Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Oncol ; 53(5): 1881-1896, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30132519

ABSTRACT

Glioblastoma, the most common primary brain tumour, is also considered one of the most lethal cancers per se. It is highly refractory to therapeutic intervention, as highlighted by the mean patient survival of only 15 months, despite an aggressive treatment approach, consisting of maximal safe surgical resection, followed by radio- and chemotherapy. Radiotherapy, in particular, can have effects on the surviving fractions of tumour cells, which are considered adverse to the desired clinical outcome: It can induce increased cellular proliferation, as well as enhanced invasion. In this study, we established that differentiated glioblastoma cells alter their DNA repair response following repeated exposure to radiation and, therefore, high single-dose irradiation (SD-IR) is not a good surrogate marker for fractionated dose irradiation (FD-IR), as used in clinical practice. Integrating irradiation into a combination therapy approach, we then investigated whether the pharmacological inhibition of PI3K signalling, the most abundantly activated survival cascade in glioblastoma, enhances the efficacy of radiotherapy. Of note, treatment with GDC-0941, which blocks PI3K-mediated signalling, did not enhance cell death upon irradiation, but both treatment modalities functioned synergistically to reduce the total cell number. Furthermore, GDC-0941 not only prevented the radiation-induced increase in the motility of the differentiated cells, but further reduced their speed below that of untreated cells. Therefore, combining radiotherapy with the pharmacological inhibition of PI3K signalling is a potentially promising approach for the treatment of glioblastoma, as it can reduce the unwanted effects on the surviving fraction of tumour cells.


Subject(s)
Antineoplastic Agents/pharmacology , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Indazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Sulfonamides/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , DNA Damage/radiation effects , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Enzyme Inhibitors/pharmacology , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Cells, Cultured
2.
Crit Rev Oncog ; 23(1-2): 119-138, 2018.
Article in English | MEDLINE | ID: mdl-29953371

ABSTRACT

The use of radiation is an essential part of both modern cancer diagnostic assessment and treatment. Next-generation imaging devices create 3D visualizations, allowing for better diagnoses and improved planning of precision treatment. This is particularly important for primary brain cancers such as diffuse intrinsic pontine glioma or the most common primary brain tumor, glioblastoma, because radiotherapy is often the only treatment modality that offers a significant improvement in survival and quality of life. In this review, we give an overview of the different imaging techniques and the historic role of radiotherapy and its place in modern cancer therapy. Finally, we discuss three key areas of risks associated with the use of ionizing radiation: (1) brain tumor induction mainly as a consequence of the diagnostic use of radiation; (2) cognitive decline as a consequence of treating childhood brain tumors as an example of long term consequences often neglected in favor of highlighting secondary primary cancers; and (3) pro-proliferative and pro-invasive alterations that occur in tumor cells that survive radiotherapy. Throughout the discussion, we highlight areas of potential future research.


Subject(s)
Brain Neoplasms/etiology , Diagnostic Imaging , Neoplasms, Second Primary/etiology , Radiotherapy , Apoptosis/radiation effects , Cell Survival/radiation effects , Diagnostic Imaging/adverse effects , Diagnostic Imaging/methods , Humans , Neoplasms/diagnosis , Neoplasms/radiotherapy , Radiation , Radiation Dosage , Radiotherapy/adverse effects , Radiotherapy/methods , Radiotherapy Dosage
3.
Oncogenesis ; 6(11): 398, 2017 Nov 29.
Article in English | MEDLINE | ID: mdl-29184057

ABSTRACT

The PI3K/Akt/mTOR signalling network is activated in almost 90% of all glioblastoma, the most common primary brain tumour, which is almost invariably lethal within 15 months of diagnosis. Despite intensive research, modulation of this signalling cascade has so far yielded little therapeutic benefit, suggesting that the role of the PI3K network as a pro-survival factor in glioblastoma and therefore a potential target in combination therapy should be re-evaluated. Therefore, we used two distinct pharmacological inhibitors that block signalling at different points of the cascade, namely, GDC-0941 (Pictilisib), a direct inhibitor of the near apical PI3K, and Rapamycin which blocks the side arm of the network that is regulated by mTOR complex 1. While both substances, at concentrations where they inhibit their primary target, have similar effects on proliferation and sensitisation for temozolomide-induced apoptosis, GDC-0941 appears to have a stronger effect on cellular motility than Rapamycin. In vivo GDC-0941 effectively retards growth of orthotopic transplanted human tumours in murine brains and significantly prolongs mouse survival. However, when looking at genetically identical cell populations that are in alternative states of differentiation, i.e. stem cell-like cells and their differentiated progeny, a more complex picture regarding the PI3K/Akt/mTOR pathway emerges. The pathway is differently regulated in the alternative cell populations and, while it contributes to the increased chemo-resistance of stem cell-like cells compared to differentiated cells, it only contributes to the motility of the latter. Our findings are the first to suggest that within a glioblastoma tumour the PI3K network can have distinct, cell-specific functions. These have to be carefully considered when incorporating inhibition of PI3K-mediated signals into complex combination therapies.

4.
Eur J Immunol ; 45(12): 3441-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26361887

ABSTRACT

The transcriptional co-activator BOB.1/OBF.1 is crucial for Octamer-driven transcription in B cells. BOB.1/OBF.1-deficiency leads to tremendous defects in B-cell development and function. Therefore, in the past research focused on the identification of BOB.1/OBF.1 target genes. However, the regulation of BOB.1/OBF.1 expression itself is poorly understood. Here we show that in B cells NF-κB as well as to some extent NFAT proteins are involved in the activation of basal as well as inducible BOB.1/OBF.1 expression by direct binding to the BOB.1/OBF.1 promoter. Moreover, the analysis of different inducers of NF-κB, like several TLR ligands, TNF-α, BAFF, or LTα1ß2, revealed that both the canonical and noncanonical NF-κB pathways are involved in the induction of BOB.1/OBF.1 gene. The identification of so far unknown inducers that regulate BOB.1/OBF.1 expression in B cells provides novel insights in the potential function of BOB.1/OBF.1 during different aspects of B-cell development and function.


Subject(s)
B-Lymphocytes/metabolism , NF-kappa B/physiology , Organic Cation Transport Proteins/genetics , Signal Transduction/physiology , Trans-Activators/genetics , Animals , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Organic Cation Transporter 2 , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...