Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Micromachines (Basel) ; 15(5)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38793132

RESUMO

Magnetic hyperthermia therapy (MHT) is a promising treatment modality for brain tumors using magnetic nanoparticles (MNPs) locally delivered to the tumor and activated with an external alternating magnetic field (AMF) to generate antitumor effects through localized heating. Magnetic particle imaging (MPI) is an emerging technology offering strong signal-to-noise for nanoparticle localization. A scoping review was performed by systematically querying Pubmed, Scopus, and Embase. In total, 251 articles were returned, 12 included. Articles were analyzed for nanoparticle type used, MHT parameters, and MPI applications. Preliminary results show that MHT is an exciting treatment modality with unique advantages over current heat-based therapies for brain cancer. Effective application relies on the further development of unique magnetic nanoparticle constructs and imaging modalities, such as MPI, that can enable real-time MNP imaging for improved therapeutic outcomes.

2.
Pharmaceuticals (Basel) ; 17(3)2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38543086

RESUMO

Magnetic hyperthermia therapy (MHT) is a re-emerging treatment modality for brain tumors where magnetic nanoparticles (MNPs) are locally delivered to the brain and then activated with an external alternating magnetic field (AMF) to generate localized heat at a site of interest. Due to the recent advancements in technology and theory surrounding the intervention, clinical and pre-clinical trials have demonstrated that MHT may enhance the effectiveness of chemotherapy and radiation therapy (RT) for the treatment of brain tumors. The future clinical success of MHT relies heavily on designing MNPs optimized for both heating and imaging, developing reliable methods for the local delivery of MNPs, and designing AMF systems with integrated magnetic particle imaging (MPI) for use in humans. However, despite the progression of technological development, the clinical progress of MHT has been underwhelming. This review aims to summarize the current state-of-the-art of MHT and offers insight into the current barriers and potential solutions for moving MHT forward.

3.
Neurosurg Clin N Am ; 34(2): 269-283, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36906333

RESUMO

Magnetic hyperthermia therapy (MHT) is a highly localized form of hyperthermia therapy (HT) that has been effective in treating various forms of cancer. Many clinical and preclinical studies have applied MHT to treat aggressive forms of brain cancer and assessed its role as a potential adjuvant to current therapies. Initial results show that MHT has a strong antitumor effect in animal studies and a positive association with overall survival in human glioma patients. Although MHT is a promising therapy with the potential to be incorporated into the future treatment of brain cancer, significant advancement of current MHT technology is required.


Assuntos
Neoplasias Encefálicas , Glioma , Hipertermia Induzida , Animais , Humanos , Hipertermia Induzida/métodos , Glioma/terapia , Neoplasias Encefálicas/terapia , Fenômenos Magnéticos
4.
Cancers (Basel) ; 15(2)2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36672278

RESUMO

We present in vivo validation of an automated magnetic hyperthermia therapy (MHT) device that uses real-time temperature input measured at the target to control tissue heating. MHT is a thermal therapy that uses heat generated by magnetic materials exposed to an alternating magnetic field. For temperature monitoring, we integrated a commercial fiber optic temperature probe containing four gallium arsenide (GaAs) temperature sensors. The controller device used temperature from the sensors as input to manage power to the magnetic field applicator. We developed a robust, multi-objective, proportional-integral-derivative (PID) algorithm to control the target thermal dose by modulating power delivered to the magnetic field applicator. The magnetic field applicator was a 20 cm diameter Maxwell-type induction coil powered by a 120 kW induction heating power supply operating at 160 kHz. Finite element (FE) simulations were performed to determine values of the PID gain factors prior to verification and validation trials. Ex vivo verification and validation were conducted in gel phantoms and sectioned bovine liver, respectively. In vivo validation of the controller was achieved in a canine research subject following infusion of magnetic nanoparticles (MNPs) into the brain. In all cases, performance matched controller design criteria, while also achieving a thermal dose measured as cumulative equivalent minutes at 43 °C (CEM43) 60 ± 5 min within 30 min.

5.
J Mater Chem B ; 10(26): 4935-4943, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35535802

RESUMO

Magnetic Fe3O4 nanoparticles "decorated" by LAPONITE® nanodisks have been materialized utilizing the Schikorr reaction following a facile approach and tested as mediators of heat for localized magnetic hyperthermia (MH) and as magnetic resonance imaging (MRI) agents. The synthetic protocol involves the interaction between two layered inorganic compounds, ferrous hydroxide, Fe(OH)2, and the synthetic smectite LAPONITE® clay Na0.7+[(Si8Mg5.5Li0.3)O20(OH)4]0.7-, towards the formation of superparamagnetic Fe3O4 nanoparticles, which are well decorated by the diamagnetic clay nanodisks. The latter imparts high negative ζ-potential values (up to -34.1 mV) to the particles, which provide stability against flocculation and precipitation, resulting in stable water dispersions. The obtained LAPONITE®-"decorated" Fe3O4 nanohybrids were characterized by powder X-ray diffraction (XRD), transmission electron microscopy (TEM), Mössbauer spectroscopy, dynamic light scattering (DLS) and vibrating sample magnetometry (VSM) at room temperature, revealing superior magnetic hyperthermia performance with specific absorption rate (SAR) values reaching 540 W gFe-1 (28 kA m-1, 150 kHz) for the hybrid material with a magnetic loading of 50 wt% Fe3O4/LAPONITE®. Toxicity studies were also performed with human glioblastoma (GBM) cells and human foreskin fibroblasts (HFF), which show negligible to no toxicity. Furthermore, T2-weighted MR imaging of rodent brain shows that the LAPONITE®-"decorated" Fe3O4 nanohybrids predominantly affected the transverse T2 relaxation time of tissue water, which resulted in a signal drop on the MRI T2-weighted imaging, allowing for imaging of the magnetic nanoparticles.


Assuntos
Hipertermia Induzida , Nanopartículas , Argila , Meios de Contraste/química , Compostos Férricos/química , Imageamento por Ressonância Magnética/métodos , Nanopartículas/química , Silicatos , Água/química
6.
Neuro Oncol ; 24(5): 694-707, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-34657158

RESUMO

BACKGROUND: Glioblastoma (GBM) remains a largely incurable disease as current therapy fails to target the invasive nature of glioma growth in disease progression and recurrence. Here, we use the FDA-approved drug and small molecule Hippo inhibitor Verteporfin (VP) to target YAP-TEAD activity, known to mediate convergent aspects of tumor invasion/metastasis, and assess the drug's efficacy and survival benefit in GBM models. METHODS: Up to 8 low-passage patient-derived GBM cell lines with distinct genomic drivers, including 3 primary/recurrent pairs, were treated with VP or vehicle (VEH) to assess in vitro effects on proliferation, migration, invasion, YAP-TEAD activity, and transcriptomics. Patient-derived orthotopic xenograft (PDX) models were used to assess VP's brain penetrance and effects on tumor burden and survival. RESULTS: VP treatment disturbed YAP/TAZ-TEAD activity; disrupted transcriptome signatures related to invasion, epithelial-to-mesenchymal, and proneural-to-mesenchymal transition, phenocopying TEAD1-knockout effects; and impaired tumor migration/invasion dynamics across primary and recurrent GBM lines. In an aggressive orthotopic PDX GBM model, short-term VP treatment consistently diminished core and infiltrative tumor burden, which was associated with decreased tumor expression of Ki67, nuclear YAP, TEAD1, and TEAD-associated targets EGFR, CDH2, and ITGB1. Finally, long-term VP treatment appeared nontoxic and conferred survival benefit compared to VEH in 2 PDX models: as monotherapy in primary (de novo) GBM and in combination with Temozolomide chemoradiation in recurrent GBM, where VP treatment associated with increased MGMT methylation. CONCLUSIONS: We demonstrate combined anti-invasive and anti-proliferative efficacy for VP with survival benefit in preclinical GBM models, indicating potential therapeutic value of this already FDA-approved drug if repurposed for GBM patients.


Assuntos
Glioblastoma , Glioma , Linhagem Celular Tumoral , Proliferação de Células , Glioblastoma/tratamento farmacológico , Humanos , Fatores de Transcrição/genética , Verteporfina/farmacologia , Verteporfina/uso terapêutico
7.
EBioMedicine ; 69: 103453, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34157482

RESUMO

Diffuse midline glioma (DMG) is an incurable malignancy with the highest mortality rate among pediatric brain tumors. While radiotherapy and chemotherapy are the most common treatments, these modalities have limited promise. Due to their diffuse nature in critical areas of the brain, the prognosis of DMG remains dismal. DMGs are characterized by unique phenotypic heterogeneity and histological features. Mutations of H3K27M, TP53, and ACVR1 drive DMG tumorigenesis. Histological artifacts include pseudopalisading necrosis and vascular endothelial proliferation. Mouse models that recapitulate human DMG have been used to study key driver mutations and the tumor microenvironment. DMG consists of a largely immunologically cold tumor microenvironment that lacks immune cell infiltration, immunosuppressive factors, and immune surveillance. While tumor-associated macrophages are the most abundant immune cell population, there is reduced T lymphocyte infiltration. Immunotherapies can stimulate the immune system to find, attack, and eliminate cancer cells. However, it is critical to understand the immune microenvironment of DMG before designing immunotherapies since differences in the microenvironment influence treatment efficacy. To this end, our review aims to overview the immune microenvironment of DMG, discuss emerging insights about the immune landscape that drives disease pathophysiology, and present recent findings and new opportunities for therapeutic discovery.


Assuntos
Neoplasias do Tronco Encefálico/imunologia , Glioma Pontino Intrínseco Difuso/imunologia , Imunoterapia/métodos , Microambiente Tumoral/imunologia , Animais , Neoplasias do Tronco Encefálico/genética , Neoplasias do Tronco Encefálico/terapia , Glioma Pontino Intrínseco Difuso/genética , Glioma Pontino Intrínseco Difuso/terapia , Humanos
8.
Neurooncol Adv ; 2(1): vdaa134, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33241215

RESUMO

BACKGROUND: Longitudinal tracking of tumor growth using noninvasive bioluminescence imaging (BLI) is a key approach for studies of in vivo cancer models, with particular relevance for investigations of malignant gliomas in rodent intracranial transplant paradigms. Akaluciferase (Akaluc) is a new BLI system with higher signal strength than standard firefly luciferase (Fluc). Here, we establish Akaluc BLI as a sensitive method for in vivo tracking of glioma expansion. METHODS: We engineered a lentiviral vector for expression of Akaluc in high-grade glioma cell lines, including patient-derived glioma stem cell (GSC) lines. Akaluc-expressing glioma cells were compared to matching cells expressing Fluc in both in vitro and in vivo BLI assays. We also conducted proof-of-principle BLI studies with intracranial transplant cohorts receiving chemoradiation therapy. RESULTS: Akaluc-expressing glioma cells produced more than 10 times higher BLI signals than Fluc-expressing counterparts when examined in vitro, and more than 100-fold higher signals when compared to Fluc-expressing counterparts in intracranial transplant models in vivo. The high sensitivity of Akaluc permitted detection of intracranial glioma transplants starting as early as 4 h after implantation and with as little as 5000 transplanted cells. The sensitivity of the system allowed us to follow engraftment and expansion of intracranial transplants of GSC lines. Akaluc was also robust for sensitive detection of in vivo tumor regression after therapy and subsequent relapse. CONCLUSION: Akaluc BLI offers superior sensitivity for in vivo tracking of glioma in the intracranial transplant paradigm, facilitating sensitive approaches for the study of glioma growth and response to therapy.

9.
Int J Hyperthermia ; 37(2): 3-19, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32672123

RESUMO

Hyperthermia therapy (HT) of cancer is a well-known treatment approach. With the advent of new technologies, HT approaches are now important for the treatment of brain tumors. We review current clinical applications of HT in neuro-oncology and ongoing preclinical research aiming to advance HT approaches to clinical practice. Laser interstitial thermal therapy (LITT) is currently the most widely utilized thermal ablation approach in clinical practice mainly for the treatment of recurrent or deep-seated tumors in the brain. Magnetic hyperthermia therapy (MHT), which relies on the use of magnetic nanoparticles (MNPs) and alternating magnetic fields (AMFs), is a new quite promising HT treatment approach for brain tumors. Initial MHT clinical studies in combination with fractionated radiation therapy (RT) in patients have been completed in Europe with encouraging results. Another combination treatment with HT that warrants further investigation is immunotherapy. HT approaches for brain tumors will continue to a play an important role in neuro-oncology.


Assuntos
Neoplasias Encefálicas , Hipertermia Induzida , Neoplasias Encefálicas/terapia , Europa (Continente) , Humanos , Terapia a Laser , Magnetismo , Nanopartículas
10.
Int J Hyperthermia ; 34(8): 1316-1328, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29353516

RESUMO

Hyperthermia therapy (HT) is the exposure of a region of the body to elevated temperatures to achieve a therapeutic effect. HT anticancer properties and its potential as a cancer treatment have been studied for decades. Techniques used to achieve a localised hyperthermic effect include radiofrequency, ultrasound, microwave, laser and magnetic nanoparticles (MNPs). The use of MNPs for therapeutic hyperthermia generation is known as magnetic hyperthermia therapy (MHT) and was first attempted as a cancer therapy in 1957. However, despite more recent advancements, MHT has still not become part of the standard of care for cancer treatment. Certain challenges, such as accurate thermometry within the tumour mass and precise tumour heating, preclude its widespread application as a treatment modality for cancer. MHT is especially attractive for the treatment of glioblastoma (GBM), the most common and aggressive primary brain cancer in adults, which has no cure. In this review, the application of MHT as a therapeutic modality for GBM will be discussed. Its therapeutic efficacy, technical details, and major experimental and clinical findings will be reviewed and analysed. Finally, current limitations, areas of improvement, and future directions will be discussed in depth.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Hipertermia Induzida , Fenômenos Magnéticos , Animais , Humanos , Resultado do Tratamento
11.
J Neurooncol ; 137(3): 653-663, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29350351

RESUMO

Cetuximab conjugated iron-oxide nanoparticles (cetuximab-IONPs) have shown both in-vitro and in-vivo anti-tumor efficacy against gliomas. The purpose of this pilot study was to evaluate the safety and potential efficacy of cetuximab-IONPs for treatment of spontaneously occurring intracranial gliomas in canines after convection-enhanced delivery (CED). The use of CED allowed for direct infusion of the cetuximab-IONPs both intratumorally and peritumorally avoiding the blood brain barrier (BBB) and limiting systemic effects. A total of eight dogs participated in the study and only two developed mild post-operative complications, which resolved with medical therapy. All canines underwent a single CED treatment of the cetuximab-IONPs over 3 days and did not receive any further adjuvant treatments. Volumetric analysis showed a median reduction in tumor size of 54.9% by MRI at 1-month (4-6 weeks) follow-up. Five dogs were euthanized due to recurrence of neurological signs other than seizures, two due to recurrent seizures, and one dog died in his sleep. Median survival time after surgery was 248 days (mean 367 days).


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Neoplasias Encefálicas/veterinária , Cetuximab/administração & dosagem , Doenças do Cão/tratamento farmacológico , Glioma/veterinária , Nanopartículas de Magnetita/administração & dosagem , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/cirurgia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/cirurgia , Convecção , Doenças do Cão/diagnóstico por imagem , Doenças do Cão/cirurgia , Cães , Feminino , Seguimentos , Glioma/diagnóstico por imagem , Glioma/tratamento farmacológico , Glioma/cirurgia , Bombas de Infusão Implantáveis , Masculino , Resultado do Tratamento , Carga Tumoral
12.
Sci Rep ; 7(1): 15593, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29142297

RESUMO

Glioblastoma (GBM) contains diverse microenvironments with uneven distributions of oncogenic alterations and signaling networks. The diffusely infiltrative properties of GBM result in residual tumor at neurosurgical resection margins, representing the source of relapse in nearly all cases and suggesting that therapeutic efforts should be focused there. To identify signaling networks and potential druggable targets across tumor microenvironments (TMEs), we utilized 5-ALA fluorescence-guided neurosurgical resection and sampling, followed by proteomic analysis of specific TMEs. Reverse phase protein array (RPPA) was performed on 205 proteins isolated from the tumor margin, tumor bulk, and perinecrotic regions of 13 previously untreated, clinically-annotated and genetically-defined high grade gliomas. Differential protein and pathway signatures were established and then validated using western blotting, immunohistochemistry, and comparable TCGA RPPA datasets. We identified 37 proteins differentially expressed across high-grade glioma TMEs. We demonstrate that tumor margins were characterized by pro-survival and anti-apoptotic proteins, whereas perinecrotic regions were enriched for pro-coagulant and DNA damage response proteins. In both our patient cohort and TCGA cases, the data suggest that TMEs possess distinct protein expression profiles that are biologically and therapeutically relevant.


Assuntos
Glioblastoma/genética , Recidiva Local de Neoplasia/genética , Neoplasia Residual/genética , Proteômica , Adulto , Idoso , Ácido Aminolevulínico/administração & dosagem , Receptores ErbB/genética , Feminino , Fluorescência , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Masculino , Margens de Excisão , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/cirurgia , Neoplasia Residual/tratamento farmacológico , Neoplasia Residual/patologia , Neoplasia Residual/cirurgia , PTEN Fosfo-Hidrolase/genética , Análise Serial de Proteínas , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/genética
13.
Neural Comput ; 29(3): 643-678, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27764592

RESUMO

The communication-through-coherence (CTC) hypothesis states that a sending group of neurons will have a particularly strong effect on a receiving group if both groups oscillate in a phase-locked ("coherent") manner (Fries, 2005 , 2015 ). Here, we consider a situation with two visual stimuli, one in the focus of attention and the other distracting, resulting in two sites of excitation at an early cortical area that project to a common site in a next area. Taking a modeler's perspective, we confirm the workings of a mechanism that was proposed by Bosman et al. ( 2012 ) in the context of providing experimental evidence for the CTC hypothesis: a slightly higher gamma frequency of the attended sending site compared to the distracting site may cause selective interareal synchronization with the receiving site if combined with a slow-rhythm gamma phase reset. We also demonstrate the relevance of a slightly lower intrinsic frequency of the receiving site for this scenario. Moreover, we discuss conditions for a transition from bottom-up to top-down driven phase locking.

14.
Anal Chem ; 88(1): 858-67, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26587976

RESUMO

Intraoperative cancer imaging and fluorescence-guided surgery have attracted considerable interest because fluorescence signals can provide real-time guidance to assist a surgeon in differentiating cancerous and normal tissues. Recent advances have led to the clinical use of a natural fluorophore called protoporphyrin IX (PpIX) for image-guided surgical resection of high-grade brain tumors (glioblastomas). However, traditional fluorescence imaging methods have only limited detection sensitivity and identification accuracy and are unable to detect low-grade or diffuse infiltrating gliomas (DIGs). Here we report a low-cost hand-held spectroscopic device that is capable of ultrasensitive detection of protoporphyrin IX fluorescence in vivo, together with intraoperative spectroscopic data obtained from both animal xenografts and human brain tumor specimens. The results indicate that intraoperative spectroscopy is at least 3 orders of magnitude more sensitive than the current surgical microscopes, allowing ultrasensitive detection of as few as 1000 tumor cells. For detection specificity, intraoperative spectroscopy allows the differentiation of brain tumor cells from normal brain cells with a contrast signal ratio over 100. In vivo animal studies reveal that protoporphyrin IX fluorescence is strongly correlated with both MRI and histological staining, confirming that the fluorescence signals are highly specific to tumor cells. Furthermore, ex vivo spectroscopic studies of excised brain tissues demonstrate that the hand-held spectroscopic device is capable of detecting diffuse tumor margins with low fluorescence contrast that are not detectable with current systems in the operating room. These results open new opportunities for intraoperative detection and fluorescence-guided resection of microscopic and low-grade glioma brain tumors with invasive or diffusive margins.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Monitorização Intraoperatória , Cirurgia Assistida por Computador , Animais , Linhagem Celular Tumoral , Fluorescência , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Camundongos , Camundongos Nus , Espectrofotometria
15.
J Neurooncol ; 124(1): 13-22, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25981803

RESUMO

The epidermal growth factor receptor deletion variant EGFRvIII is known to be expressed in a subset of patients with glioblastoma (GBM) tumors that enhances tumorigenicity and also accounts for radiation and chemotherapy resistance. Targeting the EGFRvIII deletion mutant may lead to improved GBM therapy and better patient prognosis. Multifunctional magnetic nanoparticles serve as a potential clinical tool that can provide cancer cell targeted drug delivery, imaging, and therapy. Our previous studies have shown that an EGFRvIII-specific antibody and cetuximab (an EGFR- and EGFRvIII-specific antibody), when bioconjugated to IONPs (EGFRvIII-IONPs or cetuximab-IONPs respectively), can simultaneously provide sensitive cancer cell detection by magnetic resonance imaging (MRI) and targeted therapy of experimental GBM. In this study, we investigated whether cetuximab-IONPs can additionally allow for the radiosensitivity enhancement of GBM. Cetuximab-IONPs were used in combination with single (10 Gy × 1) or multiple fractions (10 Gy × 2) of ionizing radiation (IR) for radiosensitization of EGFRvIII-overexpressing human GBM cells in vitro and in vivo after convection-enhanced delivery (CED). A significant GBM antitumor effect was observed in vitro after treatment with cetuximab-IONPs and subsequent single or fractionated IR. A significant increase in overall survival of nude mice implanted with human GBM xenografts was found after treatment by cetuximab-IONP CED and subsequent fractionated IR. Increased DNA double strands breaks (DSBs), as well as increased reactive oxygen species (ROS) formation, were felt to represent the mediators of the observed radiosensitization effect with the combination therapy of IR and cetuximab-IONPs treatment.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Cetuximab/administração & dosagem , Receptores ErbB/administração & dosagem , Glioblastoma/tratamento farmacológico , Glioblastoma/radioterapia , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Receptores ErbB/imunologia , Compostos Férricos/química , Glioblastoma/patologia , Humanos , Nanopartículas de Magnetita/química , Camundongos , Tolerância a Radiação , Espécies Reativas de Oxigênio
16.
Oncotarget ; 6(11): 8788-806, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25871395

RESUMO

Malignant gliomas remain aggressive and lethal primary brain tumors in adults. The epidermal growth factor receptor (EGFR) is frequently overexpressed in the most common malignant glioma, glioblastoma (GBM), and represents an important therapeutic target. GBM stem-like cells (GSCs) present in tumors are felt to be highly tumorigenic and responsible for tumor recurrence. Multifunctional magnetic iron-oxide nanoparticles (IONPs) can be directly imaged by magnetic resonance imaging (MRI) and designed to therapeutically target cancer cells. The targeting effects of IONPs conjugated to the EGFR inhibitor, cetuximab (cetuximab-IONPs), were determined with EGFR- and EGFRvIII-expressing human GBM neurospheres and GSCs. Transmission electron microscopy revealed cetuximab-IONP GBM cell binding and internalization. Fluorescence microscopy and Prussian blue staining showed increased uptake of cetuximab-IONPs by EGFR- as well as EGFRvIII-expressing GSCs and neurospheres in comparison to cetuximab or free IONPs. Treatment with cetuximab-IONPs resulted in a significant antitumor effect that was greater than with cetuximab alone due to more efficient, CD133-independent cellular targeting and uptake, EGFR signaling alterations, EGFR internalization, and apoptosis induction in EGFR-expressing GSCs and neurospheres. A significant increase in survival was found after cetuximab-IONP convection-enhanced delivery treatment of 3 intracranial rodent GBM models employing human EGFR-expressing GBM xenografts.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Cetuximab/farmacologia , Glioblastoma/tratamento farmacológico , Nanopartículas de Magnetita/administração & dosagem , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Células Cultivadas , Cetuximab/administração & dosagem , Cetuximab/toxicidade , Portadores de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glioblastoma/patologia , Humanos , Nanopartículas de Magnetita/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas de Neoplasias/análise , Neurônios/efeitos dos fármacos , Polietilenoglicóis/administração & dosagem , Esferoides Celulares , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Neurosurg Clin N Am ; 23(3): 439-49, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22748656

RESUMO

Glioblastoma remains one of the most difficult cancers to treat and represents the most common primary malignancy of the brain. Although conventional treatments have found modest success in reducing the initial tumor burden, infiltrating cancer cells beyond the main mass are responsible for tumor recurrence and ultimate patient demise. Targeting residual infiltrating cancer cells requires the development of new treatment strategies. The emerging field of cancer nanotechnology holds promise in the use of multifunctional nanoparticles for imaging and targeted therapy of glioblastoma. This article examines the current state of nanotechnology in the treatment of glioblastoma and directions of further study.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Nanopartículas/uso terapêutico , Braquiterapia/instrumentação , Meios de Contraste/uso terapêutico , Sistemas de Liberação de Medicamentos/instrumentação , Técnicas de Transferência de Genes/instrumentação , Humanos , Nanotecnologia/tendências , Fototerapia/instrumentação
18.
Expert Rev Clin Pharmacol ; 5(2): 173-86, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22390560

RESUMO

Magnetic nanoparticles (MNPs) represent a promising nanomaterial for the targeted therapy and imaging of malignant brain tumors. Conjugation of peptides or antibodies to the surface of MNPs allows direct targeting of the tumor cell surface and potential disruption of active signaling pathways present in tumor cells. Delivery of nanoparticles to malignant brain tumors represents a formidable challenge due to the presence of the blood-brain barrier and infiltrating cancer cells in the normal brain. Newer strategies permit better delivery of MNPs systemically and by direct convection-enhanced delivery to the brain. Completion of a human clinical trial involving direct injection of MNPs into recurrent malignant brain tumors for thermotherapy has established their feasibility, safety and efficacy in patients. Future translational studies are in progress to understand the promising impact of MNPs in the treatment of malignant brain tumors.


Assuntos
Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/terapia , Meios de Contraste/uso terapêutico , Glioblastoma/diagnóstico , Glioblastoma/terapia , Nanopartículas de Magnetita/uso terapêutico , Animais , Barreira Hematoencefálica/fisiologia , Ensaios Clínicos como Assunto , Humanos , Hipertermia Induzida/métodos , Imageamento por Ressonância Magnética/métodos , Magnetismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...