Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Neurooncol ; 147(1): 125-133, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31989489

RESUMO

INTRODUCTION: Tumor Treating Fields (TTFields) are alternating electric fields at 200 kHz that disrupt tumor cells as they undergo mitosis. Patient survival benefit has been demonstrated in randomized clinical trials but much of the data are available only for supratentorial glioblastomas. We investigated a series of alternative array configurations for the posterior fossa to determine the electric field coverage of a cerebellar glioblastoma. METHODS: Semi-automated segmentation of neuro-anatomical structures was performed while the gross tumor volume (GTV) was manually delineated. A three-dimensional finite-element mesh was generated and then solved for field distribution. RESULTS: Compared to the supratentorial array configuration, the alternative array configurations consist of posterior displacement the 2 lateral opposing arrays and inferior displacement of the posteroanterior array, resulting in an average increase of 46.6% electric field coverage of the GTV as measured by the area under the curve of the electric field-volume histogram (EAUC). Hotspots, or regions of interest with the highest 5% of TTFields intensity (E5%), had an average increase of 95.6%. Of the 6 posterior fossa configurations modeled, the PAHorizontal arrangement provided the greatest field coverage at the GTV when the posteroanterior array was placed centrally along the patient's posterior neck and horizontally parallel, along the longer axis, to the coronal plane of the patient's head. Varying the arrays also produced hotspots proportional to TTFields coverage. CONCLUSIONS: Our finite element modeling showed that the alternative array configurations offer an improved TTFields coverage to the cerebellar tumor compared to the conventional supratentorial configuration.


Assuntos
Neoplasias Cerebelares/terapia , Terapia por Estimulação Elétrica/métodos , Glioblastoma/terapia , Neoplasias Infratentoriais/terapia , Feminino , Análise de Elementos Finitos , Humanos , Pessoa de Meia-Idade
2.
BMC Neurol ; 16: 9, 2016 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-26849888

RESUMO

BACKGROUND: Patients with intravascular lymphoma (IVL) frequently have neurological signs and symptoms. Prompt diagnosis and treatment is therefore crucial for their survival. However, the spectrum of neurological presentations and their respective frequencies have not been adequately characterized. Our aim is to document the spectrum of clinical symptoms and their respective frequencies and to create a clinical framework for the prompt diagnosis of IVL. METHODS: A comprehensive meta-analysis of 654 cases of IVL published between 1957 and 2012 was performed to provide better insight into the neurological presentations of this disease. Neurologic complications were mainly divided into central nervous system (CNS) and peripheral nervous system (PNS) presentations. RESULTS: There were no differences in occurrences of CNS IVL based on gender or geographic locations (Asian Vs non-Asian). However, most patients with CNS IVL were younger than 70 years of age (p < 0.05). Our limited data do not support the treatment efficacy of methotrexate. CNS symptoms were seen in 42% of all cases. The most common CNS complications identified were cognitive impairment/dementia (60.9%), paralysis (22.2%), and seizures (13.4%). PNS complications were seen in 9.5% of cases. Out of these, muscle weakness (59.7%), neurogenic bladder (37.1%), and paresthesia (16.1%) were the most common presentations. CONCLUSIONS: CNS complications are more common among IVL patients. Out of these, dementia and seizures outnumber stroke-like presentations.


Assuntos
Doenças do Sistema Nervoso Central/etiologia , Linfoma Difuso de Grandes Células B/complicações , Doenças do Sistema Nervoso Periférico/etiologia , Neoplasias Vasculares/complicações , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
3.
Curr Neurol Neurosci Rep ; 16(1): 8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26739692

RESUMO

As with many cancer treatments, tumor treating fields (TTFields) target rapidly dividing tumor cells. During mitosis, TTFields-exposed cells exhibit uncontrolled membrane blebbing at the onset of anaphase, resulting in aberrant mitotic exit. Based on these criteria, at least two protein complexes have been proposed as TTFields' molecular targets, including α/ß-tubulin and the septin 2, 6, 7 heterotrimer. After aberrant mitotic exit, cells exhibited abnormal nuclei and signs of cellular stress, including decreased cellular proliferation and p53 dependence, and exhibit the hallmarks of immunogenic cell death, suggesting that TTFields treatment may induce an antitumor immune response. Clinical trials lead to Food and Drug Administration approval for their treatment of recurrent glioblastoma. Detailed modeling of TTFields within the brain suggests that the location of the tumor may affect treatment efficacy. These observations have a profound impact on the use of TTFields in the clinic, including what co-therapies may be best applied to boost its efficacy.


Assuntos
Terapia por Estimulação Elétrica , Glioma/terapia , Animais , Terapia por Estimulação Elétrica/métodos , Glioblastoma/terapia , Humanos , Mitose , Estresse Fisiológico , Resultado do Tratamento
4.
Curr Treat Options Oncol ; 16(8): 40, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26143265

RESUMO

OPINION STATEMENT: Glioblastoma is a deadly disease and even aggressive neurosurgical resection followed by radiation and chemotherapy only extends patient survival to a median of 1.5 years. The challenge in treating this type of tumor stems from the rapid proliferation of the malignant glioma cells, the diffuse infiltrative nature of the disease, multiple activated signal transduction pathways within the tumor, development of resistant clones during treatment, the blood brain barrier that limits the delivery of drugs into the central nervous system, and the sensitivity of the brain to treatment effect. Therefore, new therapies that possess a unique mechanism of action are needed to treat this tumor. Recently, alternating electric fields, also known as tumor treating fields (TTFields), have been developed for the treatment of glioblastoma. TTFields use electromagnetic energy at an intermediate frequency of 200 kHz as a locoregional intervention and act to disrupt tumor cells as they undergo mitosis. In a phase III clinical trial for recurrent glioblastoma, TTFields were shown to have equivalent efficacy when compared to conventional chemotherapies, while lacking the typical side effects associated with chemotherapies. Furthermore, an interim analysis of a recent clinical trial in the upfront setting demonstrated superiority to standard of care cytotoxic chemotherapy, most likely because the subjects' tumors were at an earlier stage of clonal evolution, possessed less tumor-induced immunosuppression, or both. Therefore, it is likely that the efficacy of TTFields can be increased by combining it with other anti-cancer treatment modalities.


Assuntos
Neoplasias Encefálicas/terapia , Radiação Eletromagnética , Glioma/terapia , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/mortalidade , Glioma/diagnóstico , Glioma/mortalidade , Humanos , Recidiva Local de Neoplasia , Resultado do Tratamento
5.
Adv Radiat Oncol ; 9(1): 101316, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38260214

RESUMO

Tumor treating fields (TTFields) are nonionizing alternating electric fields that have anticancer properties. After the initial approval for use in patients with recurrent glioblastoma in 2011 and newly diagnosed glioblastomas in 2015, they are now being tested in those with advanced lung cancer, ovarian carcinoma, and pancreatic cancer. Unlike ionizing radiation therapy, TTFields have nonlinear propagation characteristics; therefore, it is difficult for clinicians to recognize intuitively the location where these fields have the most impact. However, finite element analysis offers a means of delineating TTFields in the human body. Our analyses in the brain, pelvis, and thorax revealed that cerebrospinal fluid, edema, urine, ascites, pleural fluid, and necrotic core within a tumor greatly influence their distribution within these body cavities. Our observations thus provided a unified framework on the role of these compartmentalized fluids in influencing the propagation of TTFields.

6.
Neurooncol Adv ; 6(1): vdae032, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38560348

RESUMO

Tumor-treating fields (TTFields) are currently a Category 1A treatment recommendation by the US National Comprehensive Cancer Center for patients with newly diagnosed glioblastoma. Although the mechanism of action of TTFields has been partly elucidated, tangible and standardized metrics are lacking to assess antitumor dose and effects of the treatment. This paper outlines and evaluates the current standards and methodologies in the estimation of the TTFields distribution and dose measurement in the brain and highlights the most important principles governing TTFields dosimetry. The focus is on clinical utility to facilitate a practical understanding of these principles and how they can be used to guide treatment. The current evidence for a correlation between TTFields dose, tumor growth, and clinical outcome will be presented and discussed. Furthermore, we will provide perspectives and updated insights into the planning and optimization of TTFields therapy for glioblastoma by reviewing how the dose and thermal effects of TTFields are affected by factors such as tumor location and morphology, peritumoral edema, electrode array position, treatment duration (compliance), array "edge effect," electrical duty cycle, and skull-remodeling surgery. Finally, perspectives are provided on how to optimize the efficacy of future TTFields therapy.

7.
Adv Radiat Oncol ; 8(4): 101203, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37213481

RESUMO

Purpose: Tumor Treating Fields (TTFields) are alternating electric fields at 150 to 200 kHz that exert their anticancer effect by destroying tumor cells when they undergo mitosis. TTFields are currently being tested in patients with non-small cell lung cancer with advanced disease (NCT02973789) and those with brain metastasis (NCT02831959). However, the distribution of these fields within the thoracic compartment remains poorly understood. Methods and Materials: Using positron emission tomography-computed tomography image data sets obtained from a series of 4 patients with poorly differentiated adenocarcinoma, the positron emission tomography-positive gross tumor volume (GTV), clinical target volume (CTV), and structures from the chest surface to the intrathoracic compartment were manually segmented, followed by 3-dimensional physics simulation and computational modeling using finite element analysis. Electric field-volume histograms, specific absorption rate-volume histograms, and current density-volume histograms were generated to produce plan quality metrics (95%, 50%, and 5% volumes) for quantitative comparisons between models. Results: Unlike other organs in the body, the lungs have a large volume of air, which has a very low electric conductivity value. Our comprehensive and individualized models demonstrated heterogeneity in electric field penetration to the GTVs with differences upwards of 200% and yielded a diverse range of TTFields distributions. Target contact with the conductive pleura intensified TTFields at the GTV and CTV. Furthermore, in a sensitivity analysis, varying electric conductivity and mass density of the CTV altered TTFields coverage to both the CTV and GTV. Conclusions: Personalized modeling is important to accurately estimate target coverage at the tumor volumes and surrounding normal tissue structures in the thorax.

8.
Adv Radiat Oncol ; 8(1): 101046, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36483066

RESUMO

Purpose: Cerebral edema is an important component of brain metastasis, and its presence may alter the distribution of tumor-treating fields (TTFields). We therefore performed a computational study to model the extent of this alteration according to various edema conditions associated with the metastasis. Methods and Materials: Postacquisition magnetic resonance imaging data sets were obtained from 2 patients with solitary brain metastases from non-small cell lung cancer. After delineation of various anatomies, a 3-dimensional finite element mesh model was generated and then solved for the distribution of applied electric fields, rate of energy deposition, and current density at the gross tumor volume (GTV), edema, and other cranial structures. Electric field-volume histograms, specific absorption rate-volume histograms, and current density-volume histograms were generated, by which plan quality metrics were derived from and used to evaluate relative differences in field coverage between models under various conditions. Results: Changes in the conductivity of cerebral edema altered the electric fields, rate of energy deposition, and current density at the GTV region. At the cerebral edema region, increasing electric conductivity of the edema only decreased the electric fields and rate of energy deposition while the current density increased. The ratio of edema-to-tumor is also important because the plan quality metrics increased linearly when the edema-to-GTV ratio decreased, and increased vice versa. Furthermore, a conductive necrotic core additionally altered the distribution of TTFields according to the plan quality metrics. Conclusions: Our modeling study demonstrated that cerebral edema alters the distribution of applied TTFields in patients. Personalized treatment planning will need to take into account the modulating effects of cerebral edema on TTFields as well as additional effects from a necrotic core inside the GTV.

9.
J Neurooncol ; 105(2): 261-73, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21512826

RESUMO

Extracranial metastasis is a unique but rare manifestation of glioblastoma multiforme. It is thought to arise from glioblastoma cells disseminated into the blood stream. We undertook a comprehensive analysis of 88 cases of extracranial glioblastoma (5 were gliosarcomas) published between 1928 and 2009. Cases included in the analysis were primary or secondary glioblastomas that subsequently invaded organs outside the brain or spinal cord. The median age was 38 years and the median overall survival time was 10.5 months (range 0.0-60.0 months). The median time from symptom onset to diagnosis of primary glioblastoma was 2.5 months, from diagnosis to detection of extracranial metastasis was 8.5 months, and from metastasis to death was 1.5 months. From 1940 to 2009, there has been progressive lengthening of the interval from detection of extracranial metastasis to death, at a rate of 0.7 months per decade (95% confidence interval 0.5-1.0 month). Use of magnetic resonance imaging correlates with an increase in overall survival but not age, gender, or site of primary glioblastoma. Patients treated with surgery + radiation + chemotherapy + cerebrospinal fluid shunting had the longest average survival interval from metastasis to death when compared to those treated with surgery alone, radiation alone, surgery + radiation, and surgery + radiation + chemotherapy. Lung metastasis is a prognostic factor of extremely poor outcomes. We conclude that patients with glioblastoma extracranial metastasis have poor prognosis, but there has been a progressive lengthening of survival in each successive decade from 1940 to 2000.


Assuntos
Neoplasias Encefálicas/secundário , Glioblastoma/patologia , Gliossarcoma/patologia , Adolescente , Adulto , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Criança , Feminino , Glioblastoma/mortalidade , Glioblastoma/terapia , Gliossarcoma/mortalidade , Gliossarcoma/terapia , Humanos , Masculino , Metanálise como Assunto , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida , Adulto Jovem
10.
Adv Radiat Oncol ; 6(4): 100716, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34409211

RESUMO

PURPOSE: Since the inception of tumor treating fields (TTFields) therapy as a Food and Drug Administration-approved treatment with known clinical efficacy against recurrent and newly diagnosed glioblastoma, various in silico modeling studies have been performed in an effort to better understand the distribution of applied electric fields throughout the human body for various malignancies or metastases. METHODS AND MATERIALS: Postacquisition attenuation-corrected positron emission tomography-computed tomography image data sets from 2 patients with ovarian carcinoma were used to fully segment various intrapelvic and intra-abdominal gross anatomic structures. A 3-dimensional finite element mesh model was generated and then solved for the distribution of applied electric fields, rate of energy deposition, and current density at the clinical target volumes (CTVs) and other intrapelvic and intra-abdominal structures. Electric field-volume histograms, specific absorption rate-volume histograms, and current density-volume histograms were generated, by which plan quality metrics were derived from and used to evaluate relative differences in field coverage between models under various conditions. RESULTS: TTFields therapy distribution throughout the pelvis and abdomen was largely heterogeneous, where specifically the field intensity at the CTV was heavily influenced by surrounding anatomic structures as well as its shape and location. The electric conductivity of the CTV had a direct effect on the field strength within itself, as did the position of the arrays on the surface of the pelvis and/or abdomen. CONCLUSION: The combined use of electric field-volume histograms, specific absorption rate-volume histograms, current density-volume histograms, and plan quality metrics enables a personalized method to dosimetrically evaluate patients receiving TTFields therapy for ovarian carcinoma when certain patient- and tumor-specific factors are integrated with the treatment plan.

11.
J Vis Exp ; (146)2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-31058901

RESUMO

Glioblastoma is the most common and lethal form of brain cancer, with a median survival of 15 months after diagnosis and a 5 year survival rate of only 5% with current standard of care. Tumors often recur within 9 months following initial surgery, radiation and chemotherapy, at which point treatment options become limited. This highlights the pressing need for the development of better therapeutics to prolong survival and increase the quality of life for these patients. Tumor Treating Fields (TTFields) therapy was developed to take advantage of the effect of low frequency alternating electrical fields on cells for cancer therapy. TTFields have been demonstrated to disrupt cells during mitosis and slow tumor growth. There is also growing evidence that they act through stimulating immune responses within exposed tumors. The advantages of TTFields therapy include its noninvasive approach and increased quality of life compared to other treatment modalities such as cytotoxic chemotherapies. The Food and Drug Administration approved TTFields therapy for the treatment of recurrent glioblastoma in 2011 and for newly diagnosed glioblastoma in 2015. We report on the effects of TTFields during mitosis, the results of electric fields modeling, and proper transducer array placement. Our protocol outlines the clinical application of TTFields on a patient post-surgery, using the second-generation device.


Assuntos
Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica , Glioblastoma/terapia , Terapia Combinada , Condutividade Elétrica , Células HeLa , Humanos , Imageamento por Ressonância Magnética , Mitose
12.
Prog Neurol Surg ; 32: 180-195, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990984

RESUMO

Alternating electric fields of intermediate frequencies, also known as Tumor Treating Fields (TTFields or TTF) is a novel anticancer treatment modality that disrupts tumor cell mitosis at the metaphase-anaphase transition, leading to mitotic catastrophe, aberrant mitotic exit, and/or cell death. It is realized through alteration of the cytokinetic cleavage furrow by interference of proteins possessing large dipole moments, like septin heterotrimer complex and α/ß-tubulin, and that results in disordered membrane contraction and failed cytokinesis. Aberrant mitotic exit also elicits immunogenic cell death, which may potentiate an immune response against treated tumors. Notably, in patients with recurrent glioblastoma multiforme (GBM) a prospective clinical trial demonstrated comparable overall survival and progression-free survival after TTFields therapy and best physician's choice chemotherapy. Moreover, it was shown that in patients with newly diagnosed GBM initially treated with standard chemoradiotherapy with daily temozolomide (TMZ), adjuvant TTFields combined with TMZ offered better survival than adjuvant TMZ alone. Therefore, TTFields therapy can be appreciated as a standard treatment option in cases of intracranial malignant gliomas, whereas future studies should establish its optimal combination with other existing anticancer modalities, which may offer additional survival benefits for patients.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/terapia , Terapia Combinada/métodos , Dacarbazina/análogos & derivados , Terapia por Estimulação Elétrica/métodos , Campos Eletromagnéticos , Glioma/terapia , Mitose , Recidiva Local de Neoplasia/terapia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/farmacologia , Glioma/tratamento farmacológico , Humanos , Recidiva Local de Neoplasia/tratamento farmacológico , Temozolomida
13.
Am J Clin Oncol ; 41(12): 1281-1287, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29528867

RESUMO

OBJECTIVES: This systematic review aims to investigate spinal cord glioblastoma (scGBM) and correlations between patient traits and survival outcome, as well as differences in cohorts administered temozolomide or total resections, through an analysis of published cases reported up to October 2016. METHODS: We obtained patient data by querying PubMed and Google Scholar with predetermined search terms and inclusion criteria that enabled the identification of relevant case reports. Survival was compared using Kaplan-Meier curves and log-rank analyses. RESULTS: Of 153 patients with scGBM identified through a literature search, 135 met the predetermined search and inclusion criteria. Median overall survival (OS) for the resulting cohort was 12 (95% CI, 10-14) months. The female sex was found to significantly predict worse outcomes, and a sizable number of patients with long-term disease were found to have afflictions of the thoracic spinal cord. Neither the pediatric, temozolomide nor total resection subgroups had significantly improved survival characteristics, by log-rank analysis, relative to counterparts. CONCLUSIONS: These data elucidate the characteristics of patients with scGBM. For more sophisticated and in-depth analyses in the future, it is imperative that time-of-treatment information is recorded in future case reports. In addition, all case reports should be made available to prevent publication bias.


Assuntos
Glioblastoma/mortalidade , Recidiva Local de Neoplasia/mortalidade , Neoplasias da Medula Espinal/mortalidade , Terapia Combinada , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/terapia , Prognóstico , Neoplasias da Medula Espinal/patologia , Neoplasias da Medula Espinal/terapia , Taxa de Sobrevida
14.
Cancer Med ; 6(6): 1286-1300, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28544575

RESUMO

Tumor Treating Fields (TTFields) therapy is an approved treatment that has known clinical efficacy against recurrent and newly diagnosed glioblastoma. However, the distribution of the electric fields and the corresponding pattern of energy deposition in the brain are poorly understood. To evaluate the physical parameters that may influence TTFields, postacquisition MP-RAGE, T1 and T2 MRI sequences from a responder with a right parietal glioblastoma were anatomically segmented and then solved using finite-element method to determine the distribution of the electric fields and rate of energy deposition at the gross tumor volume (GTV) and other intracranial structures. Electric field-volume histograms (EVH) and specific absorption rate-volume histograms (SARVH) were constructed to numerically evaluate the relative and/or absolute magnitude volumetric differences between models. The electric field parameters EAUC , VE150 , E95% , E50% , and E20% , as well as the SAR parameters SARAUC , VSAR7.5 , SAR95% , SAR50% , and SAR20% , facilitated comparisons between models derived from various conditions. Specifically, TTFields at the GTV were influenced by the dielectric characteristics of the adjacent tissues as well as the GTV itself, particularly the presence or absence of a necrotic core. The thickness of the cerebrospinal fluid on the convexity of the brain and the geometry of the tumor were also relevant factors. Finally, the position of the arrays also influenced the electric field distribution and rate of energy deposition in the GTV. Using EVH and SARVH, a personalized approach for TTFields treatment can be developed when various patient-related and tumor-related factors are incorporated into the planning procedure.


Assuntos
Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica , Glioblastoma/terapia , Modelos Neurológicos , Neoplasias Encefálicas/diagnóstico por imagem , Condutividade Elétrica , Glioblastoma/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética
15.
Phys Med Biol ; 62(21): 8264-8282, 2017 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-29023236

RESUMO

Tumor Treating Fields (TTFields) therapy is an approved modality of treatment for glioblastoma. Patient anatomy-based finite element analysis (FEA) has the potential to reveal not only how these fields affect tumor control but also how to improve efficacy. While the automated tools for segmentation speed up the generation of FEA models, multi-step manual corrections are required, including removal of disconnected voxels, incorporation of unsegmented structures and the addition of 36 electrodes plus gel layers matching the TTFields transducers. Existing approaches are also not scalable for the high throughput analysis of large patient volumes. A semi-automated workflow was developed to prepare FEA models for TTFields mapping in the human brain. Magnetic resonance imaging (MRI) pre-processing, segmentation, electrode and gel placement, and post-processing were all automated. The material properties of each tissue were applied to their corresponding mask in silico using COMSOL Multiphysics (COMSOL, Burlington, MA, USA). The fidelity of the segmentations with and without post-processing was compared against the full semi-automated segmentation workflow approach using Dice coefficient analysis. The average relative differences for the electric fields generated by COMSOL were calculated in addition to observed differences in electric field-volume histograms. Furthermore, the mesh file formats in MPHTXT and NASTRAN were also compared using the differences in the electric field-volume histogram. The Dice coefficient was less for auto-segmentation without versus auto-segmentation with post-processing, indicating convergence on a manually corrected model. An existent but marginal relative difference of electric field maps from models with manual correction versus those without was identified, and a clear advantage of using the NASTRAN mesh file format was found. The software and workflow outlined in this article may be used to accelerate the investigation of TTFields in glioblastoma patients by facilitating the creation of FEA models derived from patient MRI datasets.


Assuntos
Neoplasias Encefálicas/patologia , Análise de Elementos Finitos , Glioblastoma/patologia , Imageamento por Ressonância Magnética/métodos , Software , Fluxo de Trabalho , Adulto , Neoplasias Encefálicas/radioterapia , Simulação por Computador , Glioblastoma/radioterapia , Humanos , Pessoa de Meia-Idade
16.
Expert Rev Med Devices ; 12(6): 717-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26513694

RESUMO

Alternating electric fields therapy, as delivered by the tumor treating fields device, is a new modality of cancer treatment that has been approved by the US FDA for recurrent glioblastoma. At a frequency of 200 kHz, these fields emanate from transducer arrays on the surface of the patient's scalp into the brain and perturb processes necessary for cytokinesis during tumor cell mitosis. In the registration Phase III trial for recurrent glioblastoma patients, the efficacy of the tumor treating fields as monotherapy was equivalent to chemotherapy, while scalp irritation was its major adverse event compared with systemic toxicities that were associated with cytotoxic chemotherapies. Alternating electric fields therapy is, therefore, an essential option for the treatment of recurrent glioblastoma. Here, we summarize our current knowledge of the physics, cell biology and clinical data supporting the use of the tumor treating fields therapy.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica/instrumentação , Fenômenos Eletrofisiológicos , Glioblastoma/fisiopatologia , Glioblastoma/terapia , Padrões de Prática Médica , Humanos , Resultado do Tratamento
17.
Cancer Med ; 4(11): 1697-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26311253

RESUMO

Tumor treating fields (TTFields) are alternating electric fields frequency tuned to 200 kHz for the treatment of recurrent glioblastoma. We report a patient treated with TTFields and determined the distribution of TTFields intracranially by computerized simulation using co-registered postgadolinium T1-weighted, T2, and MP RAGE images together with pre-specified conductivity and relative permittivity values for various cerebral structures. The distribution of the electric fields within the brain is inhomogeneous. Higher field intensities were aggregated near the ventricles, particularly at the frontal and occipital horns. The recurred tumor was found distant from the primary glioblastoma and it was located at a site of relatively lower electric field intensity. Future improvement in TTFields treatment may need to take into account the inhomogeneity of the electric field distribution within the brain.


Assuntos
Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Terapia Combinada/métodos , Simulação por Computador , Glioblastoma/patologia , Glioblastoma/terapia , Idoso , Neoplasias Encefálicas/diagnóstico , Feminino , Glioblastoma/diagnóstico , Humanos , Imageamento por Ressonância Magnética , Resultado do Tratamento
18.
Cancer Med ; 4(3): 383-91, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25620708

RESUMO

The NovoTTF-100A is a device that emits alternating electric fields and it is approved for the treatment of recurrent glioblastoma. It works by perturbing tumor cells during mitosis as they enter anaphase leading to aneuploidy, asymmetric chromosome segregation and cell death with evidence of increased immunogenicity. Clinical trial data have shown equivalent efficacy when compared to salvage chemotherapies in recurrent disease. Responders were found to have had a lower dexamethasone usage and a higher rate of prior low-grade histology. We treated a series of patients with NovoTTF-100A and bevacizumab alone (n = 34) or in combination with a regimen consisting of 6-thioguanine, lomustine, capecitabine, and celecoxib (TCCC) (n = 3). Compared to the former cohort, the latter cohort exhibited a trend for prolonged overall survival, median 4.1 (0.3-22.7) months versus 10.3 (7.7-13.6) months respectively (P = 0.0951), with one experiencing an objective response with a 50% reduction in tumor size on magnetic resonance imaging despite possessing a larger tumor size at baseline and more severe neurologic dysfunction than the median for either group. These observations illustrate the possibility of improving survival and achieving a response in patients with end-stage recurrent glioblastoma by biasing the tumor toward anti-tumor immunologic response with a combination of NovoTTF-100A and TCCC, as well as the continuation of bevacizumab in order to limit dexamethasone use due to its global immunosuppressive effect on the patient.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica , Glioblastoma/terapia , Recidiva Local de Neoplasia/terapia , Adulto , Idoso , Inibidores da Angiogênese/administração & dosagem , Antimetabólitos Antineoplásicos/administração & dosagem , Antineoplásicos Alquilantes/administração & dosagem , Bevacizumab/administração & dosagem , Neoplasias Encefálicas/patologia , Capecitabina/administração & dosagem , Celecoxib/administração & dosagem , Terapia Combinada , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Feminino , Glioblastoma/patologia , Humanos , Lomustina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Temozolomida , Tioguanina/administração & dosagem , Falha de Tratamento
19.
Melanoma Res ; 24(2): 120-30, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24463459

RESUMO

The aggressiveness of melanoma is believed to be correlated with tumor-stroma-associated immune cells. Cytokines and chemokines act to recruit and then modulate the activities of these cells, ultimately affecting disease progression. Because melanoma frequently metastasizes to the brain, we asked whether global differences in immunokine profiles could be detected in the cerebrospinal fluid (CSF) of melanoma patients and reveal aspects of tumor biology that correlate with patient outcomes. We therefore measured the levels of 12 cytokines and 12 chemokines in melanoma patient CSF and the resulting data were analyzed to develop unsupervised hierarchical clustergrams and heat maps. Unexpectedly, the overall profiles of immunokines found in these samples showed a generalized reconfiguration of their expression in melanoma patient CSF, resulting in the segregation of individuals with melanoma brain metastasis from nondisease controls. Chemokine CCL22 and cytokines IL1α, IL4, and IL5 were reduced in most samples, whereas a subset including CXCL10, CCL4, CCL17, and IL8 showed increased expression. Further, analysis of clusters identified within the melanoma patient set comparing patient outcome suggests that suppression of IL1α, IL4, IL5, and CCL22, with concomitant elevation of CXCL10, CCL4, and CCL17, may correlate with more aggressive development of brain metastasis. These results suggest that global immunokine suppression in the host, together with a selective increase in specific chemokines, constitute a predominant immunomodulatory feature of melanoma brain metastasis. These alterations likely drive the course of this disease in the brain and variations in the immune profiles of individual patients may predict outcomes.


Assuntos
Neoplasias Encefálicas/líquido cefalorraquidiano , Neoplasias Encefálicas/secundário , Quimiocinas/líquido cefalorraquidiano , Citocinas/líquido cefalorraquidiano , Melanoma/líquido cefalorraquidiano , Melanoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico
20.
Cancer Med ; 3(4): 1010-24, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24931821

RESUMO

Intravascular lymphomatosis (IVL) is a rare and clinically devastating form of extranodal B-cell non-Hodgkin's lymphoma. We performed a comprehensive analysis of the literature on IVL's published between 1959 and 2011 and evaluated the natural history as well as identified prognostic and predictive factors in patients. Nonparametric two-tailed Mann-Whitney U-test and Mantel-Cox log rank test were used to evaluate the survival intervals and prognostic factors. Multivariate analysis of variance (MANOVA) and chi-squared statistics were carried out to examine treatment-related predictive factors. Of the 740 patients with IVL, 651 (88%) had a diagnosis of B-cell lymphoma, 45 (6%) with T-cell lymphoma, and 12 patients (2%) with NK cell lymphoma. Central nervous system (CNS) IVL had the highest proportion of postmortem diagnosis, 250 (60%) compared to 21 (8%) of skin, 28 (11%) of bone marrow (BM) and spleen, and 17 (7%) of lung IVL's. Age <70 years (P = 0.0073), non-CNS site of initial diagnosis (P = 0.0014), lactate dehydrogenase (LDH) <700 (P = 0.0112), and rituximab treatment (P < 0.0001) were favorable prognostic factors. Gender, ethnicity, hemoglobin, BM biopsy, and the type of imaging studies used were not significant. Rituximab and doxorubicin treatment worked significantly better in patients with age >71 and LDH >577 compared to nonrituximab, nondoxorubicin regimens (MANOVA 2 degrees of freedom, P = 0.0345), with a median time from treatment to death of 20.0 (95% confidence interval [CI] 14.0-N/A, n = 14) months versus 2.0 (95%CI 0.5-N/A, n = 5) (χ(2) = 4.7, P = 0.0304). Patients with CNS IVL relapsed primarily in the CNS (88%) while same-organ relapse occurred less frequently in skin (23%), BM and spleen (50%) and lung (20%) IVL's. Our results indicate that IVL is primarily a disease of B-lymphoma cells. Timely diagnosis and treatment with rituximab-based chemotherapy improve patient survival. The pattern of recurrence is different between CNS IVL and IVL's in other organs.


Assuntos
Neoplasias do Sistema Nervoso Central/mortalidade , Linfoma de Células B/mortalidade , Neoplasias Cutâneas/mortalidade , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Neoplasias do Sistema Nervoso Central/metabolismo , Intervalo Livre de Doença , Humanos , Estimativa de Kaplan-Meier , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/metabolismo , Prognóstico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA