Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Int J Mol Sci ; 23(22)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36430552

ABSTRACT

Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. TTFields induce anti-mitotic effects through the disruption of the mitotic spindle and abnormal chromosome segregation, which trigger several forms of cell death, including immunogenic cell death (ICD). The efficacy of TTFields concomitant with anti-programmed death-1 (anti-PD-1) treatment was previously shown in vivo and is currently under clinical investigation. Here, the potential of TTFields concomitant with anti- PD-1/anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or anti-programmed death-ligand 1 (anti-PD-L1) immune checkpoint inhibitors (ICI) to improve therapeutic efficacy was examined in lung tumor-bearing mice. Increased circulating levels of high mobility group box 1 protein (HMGB1) and elevated intratumoral levels of phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α) were found in the TTFields-treated mice, indicative of ICD induction. The concomitant application of TTFields and ICI led to a significant decrease in tumor volume as compared to all other groups. In addition, significant increases in the number of tumor-infiltrating immune cells, specifically cytotoxic T-cells, were observed in the TTFields plus anti-PD-1/anti-CTLA-4 or anti-PD-L1 groups. Correspondingly, cytotoxic T-cells isolated from these tumors showed higher levels of IFN-γ production. Collectively, these results suggest that TTFields have an immunoactivating role that may be leveraged for concomitant treatment with ICI to achieve better tumor control by enhancing antitumor immunity.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Mice , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Cell Survival/physiology , Spindle Apparatus
3.
Biomolecules ; 12(10)2022 09 22.
Article in English | MEDLINE | ID: mdl-36291557

ABSTRACT

Despite the availability of numerous therapeutic substances that could potentially target CNS disorders, an inability of these agents to cross the restrictive blood-brain barrier (BBB) limits their clinical utility. Novel strategies to overcome the BBB are therefore needed to improve drug delivery. We report, for the first time, how Tumor Treating Fields (TTFields), approved for glioblastoma (GBM), affect the BBB's integrity and permeability. Here, we treated murine microvascular cerebellar endothelial cells (cerebEND) with 100-300 kHz TTFields for up to 72 h and analyzed the expression of barrier proteins by immunofluorescence staining and Western blot. In vivo, compounds normally unable to cross the BBB were traced in healthy rat brain following TTFields administration at 100 kHz. The effects were analyzed via MRI and immunohistochemical staining of tight-junction proteins. Furthermore, GBM tumor-bearing rats were treated with paclitaxel (PTX), a chemotherapeutic normally restricted by the BBB combined with TTFields at 100 kHz. The tumor volume was reduced with TTFields plus PTX, relative to either treatment alone. In vitro, we demonstrate that TTFields transiently disrupted BBB function at 100 kHz through a Rho kinase-mediated tight junction claudin-5 phosphorylation pathway. Altogether, if translated into clinical use, TTFields could represent a novel CNS drug delivery strategy.


Subject(s)
Blood-Brain Barrier , Glioblastoma , Animals , Mice , Rats , Blood-Brain Barrier/metabolism , rho-Associated Kinases/metabolism , Claudin-5/metabolism , Endothelial Cells/metabolism , Glioblastoma/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use
4.
Cancers (Basel) ; 14(12)2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35740624

ABSTRACT

Hepatocellular carcinoma (HCC), a highly aggressive liver cancer, is a leading cause of cancer-related death. Tumor Treating Fields (TTFields) are electric fields that exert antimitotic effects on cancerous cells. The aims of the current research were to test the efficacy of TTFields in HCC, explore the underlying mechanisms, and investigate the possible combination of TTFields with sorafenib, one of the few front-line treatments for patients with advanced HCC. HepG2 and Huh-7D12 human HCC cell lines were treated with TTFields at various frequencies to determine the optimal frequency eliciting maximal cell count reduction. Clonogenic, apoptotic effects, and autophagy induction were measured. The efficacy of TTFields alone and with concomitant sorafenib was tested in cell cultures and in an orthotopic N1S1 rat model. Tumor volume was examined at the beginning and following 5 days of treatment. At study cessation, tumors were weighed and examined by immunohistochemistry to assess autophagy and apoptosis. TTFields were found in vitro to exert maximal effect at 150 kHz, reducing cell count and colony formation, increasing apoptosis and autophagy, and augmenting the effects of sorafenib. In animals, TTFields concomitant with sorafenib reduced tumor weight and volume fold change, and increased cases of stable disease following treatment versus TTFields or sorafenib alone. While each treatment alone elevated levels of autophagy relative to control, TTFields concomitant with sorafenib induced a significant increase versus control in tumor ER stress and apoptosis levels, demonstrating increased stress under the multimodal treatment. Overall, TTFields treatment demonstrated efficacy and enhanced the effects of sorafenib for the treatment of HCC in vitro and in vivo, via a mechanism involving induction of autophagy.

5.
Lung Cancer ; 160: 99-110, 2021 10.
Article in English | MEDLINE | ID: mdl-34482104

ABSTRACT

OBJECTIVES: Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields with antimitotic effects on cancerous cells. TTFields concomitant with pemetrexed and a platinum agent are approved in the US and EU as first line therapy for unresectable, locally advanced or metastatic malignant pleural mesothelioma (MPM). The goal of the current study was to characterize the mechanism of action of TTFields in MPM cell lines and animal models. METHODS: Human MPM cell lines MSTO-211H and NCI-H2052 were treated with TTFields to determine the frequency that elicits maximal cytotoxicity. The effect of TTFields on DNA damage and repair, and the cytotoxic effect of TTFields in combination with cisplatin and/or pemetrexed were examined. Efficacy of TTFields concomitant with cisplatin and pemetrexed was evaluated in orthotopic IL-45 and subcutaneous RN5 murine models. RESULTS: TTFields at a frequency of 150 kHz demonstrated the highest cytotoxicity to MPM cells. Application of 150 kHz TTFields resulted in increased formation of DNA double strand breaks, elevated expression of DNA damage induced cell cycle arrest proteins, and reduced expression of Fanconi Anemia (FA)-BRCA DNA repair pathway proteins. Co-treatment of TTFields with cisplatin or pemetrexed significantly increased treatment efficacy versus each modality alone, with additivity and synergy exhibited by the TTFields-pemetrexed and TTFields-cisplatin combinations, respectively. In animal models, tumor volume was significantly lower for the TTFields-cisplatin-pemetrexed combination compared to control, accompanied by increased DNA damage within the tumor. CONCLUSION: This research demonstrated that the efficacy of TTFields for the treatment of MPM is associated with reduced expression of FA-BRCA pathway proteins and increased DNA damage. This mechanism of action is consistent with the observed synergism for TTFields-cisplatin vs additivity for TTFields-pemetrexed, as cisplatin-induced DNA damage is repaired via the FA-BRCA pathway.


Subject(s)
Fanconi Anemia , Lung Neoplasms , Mesothelioma, Malignant , Animals , Cisplatin , Humans , Lung Neoplasms/drug therapy , Mice , Pemetrexed
6.
Cancer Immunol Immunother ; 69(7): 1191-1204, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32144446

ABSTRACT

Tumor-treating fields (TTFields) are alternating electric fields in a specific frequency range (100-300 kHz) delivered to the human body through transducer arrays. In this study, we evaluated whether TTFields-mediated cell death can elicit antitumoral immunity and hence would be effectively combined with anti-PD-1 therapy. We demonstrate that in TTFields-treated cancer cells, damage-associated molecular patterns including high-mobility group B1 and adenosine triphosphate are released and calreticulin is exposed on the cell surface. Moreover, we show that TTFields treatment promotes the engulfment of cancer cells by dendritic cells (DCs) and DCs maturation in vitro, as well as recruitment of immune cells in vivo. Additionally, our study demonstrates that the combination of TTFields with anti-PD-1 therapy results in a significant decline of tumor volume and increase in the percentage of tumor-infiltrating leukocytes in two tumor models. In orthotopic lung tumors, these infiltrating leukocytes, specifically macrophages and DCs, showed elevated expression of PD-L1. Compatibly, cytotoxic T-cells isolated from these tumors demonstrated increased production of IFN-γ. In colon cancer tumors, T-cells infiltration was significantly increased following long treatment duration with TTFields plus anti-PD-1. Collectively, our results suggest that TTFields therapy can induce anticancer immune response. Furthermore, we demonstrate robust efficacy of concomitant application of TTFields and anti-PD-1 therapy. These data suggest that integrating TTFields with anti-PD-1 therapy may further enhance antitumor immunity, hence achieve better tumor control.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Carcinoma, Hepatocellular/therapy , Carcinoma, Lewis Lung/therapy , Electric Stimulation Therapy/methods , Immunogenic Cell Death , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Apoptosis , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Proliferation , Combined Modality Therapy , Female , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Toxicol Pathol ; 43(5): 633-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25694086

ABSTRACT

Magnetic resonance imaging (MRI) is widely used in preclinical research and drug development and is a powerful noninvasive method for assessment of phenotypes and therapeutic efficacy in murine models of disease. In vivo MRI provides an opportunity for longitudinal evaluation of tissue changes and phenotypic expression in experimental animal models. Ex vivo MRI of fixed samples permits a thorough examination of multiple digital slices while leaving the specimen intact for subsequent conventional hematoxylin and eosin (H&E) histology. With the advent of new compact MRI systems that are designed to operate in most conventional labs without the cost, complexity, and infrastructure needs of conventional MRI systems, the possibility of MRI becoming a practical modality is now viable. The purpose of this study was to investigate the capabilities of a new compact, high-performance MRI platform (M2™; Aspect Imaging, Israel) as it relates to preclinical toxicology studies. This overview will provide examples of major organ system pathologies with an emphasis on how compact MRI can serve as an important adjunct to conventional pathology by nondestructively providing 3-dimensional (3-D) digital data sets, detailed morphological insights, and quantitative information. Comparative data using compact MRI for both in vivo and ex vivo are provided as well as validation using conventional H&E.


Subject(s)
Histological Techniques/methods , Magnetic Resonance Imaging/methods , Pathology/methods , Toxicology/methods , Animals , Humans
8.
Epilepsy Res ; 105(1-2): 62-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23357721

ABSTRACT

Hyperbaric oxygen-induced seizures are classified as generalized, tonic-clonic seizures. They are believed to cause no residual neurologic damage, although this has not been investigated in depth. We used different MRI sequences to determine whether hyperbaric oxygen-induced seizures in mice caused brain structural changes. Experimental animals were exposed to a pressure of 6 atmospheres absolute breathing oxygen, and were randomly assigned to two groups in which MRI was performed immediately after the appearance of seizures or 7 days later. Control groups were not exposed to hyperbaric oxygen. Our MRI protocols included T2*-weighted images, T2 maps, diffusion-weighted echo-planar pulse sequence, and contrast-enhanced T1-weighted images. Both the cortex and the hippocampus were analyzed. T2 values of the hippocampus and the cortex in the hyperbaric oxygen-exposed groups showed a small but statistically significant decrease compared with the control groups immediately after seizures (p<0.01). One week after seizures, enhancement following contrast injection was significantly higher both in the cortex and the hippocampus in the hyperbaric oxygen-exposed groups compared with the control groups (p<0.01). Hippocampal and cortex T2 values 7 days after seizures were similar to control group values. No differences were found among the other sequences. We conclude that hyperbaric oxygen-induced seizures result in delayed injury to the blood-brain barrier. Elucidation of the mechanisms and significance of this injury will necessitate further investigation.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Hyperbaric Oxygenation/adverse effects , Magnetic Resonance Imaging/methods , Seizures/metabolism , Animals , Male , Mice , Mice, Inbred ICR , Random Allocation , Seizures/diagnosis , Seizures/etiology
9.
Neurobiol Aging ; 33(2): 432.e1-432.e13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21371785

ABSTRACT

Cerebrovascular amyloidosis is caused by amyloid accumulation in walls of blood vessel walls leading to hemorrhagic stroke and cognitive impairment. Transforming growth factor-ß1 (TGF-ß1) expression levels correlate with the degree of cerebrovascular amyloid deposition in Alzheimer's disease (AD) and TGF-ß1 immunoreactivity in such cases is increased along the cerebral blood vessels. Here we show that a nasally administered proteosome-based adjuvant activates macrophages and decreases vascular amyloid in TGF-ß1 mice. Animals were nasally treated with a proteosome-based adjuvant on a weekly basis for 3 months beginning at age 13 months. Using magnetic resonance imaging (MRI) we found that while control animals showed a significant cerebrovascular pathology, proteosome-based adjuvant prevents further brain damage and prevents pathological changes in the blood-brain barrier. Using an object recognition test and Y-maze, we found significant improvement in cognition in the treated group. Our findings support the potential use of a macrophage immunomodulator as a novel approach to reduce cerebrovascular amyloid, prevent microhemorrhage, and improve cognition.


Subject(s)
Cerebral Amyloid Angiopathy/immunology , Cerebral Amyloid Angiopathy/therapy , Cysteine Endopeptidases/administration & dosage , Disease Models, Animal , Lipopolysaccharides/administration & dosage , Macrophage Activation/drug effects , Transforming Growth Factor beta1/immunology , Administration, Inhalation , Animals , Drug Combinations , Immunotherapy/methods , Mice , Mice, Transgenic , Transforming Growth Factor beta1/genetics , Treatment Outcome
10.
Exp Neurol ; 232(2): 280-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21946269

ABSTRACT

Improvised explosive devices (IEDs) are one of the main causes for casualties among civilians and military personnel in the present war against terror. Mild traumatic brain injury from IEDs induces various degrees of cognitive, emotional and behavioral disturbances but knowledge of the exact brain pathophysiology following exposure to blast is poorly understood. The study was aimed at establishing a murine model for a mild BI-TBI that isolates low-level blast pressure effects to the brain without systemic injuries. An open-field explosives detonation was used to replicate, as closely as possible, low-level blast trauma in the battlefield or at a terror-attack site. No alterations in basic neurological assessment or brain gross pathology were found acutely in the blast-exposed mice. At 7 days post blast, cognitive and behavioral tests revealed significantly decreased performance at both 4 and 7 m distance from the blast (5.5 and 2.5 PSI, respectively). At 30 days post-blast, clear differences were found in animals at both distances in the object recognition test, and in the 7 m group in the Y maze test. Using MRI, T1 weighted images showed an increased BBB permeability 1 month post-blast. DTI analysis showed an increase in fractional anisotropy (FA) and a decrease in radial diffusivity. These changes correlated with sites of up-regulation of manganese superoxide dismutase 2 in neurons and CXC-motif chemokine receptor 3 around blood vessels in fiber tracts. These results may represent brain axonal and myelin abnormalities. Cellular and biochemical studies are underway in order to further correlate the blast-induced cognitive and behavioral changes and to identify possible underlying mechanisms that may help develop treatment- and neuroprotective modalities.


Subject(s)
Blast Injuries/pathology , Blast Injuries/physiopathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Disease Models, Animal , Mice, Inbred ICR , Animals , Brain/pathology , Brain/physiopathology , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Diffusion Tensor Imaging , Explosions , Male , Maze Learning/physiology , Mice , Motor Activity/physiology , Recognition, Psychology/physiology , Recovery of Function/physiology , Trauma Severity Indices
11.
PLoS One ; 5(4): e10282, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421983

ABSTRACT

BACKGROUND: Antiangiogenic and anti-vascular therapies present intriguing alternatives to cancer therapy. However, despite promising preclinical results and significant delays in tumor progression, none have demonstrated long-term curative features to date. Here, we show that a single treatment session of Tookad-based vascular targeted photodynamic therapy (VTP) promotes permanent arrest of tumor blood supply by rapid occlusion of the tumor feeding arteries (FA) and draining veins (DV), leading to tumor necrosis and eradication within 24-48 h. METHODOLOGY/PRINCIPAL FINDINGS: A mouse earlobe MADB106 tumor model was subjected to Tookad-VTP and monitored by three complementary, non-invasive online imaging techniques: Fluorescent intravital microscopy, Dynamic Light Scattering Imaging and photosensitized MRI. Tookad-VTP led to prompt tumor FA vasodilatation (a mean volume increase of 70%) with a transient increase (60%) in blood-flow rate. Rapid vasoconstriction, simultaneous blood clotting, vessel permeabilization and a sharp decline in the flow rates then followed, culminating in FA occlusion at 63.2 sec+/-1.5SEM. This blockage was deemed irreversible after 10 minutes of VTP treatment. A decrease in DV blood flow was demonstrated, with a slight lag from FA response, accompanied by frequent changes in flow direction before reaching a complete standstill. In contrast, neighboring, healthy tissue vessels of similar sizes remained intact and functional after Tookad-VTP. CONCLUSION/SIGNIFICANCE: Tookad-VTP selectively targets the tumor feeding and draining vessels. To the best of our knowledge, this is the first mono-therapeutic modality that primarily aims at the larger tumor vessels and leads to high cure rates, both in the preclinical and clinical arenas.


Subject(s)
Bacteriochlorophylls/therapeutic use , Neoplasms/therapy , Photochemotherapy/methods , Animals , Arteries/pathology , Blood Coagulation/drug effects , Blood Coagulation/radiation effects , Blood Flow Velocity/drug effects , Blood Flow Velocity/radiation effects , Disease Models, Animal , Ear , Mice , Necrosis , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/therapy , Permeability/drug effects , Permeability/radiation effects , Treatment Outcome , Vasoconstriction/drug effects , Vasoconstriction/radiation effects , Veins/pathology
12.
PLoS One ; 2(11): e1191, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18030327

ABSTRACT

In this study, we show how light can be absorbed by the body of a living rat due to an injected pigment circulating in the blood stream. This process is then physiologically translated in the tissue into a chemical signature that can be perceived as an image by magnetic resonance imaging (MRI). We previously reported that illumination of an injected photosynthetic bacteriochlorophyll-derived pigment leads to a generation of reactive oxygen species, upon oxygen consumption in the blood stream. Consequently, paramagnetic deoxyhemoglobin accumulating in the illuminated area induces changes in image contrast, detectable by a Blood Oxygen Level Dependent (BOLD)-MRI protocol, termed photosensitized (ps)MRI. Here, we show that laser beam pulses synchronously trigger BOLD-contrast transients in the tissue, allowing representation of the luminous spatiotemporal profile, as a contrast map, on the MR monitor. Regions with enhanced BOLD-contrast (7-61 fold) were deduced as illuminated, and were found to overlap with the anatomical location of the incident light. Thus, we conclude that luminous information can be captured and translated by typical oxygen exchange processes in the blood of ordinary tissues, and made visible by psMRI (Fig. 1). This process represents a new channel for communicating environmental light into the body in certain analogy to light absorption by visual pigments in the retina where image perception takes place in the central nervous system. Potential applications of this finding may include: non-invasive intra-operative light guidance and follow-up of photodynamic interventions, determination of light diffusion in opaque tissues for optical imaging and possible assistance to the blind.


Subject(s)
Light , Magnetic Resonance Imaging/methods , Animals , Female , Rats , Rats, Wistar , Tumor Cells, Cultured
13.
Mol Cell Endocrinol ; 187(1-2): 19-22, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11988307

ABSTRACT

Magnetic resonance imaging (MRI) was employed for non-invasive analysis of vascular remodeling during follicular maturation in the PMSG/hCG rat ovary model. Changes in water diffusion and in perfusion led us to suggest that hypoxic stress may be a component in the regulation of angiogenesis in the growing follicle. However, in contrast with solid tumors of similar size, the spatial and temporal pattern of expression of vascular endothelial growth factor (VEGF), did not match the angiogenic response. The mismatch could be explained by the role of hyaluronan as a high molecular weight suppressor of angiogenesis maintaining an avascular follicular antrum.


Subject(s)
Magnetic Resonance Imaging , Neovascularization, Physiologic , Ovary/blood supply , Animals , Diffusion , Female , Hyaluronic Acid/physiology , Ovary/growth & development , Rats , Water
SELECTION OF CITATIONS
SEARCH DETAIL
...