Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Fluids Barriers CNS ; 20(1): 67, 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37737197

ABSTRACT

BACKGROUND: Pharmacological treatment of CNS diseases is limited due to the presence of the blood-brain barrier (BBB). Recent years showed significant advancement in the field of CNS drug delivery enablers, with technologies such as MR-guided focused ultrasound reaching clinical trials. This have inspired researchers in the field to invent novel brain barriers opening (BBo) technologies that are required to be simple, fast, safe and efficient. One such technology, recently developed by us, is BDF (Barrier Disrupting Fields), based on low pulsed electric fields (L-PEFs) for opening the BBB in a controlled, safe, reversible and non-invasive manner. Here, we conducted an in vivo study to show that BDF is a feasible technology for delivering Doxorubicin (Doxo) into mice brain. Means for depicting BBBo levels were developed and applied for monitoring the treatment and predicting response. Overall, the goals of the presented study were to demonstrate the feasibility for delivering therapeutic Doxo doses into naïve and tumor-bearing mice brains and applying delayed-contrast MRI (DCM) for monitoring the levels of BBBo. METHODS: L-PEFs were applied using plate electrodes placed on the intact skull of naïve mice. L-PEFs/Sham mice were scanned immediately after the procedure by DCM ("MRI experiment"), or injected with Doxo and Trypan blue followed by delayed (4 h) perfusion and brain extraction ("Doxo experiment"). Doxo concentrations were measured in brain samples using confocal microscopy and compared to IC50 of Doxo in glioma cell lines in vitro. In order to map BBBo extent throughout the brain, pixel by pixel MR image analysis was performed using the DCM data. Finally, the efficacy of L-PEFs in combination with Doxo was tested in nude mice bearing intracranial human glioma tumors. RESULTS: Significant amount of Doxo was found in cortical regions of all L-PEFs-treated mice brains (0.50 ± 0.06 µg Doxo/gr brain) while in Sham brains, Doxo concentrations were below or on the verge of detection limit (0.03 ± 0.02 µg Doxo/gr brain). This concentration was x97 higher than IC50 of Doxo calculated in gl261 mouse glioma cells and x8 higher than IC50 of Doxo calculated in U87 human glioma cells. DCM analysis revealed significant BBBo levels in the cortical regions of L-PEFs-treated mice; the average volume of BBBo in the L-PEFs-treated mice was x29 higher than in the Sham group. The calculated BBBo levels dropped exponentially as a function of BBBo threshold, similarly to the electric fields distribution in the brain. Finally, combining non-invasive L-PEFs with Doxo significantly decreased brain tumors growth rates in nude mice. CONCLUSIONS: Our results demonstrate significant BBBo levels induced by extra-cranial L-PEFs, enabling efficient delivery of therapeutic Doxo doses into the brain and reducing tumor growth. As BBBo was undetectable by standard contrast-enhanced MRI, DCM was applied to generate maps depicting the BBBo levels throughout the brain. These findings suggest that BDF is a promising technology for efficient drug delivery into the brain with important implications for future treatment of brain cancer and additional CNS diseases.


Subject(s)
Brain Neoplasms , Glioma , Humans , Animals , Mice , Blood-Brain Barrier , Mice, Nude , Brain/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Glioma/diagnostic imaging , Glioma/drug therapy , Doxorubicin/pharmacology
2.
Sci Rep ; 12(1): 18078, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302937

ABSTRACT

Organophosphates (OPs) are inhibitors of acetylcholinesterase and have deleterious effects on the central nervous system. Clinical manifestations of OP poisoning include convulsions, which represent an underlying toxic neuro-pathological process, leading to permanent neuronal damage. This neurotoxicity is mediated through the cholinergic, GABAergic and glutamatergic (NMDA) systems. Pharmacological interventions in OP poisoning are designed to mitigate these specific neuro-pathological pathways, using anticholinergic drugs and GABAergic agents. Benactyzine is a combined anticholinergic, anti-NMDA compound. Based on previous development of novel GABA derivatives (such as prodrugs based on perphenazine for the treatment of schizophrenia and nortriptyline against neuropathic pain), we describe the synthesis and preliminary testing of a mutual prodrug ester of benactyzine and GABA. It is assumed that once the ester crosses the blood-brain-barrier it will undergo hydrolysis, releasing benactyzine and GABA, which are expected to act synergistically. The combined release of both compounds in the brain offers several advantages over the current OP poisoning treatment protocol: improved efficacy and safety profile (where the inhibitory properties of GABA are expected to counteract the anticholinergic cognitive adverse effects of benactyzine) and enhanced chemical stability compared to benactyzine alone. We present here preliminary results of animal studies, showing promising results with early gabactyzine administration.


Subject(s)
Chemical Warfare Agents , Organophosphate Poisoning , Prodrugs , Animals , Benactyzine , Antidotes/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use , Organophosphates , Acetylcholinesterase/metabolism , Cholinergic Antagonists/pharmacology , Esters , gamma-Aminobutyric Acid , Organophosphate Poisoning/drug therapy , Cholinesterase Inhibitors/pharmacology
3.
Isr Med Assoc J ; 23(10): 625-630, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34672443

ABSTRACT

BACKGROUND: Only a small proportion of schizophrenia patients present with catatonic symptoms. Imaging studies suggest that brain motor circuits are involved in the underlying pathology of catatonia. However, data about diffusivity dysregulation of these circuits in catatonic schizophrenia are scarce. OBJECTIVES: To assess the involvement of brain motor circuits in schizophrenia patients with catatonia. METHODS: Diffusion tensor imaging (DTI) was used to measure white matter signals in selected brain regions linked to motor circuits. Relevant DTI data of seven catatonic schizophrenia patients were compared to those of seven non-catatonic schizophrenia patients, matched for sex, age, and education level. RESULTS: Significantly elevated fractional anisotropy values were found in the splenium of the corpus callosum, the right peduncle of the cerebellum, and the right internal capsule of the schizophrenia patients with catatonia compared to those without catatonia. This finding showed altered diffusivity in selected motor-related brain areas. CONCLUSIONS: Catatonic schizophrenia is associated with dysregulation of the connectivity in specific motoric brain regions and corresponding circuits. Future DTI studies are needed to address the neural correlates of motor abnormalities in schizophrenia-related catatonia during the acute and remitted state of the illness to identify the specific pathophysiology of this disorder.


Subject(s)
Diffusion Tensor Imaging/methods , Motor Cortex , Schizophrenia, Catatonic , Adult , Anisotropy , Cerebellum/diagnostic imaging , Cerebellum/physiopathology , Connectome/methods , Corpus Callosum/diagnostic imaging , Corpus Callosum/physiopathology , Correlation of Data , Diagnostic and Statistical Manual of Mental Disorders , Female , Humans , Internal Capsule/diagnostic imaging , Internal Capsule/physiopathology , Male , Motor Cortex/diagnostic imaging , Motor Cortex/physiopathology , Psychiatric Status Rating Scales , Schizophrenia, Catatonic/diagnosis , Schizophrenia, Catatonic/physiopathology
4.
Pharmaceutics ; 13(2)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513968

ABSTRACT

The blood-brain barrier (BBB) is a major hurdle for the treatment of central nervous system disorders, limiting passage of both small and large therapeutic agents from the blood stream into the brain. Thus, means for inducing BBB disruption (BBBd) are urgently needed. Here, we studied the application of low pulsed electrical fields (PEFs) for inducing BBBd in mice. Mice were treated by low PEFs using electrodes pressed against both sides of the skull (100-400 square 50 µs pulses at 4 Hz with different voltages). BBBd as a function of treatment parameters was evaluated using MRI-based treatment response assessment maps (TRAMs) and Evans blue extravasation. A 3D numerical model of the mouse brain and electrodes was constructed using finite element software, simulating the electric fields distribution in the brain and ensuring no significant temperature elevation. BBBd was demonstrated immediately after treatment and significant linear regressions were found between treatment parameters and the extent of BBBd. The maximal induced electric field in the mice brains, calculated by the numerical model, ranged between 62.4 and 187.2 V/cm for the minimal and maximal applied voltages. These results demonstrate the feasibility of inducing significant BBBd using non-invasive low PEFs, well below the threshold for electroporation.

5.
Bioelectrochemistry ; 134: 107521, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32251984

ABSTRACT

When applying electroporation to the brain, it is important to understand the effects on the blood-brain barrier (BBB) and brain vasculature. Here we studied the effects of point-source electroporation on rats' brains as a function of time from treatment using conventional contrast-enhanced MRI and treatment response assessment maps (TRAMs), enabling depiction of subtle BBB disruption and differentiating contrast agent clearance from accumulation. Effects on vessels were also studied using Lectin staining. The TRAMs revealed that conventional contrast-enhanced MRI underestimates BBB disruption volume by nearly a factor of two, and that despite significant enhancement on standard MRI immediately post electroporation, there was no contrast accumulation in the tissue (clearance was faster than accumulation). Histology revealed significant increased vessel coverage in the treated striatum (40 ± 24% p < 0.03) immediately post electroporation, suggesting vasodilatation. Two-three hours post electroporation, both conventional MRI and TRAMs showed minor BBB disruption and histology showed decreased vessel coverage (56 ± 16%, p < 0.01), suggesting vasoconstriction. Four hours post electroporation, despite minor enhancement, the TRAMs showed significant BBB disruption with contrast accumulation, lasting over 24 h, with decreasing volumes. These results suggest that electroporation triggers several unique brain vascular mechanisms and that the optimal time window for drug administration is 4-6 h after electroporation.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/diagnostic imaging , Electroporation , Animals , Blood-Brain Barrier/cytology , Brain/cytology , Brain/metabolism , Magnetic Resonance Imaging , Male , Rats
6.
Sci Rep ; 10(1): 2178, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034261

ABSTRACT

The prognosis of Glioblastoma Multiforme patients is poor despite aggressive therapy. Reasons include poor chemotherapy penetration across the blood-brain barrier and tumor infiltration into surrounding tissues. Here we studied the effects of combined point-source electroporation (EP) and systemic chemotherapy in glioma-bearing rats. 128 rats were studied. Treatment groups were administered systemic Cisplatin/Methotrexate before EP (either 90 or 180 pulses). Control groups were treated by EP, chemotherapy, or no treatment. Tumor volumes were determined by MRI. Tumors growth rates of the EP + Methotrexate group (1.02 ± 0.77) were significantly lower (p < 0.01) than the control (5.2 ± 1.0) 1-week post treatment. No significant difference was found compared to Methotrexate (1.7 ± 0.5). Objective response rates (ORR) were 40% and 57% for the Methotrexate and EP + Methotrexate groups respectively. Tumor growth rates and ORR of the EP + Cisplatin groups (90 pulses 0.98 ± 0.2, 57%, 180 pulses 1.2 ± 0.1, 33%) were significantly smaller than the control (6.4 ± 1.0, p < 0.01, p < 0.02, 0%) and Cisplatin (3.9 ± 1.0, p < 0.04, p < 0.05, 13%) groups. No significant differences were found between the control groups. Increased survival was found in the EP + Cisplatin group, Χ2 = 7.54, p < 0.006 (Log Rank). Point-source EP with systemic chemotherapy is a rapid, minimal-invasive treatment that was found to induce significant antineoplastic effects in a rat glioma model.


Subject(s)
Brain Neoplasms/drug therapy , Electroporation/methods , Glioma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Electroporation/instrumentation , Male , Methotrexate/administration & dosage , Methotrexate/therapeutic use , Random Allocation , Rats , Rats, Inbred Lew
7.
Drug Deliv ; 26(1): 459-469, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30957567

ABSTRACT

The blood-brain barrier (BBB) is limiting transcellular and paracellular movement of molecules and cells, controls molecular traffic, and keeps out toxins. However, this protective function is the major hurdle for treating brain diseases such as brain tumors, Parkinson's disease, Alzheimer's disease, etc. It was previously demonstrated that high pulsed electrical fields (PEFs) can disrupt the BBB by inducing electroporation (EP) which increases the permeability of the transcellular route. Our goal was to study the effects of low PEFs, well below the threshold of EP on the integrity and function of the BBB. Ten low voltage pulses (5-100 V) were applied to a human in vitro BBB model. Changes in permeability to small molecules (NaF) were studied as well as changes in impedance spectrum and trans-endothelial electric resistivity. Viability and EP were evaluated by Presto-Blue and endogenous Lactate dehydrogenase release assays. The effect on tight junction and adherent junction protein was also studied. The results of low voltage experiments were compared to high voltage experiments (200-1400 V). A significant increase in permeability was found at voltages as low as 10 V despite EP only occurring from 100 V. The changes in permeability as a function of applied voltage were fitted to an inverse-exponential function, suggesting a plateau effect. Staining of VE-cadherin showed specific changes in protein expression. The results indicate that low PEFs can transiently disrupt the BBB by affecting the paracellular route, although the mechanism remains unclear.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Electroporation/methods , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Models, Biological , Pericytes/metabolism , Animals , Cattle , Cells, Cultured , Coculture Techniques , Electric Impedance , Humans
8.
Brain Stimul ; 12(1): 1-8, 2019.
Article in English | MEDLINE | ID: mdl-30181107

ABSTRACT

BACKGROUND: Essential tremor (ET) is one of the most common movement disorders of adults, characterized by postural and kinetic tremor. With drug treatment only partially efficient, new treatments are being developed. OBJECTIVES: The goal of this study was to demonstrate the feasibility of non-thermal focused-ultrasound (FUS) to induce tremor-suppression in an ET rat model. METHODS: Harmaline-induced tremor rats were treated with FUS along the inferior olivary (IO) system. EMG was recorded continuously during treatment in order to quantify FUS-induced tremor suppression. T2-weighted MRI was performed immediately following treatment and periodically thereafter. RESULTS: FUS treatment at an intensity of 27.2 W/cm2 (Isppa) induced significant reduction of tremor in 12 out of 13 ET rats. Tremor frequency was reduced from 6.2 ±â€¯2.8 to 2 ±â€¯1 Hz, p < 0.0003. In 6 of the 12 responding rats, tremor was completely suppressed. Response duration was 70 ±â€¯61s, on average. FUS induced motor response, depicted as movement of the tail and/or the limbs synchronized with the FUS sonication, was also demonstrated both in ET rats and in naïve rats when treated in the medulla oblongata region. CONCLUSIONS: These results demonstrate the feasibly for obtaining significant tremor reduction or tremor suppression induced by non-thermal, non-invasive, reversible focused-ultrasound.


Subject(s)
Essential Tremor/therapy , Ultrasonic Therapy/methods , Animals , Essential Tremor/etiology , Harmaline/toxicity , Male , Rats
9.
Clin Transl Radiat Oncol ; 15: 1-6, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30547098

ABSTRACT

•Of 310 brain tumors patients recruited, histology of 99 lesions was available.•Of those, 5 were histologically confirmed as radiation-induced malformations.•TRAMs cannot differentiate active tumor from vascular malformation.

10.
Ultrasound Med Biol ; 44(5): 1022-1030, 2018 05.
Article in English | MEDLINE | ID: mdl-29501283

ABSTRACT

The goal of this study was to determine the feasibility of focused ultrasound-based neuromodulation affecting auditory evoked potentials (AEPs) in animals. Focused ultrasound-induced suppression of AEPs was performed in 22 rats and 5 pigs: Repetitive sounds were produced, and the induced AEPs were recorded before and repeatedly after FUS treatment of the auditory pathway. All treated animals exhibited a decrease in AEP amplitude post-treatment in contrast to animals undergoing the sham treatment. Suppression was weaker for rats treated at 2.3 W/cm2 (amplitudes decreased to 59.8 ± 3.3% of baseline) than rats treated at 4.6 W/cm2 (36.9 ± 7.5%, p <0.001). Amplitudes of the treated pigs decreased to 27.7 ± 5.9% of baseline. This effect lasted between 30 min and 1 mo in most treated animals. No evidence of heating during treatment or later brain damage/edema was observed. These results demonstrate the feasibility of inducing significant neuromodulation with non-thermal, non-invasive, reversible focused ultrasound. The long recovery times may have clinical implications.


Subject(s)
Auditory Pathways/physiopathology , Evoked Potentials, Auditory , Ultrasonic Waves , Acoustic Stimulation , Animals , Feasibility Studies , Female , Male , Models, Animal , Rats , Rats, Sprague-Dawley , Swine
11.
Front Neurol ; 9: 1087, 2018.
Article in English | MEDLINE | ID: mdl-30619047

ABSTRACT

Data from human biopsies, in-vitro and in-vivo models, strongly supports the role of thrombin, and its protease-activated receptor (PAR1) in the pathology and progression of glioblastoma (GBM), a high-grade glial tumor. Activation of PAR1 by thrombin stimulates vasogenic edema, tumor adhesion and tumor growth. We here present a novel six amino acid chloromethyl-ketone compound (SIXAC) which specifically inhibits PAR1 proteolytic activation and counteracts the over-activation of PAR1 by tumor generated thrombin. SIXAC effects were demonstrated in-vitro utilizing 3 cell-lines, including the highly malignant CNS-1 cell-line which was also used as a model for GBM in-vivo. The in-vitro effects of SIXAC on proliferation rate, invasion and thrombin activity were measured by XTT, wound healing, colony formation and fluorescent assays, respectively. The effect of SIXAC on GBM in-vivo was assessed by measuring tumor and edema size as quantified by MRI imaging, by survival follow-up and brain histopathology. SIXAC was found in-vitro to inhibit thrombin-activity generated by CNS-1 cells (IC50 = 5 × 10-11M) and significantly decrease proliferation rate (p < 0.03) invasion (p = 0.02) and colony formation (p = 0.03) of these cells. In the CNS-1 GBM rat animal model SIXAC was found to reduce edema volume ratio (8.8 ± 1.9 vs. 4.9 ± 1, p < 0.04) and increase median survival (16 vs. 18.5 days, p < 0.02 by Log rank Mental-Cox test). These results strengthen the important role of thrombin/PAR1 pathway in glioblastoma progression and suggest SIXAC as a novel therapeutic tool for this fatal disease.

12.
Oncotarget ; 7(46): 74860-74871, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27556862

ABSTRACT

Temozolomide (TMZ) is an alkylating agent that has become the mainstay treatment of the most malignant brain cancer, glioblastoma multiforme (GBM). Unfortunately only a limited number of patients positively respond to it. It has been shown that zinc metal reestablishes chemosensitivity but this effect has not been tested with TMZ. Using both in vitro and in vivo experimental approaches, we investigated whether addition of zinc to TMZ enhances its cytotoxicity against GBM. In vitro cell viability analysis showed that the cytotoxic activity of TMZ was substantially increased with addition of zinc and this response was accompanied by an elevation of p21, PUMA, BAX and Caspase-3 expression and a decrease in growth fraction as manifested by low ki67 and lower colony formation. Analysis of GBM as intracranial xenografts in athymic mice and administration of concurrent TMZ and zinc yielded results consistent with those of the in vitro analyses. The co-treatment resulted in significant reduction in tumor volume in TMZ/zinc treated mice relative to treatment with TMZ alone. Our results suggest that zinc may serve as a potentiator of TMZ therapy in GBM patients.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Glioblastoma/pathology , Zinc/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Caspase 3/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Dacarbazine/pharmacology , Disease Models, Animal , Drug Synergism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Temozolomide , Tumor Burden , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Radiol Oncol ; 50(1): 28-38, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-27069447

ABSTRACT

BACKGROUND: Electroporation-based therapies such as electrochemotherapy (ECT) and irreversible electroporation (IRE) are emerging as promising tools for treatment of tumors. When applied to the brain, electroporation can also induce transient blood-brain-barrier (BBB) disruption in volumes extending beyond IRE, thus enabling efficient drug penetration. The main objective of this study was to develop a statistical model predicting cell death and BBB disruption induced by electroporation. This model can be used for individual treatment planning. MATERIAL AND METHODS: Cell death and BBB disruption models were developed based on the Peleg-Fermi model in combination with numerical models of the electric field. The model calculates the electric field thresholds for cell kill and BBB disruption and describes the dependence on the number of treatment pulses. The model was validated using in vivo experimental data consisting of rats brains MRIs post electroporation treatments. RESULTS: Linear regression analysis confirmed that the model described the IRE and BBB disruption volumes as a function of treatment pulses number (r(2) = 0.79; p < 0.008, r(2) = 0.91; p < 0.001). The results presented a strong plateau effect as the pulse number increased. The ratio between complete cell death and no cell death thresholds was relatively narrow (between 0.88-0.91) even for small numbers of pulses and depended weakly on the number of pulses. For BBB disruption, the ratio increased with the number of pulses. BBB disruption radii were on average 67% ± 11% larger than IRE volumes. CONCLUSIONS: The statistical model can be used to describe the dependence of treatment-effects on the number of pulses independent of the experimental setup.

14.
J Cereb Blood Flow Metab ; 35(6): 967-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25669901

ABSTRACT

Despite aggressive therapy, existing treatments offer poor prognosis for glioblastoma multiforme patients, in part due to poor penetration of most drugs across the blood-brain barrier (BBB). We propose a minimal-invasive combined treatment approach consisting of local BBB disruption in the tumor in parallel to systemic drug administration. Local BBB disruption is obtained by convection-enhanced delivery of a novel BBB disruption agent, enabling efficient/targeted delivery of the systemically administered drug by the tumors own vasculature. Various human serum albumin (HSA) analogs were synthesized and screened for BBB disruption efficacy in custom in vitro systems. The candidate analogs were then delivered into naïve rat brains by convection-enhanced delivery and screened for maximal BBB disruption and minimal brain toxicity. These studies found a noncationized/neutralized analog, ethylamine (EA)-HSA, to be the optimal BBB-opening agent. Immunocytochemical studies suggested that BBB disruption by EA-HSA may be explained by alterations in occludin expression. Finally, an efficacy study in rats bearing intracranial gliomas was performed. The rats were treated by convection-enhanced delivery of EA-HSA in parallel to systemic administration of Methotrexate, showing significant antineoplastic effects of the combined approached reflected in suppressed tumor growth and significantly (~x3) prolonged survival.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Brain Neoplasms/drug therapy , Brain/pathology , Drug Delivery Systems/methods , Glioma/drug therapy , Methotrexate/administration & dosage , Animals , Antimetabolites, Antineoplastic/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/drug effects , Brain Neoplasms/pathology , Cell Line , Convection , Ethylamines/adverse effects , Ethylamines/chemical synthesis , Ethylamines/chemistry , Glioma/pathology , Humans , Male , Methotrexate/therapeutic use , Rats , Rats, Inbred Lew , Serum Albumin/adverse effects , Serum Albumin/chemical synthesis , Serum Albumin/chemistry , Swine
15.
J Mol Neurosci ; 56(3): 644-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25691153

ABSTRACT

High-grade gliomas constitute a group of aggressive CNS cancers that have high morbidity and mortality rates. Despite extensive research, current therapeutic approaches enable survival beyond 2 years in rare cases only. Thrombin and its main CNS target, protease-activated receptor-1, have been implicated in tumor progression and brain edema. Our aim was to study protease-activated receptor-1 (PAR-1) protein expression and thrombin-like activity levels in both in vitro and in vivo models of glioblastoma and correlate them with the volume of the surrounding edema. We measured the presence of PAR-1 protein using fluorescence immunohistochemistry and assessed thrombin activity in various glial and non-glial cell lines and in a CNS-1 glioma rat model using a thrombin-specific fluorescent assay. Thrombin activity was found to be highly elevated in various high-grade glioma cell lines as well as in non-glial malignant cell lines. In the CNS-1 glioma model, the level of PAR-1 fluorescence in the tumor was significantly elevated compared to adjacent regions of reactive gliosis or distant brain areas. The elevated level of thrombin activity observed in the high-grade glioma positively correlated with tumor-induced brain edema. In conclusion, thrombin is secreted from glioma cells and PAR-1 may be a new biological marker for high-grade gliomas.


Subject(s)
Biomarkers, Tumor/metabolism , Brain/metabolism , Glioblastoma/metabolism , Receptor, PAR-1/metabolism , Thrombin/metabolism , Animals , Brain/pathology , Cell Line, Tumor , Glioblastoma/pathology , Male , Neuroglia/metabolism , Rats , Rats, Inbred Lew
16.
Neuro Oncol ; 17(3): 457-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25452395

ABSTRACT

BACKGROUND: Conventional magnetic resonance imaging (MRI) is unable to differentiate tumor/nontumor enhancing tissues. We have applied delayed-contrast MRI for calculating high resolution treatment response assessment maps (TRAMs) clearly differentiating tumor/nontumor tissues in brain tumor patients. METHODS: One hundred and fifty patients with primary/metastatic tumors were recruited and scanned by delayed-contrast MRI and perfusion MRI. Of those, 47 patients underwent resection during their participation in the study. Region of interest/threshold analysis was performed on the TRAMs and on relative cerebral blood volume maps, and correlation with histology was studied. Relative cerebral blood volume was also assessed by the study neuroradiologist. RESULTS: Histological validation confirmed that regions of contrast agent clearance in the TRAMs >1 h post contrast injection represent active tumor, while regions of contrast accumulation represent nontumor tissues with 100% sensitivity and 92% positive predictive value to active tumor. Significant correlation was found between tumor burden in the TRAMs and histology in a subgroup of lesions resected en bloc (r(2) = 0.90, P < .0001). Relative cerebral blood volume yielded sensitivity/positive predictive values of 51%/96% and there was no correlation with tumor burden. The feasibility of applying the TRAMs for differentiating progression from treatment effects, depicting tumor within hemorrhages, and detecting residual tumor postsurgery is demonstrated. CONCLUSIONS: The TRAMs present a novel model-independent approach providing efficient separation between tumor/nontumor tissues by adding a short MRI scan >1 h post contrast injection. The methodology uses robust acquisition sequences, providing high resolution and easy to interpret maps with minimal sensitivity to susceptibility artifacts. The presented results provide histological validation of the TRAMs and demonstrate their potential contribution to the management of brain tumor patients.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Contrast Media , Magnetic Resonance Imaging/methods , Adolescent , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Angiography/methods , Male , Middle Aged , Neoplasm, Residual/pathology , Time Factors , Young Adult
17.
Bioelectrochemistry ; 99: 30-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24976141

ABSTRACT

In spite of aggressive therapy, existing treatments offer poor prognosis for glioblastoma multiforme due to tumor infiltration into the surrounding brain as well as poor blood-brain barrier penetration of most therapeutic agents. In this paper we present a novel approach for a minimally invasive treatment and a non-invasive response assessment methodology consisting of applying intracranial point-source electroporation and assessing treatment effect volumes using magnetic resonance imaging. Using a unique setup of a single intracranial electrode and an external surface electrode we treated rats' brains with various electroporation protocols and applied magnetic resonance imaging to study the dependence of the physiological effects on electroporation treatment parameters. The extent of blood-brain barrier disruption and later volumes of permanent brain tissue damage were found to correlate significantly with the treatment voltages (r(2)=0.99, p<0.001) and the number of treatment pulses (r(2)=0.94, p<0.002). Blood-brain barrier disruption depicted 3.2±0.3 times larger volumes than the final permanent damage volumes (p<0.0001). These results indicate that it may be beneficial to use more than one modality of electroporation when planning a treatment for brain tumors.


Subject(s)
Brain Neoplasms/therapy , Brain/pathology , Electroporation/methods , Glioblastoma/therapy , Animals , Blood-Brain Barrier/pathology , Brain Neoplasms/pathology , Electroporation/instrumentation , Glioblastoma/pathology , Magnetic Resonance Imaging , Male , Rats , Rats, Sprague-Dawley
18.
Sci Rep ; 3: 3088, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24169528

ABSTRACT

To generate an understanding of the physiological significance of MR images of Non-Thermal Irreversible Electroporation (NTIRE) we compared the following MR imaging sequences: T1W, T2W, PD, GE, and T2 SPAIR acquired after NTIRE treatment in a rodent liver model. The parameters that were studied included the presence or absence of a Gd-based contrast agent, and in vivo and ex-vivo NTIRE treatments in the same liver. NTIRE is a new minimally invasive tissue ablation modality in which pulsed electric fields cause molecularly selective cell death while, the extracellular matrix and large blood vessels remain patent. This attribute of NTIRE is of major clinical importance as it allows treatment of undesirable tissues near critical blood vessels. The presented study results suggest that MR images acquired following NTIRE treatment are all directly related to the unique pattern of blood flow after NTIRE treatment and are not produced in the absence of blood flow.


Subject(s)
Electroporation , Magnetic Resonance Imaging , Regional Blood Flow , Animals , Electroporation/methods , Liver/blood supply , Liver/pathology , Male , Rats
19.
PLoS One ; 7(12): e52008, 2012.
Article in English | MEDLINE | ID: mdl-23251672

ABSTRACT

The current standard of care for newly diagnosed glioblastoma multiforme (GBM) is resection followed by radiotherapy with concomitant and adjuvant temozolomide. Recent studies suggest that nearly half of the patients with early radiological deterioration post treatment do not suffer from tumor recurrence but from pseudoprogression. Similarly, a significant number of patients with brain metastases suffer from radiation necrosis following radiation treatments. Conventional MRI is currently unable to differentiate tumor progression from treatment-induced effects. The ability to clearly differentiate tumor from non-tumoral tissues is crucial for appropriate patient management. Ten patients with primary brain tumors and 10 patients with brain metastases were scanned by delayed contrast extravasation MRI prior to surgery. Enhancement subtraction maps calculated from high resolution MR images acquired up to 75 min after contrast administration were used for obtaining stereotactic biopsies. Histological assessment was then compared with the pre-surgical calculated maps. In addition, the application of our maps for prediction of progression was studied in a small cohort of 13 newly diagnosed GBM patients undergoing standard chemoradiation and followed up to 19.7 months post therapy. The maps showed two primary enhancement populations: the slow population where contrast clearance from the tissue was slower than contrast accumulation and the fast population where clearance was faster than accumulation. Comparison with histology confirmed the fast population to consist of morphologically active tumor and the slow population to consist of non-tumoral tissues. Our maps demonstrated significant correlation with perfusion-weighted MR data acquired simultaneously, although contradicting examples were shown. Preliminary results suggest that early changes in the fast volumes may serve as a predictor for time to progression. These preliminary results suggest that our high resolution MRI-based delayed enhancement subtraction maps may be applied for clear depiction of tumor and non-tumoral tissues in patients with primary brain tumors and patients with brain metastases.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , Magnetic Resonance Imaging/methods , Adult , Aged , Aged, 80 and over , Brain Neoplasms/diagnosis , Brain Neoplasms/surgery , Contrast Media , Disease Progression , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Metastasis
20.
PLoS One ; 7(8): e42817, 2012.
Article in English | MEDLINE | ID: mdl-22900052

ABSTRACT

Electroporation, is known to induce cell membrane permeabilization in the reversible (RE) mode and cell death in the irreversible (IRE) mode. Using an experimental system designed to produce a continuum of IRE followed by RE around a single electrode we used MRI to study the effects of electroporation on the brain. Fifty-four rats were injected with Gd-DOTA and treated with a G25 electrode implanted 5.5 mm deep into the striata. MRI was acquired immediately after treatment, 10 min, 20 min, 30 min, and up to three weeks following the treatment using: T1W, T2W, Gradient echo (GE), serial SPGR (DCE-MRI) with flip angles ranging over 5-25°, and diffusion-weighted MRI (DWMRI). Blood brain barrier (BBB) disruption was depicted as clear enhancement on T1W images. The average signal intensity in the regions of T1-enhancement, representing BBB disruption, increased from 1887±83 (arbitrary units) immediately post treatment to 2246±94 20 min post treatment, then reached a plateau towards the 30 min scan where it reached 2289±87. DWMRI at 30 min showed no significant effects. Early treatment effects and late irreversible damage were clearly depicted on T2W. The enhancing volume on T2W has increased by an average of 2.27±0.27 in the first 24-48 hours post treatment, suggesting an inflammatory tissue response. The permanent tissue damage, depicted as an enhancing region on T2W, 3 weeks post treatment, decreased to an average of 50±10% of the T2W enhancing volumes on the day of the treatment which was 33±5% of the BBB disruption volume. Permanent tissue damage was significantly smaller than the volume of BBB disruption, suggesting, that BBB disruption is associated with RE while tissue damage with IRE. These results demonstrate the feasibility of applying reversible and irreversible electroporation for transient BBB disruption or permanent damage, respectively, and applying MRI for planning/monitoring disruption volume/shape by optimizing electrode positions and treatment parameters.


Subject(s)
Blood-Brain Barrier/injuries , Electroporation , Magnetic Resonance Imaging , Animals , Brain/pathology , Electroporation/methods , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...