Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Adv Healthc Mater ; 13(19): e2400421, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38576069

RESUMEN

Glioblastoma (GBM), the most prevalent and aggressive primary malignant brain tumor, exhibits profound immunosuppression and demonstrates a low response rate to current immunotherapy strategies. Manganese cations (Mn2+) directly activate the cGAS/STING pathway and induce the unique catalytic synthesis of 2'3'-cGAMP to facilitate type I IFN production, thereby enhancing innate immunity. Here, a telodendrimer and Mn2+-based nanodriver (PLHM) with a small size is developed, which effectively target lymph nodes through the blood circulation and exhibit tumor-preventive effects at low doses of Mn2+ (3.7 mg kg-1). On the other hand, the PLHM nanodriver also exhibits apparent antitumor effects in GBM-bearing mice via inducing in vivo innate immune responses. The combination of PLHM with doxorubicin nanoparticles (PLHM-DOX NPs) results in superior inhibition of tumor growth in GBM-bearing mice due to the synergistic potentiation of STING pathway functionality by Mn2+ and the presence of cytoplasmic DNA. These findings demonstrate that PLHM-DOX NPs effectively stimulate innate immunity, promote dendritic cell maturation, and orchestrate cascaded infiltration of CD8 cytotoxic T lymphocytes within glioblastomas characterized by low immunogenicity. These nanodivers chelated with Mn2+ show promising potential for tumor prevention and antitumor effects on glioblastoma by activating the STING pathway.


Asunto(s)
Doxorrubicina , Glioblastoma , Manganeso , Proteínas de la Membrana , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/metabolismo , Glioblastoma/prevención & control , Animales , Manganeso/química , Manganeso/farmacología , Ratones , Doxorrubicina/farmacología , Doxorrubicina/química , Humanos , Línea Celular Tumoral , Proteínas de la Membrana/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Neoplasias Encefálicas/metabolismo , Nanopartículas/química , Inmunidad Innata/efectos de los fármacos , Ratones Endogámicos C57BL
2.
Folia Neuropathol ; 61(3): 235-241, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37818684

RESUMEN

Brain tumours are heterogeneous and are classified comprehensively into molecular subtypes based on genetic alterations. Glioblastoma rapid progression, drug resistance, and recurrence have been scientifically linked to several factors, including its rapid growth rate, loss of apoptosis, pro-survival signalling, molecular heterogeneities and hallmark features to infiltrate vital brain structures. Because of the growing demand for design and development of delivery systems to overcome the existing limitations with the current therapeutic strategies, researchers are exploiting multifaceted aspects of nanotechnology to improve delivery of the drug payload. Firstly, nanotechnology procedures can improve the drug delivery methods with the help of nanoparticles (NPs) based nanovectors that can efficiently cross blood-brain barrier. Secondly, NPs also improve the cellular uptake of the drug as they can efficiently bind with the cell surface. Thirdly, NPs make the delivery of siRNAs and peptides possible, which can suppress the resistance of glioblastoma against TMZ or other chemo-preventive drugs. Fourthly, the use of metal NPs increases the efficiency of scanning or magnetic resonance imaging (MRI) procedures as they can produce contrasts in it. Lastly, NPs make it possible to use highly targeted co-administered strategies like chemoprevention and near infrared (NIR) or radiotherapy (RT). Hence, nanotechnology offers several promising solutions against glioblastoma by countering it on many fronts.


Asunto(s)
Antineoplásicos , Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/prevención & control , Glioblastoma/patología , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Nanotecnología , Quimioprevención , Neoplasias Encefálicas/prevención & control , Neoplasias Encefálicas/patología , Línea Celular Tumoral
3.
PLoS One ; 18(5): e0285536, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37228120

RESUMEN

Tetra-O-methyl-nordihydroguaiaretic acid (terameprocol; M4N), a global transcription inhibitor, in combination with a second anticancer drug induces strong tumoricidal activity and has the ability to suppress energy metabolism in cultured cancer cells. In this study, we showed that after continuous oral consumption of high-fat (HF) diets containing M4N, the M4N concentration in most of the organs in mice reached ~1 µM (the M4N concentration in intestines and fat pads was as high as 20-40 µM) and treatment with the combination of M4N with temozolomide (TMZ) suppressed glycolysis and the tricarboxylic acid cycle in LN229 human glioblastoma implanted in xenograft mice. Combination treatment of M4N with TMZ also reduced the levels of lactate dehydrogenase A (LDHA), a key enzyme for glycolysis; lactate, a product of LDHA-mediated enzymatic activity; nicotinamide phosphoribosyltransferase, a rate-limiting enzyme for nicotinamide adenine dinucleotide plus hydrogen (NADH)/NAD+ salvage pathway; and NAD+, a redox electron carrier essential for energy metabolism. It was also shown that M4N suppressed oxygen consumption in cultured LN229 cells, indicating that M4N inhibited oxidative phosphorylation. Treatment with M4N and TMZ also decreased the level of hypoxia-inducible factor 1A, a major regulator of LDHA, under hypoxic conditions. The ability of M4N to suppress energy metabolism resulted in induction of the stress-related proteins activating transcription factor 4 and cation transport regulator-like protein 1, and an increase in reactive oxygen species production. In addition, the combination treatment of M4N with TMZ reduced the levels of oncometabolites such as 2-hydroxyglutarate as well as the aforementioned lactate. M4N also induced methylidenesuccinic acid (itaconate), a macrophage-specific metabolite with anti-inflammatory activity, in tumor microenvironments. Meanwhile, the ability of M4N to suppress energy metabolism prevented obesity in mice consuming HF diets, indicating that M4N has beneficial effects on normal tissues. The dual ability of combination treatment with M4N to suppress both energy metabolism and oncometabolites shows that it is potentially an effective therapy for cancer.


Asunto(s)
Glioblastoma , Humanos , Animales , Ratones , Masoprocol/farmacología , Temozolomida/farmacología , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/prevención & control , Glioblastoma/patología , Dieta Alta en Grasa/efectos adversos , NAD , Línea Celular Tumoral , Metabolismo Energético , Microambiente Tumoral
4.
Front Immunol ; 12: 770390, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34795675

RESUMEN

Glioblastomas (GBM) are the most frequent and aggressive malignant primary brain tumor and remains a therapeutic challenge: even after multimodal therapy, median survival of patients is only 15 months. Dendritic cell vaccination (DCV) is an active immunotherapy that aims at inducing an antitumoral immune response. Numerous DCV trials have been performed, vaccinating hundreds of GBM patients and confirming feasibility and safety. Many of these studies reported induction of an antitumoral immune response and indicated improved survival after DCV. However, two controlled randomized trials failed to detect a survival benefit. This raises the question of whether the promising concept of DCV may not hold true or whether we are not yet realizing the full potential of this therapeutic approach. Here, we discuss the results of recent vaccination trials, relevant parameters of the vaccines themselves and of their application, and possible synergies between DCV and other therapeutic approaches targeting the immunosuppressive microenvironment of GBM.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Glioblastoma/terapia , Inmunoterapia/métodos , Vacunación/métodos , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/prevención & control , Vacunas contra el Cáncer/administración & dosificación , Terapia Combinada , Glioblastoma/inmunología , Glioblastoma/prevención & control , Humanos , Evaluación de Resultado en la Atención de Salud , Supervivencia sin Progresión , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
5.
Med Oncol ; 38(11): 132, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34581882

RESUMEN

WAC is closely related to the occurrence and development of tumors. However, its role in human glioblastoma (GBM) and its potential regulatory mechanisms have not been investigated. This study demonstrated that WAC is downregulated in GBM, and its low expression predicts a poor prognosis. We investigated the effect of WAC on the proliferation of glioma cells through a CCK-8 assay, EdU incorporation, and cell formation. The effects of WAC on apoptosis and autophagy in glioma were determined by flow cytometry, TUNEL detection, immunofluorescence, q-PCR, WB, and scanning electron microscopy. We found that overexpression of WAC inhibited the proliferation of glioma cells, promoted apoptosis, and induced autophagy. Therefore, WAC is likely to play a role as a new regulatory molecule in glioma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Apoptosis , Autofagia/fisiología , Neoplasias Encefálicas/patología , Glioblastoma/patología , Proteínas Supresoras de Tumor/fisiología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/prevención & control , Línea Celular Tumoral , Proliferación Celular , Glioblastoma/mortalidad , Glioblastoma/prevención & control , Humanos , Transducción de Señal
6.
Pharmacol Res ; 169: 105638, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33933637

RESUMEN

Glioblastoma (GBM) is the most common primary tumor of the central nervous system. Current treatments available for GBM entails surgical resection followed by temozolomide chemotherapy and/or radiotherapy, which are associated with multidrug resistance and severe side effects. While this treatment could yield good results, in almost all cases, patients suffer from relapse, which leads to reduced survival rates. Thus, therapeutic approaches with improved efficiency and reduced off-target risks are needed to overcome these problems. Regarding this, natural products appear as a safe and attractive strategy as chemotherapeutic agents or adjuvants in the treatment of GBM. Besides the increasing role of natural compounds for chemoprevention of GBM, it has been proposed to prevent carcinogenesis and metastasis of GBM. Numerous investigations showed that natural products are able to inhibit proliferation and angiogenesis, to induce apoptosis, and to target GBM stem cells, which are associated with tumor development and recurrence. This review gives a timely and comprehensive overview of the current literature regarding chemoprevention and therapy of GBM by natural products with a focus on essential oils and phenolic compounds and their molecular mechanisms.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Aceites Volátiles/uso terapéutico , Fenoles/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Animales , Productos Biológicos/uso terapéutico , Neoplasias Encefálicas/prevención & control , Glioblastoma/prevención & control , Humanos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/prevención & control
7.
Life Sci ; 279: 119643, 2021 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-34048811

RESUMEN

BACKGROUND: Glioblastoma multiform (GBM) is the most belligerent and prevalent brain malignancy among adults. Due to the blood-brain barrier (BBB), drug administration is confronted by massive challenges, making resectional surgery the only treatment pipeline. MicroRNAs have recently absorbed the attention of studies for correlating with the progression of various malignancies. miR-30c has been reported to play a role in cell proliferation, metabolism, and apoptosis process. For instance, miR-30c has been reported to regulate apoptosis through the TNF-related apoptosis-inducing ligand (TRAIL). miR-30c also targets IL-6, which further induces apoptosis. Besides, miR-30c inhibits glioma proliferation and its migratory ability. Besides, the overexpression of miR-30c arrested cells at G0 as well as dampening their migration and invasion. However, it has been shown that the expression level of miR-30c was low in glioma. MSCs can migrate toward tumor cells which is called tumor-tropism, in which they are capable of delivering engineered miR-30c based on gap junction and non-intimacy mechanisms. MATERIAL AND METHODS: MiR-30c was cloned into pCDH-CMV-MCS-EF1-copGFP vector utilizing XbaI and EcoRI in order to construct pCDH-miR-30c. Then psPAX2, pMD2.G, and pCDH-miR-30c were co-transfected into Hek-293T to yield lenti-miR-30c virus particles. Next, bone marrow-mesenchymal stem cells (BM-MSCs) were Transduced with lenti-miR-30c. Thereafter, we co-cultured U-251 cell line with BM-MCSs-miR-30c and evaluated the apoptosis rate and the relative expression level of IL-6, Klf4, Sox2, c-Myc, and Oct4 using Real-Time PCR and flow cytometry. RESULTS: Wound healing assays represented low migratory ability in U-251 cells treated with BM-MSCs-miR-30c. Plus, apoptosis assay using Annexin V/7AAD showed an increased number of apoptotic U-251 cells following the treatment. miR-30 targeted IL-6 and induced apoptosis. It also impacted on the self-renewal and the anti-apoptotic cluster of genes, namely Klf4, Sox2, c-Myc, and Oct4, to induce apoptosis and dwindle the migration and invasion.


Asunto(s)
Apoptosis , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/prevención & control , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Glioblastoma/genética , Glioblastoma/patología , Humanos , Factor 4 Similar a Kruppel , Células Madre Mesenquimatosas/citología , Invasividad Neoplásica , Células Tumorales Cultivadas
8.
J Neurooncol ; 148(2): 231-244, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32342332

RESUMEN

PURPOSE: Glioblastoma (GBM) carries a dismal prognosis despite standard multimodal treatment with surgery, chemotherapy and radiation. Immune checkpoint inhibitors, such as PD1 blockade, for treatment of GBM failed to show clinical benefit. Rational combination strategies to overcome resistance of GBM to checkpoint monotherapy are needed to extend the promise of immunotherapy to GBM management. Emerging evidence suggests that protein phosphatase 2A (PP2A) plays a critical role in the signal transduction pathways of both adaptive and innate immune cells and that inhibition of PP2A could enhance cancer immunity. We investigated the use of a PP2A inhibitor, LB-100, to enhance antitumor efficacy of PD1 blockade in a syngeneic glioma model. METHODS: C57BL/6 mice were implanted with murine glioma cell line GL261-luc or GL261-WT and randomized into 4 treatment arms: (i) control, (ii) LB-100, (iii) PD1 blockade and (iv) combination. Survival was assessed and detailed profiling of tumor infiltrating leukocytes was performed. RESULTS: Dual PP2A and PD1 blockade significantly improved survival compared with monotherapy alone. Combination therapy resulted in complete regression of tumors in about 25% of mice. This effect was dependent on CD4 and CD8 T cells and cured mice established antigen-specific secondary protective immunity. Analysis of tumor lymphocytes demonstrated enhanced CD8 infiltration and effector function. CONCLUSION: This is the first preclinical investigation of the effect of combining PP2A inhibition with PD1 blockade for GBM. This novel combination provided effective tumor immunotherapy and long-term survival in our animal GBM model.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Piperazinas/administración & dosificación , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína Fosfatasa 2/antagonistas & inhibidores , Animales , Neoplasias Encefálicas/prevención & control , Línea Celular Tumoral , Quimioterapia Combinada/métodos , Femenino , Glioblastoma/prevención & control , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Proteína Fosfatasa 2/inmunología
9.
FASEB J ; 33(12): 13476-13491, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31570001

RESUMEN

Glioblastoma (GBM; grade 4 glioma) is a highly aggressive and incurable tumor. GBM has recently been characterized as highly dependent on alternative splicing, a critical driver of tumor heterogeneity and plasticity. Estrogen-related receptor ß (ERR-ß) is an orphan nuclear receptor expressed in the brain, where alternative splicing of the 3' end of the pre-mRNA leads to the production of 3 validated ERR-ß protein products: ERR-ß short form (ERR-ßsf), ERR-ß2, and ERR-ß exon 10 deleted. Our prior studies have shown the ERR-ß2 isoform to play a role in G2/M cell cycle arrest and induction of apoptosis, in contrast to the function of the shorter ERR-ßsf isoform in senescence and G1 cell cycle arrest. In this study, we sought to better define the role of the proapoptotic ERR-ß2 isoform in GBM. We show that the ERR-ß2 isoform is located not only in the nucleus but also in the cytoplasm. ERR-ß2 suppresses GBM cell migration and interacts with the actin nucleation-promoting factor cortactin, and an ERR-ß agonist is able to remodel the actin cytoskeleton and similarly suppress GBM cell migration. We further show that inhibition of the splicing regulatory cdc2-like kinases in combination with an ERR-ß agonist shifts isoform expression in favor of ERR-ß2 and potentiates inhibition of growth and migration in GBM cells and intracranial tumors.-Tiek, D. M., Khatib, S. A., Trepicchio, C. J., Heckler, M. M., Divekar, S. D., Sarkaria, J. N., Glasgow, E., Riggins, R. B. Estrogen-related receptor ß activation and isoform shifting by cdc2-like kinase inhibition restricts migration and intracranial tumor growth in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Movimiento Celular , Glioblastoma/prevención & control , Hidrazinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Tiazoles/farmacología , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Ciclo Celular , Proliferación Celular , Quimioterapia Combinada , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Isoformas de Proteínas , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
10.
J Neurooncol ; 144(3): 489-499, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31482266

RESUMEN

PURPOSE: Glioblastoma is the most aggressive malignant brain tumor. Despite multimodal treatments, median survival is only 15 months for glioblastoma patients, with tumor recurring in the resection margins after surgical removal. Hypothermia is emerging as an interesting and safe treatment for several conditions. In the context of glioblastoma, we propose that moderate hypothermia could inhibit both cell proliferation and migration, and thus help prevent secondary tumor growth. METHODS: In vitro experiments on A172, U251, U87 and T98G human glioblastoma cell lines explored the effects of severe (23 °C), moderate (28 °C), and mild (33 °C) hypothermia. We further investigated the effects of moderate hypothermia on cell proliferation, migration, morphology, and cell cycle distribution. RESULTS: Similar results were obtained with all four cell lines, indicating a consistent and broad effect of moderate hypothermia. Hypothermia inhibited both cell proliferation and non-oriented migration in a dose-dependent manner, with a significant reduction at 33 °C and almost total arrest at 28 °C. Cell proliferation arrest was long-lasting and oriented cell migration was also reduced at 28 °C. Moreover, moderate hypothermia significantly altered cell cycle distribution, with cells accumulating in the G2/M phase, leading to cell cycle arrest. Lastly, hypothermia at 28 °C also affected cell morphology by deteriorating cell membranes and altering cell shape. CONCLUSIONS: The presented results demonstrate that moderate hypothermia could be a promising adjuvant therapy for glioblastoma treatment as it strongly inhibits both cell proliferation and migration. If in vivo preclinical results corroborate our findings, therapeutic hypothermia applied at the resection margins could probably delay tumor recurrence, combined with current treatments.


Asunto(s)
Puntos de Control del Ciclo Celular , Movimiento Celular , Proliferación Celular , Glioblastoma/prevención & control , Hipotermia , Apoptosis , Glioblastoma/patología , Humanos , Células Tumorales Cultivadas , Cicatrización de Heridas
11.
J Neurooncol ; 142(3): 423-434, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30838489

RESUMEN

PURPOSE: Both IDH1-mutated and wild-type gliomas abundantly display aberrant CpG island hypermethylation. However, the potential role of hypermethylation in promoting gliomas, especially the most aggressive form, glioblastoma (GBM), remains poorly understood. METHODS: We analyzed RRBS-generated methylation profiles for 11 IDH1WT gliomas (including 7 GBMs), 24 IDH1MUT gliomas (including 6 GBMs), and 5 normal brain samples and employed TCGA GBM methylation profiles as a validation set. Upon classification of differentially methylated CpG islands by IDH1 status, we used integrated analysis of methylation and gene expression to identify SPINT2 as a top cancer related gene. To explore functional consequences of SPINT2 methylation in GBM, we validated SPINT2 methylation status using targeted bisulfite sequencing in a large cohort of GBM samples. We assessed DNA methylation-mediated SPINT2 gene regulation using 5-aza-2'-deoxycytidine treatment, DNMT1 knockdown and luciferase reporter assays. We conducted functional analyses of SPINT2 in GBM cell lines in vitro and in vivo. RESULTS: We identified SPINT2 as a candidate tumor-suppressor gene within a group of CpG islands (designated GT-CMG) that are hypermethylated in both IDH1MUT and IDH1WT gliomas but not in normal brain. We established that SPINT2 downregulation results from promoter hypermethylation, and that restoration of SPINT2 expression reduces c-Met activation and tumorigenic properties of GBM cells. CONCLUSIONS: We defined a previously under-recognized group of coordinately methylated CpG islands common to both IDH1WT and IDH1MUT gliomas (GT-CMG). Within GT-CMG, we identified SPINT2 as a top cancer-related candidate and demonstrated that SPINT2 suppressed GBM via down-regulation of c-Met activation.


Asunto(s)
Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Glioblastoma/prevención & control , Isocitrato Deshidrogenasa/genética , Glicoproteínas de Membrana/genética , Mutación , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Apoptosis , Proliferación Celular , Islas de CpG , Glioblastoma/genética , Glioblastoma/patología , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-met/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Rep ; 8(1): 15383, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337641

RESUMEN

XPO1 has recently emerged as a viable treatment target for solid malignancies, including glioblastoma (GBM), the most common primary malignant brain tumor in adults. However, given that tumors become commonly resistant to single treatments, the identification of combination therapies is critical. Therefore, we tested the hypothesis that inhibition of anti-apoptotic Bcl-2 family members and XPO1 are synthetically lethal. To this purpose, two clinically validated drug compounds, the BH3-mimetic, ABT263, and the XPO1 inhibitor, Selinexor, were used in preclinical GBM model systems. Our results show that inhibition of XPO1 reduces cellular viability in glioblastoma cell cultures. Moreover, addition of ABT263 significantly enhances the efficacy of XPO1 inhibition on the reduction of cellular viability, which occurs in a synergistic manner. While selinexor inhibits the proliferation of glioblastoma cells, the combination treatment of ABT263 and selinexor results in substantial induction of cell death, which is accompanied by activation of effector- initiator caspases and cleavage of PARP. Mechanistically we find that XPO1 inhibition results in down-regulation of anti-apoptotic Mcl-1 and attenuates ABT263 driven Mcl-1 up-regulation. Consistently, siRNA mediated silencing of Mcl-1 sensitizes for ABT263 mediated cell death and partially for the combination treatment. By using a human patient-derived xenograft model of glioblastoma in mice, we demonstrate that the combination treatment of ABT263 and Selinexor reduces tumor growth significantly more than each compound alone. Collectively, these results suggest that inhibition of XPO1 and Bcl-2/Bcl-xL might be a potential strategy for the treatment of malignant glial tumors.


Asunto(s)
Compuestos de Anilina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/prevención & control , Hidrazinas/farmacología , Carioferinas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Sulfonamidas/farmacología , Triazoles/farmacología , Proteína bcl-X/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
13.
Cancer Immunol Immunother ; 67(10): 1491-1503, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30030559

RESUMEN

High-grade gliomas harbor abundant myeloid cells that suppress anti-tumor immunity and support tumor growth. Targeting transcription factors, such as NF-κB p50, that mediate suppressive myeloid M2 polarization may prove therapeutic. GL261-Luc glioblastoma cells were inoculated into wild-type and p50-/- mice, followed by analysis of tumor growth, survival, tumor myeloid cells, and T cells. The absence of host p50 slows tumor growth and enables regression in 30% of recipients, leading to prolonged survival. Tumors developing in p50-/- mice possess a greater concentration of tumor-infiltrating myeloid cells (TIMs) than those in wild-type mice. TIMs are predominantly F4/80hi macrophages which, along with tumor-associated microglia, express increased pro-inflammatory M1 and reduced immune-suppressive M2 markers. In p50-/- mice, total tumor CD4 T cells are threefold more abundant, whereas CD8 T-cell numbers are unchanged, and both produce increased IFNγ and Granzyme B. Naïve splenic p50-/- CD8 T cells manifest increased activation, whereas naïve p50-/- and WT CD4 T cells show similar Th1, Th2, and Th17 polarization. Antibody targeting CD4, but not CD8, fully obviates the p50-/- survival advantage. Combined CD4 and CD8 T-cell depletion reverses myeloid M2 polarization in wild-type hosts, without affecting myeloid M1 polarization in p50-/- hosts. Finally, gliomas grow similarly in p50(f/f) and p50(f/f);Lysozyme-Cre mice, the latter having reduced p50 specifically in myeloid cells and tumor microglia. Thus, high-grade glioma T cells play a key role in directing M2 polarization of tumor myeloid cells, and reducing NF-κB p50 in both tumor myeloid cells and T cells may contribute to glioma therapy.


Asunto(s)
Glioblastoma/prevención & control , Macrófagos/inmunología , Células Mieloides/inmunología , Subunidad p50 de NF-kappa B/fisiología , Linfocitos T/inmunología , Animales , Células Cultivadas , Glioblastoma/inmunología , Glioblastoma/mortalidad , Activación de Linfocitos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Tasa de Supervivencia , Linfocitos T/metabolismo
14.
Neuro Oncol ; 20(7): 954-965, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29294080

RESUMEN

Background: Targeted approaches for treating glioblastoma (GBM) attempted to date have consistently failed, highlighting the imperative for treatment strategies that operate on different mechanistic principles. Bioenergetics deprivation has emerged as an effective therapeutic approach for various tumors. We have previously found that cancer cells preferentially utilize cytosolic NADH supplied by aldehyde dehydrogenase (ALDH) for ATP production through oxidative phosphorylation (OxPhos). This study is aimed at examining therapeutic responses and underlying mechanisms of dual inhibition of ALDH and OxPhos against GBM. Methods: For inhibition of ALDH and OxPhos, the corresponding inhibitors, gossypol and phenformin were used. Biological functions, including ATP levels, stemness, invasiveness, and viability, were evaluated in GBM tumorspheres (TSs). Gene expression profiles were analyzed using microarray data. In vivo anticancer efficacy was examined in a mouse orthotopic xenograft model. Results: Combined treatment of GBM TSs with gossypol and phenformin significantly reduced ATP levels, stemness, invasiveness, and cell viability. Consistently, this therapy substantially decreased expression of genes associated with stemness, mesenchymal transition, and invasion in GBM TSs. Supplementation of ATP using malate abrogated these effects, whereas knockdown of ALDH1L1 mimicked them, suggesting that disruption of ALDH-mediated ATP production is a key mechanism of this therapeutic combination. In vivo efficacy confirmed remarkable therapeutic responses to combined treatment with gossypol and phenformin. Conclusion: Our findings suggest that dual inhibition of tumor bioenergetics is a novel and effective strategy for the treatment of GBM.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Neoplasias Encefálicas/prevención & control , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Metabolismo Energético/efectos de los fármacos , Glioblastoma/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Anticonceptivos Masculinos/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/patología , Gosipol/farmacología , Humanos , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenformina/farmacología , Pronóstico , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cell Mol Neurobiol ; 38(2): 559-573, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28600709

RESUMEN

Glioblastoma is one of the most malignant and aggressive types of brain tumors. 5-lipoxygenase and cysteinyl leukotriene receptor 1 (CysLT1) play a role in human carcinogenesis. Leukotriene receptor antagonists (LTRAs), anti-asthmatic drugs with mild side effects, have anti-metastatic activity in epidermoid carcinoma, lung carcinoma, and colon cancers as well as neuroprotective effects. Herein, anti-migratory effects of two LTRAs, montelukast and zafirlukast, were investigated in glioblastoma cells. The level of CysLT1 in A172 cells was increased by 3.13 folds after IL-1ß treatment. The median toxic concentration of LTRAs in A172, U373, and primary astrocytes ranged from 7.17 to 26.28 µM at 24-h post-exposure. Both LTRAs inhibited migration and invasion of glioma. Additionally, both drugs significantly inhibited the expression and activities of MMP-2 and MMP-9 in A172 and U373 glioblastoma cells and primary human astrocytes, suggesting that CysLT1 plays a role in migration and invasion of glioma, and LTRAs are potential drugs to reduce migration and invasion.


Asunto(s)
Neoplasias Encefálicas/enzimología , Movimiento Celular/fisiología , Glioblastoma/enzimología , Antagonistas de Leucotrieno/farmacología , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Acetatos/farmacología , Acetatos/uso terapéutico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ciclopropanos , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica , Glioblastoma/patología , Glioblastoma/prevención & control , Humanos , Antagonistas de Leucotrieno/uso terapéutico , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Invasividad Neoplásica/genética , Quinolinas/farmacología , Quinolinas/uso terapéutico , Receptores de Leucotrienos/metabolismo , Sulfuros
17.
JCI Insight ; 2(24)2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29263302

RESUMEN

Tumor microenvironments can promote stem cell maintenance, tumor growth, and therapeutic resistance, findings linked by the tumor-initiating cell hypothesis. Standard of care for glioblastoma (GBM) includes temozolomide chemotherapy, which is not curative, due, in part, to residual therapy-resistant brain tumor-initiating cells (BTICs). Temozolomide efficacy may be increased by targeting carbonic anhydrase 9 (CA9), a hypoxia-responsive gene important for maintaining the altered pH gradient of tumor cells. Using patient-derived GBM xenograft cells, we explored whether CA9 and CA12 inhibitor SLC-0111 could decrease GBM growth in combination with temozolomide or influence percentages of BTICs after chemotherapy. In multiple GBMs, SLC-0111 used concurrently with temozolomide reduced cell growth and induced cell cycle arrest via DNA damage in vitro. In addition, this treatment shifted tumor metabolism to a suppressed bioenergetic state in vivo. SLC-0111 also inhibited the enrichment of BTICs after temozolomide treatment determined via CD133 expression and neurosphere formation capacity. GBM xenografts treated with SLC-0111 in combination with temozolomide regressed significantly, and this effect was greater than that of temozolomide or SLC-0111 alone. We determined that SLC-0111 improves the efficacy of temozolomide to extend survival of GBM-bearing mice and should be explored as a treatment strategy in combination with current standard of care.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/prevención & control , Glioblastoma/prevención & control , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Daño del ADN , ADN de Neoplasias/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Temozolomida/administración & dosificación , Temozolomida/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Sci Rep ; 7(1): 5609, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28717188

RESUMEN

The R5 subfamily of receptor-type protein tyrosine phosphatases (RPTPs) comprises PTPRZ and PTPRG. A recent study on primary human glioblastomas suggested a close association between PTPRZ1 (human PTPRZ) expression and cancer stemness. However, the functional roles of PTPRZ activity in glioma stem cells have remained unclear. In the present study, we found that sphere-forming cells from the rat C6 and human U251 glioblastoma cell lines showed high expression levels of PTPRZ-B, the short receptor isoform of PTPRZ. Stable PTPRZ knockdown altered the expression levels of stem cell transcription factors such as SOX2, OLIG2, and POU3F2 and decreased the sphere-forming abilities of these cells. Suppressive effects on the cancer stem-like properties of the cells were also observed following the knockdown of PTPRG. Here, we identified NAZ2329, a cell-permeable small molecule that allosterically inhibits both PTPRZ and PTPRG. NAZ2329 reduced the expression of SOX2 in C6 and U251 cells and abrogated the sphere-forming abilities of these cells. Tumor growth in the C6 xenograft mouse model was significantly slower with the co-treatment of NAZ2329 with temozolomide, an alkylating agent, than with the individual treatments. These results indicate that pharmacological inhibition of R5 RPTPs is a promising strategy for the treatment of malignant gliomas.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glioblastoma/prevención & control , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Temozolomida/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Femenino , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Ratas , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
BMC Cancer ; 17(1): 126, 2017 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-28193190

RESUMEN

BACKGROUND: Antiangiogenic therapies are considered promising for the treatment of glioblastoma (GB). The non-collagenous C-terminal globular NC1 domain of type VIII collagen a1 chain, Vastatin, is an endogenous antiangiogenic polypeptide. Sustained enhanced expression of Vastatin was shown to inhibit tumour growth and metastasis in murine hepatocellular carcinoma models. In this study, we further explored the efficacy of Vastatin in the treatment of GB xenografts. METHOD: Treatment of Vastatin was carried out using a nanopolymer gene vector PEI600-CyD-Folate (H1). Antiangiogenic effect of Vastatin was tested in vitro by using co-culture system and conditioned medium. An orthotopic GB murine model was established to examine the in vivo therapeutic effect of Vastatin alone treatment and its combination with temozolomide. RESULTS: Vastatin gene transfection mediated by H1 could target tumour cells specifically and suppress the proliferation of microvessel endothelial cells (MECs) through a paracrine inhibition manner. Enhancing Vastatin expression by intracerebral injection of H1-Vastatin significantly prolonged animal survival from 48 to 75 days in GB murine model, which was comparable to the effect of Endostatin, the most studied endogenous antiangiogenic polypeptide. The diminished presence of CD34 positive cells in the GB xenografts suggested that Vastatin induced significant antiangiogenesis. Moreover, a synergistic effect in extending survival was detected when H1-Vastatin was administered with temozolomide (TMZ) in GB chemoresistant murine models. CONCLUSION: Our results suggest, for the first time, that Vastatin is an antiangiogenic polypeptide with significant potential therapeutic benefit for GB. H1-Vastatin gene therapy may have important implications in re-sensitizing recurrent GB to standard chemotherapeutic agents.


Asunto(s)
Neoplasias Encefálicas/mortalidad , Proliferación Celular , Colágeno Tipo VIII/metabolismo , Glioblastoma/mortalidad , Neovascularización Patológica/prevención & control , Animales , Apoptosis , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Colágeno Tipo VIII/genética , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/prevención & control , Humanos , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Neuro Oncol ; 19(6): 796-807, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115578

RESUMEN

Background: Adaptive immune resistance in the tumor microenvironment appears to attenuate the immunotherapeutic targeting of glioblastoma (GBM). In this study, we identified a tumor-infiltrating myeloid cell (TIM) population that expands in response to dendritic cell (DC) vaccine treatment. The aim of this study was to understand how this programmed death ligand 1 (PD-L1)-expressing population restricts activation and tumor-cytolytic function of vaccine-induced tumor-infiltrating lymphocytes (TILs). Methods: To test this hypothesis in our in vivo preclinical model, we treated mice bearing intracranial gliomas with DC vaccination ± murine anti-PD-1 monoclonal antibody (mAb) blockade or a colony stimulating factor 1 receptor inhibitor (CSF-1Ri) (PLX3397) and measured overall survival. We then harvested and characterized the PD-L1+ TIM population and its role in TIL activation and tumor cytolysis in vitro. Results: Our data indicated that the majority of PD-L1 expression in the GBM environment is contributed by TIMs rather than by tumor cells themselves. While PD-1 blockade partially reversed the TIL dysfunction, targeting TIMs directly with CSF-1Ri altered TIM expression of key chemotactic factors associated with promoting increased TIL infiltration after vaccination. Neither PD-1 mAb nor CSF-1Ri had a demonstrable therapeutic benefit alone, but when combined with DC vaccination, a significant survival benefit was observed. When the tripartite regimen was given (DC vaccine, PD-1 mAb, PLX3397), long-term survival was noted together with an increase in the number of TILs and TIL activation. Conclusion: Together, these studies elucidate the role that TIMs play in mediating adaptive immune resistance in the GBM microenvironment and provide evidence that they can be manipulated pharmacologically with agents that are clinically available. Development of immune resistance in response to active vaccination in GBM can be reversed with dual administration of CSF-1Ri and PD-1 mAb.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Vacunas contra el Cáncer/administración & dosificación , Glioblastoma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Células Mieloides/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...