Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Phys Med Biol ; 69(8)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38417178

ABSTRACT

Objective.Alternating electric fields (AEF) therapy is a treatment modality for patients with glioblastoma. Tumor characteristics such as size, location, and extent of peritumoral edema may affect the AEF strength and distribution. We evaluated the sensitivity of the AEFs in a realistic 3D rat glioma model with respect to these properties.Approach.The electric properties of the peritumoral edema were varied based on calculated and literature-reported values. Models with different tumor composition, size, and location were created. The resulting AEFs were evaluated in 3D rat glioma models.Main results.In all cases, a pair of 5 mm diameter electrodes induced an average field strength >1 V cm-1. The simulation results showed that a negative relationship between edema conductivity and field strength was found. As the tumor core size was increased, the average field strength increased while the fraction of the shell achieving >1.5 V cm-1decreased. Increasing peritumoral edema thickness decreased the shell's mean field strength. Compared to rostrally/caudally, shifting the tumor location laterally/medially and ventrally (with respect to the electrodes) caused higher deviation in field strength.Significance.This study identifies tumor properties that are key drivers influencing AEF strength and distribution. The findings might be potential preclinical implications.


Subject(s)
Brain Neoplasms , Electric Stimulation Therapy , Glioblastoma , Glioma , Lymphokines , Humans , Rats , Animals , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Electric Stimulation Therapy/methods , Glioma/therapy , Glioblastoma/pathology
2.
Phys Med Biol ; 68(20)2023 10 06.
Article in English | MEDLINE | ID: mdl-37703902

ABSTRACT

Objective.Application of alternating electrical fields (AEFs) in the kHz range is an established treatment modality for primary and recurrent glioblastoma. Preclinical studies would enable innovations in treatment monitoring and efficacy, which could then be translated to benefit patients. We present a practical translational process converting image-based data into 3D rat head models for AEF simulations and study its sensitivity to parameter choices.Approach.Five rat head models composed of up to 7 different tissue types were created, and relative permittivity and conductivity of individual tissues obtained from the literature were assigned. Finite element analysis was used to model the AEF strength and distribution in the models with different combinations of head tissues, a virtual tumor, and an electrode pair.Main results.The simulations allowed for a sensitivity analysis of the AEF distribution with respect to different tissue combinations and tissue parameter values.Significance.For a single pair of 5 mm diameter electrodes, an average AEF strength inside the tumor exceeded 1.5 V cm-1, expected to be sufficient for a relevant therapeutic outcome. This study illustrates a robust and flexible approach for simulating AEF in different tissue types, suitable for preclinical studies in rodents and translatable to clinical use.


Subject(s)
Electric Stimulation Therapy , Glioblastoma , Humans , Rats , Animals , Glioblastoma/pathology , Electricity , Electric Conductivity , Electric Stimulation Therapy/methods
3.
Bioelectrochemistry ; 149: 108287, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36306728

ABSTRACT

Exposing cancer cells to alternating electric fields of 100-300 kHz frequency and 1-4 V/cm strength has been shown to significantly reduce cancer growth in cell culture and in human patients. This form of anti-cancer therapy is more commonly referred to as tumor treating fields (TTFields), a novel treatment modality that has been approved by the U.S. Food and Drug Administration for use in patients with glioblastoma and malignant pleural mesothelioma. Pivotal trials in other solid organ cancer trials are underway. In regards to overall survival, TTFields alone is comparable to chemotherapy alone in recurrent glioblastoma. However, when combined with adjuvant chemotherapy, TTFields prolong median survival by 4.9 months in newly-diagnosed glioblastoma. TTFields hold promise as a therapeutic approach to numerous solid organ cancers. This review summarizes the current status of TTFields research at the preclinical level, highlighting recent aspects of a relatively complex working hypothesis. In addition, we point out the gaps between limited preclinical in vivo studies and the available clinical data. To date, no customized system for TTFields delivery in rodent models of glioblastoma has been presented. We aim to motivate the expansion of TTFields preclinical research and facilitate the availability of suitable hardware, to ultimately improve outcomes in patients with cancer.


Subject(s)
Brain Neoplasms , Electric Stimulation Therapy , Glioblastoma , Humans , Glioblastoma/therapy , Combined Modality Therapy , Electricity
4.
Cell Death Discov ; 4: 113, 2018.
Article in English | MEDLINE | ID: mdl-30534421

ABSTRACT

Glioblastoma is the most common yet most lethal of primary brain cancers with a one-year post-diagnosis survival rate of 65% and a five-year survival rate of barely 5%. Recently the U.S. Food and Drug Administration approved a novel fourth approach (in addition to surgery, radiation therapy, and chemotherapy) to treating glioblastoma; namely, tumor treating fields (TTFields). TTFields involves the delivery of alternating electric fields to the tumor but its mechanisms of action are not fully understood. Current theories involve TTFields disrupting mitosis due to interference with proper mitotic spindle assembly. We show that TTFields also alters cellular membrane structure thus rendering it more permeant to chemotherapeutics. Increased membrane permeability through the imposition of TTFields was shown by several approaches. For example, increased permeability was indicated through increased bioluminescence with TTFields exposure or with the increased binding and ingress of membrane-associating reagents such as Dextran-FITC or ethidium D or with the demonstration by scanning electron microscopy of augmented number and sizes of holes on the cellular membrane. Further investigations showed that increases in bioluminescence and membrane hole production with TTFields exposure disappeared by 24 h after cessation of alternating electric fields thus demonstrating that this phenomenom is reversible. Preliminary investigations showed that TTFields did not induce membrane holes in normal human fibroblasts thus suggesting that the phenomenom was specific to cancer cells. With TTFields, we present evidence showing augmented membrane accessibility by compounds such as 5-aminolevulinic acid, a reagent used intraoperatively to delineate tumor from normal tissue in glioblastoma patients. In addition, this mechanism helps to explain previous reports of additive and synergistic effects between TTFields and other chemotherapies. These findings have implications for the design of combination therapies in glioblastoma and other cancers and may significantly alter standard of care strategies for these diseases.

5.
Small ; 14(19): e1703683, 2018 05.
Article in English | MEDLINE | ID: mdl-29635739

ABSTRACT

Raman microspectroscopy provides chemo-selective image contrast, sub-micrometer resolution, and multiplexing capabilities. However, it suffers from weak signals resulting in image-acquisition times of up to several hours. Surface-enhanced Raman scattering (SERS) can dramatically enhance signals of molecules in close vicinity of metallic surfaces and overcome this limitation. Multimodal, SERS-active nanoparticles are usually labeled with Raman marker molecules, limiting SERS to the coating material. In order to realize multimodal imaging while acquiring the rich endogenous vibronic information of the specimen, a core-shell particle based on "Nanorice", where a spindle-shaped iron oxide core is encapsulated by a closed gold shell, is developed. An ultrathin layer of silica prevents agglomeration and unwanted chemical interaction with the specimen. This approach provides Raman signal enhancement due to plasmon resonance effects of the shell while the optical absorption in the near-infrared spectral region provides contrast in photoacoustic tomography. Finally, T2-relaxation of a magnetic resonance imaging (MRI) experiment is altered by taking advantage of the iron oxide core. The feasibility for Raman imaging is evaluated by nearfield simulations and experimental studies on the primate cell line COS1. MRI and photoacoustics are demonstrated in agarose phantoms illustrating the promising translational nature of this strategy for clinical applications in radiology.


Subject(s)
Contrast Media/chemistry , Dust , Magnetic Resonance Imaging/methods , Nanoparticles/chemistry , Photoacoustic Techniques/methods , Spectrum Analysis, Raman , Animals , COS Cells , Chlorocebus aethiops , Computer Simulation , Nanoparticles/ultrastructure , Phantoms, Imaging
6.
J Neurooncol ; 134(2): 259-268, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28681243

ABSTRACT

Glioblastoma (GBM) is the most aggressive and lethal form of brain cancer. Standard therapies are non-specific and often of limited effectiveness; thus, efforts are underway to uncover novel, unorthodox therapies against GBM. In previous studies, we investigated Withaferin A, a steroidal lactone from Ayurvedic medicine that inhibits proliferation in cancers including GBM. Another novel approach, tumor treating fields (TTFields), is thought to disrupt mitotic spindle formation and stymie proliferation of actively dividing cells. We hypothesized that combining TTFields with Withaferin A would synergistically inhibit proliferation in glioblastoma. Human glioblastoma cells (GBM2, GBM39, U87-MG) and human breast adenocarcinoma cells (MDA-MB-231) were isolated from primary tumors. The glioma cell lines were genetically engineered to express firefly luciferase. Proliferative potential was assessed either by bioluminescence imaging or cell counting via hemocytometer. TTFields (4 V/cm) significantly inhibited growth of the four cancer cell lines tested (n = 3 experiments per time point, four measurements per sample, p < 0.02 at least; 2-way ANOVA, control vs. treatment). The combination of Withaferin A (10-100 nM) with TTFields significantly inhibited the growth of the glioma cells to a degree beyond that of Withaferin A or TTFields alone. The interaction of the Withaferin A and TTFields on glioma cells was found to be synergistic in nature (p < 0.01, n = 3 experiments). These findings were validated by both bioluminescence and hemocytometric measurements. The combination of Withaferin A with TTFields represents a novel approach to treat GBM in a manner that is likely better than either treatment alone and that is synergistic.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Cell Proliferation , Electric Stimulation Therapy , Glioma/therapy , Withanolides/therapeutic use , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Adenocarcinoma/therapy , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Doxorubicin/therapeutic use , Electric Stimulation Therapy/methods , Glioma/pathology , Glioma/physiopathology , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Temperature
7.
J Biomed Opt ; 22(6): 66005, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28613345

ABSTRACT

Multiplex coherent anti-Stokes Raman scattering (MCARS) microscopy was carried out to map a solid tumor in mouse brain tissue. The border between normal and tumor tissue was visualized using support vector machines (SVM) as a higher ranking type of data classification. Training data were collected separately in both tissue types, and the image contrast is based on class affiliation of the single spectra. Color coding in the image generated by SVM is then related to pathological information instead of single spectral intensities or spectral differences within the data set. The results show good agreement with the H&E stained reference and spontaneous Raman microscopy, proving the validity of the MCARS approach in combination with SVM.


Subject(s)
Brain/diagnostic imaging , Diagnostic Imaging/methods , Spectrum Analysis, Raman , Animals , Diagnostic Imaging/instrumentation , Mice
8.
J Neurooncol ; 126(2): 253-64, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26650066

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive, malignant cancer Johnson and O'Neill (J Neurooncol 107: 359-364, 2012). An extract from the winter cherry plant (Withania somnifera ), AshwaMAX, is concentrated (4.3 %) for Withaferin A; a steroidal lactone that inhibits cancer cells Vanden Berghe et al. (Cancer Epidemiol Biomark Prev 23: 1985-1996, 2014). We hypothesized that AshwaMAX could treat GBM and that bioluminescence imaging (BLI) could track oral therapy in orthotopic murine models of glioblastoma. Human parietal-cortical glioblastoma cells (GBM2, GBM39) were isolated from primary tumors while U87-MG was obtained commercially. GBM2 was transduced with lentiviral vectors that express Green Fluorescent Protein (GFP)/firefly luciferase fusion proteins. Mutational, expression and proliferative status of GBMs were studied. Intracranial xenografts of glioblastomas were grown in the right frontal regions of female, nude mice (n = 3-5 per experiment). Tumor growth was followed through BLI. Neurosphere cultures (U87-MG, GBM2 and GBM39) were inhibited by AshwaMAX at IC50 of 1.4, 0.19 and 0.22 µM equivalent respectively and by Withaferin A with IC50 of 0.31, 0.28 and 0.25 µM respectively. Oral gavage, every other day, of AshwaMAX (40 mg/kg per day) significantly reduced bioluminescence signal (n = 3 mice, p < 0.02, four parameter non-linear regression analysis) in preclinical models. After 30 days of treatment, bioluminescent signal increased suggesting onset of resistance. BLI signal for control, vehicle-treated mice increased and then plateaued. Bioluminescent imaging revealed diffuse growth of GBM2 xenografts. With AshwaMAX, GBM neurospheres collapsed at nanomolar concentrations. Oral treatment studies on murine models confirmed that AshwaMAX is effective against orthotopic GBM. AshwaMAX is thus a promising candidate for future clinical translation in patients with GBM.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Plant Extracts/administration & dosage , Withania/chemistry , Withanolides/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Luminescent Measurements , Mice , Mice, Nude , Neural Stem Cells/drug effects , Plant Extracts/chemistry , Withanolides/pharmacology , Xenograft Model Antitumor Assays
9.
Sci Transl Med ; 7(310): 310ra169, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26491079

ABSTRACT

Cancer cells reprogram their metabolism to meet increased biosynthetic demands, commensurate with elevated rates of replication. Pyruvate kinase M2 (PKM2) catalyzes the final and rate-limiting step in tumor glycolysis, controlling the balance between energy production and the synthesis of metabolic precursors. We report here the synthesis and evaluation of a positron emission tomography (PET) radiotracer, [(11)C]DASA-23, that provides a direct noninvasive measure of PKM2 expression in preclinical models of glioblastoma multiforme (GBM). In vivo, orthotopic U87 and GBM39 patient-derived tumors were clearly delineated from the surrounding normal brain tissue by PET imaging, corresponding to exclusive tumor-associated PKM2 expression. In addition, systemic treatment of mice with the PKM2 activator TEPP-46 resulted in complete abrogation of the PET signal in intracranial GBM39 tumors. Together, these data provide the basis for the clinical evaluation of imaging agents that target this important gatekeeper of tumor glycolysis.


Subject(s)
Hexokinase/metabolism , Positron-Emission Tomography , Pyruvate Kinase/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/enzymology , Carbon Radioisotopes , Glycolysis , Humans
10.
J Biophotonics ; 7(1-2): 126-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23836627

ABSTRACT

Chemical and structural composition of wood biomass is studied by label-free and chemically specific Coherent Anti-Stokes Raman Scattering (CARS) microscopy. A concept developed for assignment and semi-quantitative imaging of sample components; cellulose, hemicellulose, and lignin; by multiplex CARS microspectroscopy and subsequent data analysis is presented. Specific imaging without fluorescence backround is achieved an order of magnitude faster compared with conventional Raman microscopy. Laser polarization control yield information on molecular arrangement in wood fibers. Narrowband CARS excitation of single vibrations allows for three-dimensional volume imaging. Thus, CARS microscopy has potential as an important instrument for characterization of lignocellulosic materials.


Subject(s)
Biomass , Lignin/chemistry , Microscopy/methods , Spectrum Analysis, Raman/methods , Imaging, Three-Dimensional , Time Factors , Wood/chemistry
11.
MRS Bull ; 38(8)2013 Aug.
Article in English | MEDLINE | ID: mdl-24293809

ABSTRACT

Molecular imaging scans cellular and molecular targets in living subjects through the introduction of imaging agents that bind to these targets and report their presence through a measurable signal. The picomolar sensitivity, signal stability, and high multiplexing capacity of Raman spectroscopy satisfies important needs within the field of molecular imaging, and several groups now utilize Raman and surface-enhanced Raman spectroscopy to image molecular targets in small animal models of human disease. This article details the role of Raman spectroscopy in molecular imaging, describes some substrates and imaging agents used in animal models, and illustrates some examples.

12.
Biomed Opt Express ; 2(8): 2110-6, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21833351

ABSTRACT

The fast and reliable characterization of pathological tissue is a debated topic in the application of vibrational spectroscopy in medicine. In the present work we apply multiplex coherent anti-Stokes Raman scattering (MCARS) to the investigation of fresh mouse brain tissue. The combination of imaginary part extraction followed by principal component analysis led to color contrast between grey and white matter as well as layers of granule and Purkinje cells. Additional quantitative information was obtained by using a decomposition algorithm. The results perfectly agree with HE stained references slides prepared separately making multiplex CARS an ideal approach for chemoselective imaging.

13.
J Biomed Opt ; 16(2): 021105, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21361668

ABSTRACT

Multiplex coherent anti-Stokes Raman scattering (MCARS) provides labeling free and fast characterization of materials and biological samples in nonlinear microscopy. In spite of its success, remaining challenges regarding the data analysis for chemoselective imaging still have to be solved. In general, image contrast has been realized by using only one spectral feature directly taken from the unprocessed raw data. This procedure is limited to strong and well separated Raman resonances like the saturated CH-stretching vibration of lipids in the case of biological samples. In order to overcome this limitation, we present a new method of MCARS data processing that exploits the whole measured spectrum to disentangle overlapping contributions of different (bio-) chemical components. Our "two-step" approach is based on the combination of imaginary part extraction followed by global fitting of the hyperspectral data set. Previous knowledge about the sample, e.g., pure spectra of the individual components is no longer necessary. The result is a highly contrasted image, where the patterns and differences between the sample components can be represented in different colors. We successfully applied this method to complex structured polymer samples and biological tissues.


Subject(s)
Algorithms , Biopolymers/analysis , Microscopy/methods , Spectrum Analysis, Raman/methods , Sensitivity and Specificity
14.
Opt Lett ; 35(22): 3721-3, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21081975

ABSTRACT

We combined the ultrabroadband supercontinuum of a photonic crystal fiber with a pulse shaper, resulting in a highly flexible light source for multiplex coherent anti-Stokes Raman microscopy. Implemented as the Stokes pulse, it provides tailored selection of the relevant Raman transitions, resulting in a reduced photon load and partial suppression of the nonresonant background. This experiment exploits the advantages of multiplex excitation with the increased acquisition speed of single-channel detection. The molecule-specific Stokes pulses are demonstrated for chemical mapping of a polymer blend.


Subject(s)
Microscopy/methods , Spectrum Analysis, Raman , Light , Optics and Photonics , Photons , Polyethylene/chemistry , Polymers/chemistry , Polymethyl Methacrylate/chemistry , Vibration
SELECTION OF CITATIONS
SEARCH DETAIL
...