12 to 18 months with standard therapy and 3 to 6 months without therapy. The prognosis in children is better compared to adult tumor onset with a mean survival of approximately 4 years following gross total surgical resection and chemotherapy. There have been few advances in the treatment of glioblastoma multiforme in the past 40 years beyond surgery, radiotherapy, chemotherapy, and corticosteroids. For this reason a restrictive calorie ketogenic diet, similar to that used in children to control drug resistant seizure activity, has been advanced as an alternative adjunctive treatment to help prolonged survival. This article reviews the science of tumor metabolism and discusses the mechanism of calorie restriction, cellular energy metabolism, and how dietary induced ketosis can inhibit cancer cell's energy supply to slow tumor growth.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Restricción Calórica/métodos , Dieta Cetogénica/métodos , Glioblastoma/dietoterapia , Animales , Neoplasias Encefálicas/patología , Terapia Combinada/métodos , Glioblastoma/patología , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Humanos , Estados Unidos
17.
Cancer Res ; 72(5): 1103-15, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22237627

RESUMEN

In metronomic chemotherapy, frequent drug administration at lower than maximally tolerated doses can improve activity while reducing the dose-limiting toxicity of conventional dosing schedules. Although the antitumor activity produced by metronomic chemotherapy is attributed widely to antiangiogenesis, the significance of this mechanism remains somewhat unclear. In this study, we show that a 6-day repeating metronomic schedule of cyclophosphamide administration activates a potent antitumor immune response associated with brain tumor recruitment of natural killer (NK) cells, macrophages, and dendritic cells that leads to marked tumor regression. Tumor regression was blocked in nonobese diabetic/severe combined immunodeficient (NOD/SCID-γ) mice, which are deficient or dysfunctional in all these immune cell types. Furthermore, regression was blunted by NK cell depletion in immunocompetent syngeneic mice or in perforin-deficient mice, which are compromised for NK, NKT, and T-cell cytolytic functions. Unexpectedly, we found that VEGF receptor inhibitors blocked both innate immune cell recruitment and the associated tumor regression response. Cyclophosphamide administered at a maximum tolerated dose activated a transient, weak innate immune response, arguing that persistent drug-induced cytotoxic damage or associated cytokine and chemokine responses are required for effective innate immunity-based tumor regression. Together, our results reveal an innate immunity-based mechanism of tumor regression that can be activated by a traditional cytotoxic chemotherapy administered on a metronomic schedule. These findings suggest the need to carefully evaluate the clinical effects of combination chemotherapies that incorporate antiangiogenesis drugs targeting VEGF receptor.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Ciclofosfamida/farmacología , Glioblastoma/dietoterapia , Glioblastoma/inmunología , Imidazoles/farmacología , Inmunidad Innata/efectos de los fármacos , Indazoles/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Administración Metronómica , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/administración & dosificación , Axitinib , Línea Celular Tumoral , Ciclofosfamida/administración & dosificación , Humanos , Masculino , Dosis Máxima Tolerada , Ratones , Ratones Endogámicos NOD , Ratones SCID , Perforina/deficiencia , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Epilepsy Res ; 100(3): 310-26, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21885251

RESUMEN

Malignant brain cancer persists as a major disease of morbidity and mortality. The failure to recognize brain cancer as a disease of energy metabolism has contributed in large part to the failure in management. As long as brain tumor cells have access to glucose and glutamine, the disease will progress. The current standard of care provides brain tumors with access to glucose and glutamine. The high fat low carbohydrate ketogenic diet (KD) will target glucose availability and possibly that of glutamine when administered in carefully restricted amounts to reduce total caloric intake and circulating levels of glucose. The restricted KD (RKD) targets major signaling pathways associated with glucose and glutamine metabolism including the IGF-1/PI3K/Akt/Hif pathway. The RKD is anti-angiogenic, anti-invasive, anti-inflammatory, and pro-apoptotic when evaluated in mice with malignant brain cancer. The therapeutic efficacy of the restricted KD can be enhanced when combined with drugs that also target glucose and glutamine. Therapeutic efficacy of the RKD was also seen against malignant gliomas in human case reports. Hence, the RKD can be an effective non-toxic therapeutic option to the current standard of care for inhibiting the growth and invasive properties of malignant brain cancer.


Asunto(s)
Neoplasias Encefálicas/dietoterapia , Restricción Calórica , Dieta Cetogénica , Metabolismo Energético/efectos de los fármacos , Glioblastoma/dietoterapia , Animales , Glucosa/metabolismo , Humanos
19.
Neurochem Res ; 36(8): 1566-73, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21538079

RESUMEN

This study was undertaken to determine if the ketogenic diet could be useful for glioblastoma patients. The hypothesis tested was whether glioblastoma cells can metabolize ketone bodies. Cerebellar astrocytes and C6 glioblastoma cells were incubated in glutamine and serum free medium containing [2,4-(13)C]ß-hydroxybutyrate (BHB) with and without glucose. Furthermore, C6 cells were incubated with [1-(13)C]glucose in the presence and absence of BHB. Cell extracts were analyzed by mass spectrometry and media by (1)H magnetic resonance spectroscopy and HPLC. Using [2,4-(13)C]BHB and [1-(13)C]glucose it could be shown that C6 cells, in analogy to astrocytes, had efficient mitochondrial activity, evidenced by (13)C labeling of glutamate, glutamine and aspartate. However, in the presence of glucose, astrocytes were able to produce and release glutamine, whereas this was not accomplished by the C6 cells, suggesting lack of anaplerosis in the latter. We hypothesize that glioblastoma cells kill neurons by not supplying the necessary glutamine, and by releasing glutamate.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , Astrocitos/metabolismo , Neoplasias Encefálicas/dietoterapia , Neoplasias Encefálicas/metabolismo , Dieta Cetogénica , Glioblastoma/dietoterapia , Glioblastoma/metabolismo , Ácido 3-Hidroxibutírico/química , Animales , Ácido Aspártico/metabolismo , Astrocitos/citología , Ciclo del Ácido Cítrico/fisiología , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Cuerpos Cetónicos/metabolismo , Ácido Láctico/metabolismo , Ratones , Células Tumorales Cultivadas
20.
Biochim Biophys Acta ; 1807(6): 577-94, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20804725

RESUMEN

Malignant brain tumors are a significant health problem in children and adults. Conventional therapeutic approaches have been largely unsuccessful in providing long-term management. As primarily a metabolic disease, malignant brain cancer can be managed through changes in metabolic environment. In contrast to normal neurons and glia, which readily transition to ketone bodies (ß-hydroxybutyrate) for energy under reduced glucose, malignant brain tumors are strongly dependent on glycolysis for energy. The transition from glucose to ketone bodies as a major energy source is an evolutionary conserved adaptation to food deprivation that permits the survival of normal cells during extreme shifts in nutritional environment. Only those cells with a flexible genome and normal mitochondria can effectively transition from one energy state to another. Mutations restrict genomic and metabolic flexibility thus making tumor cells more vulnerable to energy stress than normal cells. We propose an alternative approach to brain cancer management that exploits the metabolic flexibility of normal cells at the expense of the genetically defective and metabolically challenged tumor cells. This approach to brain cancer management is supported from recent studies in mice and humans treated with calorie restriction and the ketogenic diet. Issues of implementation and use protocols are presented for the metabolic management of brain cancer.


Asunto(s)
Neoplasias Encefálicas/terapia , Metabolismo Energético/fisiología , Glioblastoma/terapia , Adulto , Animales , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/dietoterapia , Neoplasias Encefálicas/metabolismo , Restricción Calórica , Niño , Dieta Cetogénica , Modelos Animales de Enfermedad , Glioblastoma/complicaciones , Glioblastoma/dietoterapia , Glioblastoma/metabolismo , Humanos , Ratones , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/terapia , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...