Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Antioxidants (Basel) ; 11(6)2022 May 28.
Article in English | MEDLINE | ID: mdl-35739974

ABSTRACT

Flow (shear stress)-mediated dilation (FMD) of resistance arteries is a rapid endothelial response involved in tissue perfusion. FMD is reduced early in cardiovascular diseases, generating a major risk factor for atherosclerosis. As alteration of mitochondrial fusion reduces endothelial cells' (ECs) sprouting and angiogenesis, we investigated its role in ECs responses to flow. Opa1 silencing reduced ECs (HUVECs) migration and flow-mediated elongation. In isolated perfused resistance arteries, FMD was reduced in Opa1+/- mice, a model of the human disease due to Opa1 haplo-insufficiency, and in mice with an EC specific Opa1 knock-out (EC-Opa1). Reducing mitochondrial oxidative stress restored FMD in EC-Opa1 mice. In isolated perfused kidneys from EC-Opa1 mice, flow induced a greater pressure, less ATP, and more H2O2 production, compared to control mice. Opa1 expression and mitochondrial length were reduced in ECs submitted in vitro to disturbed flow and in vivo in the atheroprone zone of the mouse aortic cross. Aortic lipid deposition was greater in Ldlr-/--Opa1+/- and in Ldlr-/--EC-Opa1 mice than in control mice fed with a high-fat diet. In conclusion, we found that reduction in mitochondrial fusion in mouse ECs altered the dilator response to shear stress due to excessive superoxide production and induced greater atherosclerosis development.

2.
Drug Deliv ; 25(1): 1756-1765, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30338715

ABSTRACT

Anticancer agents that target both tumor cells and angiogenesis are of potential interest for glioblastoma (GB) therapy. One such agent is sorafenib (SFN), a tyrosine kinase inhibitor. However, poor aqueous solubility and undesirable side effects limit its clinical application, including local treatment. We encapsulated SFN in lipid nanocapsules (LNCs) to overcome these drawbacks. LNCs are nanocarriers formulated according to a solvent-free process, using only components that have received regulatory approval. SFN-LNCs had a diameter of 54 ± 1 nm, high encapsulation efficiency (>90%), and a drug payload of 2.11 ± 0.03 mg/g of LNC dispersion. They inhibited in vitro angiogenesis and decreased human U87MG GB cell viability similarly to free SFN. In vivo studies showed that the intratumoral administration of SFN-LNCs or free SFN in nude mice bearing an orthotopic U87MG human GB xenograft decreased the proportion of proliferating cells in the tumor relative to control groups. SFN-LNCs were more effective than free SFN for inducing early tumor vascular normalization, characterized by increases in tumor blood flow and decreases in tumor vessel area. These results highlight the potential of LNCs as delivery systems for SFN. The vascular normalization induced by SFN-LNCs could be used to improve the efficacy of chemotherapy or radiotherapy for treating GB.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioblastoma/drug therapy , Lipids , Nanocapsules , Sorafenib/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Compounding/methods , Humans , Lipids/chemistry , Mice , Mice, Nude , Nanocapsules/chemistry , Sorafenib/therapeutic use
3.
Biomater Sci ; 6(9): 2386-2409, 2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30023990

ABSTRACT

Glioblastoma is the most frequent and aggressive primary malignant tumor of the central nervous system with a gloomy prognosis. Platinum derivatives and one among them, cisplatin, exhibited promising results when locally administered into the brain of glioblastoma bearing rats. Nanovectorization of anticancer agents through polymeric nanoparticles may even promote drug accumulation within cells, thus concentrating the drug efficiently at its target. Anchorage of gadolinium complexes on the corona of such smart drug delivery systems could further allow magnetic resonance imaging (MRI) monitoring of the nanoplatform biodistribution in the damaged parenchyma and its therapeutic benefit. For this purpose, a biocompatible amphiphilic triblock copolymer, made of degradable polyester and polycarbonate and bioeliminable polyethylene oxide (PEO), was synthesized by successive ring-opening polymerizations. After micellization in water, gadolinium complexes were grafted onto the PEO micelle corona and the carboxylate functions, located at the surface of the micelle's core, were able to cross-link with Pt(ii) complexes. A macromolecular prodrug was therefore recovered in which more than one third of the carboxylate functions were linked to a platinum atom. By this strategy, stable cisplatin cross-linked nanoparticles were formulated with a mean size in the range of 100.63 ± 12.04 nm consistent with biological investigations. Relaxometry measurements both in water and in plasma at 7 T, 25 °C, confirmed the intrinsic potential of these hybrid nanoparticles as alternative MRI contrast agents with a substantial increase in the r2/r1 ratio by a factor of 3.3 and 2.7, respectively, compared to the conventional low molar mass Gd-DTPA. As a result, their infusion within the striatum of glioblastoma-bearing mice resulted in a hypersignal on T2-weighted MR images that persisted over time. Ultimately, the formulated prodrug exhibited up to 50-fold increased accumulation in human glioblastoma cell lines and up to 32-fold enhanced subsequent Pt-DNA adduct formation in comparison with free cisplatin, thus supporting the potential of this innovative bimodal tool for further applications.


Subject(s)
Antineoplastic Agents , Cisplatin , Gadolinium DTPA , Nanoparticles , Prodrugs , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/chemistry , DNA Adducts/metabolism , Drug Liberation , Female , Gadolinium DTPA/administration & dosage , Gadolinium DTPA/chemistry , Glioblastoma/drug therapy , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Platinum/metabolism , Polycarboxylate Cement/chemistry , Polyesters/administration & dosage , Polyesters/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry
4.
Theranostics ; 7(18): 4517-4536, 2017.
Article in English | MEDLINE | ID: mdl-29158842

ABSTRACT

PURPOSE: Gold standard beam radiation for glioblastoma (GBM) treatment is challenged by resistance phenomena occurring in cellular populations well prepared to survive or to repair damage caused by radiation. Among signals that have been linked with radio-resistance, the SDF1/CXCR4 axis, associated with cancer stem-like cell, may be an opportune target. To avoid the problem of systemic toxicity and blood-brain barrier crossing, the relevance and efficacy of an original system of local brain internal radiation therapy combining a radiopharmaceutical with an immuno-nanoparticle was investigated. EXPERIMENT DESIGN: The nanocarrier combined lipophilic thiobenzoate complexes of rhenium-188 loaded in the core of a lipid nanocapsule (LNC188Re) with a function-blocking antibody, 12G5 directed at the CXCR4, on its surface. The efficiency of 12G5-LNC188Re was investigated in an orthotopic and xenogenic GBM model of CXCR4-positive U87MG cells implanted in the striatum of Scid mice. RESULTS: We demonstrated that 12G5-LNC188Re single infusion treatment by convection-enhanced delivery resulted in a major clinical improvement in median survival that was accompanied by locoregional effects on tumor development including hypovascularization and stimulation of the recruitment of bone marrow derived CD11b- or CD68-positive cells as confirmed by immunohistochemistry analysis. Interestingly, thorough analysis by spectral imaging in a chimeric U87MG GBM model containing CXCR4-positive/red fluorescent protein (RFP)-positive- and CXCR4-negative/RFP-negative-GBM cells revealed greater confinement of DiD-labeled 12G5-LNCs than control IgG2a-LNCs in RFP compartments. Main conclusion: These findings on locoregional impact and targeting of disseminated cancer cells in tumor margins suggest that intracerebral active targeting of nanocarriers loaded with radiopharmaceuticals may have considerable benefits in clinical applications.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Nanoparticles/administration & dosage , Radioisotopes/administration & dosage , Radiopharmaceuticals/administration & dosage , Receptors, CXCR4/administration & dosage , Rhenium/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Brain/radiation effects , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Lipids/administration & dosage , Mice , Nanocapsules/administration & dosage , Neoplastic Stem Cells/radiation effects , Xenograft Model Antitumor Assays/methods
5.
J Neurooncol ; 131(1): 49-58, 2017 01.
Article in English | MEDLINE | ID: mdl-27783195

ABSTRACT

Internal radiation strategies hold great promise for glioblastoma (GB) therapy. We previously developed a nanovectorized radiotherapy that consists of lipid nanocapsules loaded with a lipophilic complex of Rhenium-188 (LNC188Re-SSS). This approach resulted in an 83 % cure rate in the 9L rat glioma model, showing great promise. The efficacy of LNC188Re-SSS treatment was optimized through the induction of a T-cell immune response in this model, as it is highly immunogenic. However, this is not representative of the human situation where T-cell suppression is usually encountered in GB patients. Thus, in this study, we investigated the efficacy of LNC188Re-SSS in a human GB model implanted in T-cell deficient nude mice. We also analyzed the distribution and tissue retention of LNC188Re-SSS. We observed that intratumoral infusion of LNCs by CED led to their complete distribution throughout the tumor and peritumoral space without leakage into the contralateral hemisphere except when large volumes were used. Seventy percent of the 188Re-SSS activity was present in the tumor region 24 h after LNC188Re-SSS injection and no toxicity was observed in the healthy brain. Double fractionated internal radiotherapy with LNC188Re-SSS triggered survival responses in the immunocompromised human GB model with a cure rate of 50 %, which was not observed with external radiotherapy. In conclusion, LNC188Re-SSS can induce long-term survival in an immunosuppressive environment, highlighting its potential for GB therapy.


Subject(s)
Brain Neoplasms/radiotherapy , Glioblastoma/radiotherapy , Nanocapsules/therapeutic use , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Rhenium/therapeutic use , Animals , Autoradiography , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Nanocapsules/administration & dosage , Radioisotopes/administration & dosage , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry , Rhenium/administration & dosage , Rhenium/pharmacokinetics , T-Lymphocytes/pathology , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Int J Nanomedicine ; 10: 1259-71, 2015.
Article in English | MEDLINE | ID: mdl-25709447

ABSTRACT

Recently developed drug delivery nanosystems, such as lipid nanocapsules (LNCs), hold great promise for the treatment of glioblastomas (GBs). In this study, we used a subpopulation of human mesenchymal stem cells, "marrow-isolated adult multilineage inducible" (MIAMI) cells, which have endogenous tumor-homing activity, to deliver LNCs containing an organometallic complex (ferrociphenol or Fc-diOH), in the orthotopic U87MG GB model. We determined the optimal dose of Fc-diOH-LNCs that can be carried by MIAMI cells and compared the efficacy of Fc-diOH-LNC-loaded MIAMI cells with that of the free-standing Fc-diOH-LNC system. We showed that MIAMI cells entrapped an optimal dose of about 20 pg Fc-diOH per cell, with no effect on cell viability or migration capacity. The survival of U87MG-bearing mice was longer after the intratumoral injection of Fc-diOH-LNC-loaded MIAMI cells than after the injection of Fc-diOH-LNCs alone. The greater effect of the Fc-diOH-LNC-loaded MIAMI cells may be accounted for by their peritumoral distribution and a longer residence time of the drug within the tumor. These results confirm the potential of combinations of stem cell therapy and nanotechnology to improve the local tissue distribution of anticancer drugs in GB.


Subject(s)
Antineoplastic Agents , Ferrous Compounds , Glioblastoma/therapy , Lipids , Mesenchymal Stem Cell Transplantation , Nanocapsules , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Ferrous Compounds/administration & dosage , Ferrous Compounds/chemistry , Ferrous Compounds/therapeutic use , Ferrous Compounds/toxicity , Humans , Lipids/administration & dosage , Lipids/chemistry , Lipids/therapeutic use , Lipids/toxicity , Mice , Nanocapsules/administration & dosage , Nanocapsules/chemistry , Nanocapsules/therapeutic use , Nanocapsules/toxicity , Xenograft Model Antitumor Assays
7.
J Neurooncol ; 122(1): 1-10, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25503303

ABSTRACT

Glioblastoma (GB) is a highly infiltrative tumor recurring within a few centimeters of the resection cavity in 85 % of cases, even in cases of complete tumor resection and adjuvant chemo/radiotherapy. We recently isolated GB-associated stromal cells (GASCs) from the GB peritumoral zone, with phenotypic and functional properties similar to those of the cancer-associated fibroblasts present in the stroma of carcinomas. In particular, GASCs promote blood vessel development and have tumor-promoting effects on glioma cells in vitro and in vivo. In this study, we characterized these cells further, by analyzing the transcriptome and methylome of 14 GASC and five control stromal cell preparations derived from non-GB peripheral brain tissues. We identified two subtypes of GASCs in surgical margins in GB patients: GASC-A and GASC-B. GASC-B promoted the development of tumors and endothelium, whereas GASC-A did not. A difference in DNA methylation may underlie these two subtypes. We identified various proteins as being produced in the procarcinogenic GASC-B. Some of these proteins may serve as prognostic factors for GB and/or targets for anti-glioma treatment. In conclusion, in this era of personalized therapy, the status of GASCs in GB-free surgical margins should be taken into account, to improve treatment and the prevention of recurrence.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Cell Transformation, Neoplastic/pathology , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells/pathology , Stromal Cells/pathology , Animals , Apoptosis , Biomarkers, Tumor , Blotting, Western , Brain/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , DNA Methylation , Female , Flow Cytometry , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Stromal Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Nanomedicine ; 10(8): 1667-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24842766

ABSTRACT

In this work, a novel ferrocenyl complex (ansa-FcdiOH) was assessed for brain tumor therapy through stealth lipid nanocapsules (LNCs). Stealth LNCs, prepared according to a one-step process, showed rapid uptake by cancer cells and extended blood circulation time. The ferrocenyl complex was successfully encapsulated into these LNCs measuring 40 nm with a high loading capacity (6.4%). In vitro studies showed a potent anticancer effect of ansa-FcdiOH on 9L cells with a low IC50 value (0.1 µM) associated with an oxidative stress and a dose-dependent alteration of the cell cycle. Repeated intravenous injections of stealth ansa-FcdiOH LNCs in ectopic glioma bearing rats induced a significant tumor growth inhibition, supported by a reduced number of proliferative cells in tumors compared to control group. Additionally, no liver damage was observed in treated animals. These results indicated that stealth ansa-FcdiOH LNCs might be considered as a potential new approach for cancer chemotherapy. FROM THE CLINICAL EDITOR: In this study, a novel ferrocenyl complex was assessed for brain tumor therapy through stealth lipid nanocapsules, demonstrating no liver damage, and superior tumor volume reduction compared to saline and stealth lipid nanocapsules alone in an ectopic glioma model.


Subject(s)
Ferrous Compounds/chemistry , Ferrous Compounds/therapeutic use , Glioma/drug therapy , Nanocapsules/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Drug Carriers/chemistry , Female , Nanomedicine , Rats , Rats, Inbred F344 , Reactive Oxygen Species
9.
J Pathol ; 233(1): 74-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24481573

ABSTRACT

Glioblastoma (GB) displays diffusely infiltrative growth patterns. Dispersive cells escape surgical resection and contribute to tumour recurrence within a few centimeters of the resection cavity in 90% of cases. We know that the non-neoplastic stromal compartment, in addition to infiltrative tumour cells, plays an active role in tumour recurrence. We isolated a new stromal cell population from the histologically normal surgical margins of GB by computer-guided stereotaxic biopsies and primary culture. These GB-associated stromal cells (GASCs) share phenotypic and functional properties with the cancer-associated fibroblasts (CAFs) described in the stroma of carcinomas. In particular, GASCs have tumour-promoting effects on glioma cells in vitro and in vivo. Here, we describe a quantitative proteomic analysis, using iTRAQ labelling and mass spectrometry, to compare GASCs with control stromal cells derived from non-GB peripheral brain tissues. A total of 1077 proteins were quantified and 67 proteins were found to differ between GASCs and control stromal cells. Several proteins changed in GASCs are related to a highly motile myofibroblast phenotype, and to wound healing and angiogenesis. The results for several selected proteins were validated by western blotting or flow cytometry. Furthermore, the effect of GASCs on angiogenesis was confirmed using the orthotopic U87MG glioma model. In conclusion, GASCs, isolated from GB histologically normal surgical margins and found mostly near blood vessels, could be a vascular niche constituent establishing a permissive environment, facilitating angiogenesis and possibly colonization of recurrence-initiating cells. We identify various proteins as being expressed in GASCs: some of these proteins may serve as prognostic factors for GB and/or targets for anti-glioma treatment.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Myofibroblasts/pathology , Neovascularization, Pathologic , Stromal Cells/pathology , Biomarkers, Tumor/metabolism , Biopsy , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/surgery , Cell Communication , Cell Separation , Chemokine CXCL12/metabolism , Coculture Techniques , Flow Cytometry , Glioblastoma/metabolism , Glioblastoma/surgery , Hepatocyte Growth Factor/metabolism , Humans , Myofibroblasts/metabolism , Neoplasm, Residual , Phenotype , Primary Cell Culture , Proteomics/methods , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stromal Cells/metabolism , Tandem Mass Spectrometry , Tumor Cells, Cultured , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...