Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
NMR Biomed ; 36(3): e4858, 2023 03.
Article in English | MEDLINE | ID: mdl-36285719

ABSTRACT

Acute ischemic stroke results in an ischemic core surrounded by a tissue at risk, named the penumbra, which is potentially salvageable. One way to differentiate the tissues is to measure the hypoxia status. The purpose of the current study is to correlate the abnormal brain tissue volume derived from magnetic resonance-based imaging of brain oxygen saturation (St O2 -MRI) to the fluorine-18 fluoromisonidazole ([18 F]FMISO) positron emission tomography (PET) volume for hypoxia imaging validation, and to analyze the ability of St O2 -MRI to depict the different hypoxic tissue types in the acute phase of stroke. In a pertinent model of stroke in the rat, the volume of tissue with decreased St O2 -MRI signal and that with increased uptake of [18 F]FMISO were equivalent and correlated (r = 0.706; p = 0.015). The values of St O2 in the tissue at risk were significantly greater than those quantified in the core of the lesion, and were less than those for healthy tissue (52.3% ± 2.0%; 43.3% ± 1.9%, and 67.9 ± 1.4%, respectively). A threshold value for St O2 of ≈60% as the cut-off for the identification of the tissue at risk was calculated. Tissue volumes with reduced St O2 -MRI correlated with the final lesion (r = 0.964, p < 0.0001). The findings show that the St O2 -MRI approach is sensitive for the detection of hypoxia and for the prediction of the final lesion after stroke. Once validated in acute clinical settings, this approach might be used to enhance the stratification of patients for potential therapeutic interventions.


Subject(s)
Ischemic Stroke , Stroke , Rats , Animals , Positron-Emission Tomography , Stroke/diagnostic imaging , Misonidazole , Hypoxia/diagnostic imaging , Magnetic Resonance Imaging , Radiopharmaceuticals
2.
Sci Rep ; 11(1): 11239, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045576

ABSTRACT

Lung cancer patients frequently develop brain metastases (BM). Despite aggressive treatment including neurosurgery and external-radiotherapy, overall survival remains poor. There is a pressing need to further characterize factors in the microenvironment of BM that may confer resistance to radiotherapy (RT), such as hypoxia. Here, hypoxia was first evaluated in 28 biopsies from patients with non­small cell lung cancer (NSCLC) BM, using CA-IX immunostaining. Hypoxia characterization (pimonidazole, CA-IX and HIF-1α) was also performed in different preclinical NSCLC BM models induced either by intracerebral injection of tumor cells (H2030-Br3M, H1915) into the cortex and striatum, or intracardial injection of tumor cells (H2030-Br3M). Additionally, [18F]-FMISO-PET and oxygen-saturation-mapping-MRI (SatO2-MRI) were carried out in the intracerebral BM models to further characterize tumor hypoxia and evaluate the potential of Hypoxia-image-guided-RT (HIGRT). The effect of RT on proliferation of BM ([18F]-FLT-PET), tumor volume and overall survival was determined. We showed that hypoxia is a major yet heterogeneous feature of BM from lung cancer both preclinically and clinically. HIGRT, based on hypoxia heterogeneity observed between cortical and striatal metastases in the intracerebrally induced models, showed significant potential for tumor control and animal survival. These results collectively highlight hypoxia as a hallmark of BM from lung cancer and the value of HIGRT in better controlling tumor growth.


Subject(s)
Brain Neoplasms/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Radiotherapy, Image-Guided , Tumor Hypoxia , Aged , Animals , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Magnetic Resonance Imaging , Middle Aged , Rats , Registries
3.
Neuro Oncol ; 22(3): 357-368, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31538194

ABSTRACT

BACKGROUND: Brain metastases (BM) develop frequently in patients with breast cancer. Despite the use of external beam radiotherapy (EBRT), the average overall survival is short (6 months from diagnosis). The therapeutic challenge is to deliver molecularly targeted therapy at an early stage when relatively few metastatic tumor cells have invaded the brain. Vascular cell adhesion molecule 1 (VCAM-1), overexpressed by nearby endothelial cells during the early stages of BM development, is a promising target. The aim of this study was to investigate the therapeutic value of targeted alpha-particle radiotherapy, combining lead-212 (212Pb) with an anti-VCAM-1 antibody (212Pb-αVCAM-1). METHODS: Human breast carcinoma cells that metastasize to the brain, MDA-231-Br-GFP, were injected into the left cardiac ventricle of nude mice. Twenty-one days after injection, 212Pb-αVCAM-1 uptake in early BM was determined in a biodistribution study and systemic/brain toxicity was evaluated. Therapeutic efficacy was assessed using MR imaging and histology. Overall survival after 212Pb-αVCAM-1 treatment was compared with that observed after standard EBRT. RESULTS: 212Pb-αVCAM-1 was taken up into early BM with a tumor/healthy brain dose deposition ratio of 6 (5.52e108 and 0.92e108) disintegrations per gram of BM and healthy tissue, respectively. MRI analyses showed a statistically significant reduction in metastatic burden after 212Pb-αVCAM-1 treatment compared with EBRT (P < 0.001), translating to an increase in overall survival of 29% at 40 days post prescription (P < 0.01). No major toxicity was observed. CONCLUSIONS: The present investigation demonstrates that 212Pb-αVCAM-1 specifically accumulates at sites of early BM causing tumor growth inhibition.


Subject(s)
Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Radiotherapy/methods , Vascular Cell Adhesion Molecule-1/immunology , Alpha Particles , Animals , Antibodies/administration & dosage , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Female , Humans , Lead Radioisotopes/administration & dosage , Mice , Mice, Nude
4.
EJNMMI Res ; 9(1): 114, 2019 Dec 19.
Article in English | MEDLINE | ID: mdl-31858290

ABSTRACT

BACKGROUND: Diacetyl-bis(N4-methylthiosemicarbazone), labeled with 64Cu (64Cu-ATSM) has been suggested as a promising tracer for imaging hypoxia. However, various controversial studies highlighted potential pitfalls that may disable its use as a selective hypoxic marker. They also highlighted that the results may be tumor location dependent. Here, we first analyzed uptake of Cu-ATSM and its less lipophilic counterpart Cu-Cl2 in the tumor over time in an orthotopic glioblastoma model. An in vitro study was also conducted to investigate the hypoxia-dependent copper uptake in tumor cells. We then further performed a comprehensive ex vivo study to compare 64Cu uptake to hypoxic markers, specific cellular reactions, and also transporter expression. METHODS: µPET was performed 14 days (18F-FMISO), 15 days (64Cu-ATSM and 64Cu-Cl2), and 16 days (64Cu-ATSM and 64Cu-Cl2) after C6 cell inoculation. Thereafter, the brains were withdrawn for further autoradiography and immunohistochemistry. C6 cells were also grown in hypoxic workstation to analyze cellular uptake of Cu complexes in different oxygen levels. RESULTS: In vivo results showed that Cu-ASTM and Cu-Cl2 accumulated in hypoxic areas of the tumors. Cu-ATSM also stained, to a lesser extent, non-hypoxic regions, such as regions of astrogliosis, with high expression of copper transporters and in particular DMT-1 and CTR1, and also characterized by the expression of elevated astrogliosis. In vitro results show that 64Cu-ATSM showed an increase in the uptake only in severe hypoxia at 0.5 and 0.2% of oxygen while for 64Cu-Cl2, the cell retention was significantly increased at 5% and 1% of oxygen with no significant rise at lower oxygen percentages. CONCLUSION: In the present study, we show that Cu-complexes undoubtedly accumulate in hypoxic areas of the tumors. This uptake may be the reflection of a direct dependency to a redox metabolism and also a reflection of hypoxic-induced overexpression of transporters. We also show that Cu-ATSM also stained non-hypoxic regions such as astrogliosis.

5.
Theranostics ; 8(21): 5814-5827, 2018.
Article in English | MEDLINE | ID: mdl-30613264

ABSTRACT

Alteration of the extracellular matrix (ECM) is one of the major events in the pathogenesis of brain lesions following ischemic stroke. Heparan sulfate mimetics (HSm) are synthetic pharmacologically active polysaccharides that promote ECM remodeling and tissue regeneration in various types of lesions. HSm bind to growth factors, protect them from enzymatic degradation and increase their bioavailability, which promotes tissue repair. As the ECM is altered during stroke and HSm have been shown to restore the ECM, we investigated the potential of HSm4131 (also named RGTA-4131®) to protect brain tissue and promote regeneration and plasticity after a stroke. Methods: Ischemic stroke was induced in rats using transient (1 h) intraluminal middle cerebral artery occlusion (MCAo). Animals were assigned to the treatment (HSm4131; 0.1, 0.5, 1.5, or 5 mg/kg) or vehicle control (saline) groups at different times (1, 2.5 or 6 h) after MCAo. Brain damage was assessed by MRI for the acute (2 days) and chronic (14 days) phases post-occlusion. Functional deficits were evaluated with a battery of sensorimotor behavioral tests. HSm4131-99mTc biodistribution in the ischemic brain was analyzed between 5 min and 3 h following middle cerebral artery reperfusion. Heparan sulfate distribution and cellular reactions, including angiogenesis and neurogenesis, were evaluated by immunohistochemistry, and growth factor gene expression (VEGF-A, Ang-2) was quantified by RT-PCR. Results: HSm4131, administered intravenously after stroke induction, located and remained in the ischemic hemisphere. HSm4131 conferred long-lasting neuroprotection, and significantly reduced functional deficits with no alteration of physiological parameters. It also restored the ECM, and increased brain plasticity processes, i.e., angiogenesis and neurogenesis, in the affected brain hemisphere. Conclusion: HSm represent a promising ECM-based therapeutic strategy to protect and repair the brain after a stroke and favor functional recovery.


Subject(s)
Brain Ischemia/therapy , Brain/pathology , Brain/physiology , Extracellular Matrix/metabolism , Glucans/administration & dosage , Heparitin Sulfate/administration & dosage , Stroke/therapy , Animals , Behavior, Animal , Brain/diagnostic imaging , Disease Models, Animal , Immunohistochemistry , Magnetic Resonance Imaging , Neovascularization, Physiologic , Neurogenesis , Rats , Real-Time Polymerase Chain Reaction , Regeneration , Treatment Outcome
6.
Oncotarget ; 8(42): 72597-72612, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-29069812

ABSTRACT

In some highly inflammatory tumors, such as glioblastoma (GB), macrophages (MΦ) represent the most abundant population of reactive cells. MΦ, initially denoted as M0 MΦ, can be polarized into two further phenotypes: the antitumor M1 MΦ, and the protumor M2 MΦ. The three phenotypes can reside simultaneously in the tumor mass and various external factors may influence MΦ polarization. Radiotherapy is a common modality of cancer treatment aiming to target tumor cells. However, the specific effects of X-ray radiation on the inflammatory cells are, so far, controversial and not fully understood. In the present investigation, we have first analyzed, in vivo, the effect of X-ray radiation on MΦ present in GB tumors. We have observed a decrease in MΦ number paralleled by an increase in the proportion of M2 MΦ. To understand this phenomenon, we then evaluated, in vitro, the effects of X-rays on the MΦ phenotypes and survival. We have found that X-ray radiation failed to modify the phenotype of the different MΦ. However, M1 MΦ were more sensitive to ionizing radiation than M2 MΦ, both in normoxia and in hypoxia, which could explain the in vivo observations. To conclude, M2 MΦ are more radioresistant than M0 and M1 MΦ and the present study allows us to propose that X-ray radiotherapy could contribute, along with other phenomena, to the increased density in the protumor M2 MΦ in GB.

7.
Sci Rep ; 7(1): 14000, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070788

ABSTRACT

In stroke patients, loss of skeletal muscle mass leads to prolonged weakness and less efficient rehabilitation. We previously showed that expression of myostatin, a master negative regulator of skeletal muscle mass, was strongly increased in skeletal muscle in a mouse model of stroke. We therefore tested the hypothesis that myostatin inhibition would improve recovery of skeletal muscle mass and function after cerebral ischemia. Cerebral ischemia (45 minutes) was induced by intraluminal right middle cerebral artery occlusion (MCAO). Swiss male mice were randomly assigned to Sham-operated mice (n = 10), MCAO mice receiving the vehicle (n = 15) and MCAO mice receiving an anti-myostatin PINTA745 (n = 12; subcutaneous injection of 7.5 mg.kg-1 PINTA745 immediately after surgery, 3, 7 and 10 days after MCAO). PINTA745 reduced body weight loss and improved body weight recovery after cerebral ischemia, as well as muscle strength and motor function. PINTA745 also increased muscle weight recovery 15 days after cerebral ischemia. Mechanistically, the better recovery of skeletal muscle mass in PINTA745-MCAO mice involved an increased expression of genes encoding myofibrillar proteins. Therefore, an anti-myostatin strategy can improve skeletal muscle recovery after cerebral ischemia and may thus represent an interesting strategy to combat skeletal muscle loss and weakness in stroke patients.


Subject(s)
Muscle Development/drug effects , Muscular Atrophy/drug therapy , Myostatin/antagonists & inhibitors , Peptide Fragments/pharmacology , Stroke/physiopathology , Animals , Disease Models, Animal , Male , Mice , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Stroke/complications
8.
J Cereb Blood Flow Metab ; 37(7): 2584-2597, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27702880

ABSTRACT

The partial pressure in oxygen remains challenging to map in the brain. Two main strategies exist to obtain surrogate measures of tissue oxygenation: the tissue saturation studied by magnetic resonance imaging (StO2-MRI) and the identification of hypoxia by a positron emission tomography (PET) biomarker with 3-[18F]fluoro-1-(2-nitro-1-imidazolyl)-2-propanol ([18F]-FMISO) as the leading radiopharmaceutical. Nonetheless, a formal validation of StO2-MRI against FMISO-PET has not been performed. The objective of our studies was to compare the two approaches in (a) the normal rat brain when the rats were submitted to hypoxemia; (b) animals implanted with four tumour types differentiated by their oxygenation. Rats were submitted to normoxic and hypoxemic conditions. For the brain tumour experiments, U87-MG, U251-MG, 9L and C6 glioma cells were orthotopically inoculated in rats. For both experiments, StO2-MRI and [18F]-FMISO PET were performed sequentially. Under hypoxemia conditions, StO2-MRI revealed a decrease in oxygen saturation in the brain. Nonetheless, [18F]-FMISO PET, pimonidazole immunohistochemistry and molecular biology were insensitive to hypoxia. Within the context of tumours, StO2-MRI was able to detect hypoxia in the hypoxic models, mimicking [18F]-FMISO PET with high sensitivity/specificity. Altogether, our data clearly support that, in brain pathologies, StO2-MRI could be a robust and specific imaging biomarker to assess hypoxia.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain/blood supply , Glioma/diagnostic imaging , Hypoxia, Brain/diagnostic imaging , Oxygen/blood , Animals , Brain/diagnostic imaging , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cerebrovascular Circulation/physiology , Glioma/metabolism , Glioma/pathology , Hypoxia, Brain/metabolism , Magnetic Resonance Imaging , Male , Neoplasm Transplantation , Positron-Emission Tomography , Rats, Inbred F344 , Rats, Nude , Rats, Wistar
9.
J Cereb Blood Flow Metab ; 37(6): 2270-2282, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27496553

ABSTRACT

The alleviation of hypoxia in glioblastoma with carbogen to improve treatment has met with limited success. Our hypothesis is that the eventual benefits of carbogen depend on the capacity for vasodilation. We examined, with MRI, changes in fractional cerebral blood volume, blood oxygen saturation, and blood oxygenation level dependent signals in response to carbogen. The analyses were performed in two xenograft models of glioma (U87 and U251) recognized to have different vascular patterns. Carbogen increased fractional cerebral blood volume, blood oxygen saturation, and blood oxygenation level dependent signals in contralateral tissues. In the tumor core and peritumoral regions, changes were dependent on the capacity to vasodilate rather than on resting fractional cerebral blood volume. In the highly vascularised U87 tumor, carbogen induced a greater increase in fractional cerebral blood volume and blood oxygen saturation in comparison to the less vascularized U251 tumor. The blood oxygenation level dependent signal revealed a delayed response in U251 tumors relative to the contralateral tissue. Additionally, we highlight the considerable heterogeneity of fractional cerebral blood volume, blood oxygen saturation, and blood oxygenation level dependent within U251 tumor in which multiple compartments co-exist (tumor core, rim and peritumoral regions). Finally, our study underlines the complexity of the flow/metabolism interactions in different models of glioblastoma. These irregularities should be taken into account in order to palliate intratumoral hypoxia in clinical trials.


Subject(s)
Brain Neoplasms/blood supply , Carbon Dioxide/pharmacology , Cerebrovascular Circulation/drug effects , Glioblastoma/blood supply , Magnetic Resonance Imaging/methods , Oxygen/blood , Animals , Brain Neoplasms/diagnostic imaging , Carbon Dioxide/administration & dosage , Glioblastoma/diagnostic imaging , Humans , Oxygen/administration & dosage , Oxygen/pharmacology , Rats, Nude , Xenograft Model Antitumor Assays
10.
Eur J Nucl Med Mol Imaging ; 43(4): 682-94, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26537287

ABSTRACT

PURPOSE: The primary objective of this study was to compare the ability of PET and MRI biomarkers to predict treatment efficacy in a preclinical model of recurrent glioblastoma multiforme. METHODS: MRI (anatomical, diffusion, vasculature and oxygenation) and PET ([(18)F]FDG and [(18)F]FLT) parameters were obtained 3 days after the end of treatment and compared with late tumour growth and survival. RESULTS: Early after tumour recurrence, no effect of treatment with temozolomide combined with bevacizumab was observed on tumour volume as assessed by T2-W MRI. At later times, the treatment decreased tumour volume and increased survival. Interestingly, at the earlier time, temozolomide + bevacizumab decreased [(18)F]FLT uptake, cerebral blood volume and oedema. [(18)F]FLT uptake, oedema and cerebral blood volume were correlated with overall survival but [(18)F]FLT uptake had the highest specificity and sensitivity for the early prediction of treatment efficacy. CONCLUSION: The present investigation in a preclinical model of glioblastoma recurrence underscores the importance of multimodal imaging in the assessment of oedema, tumour vascular status and cell proliferation. Finally, [(18)F]FLT holds the greatest promise for the early assessment of treatment efficacy. These findings may translate clinically in that individualized treatment for recurrent glioma could be prescribed for patients selected after PET/MRI examinations.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioblastoma/diagnostic imaging , Magnetic Resonance Imaging , Multimodal Imaging , Positron-Emission Tomography , Animals , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Cell Line, Tumor , Dideoxynucleosides , Glioblastoma/diagnosis , Glioblastoma/drug therapy , Humans , Male , Radiopharmaceuticals , Rats
11.
Stroke ; 46(6): 1673-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25953371

ABSTRACT

BACKGROUND AND PURPOSE: Loss of muscle mass and function is a severe complication in patients with stroke that contributes to promoting physical inactivity and disability. The deleterious consequences of skeletal muscle mass loss underline the necessity to identity the molecular mechanisms involved in skeletal muscle atrophy after cerebral ischemia. METHODS: Transient focal cerebral ischemia (60 minutes) was induced by occlusion of the right middle cerebral artery in C57BL/6J male mice. Skeletal muscles were removed 3 days later and analyzed for the regulation of critical determinants of muscle mass homeostasis (Akt/mammalian target of rapamycin pathway, myostatin-Smad2/3 and bone morphogenetic protein-Smad1/5/8 signaling pathways, ubiquitin-proteasome and autophagy-lysosome proteolytic pathways). RESULTS: Cerebral ischemia induced severe sensorimotor deficits associated with muscle mass loss of the paretic limbs. Mechanistically, cerebral ischemia repressed Akt/mammalian target of rapamycin pathway and increased expression of key players of ubiquitin-proteasome pathway (MuRF1 [muscle RING finger-1], MAFbx [muscle atrophy F-box], Musa1 [muscle ubiquitin ligase of SCF complex in atrophy-1]), together with a marked increase in myostatin expression, in both paretic and nonparetic skeletal muscles. The Smad1/5/8 pathway was also activated. CONCLUSIONS: Our data fit with a model in which a repression of Akt/mammalian target of rapamycin pathway and an increase in the expression of key players of ubiquitin-proteasome pathway are critically involved in skeletal muscle atrophy after cerebral ischemia. Cerebral ischemia also caused an activation of bone morphogenetic protein-Smad1/5/8 signaling pathway, suggesting that compensatory mechanisms are also concomitantly activated to limit the extent of skeletal muscle atrophy.


Subject(s)
Brain Ischemia/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Signal Transduction , Animals , Brain Ischemia/complications , Brain Ischemia/pathology , Disease Models, Animal , Male , Mice , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/pathology
12.
Acta Biomater ; 15: 77-88, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25556361

ABSTRACT

Few effective therapeutic interventions are available to limit brain damage and functional deficits after ischaemic stroke. Within this context, mesenchymal stem cell (MSC) therapy carries minimal risks while remaining efficacious through the secretion of trophic, protective, neurogenic and angiogenic factors. The limited survival rate of MSCs restricts their beneficial effects. The usefulness of a three-dimensional support, such as a pharmacologically active microcarrier (PAM), on the survival of MSCs during hypoxia has been shown in vitro, especially when the PAMs were loaded with vascular endothelial growth factor (VEGF). In the present study, the effect of MSCs attached to laminin-PAMs (LM-PAMs), releasing VEGF or not, was evaluated in vivo in a model of transient stroke. The parameters assessed were infarct volume, functional recovery and endogenous cellular reactions. LM-PAMs induced the expression of neuronal markers by MSCs both in vitro and in vivo. Moreover, the prolonged release of VEGF increased angiogenesis around the site of implantation of the LM-PAMs and facilitated the migration of immature neurons towards the ischaemic tissue. Nonetheless, MSCs/LM-PAMs-VEGF failed to improve sensorimotor functions. The use of LM-PAMs to convey MSCs and to deliver growth factors could be an effective strategy to repair the brain damage caused by a stroke.


Subject(s)
Brain Ischemia/complications , Drug Carriers/chemistry , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Stroke/drug therapy , Stroke/etiology , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Behavior, Animal , Blood Vessels/drug effects , Brain Ischemia/physiopathology , Disease Models, Animal , Doublecortin Domain Proteins , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Laminin/pharmacology , Magnetic Resonance Imaging , Male , Mesenchymal Stem Cells/drug effects , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects , Stroke/physiopathology , Treatment Outcome , Vascular Endothelial Growth Factor A/pharmacology
13.
Oncotarget ; 6(4): 2101-19, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25544764

ABSTRACT

Hypoxia-inducible genes may contribute to therapy resistance in glioblastoma (GBM), the most aggressive and hypoxic brain tumours. It has been recently reported that erythropoietin (EPO) and its receptor (EPOR) are involved in glioma growth. We now investigated whether EPOR signalling may modulate the efficacy of the GBM current treatment based on chemotherapy (temozolomide, TMZ) and radiotherapy (X-rays). Using RNA interference, we showed on glioma cell lines (U87 and U251) that EPOR silencing induces a G2/M cell cycle arrest, consistent with the slowdown of glioma growth induced by EPOR knock-down. In vivo, we also reported that EPOR silencing combined with TMZ treatment is more efficient to delay tumour recurrence and to prolong animal survival compared to TMZ alone. In vitro, we showed that EPOR silencing not only increases the sensitivity of glioma cells to TMZ as well as X-rays but also counteracts the hypoxia-induced chemo- and radioresistance. Silencing EPOR on glioma cells exposed to conventional treatments enhances senescence and induces a robust genomic instability that leads to caspase-dependent mitotic death by increasing the number of polyploid cells and cyclin B1 expression. Overall these data suggest that EPOR could be an attractive target to overcome therapeutic resistance toward ionising radiation or temozolomide.


Subject(s)
Brain Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioma/therapy , Receptors, Erythropoietin/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Chemoradiotherapy , Cyclin B1/metabolism , Dacarbazine/pharmacology , Glioma/genetics , Glioma/metabolism , Humans , Immunohistochemistry , Male , Mice, Nude , Mitosis/drug effects , Mitosis/radiation effects , Polyploidy , RNA Interference , RNAi Therapeutics , Receptors, Erythropoietin/genetics , Temozolomide , Treatment Outcome , Xenograft Model Antitumor Assays
14.
Biol Chem ; 394(4): 529-39, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23399636

ABSTRACT

Despite multiple advances in cancer therapies, patients with glioblastoma (GBM) still have a poor prognosis. Numerous glioma models are used not only for the development of innovative therapies but also to optimize conventional ones. Given the significance of hypoxia in drug and radiation resistance and that hypoxia is widely observed among GBM, the establishment of a reliable method to map hypoxia in preclinical human models may contribute to the discovery and translation of future and more targeted therapies. The aim of this study was to compare the hypoxic status of two commonly used human orthotopic glioma models (U87 and U251) developed in rats and studied by noninvasive hypoxia imaging with 3-[18F]fluoro-1-(2-nitro-1-imidazolyl)-2-propanol-micro-positron emission tomography ([18F]-FMISO-µPET). In parallel, because of the relationships between angiogenesis and hypoxia, we used magnetic resonance imaging (MRI), histology, and immunohistochemistry to characterize the tumoral vasculature. Although all tumors were detectable in T2-weighted MRI and 2-deoxy-2-[18F]fluoro-d-glucose-µPET, only the U251 model exhibited [18F]-FMISO uptake. Additionally, the U251 tumors were less densely vascularized than U87 tumors. Our study demonstrates the benefits of noninvasive imaging of hypoxia in preclinical models to define the most reliable one for translation of future therapies to clinic based on the importance of intratumoral oxygen tension for the efficacy of chemotherapy and radiotherapy.


Subject(s)
Glioma/pathology , Hypoxia/diagnosis , Misonidazole/analogs & derivatives , Positron-Emission Tomography/methods , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy
15.
Neuro Oncol ; 15(1): 41-56, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23115160

ABSTRACT

The individualized care of glioma patients ought to benefit from imaging biomarkers as precocious predictors of therapeutic efficacy. Contrast enhanced MRI and [(18)F]-fluorodeoxyglucose (FDG)-PET are routinely used in clinical settings; their ability to forecast the therapeutic response is controversial. The objectives of our preclinical study were to analyze sensitive µMRI and/or µPET imaging biomarkers to predict the efficacy of anti-angiogenic and/or chemotherapeutic regimens. Human U87 and U251 orthotopic glioma models were implanted in nude rats. Temozolomide and/or bevacizumab were administered. µMRI (anatomical, diffusion, and microrheological parameters) and µPET ([(18)F]-FDG and [(18)F]-fluoro-l-thymidine [FLT]-PET) studies were undertaken soon (t(1)) after treatment initiation compared with late anatomical µMRI evaluation of tumor volume (t(2)) and overall survival. In both models, FDG and FLT uptakes were attenuated at t(1) in response to temozolomide alone or with bevacizumab. The distribution of FLT, reflecting intratumoral heterogeneity, was also modified. FDG was less predictive for treatment efficacy than was FLT (also highly correlated with outcome, P < .001 for both models). Cerebral blood volume was significantly decreased by temozolomide + bevacizumab and was correlated with survival for rats with U87 implants. While FLT was highly predictive of treatment efficacy, a combination of imaging biomarkers was superior to any one alone (P < .0001 in both tumors with outcome). Our results indicate that FLT is a sensitive predictor of treatment efficacy and that predictability is enhanced by a combination of imaging biomarkers. These findings may translate clinically in that individualized glioma treatments could be decided in given patients after PET/MRI examinations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/diagnostic imaging , Fluorodeoxyglucose F18 , Glioblastoma/diagnostic imaging , Magnetic Resonance Imaging , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Bevacizumab , Biomarkers/analysis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Immunoenzyme Techniques , Rats , Rats, Nude , Survival Rate , Temozolomide , Treatment Outcome , Tumor Burden , Xenograft Model Antitumor Assays
16.
J Cereb Blood Flow Metab ; 33(3): 389-95, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23211963

ABSTRACT

Most forms of cerebral ischemia are characterized by damage to the entire neurovascular unit, which leads to an increase in the permeability of the blood-brain barrier (BBB). In response to permanent focal cerebral ischemia in mice, we detected an early concomitant increase in the expression of the vascular endothelial growth factor (VEGF), a key inducer of vascular leakage and pathological blood vessel growth, and of angiopoietin-2 (Ang2), which is closely associated with VEGF in vascular remodeling. Thus, the aim of this study was to evaluate the role of Ang2 alone, or in combination with VEGF, in the acute phase of cerebral ischemia. The effect of these angiogenic factors on the ischemic lesion volume was evaluated by magnetic resonance imaging. We observed that timely administration of VEGF exacerbates ischemic damage. In contrast, Ang2 decreases the ischemic volume and this beneficial effect is maintained in the presence of VEGF. This investigation reports, for the first time, a protective role of Ang2 following cerebral ischemia, an action associated with a reduced BBB permeability. We propose that Ang2 represents a pertinent molecular target for the treatment of cerebral ischemia since acute brain damage may be limited by a pharmacological protection of the vascular compartment.


Subject(s)
Angiopoietin-2/biosynthesis , Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Animals , Blood-Brain Barrier/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Brain Ischemia/therapy , Capillary Permeability/drug effects , Gene Expression Regulation/drug effects , Magnetic Resonance Angiography , Mice , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
17.
Exp Neurol ; 233(1): 264-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22051176

ABSTRACT

Hypoxic-ischemic (HI) brain injury occurring during the perinatal period is still a major cause of mortality and morbidity. We assessed the impact of maternal hypertension, the most common medical disorder of pregnancy, on the anatomical and functional consequences of HI insult in the immature brain. Rat pups from spontaneously hypertensive (SHR) and normotensive (Wistar Kyoto - WKY) dams were subjected to HI brain damage at post-natal day 7 (P7). Brain lesion and functional deficits were analyzed from 10 min to 35 days after HI, using magnetic resonance imaging (MRI), sensorimotor and cognitive tests. MRI data revealed that SHR pups displayed less brain damage than WKY, attested by an initial smaller lesion followed by a reduced tissue loss at chronic stage (57.1±21.6 and 31.1±27% ipsilateral hemisphere atrophy in WKY and SHR, respectively). Behavioral analyses showed less HI-induced behavioral deficits in motor coordination (rotarod test) and spatial learning (Morris water maze test) in pups from hypertensive dams compared to those from normotensive ones. The data suggest that maternal hypertension causes prenatal stress that may render the immature brain more resistant to subsequent hypoxia-ischemia, related to a preconditioning phenomenon.


Subject(s)
Behavior, Animal/physiology , Hypertension/physiopathology , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/diagnosis , Ischemic Preconditioning , Maternal-Fetal Exchange/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Blood Pressure , Cognition Disorders/etiology , Disease Models, Animal , Female , Hand Strength/physiology , Hypertension/diagnosis , Magnetic Resonance Imaging , Male , Maze Learning , Motor Activity , Pregnancy , Psychomotor Performance/physiology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rotarod Performance Test , Spatial Behavior
18.
Nucl Med Biol ; 38(6): 781-93, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21843775

ABSTRACT

INTRODUCTION: No direct proof has been brought to light in a link between hypoxic changes in glioma models and the effects of antiangiogenic treatments. Here, we assessed the sensitivity of the detection of hypoxia through the use of (18)F-fluoromisonidazole positron emission tomography ([(18)F]-FMISO PET) in response to the evolution of the tumor and its vasculature. METHODS: Orthotopic glioma tumors were induced in rats after implantation of C6 or 9L cells. Sunitinib was administered from day (D) 17 to D24. At D17 and D24, multiparametric magnetic resonance imaging was performed to characterize tumor growth and vasculature. Hypoxia was assessed by [(18)F]-FMISO PET. RESULTS: We showed that brain hypoxic volumes are related to glioma volume and its vasculature and that an antiangiogenic treatment, leading to an increase in cerebral blood volume and a decrease in vessel permeability, is accompanied by a decrease in the degree of hypoxia. CONCLUSIONS: We propose that [(18)F]-FMISO PET and multiparametric magnetic resonance imaging are pertinent complementary tools in the evaluation of the effects of an antiangiogenic treatment in glioma.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Indoles/pharmacology , Magnetic Resonance Imaging , Misonidazole/analogs & derivatives , Positron-Emission Tomography , Pyrroles/pharmacology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Biological Transport , Brain/blood supply , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Hypoxia/drug effects , Cell Line, Tumor , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Glioma/blood supply , Glioma/diagnosis , Glioma/drug therapy , Glioma/pathology , Indoles/therapeutic use , Kinetics , Misonidazole/metabolism , Neovascularization, Pathologic/drug therapy , Permeability/drug effects , Pyrroles/therapeutic use , Rats , Sunitinib , Treatment Outcome
19.
Eur J Nucl Med Mol Imaging ; 37(12): 2371-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20814674

ABSTRACT

PURPOSE: Neuroinflammation is involved in stroke pathophysiology and might be imaged using radioligands targeting the 18 kDa translocator protein (TSPO). METHODS: We studied microglial reaction in brain areas remote from the primary lesion site in two rodent models of focal cerebral ischaemia (permanent or transient) using [125I]-CLINDE, a promising TSPO single photon emission computed tomography radioligand. RESULTS: In a mouse model of permanent middle cerebral artery occlusion (MCAO), ex vivo autoradiographic studies demonstrated, besides in the ischaemic territory, accumulation of [125I]-CLINDE in the ipsilateral thalamus with a binding that progressed up to 3 weeks after MCAO. [125I]-CLINDE binding markedly decreased in animals pre-injected with either unlabelled CLINDE or PK11195, while no change was observed with flumazenil pre-treatment, demonstrating TSPO specificity. In rats subjected to transient MCAO, [125I]-CLINDE binding in the ipsilateral thalamus and substantia nigra pars reticulata (SNr) was significantly higher than that in contralateral tissue. Moreover, [125I]-CLINDE binding in the thalamus and SNr was quantitatively correlated to the ischaemic volume assessed by MRI in the cortex and striatum, respectively. CONCLUSION: Clinical consequences of secondary neuronal degeneration in stroke might be better treated thanks to the discrimination of neuronal processes using in vivo molecular imaging and potent TSPO radioligands like CLINDE to guide therapeutic interventions.


Subject(s)
Brain Ischemia/diagnostic imaging , Brain Ischemia/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Microglia/diagnostic imaging , Microglia/metabolism , Receptors, GABA/metabolism , Animals , Disease Models, Animal , Humans , Male , Mice , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
20.
Nat Protoc ; 4(10): 1560-4, 2009.
Article in English | MEDLINE | ID: mdl-19798088

ABSTRACT

Long-term functional deficits after a brain injury are difficult to assess in the mouse. If no deficit is observed, researchers could conclude either that the animal has fully recovered or that the tests they used were not appropriate or sensitive enough to the modality of the deficits. We present here a detailed protocol describing how to conduct an adhesive removal test for this species. It consists of applying adhesive tape on each forepaw of the animal and measuring the time-to-contact and the time-to-remove them. This behavior implies correct paw and mouth sensitivity (time-to-contact) and correct dexterity (time-to-remove). To decrease interindividual differences, we recommend a training session (1 week, 1 trial per day) before surgical procedures so that mice to reach optimal performances.


Subject(s)
Behavior, Animal , Neuropsychological Tests , Surgical Tape , Animals , Brain Ischemia/physiopathology , Functional Laterality , Mice , Psychomotor Performance
SELECTION OF CITATIONS
SEARCH DETAIL
...