Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cells ; 10(9)2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34571851

RESUMO

Solid tumors in advanced cancer often feature a structurally and functionally abnormal vasculature through tumor angiogenesis, which contributes to cancer progression, metastasis, and therapeutic resistances. Hypoxia is considered a major driver of angiogenesis in tumor microenvironments. However, there remains a lack of in vitro models that recapitulate both the vasculature and hypoxia in the same model with physiological resemblance to the tumor microenvironment, while allowing for high-content spatiotemporal analyses for mechanistic studies and therapeutic evaluations. We have previously constructed a hypoxia microdevice that utilizes the metabolism of cancer cells to generate an oxygen gradient in the cancer cell layer as seen in solid tumor sections. Here, we have engineered a new composite microdevice-microfluidics platform that recapitulates a vascularized hypoxic tumor. Endothelial cells were seeded in a collagen channel formed by viscous fingering, to generate a rounded vascular lumen surrounding a hypoxic tumor section composed of cancer cells embedded in a 3-D hydrogel extracellular matrix. We demonstrated that the new device can be used with microscopy-based high-content analyses to track the vascular phenotypes, morphology, and sprouting into the hypoxic tumor section over a 7-day culture, as well as the response to different cancer/stromal cells. We further evaluated the integrity/leakiness of the vascular lumen in molecular delivery, and the potential of the platform to study the movement/trafficking of therapeutic immune cells. Therefore, our new platform can be used as a model for understanding tumor angiogenesis and therapeutic delivery/efficacy in vascularized hypoxic tumors.


Assuntos
Microfluídica/instrumentação , Neoplasias/irrigação sanguínea , Microambiente Tumoral/fisiologia , Vasos Sanguíneos/fisiologia , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Humanos , Hipóxia/patologia , Microfluídica/métodos , Modelos Biológicos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Oxigênio/metabolismo , Células Estromais/metabolismo
2.
IEEE Trans Med Imaging ; 38(1): 1-10, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-28796613

RESUMO

Vasculature is known to be of key biological significance, especially in the study of tumors. As such, considerable effort has been focused on the automated segmentation of vasculature in medical and pre-clinical images. The majority of vascular segmentation methods focus on bloodpool labeling methods; however, particularly, in the study of tumors, it is of particular interest to be able to visualize both the perfused and the non-perfused vasculature. Imaging vasculature by highlighting the endothelium provides a way to separate the morphology of vasculature from the potentially confounding factor of perfusion. Here, we present a method for the segmentation of tumor vasculature in 3D fluorescence microscopic images using signals from the endothelial and surrounding cells. We show that our method can provide complete and semantically meaningful segmentations of complex vasculature using a supervoxel-Markov random field approach. We show that in terms of extracting meaningful segmentations of the vasculature, our method outperforms both state-of-the-art method, specific to these data, as well as more classical vasculature segmentation methods.


Assuntos
Vasos Sanguíneos/diagnóstico por imagem , Células Endoteliais/citologia , Imageamento Tridimensional/métodos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Algoritmos , Animais , Aprendizado de Máquina , Cadeias de Markov , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico por imagem , Neovascularização Patológica/diagnóstico por imagem
3.
Cancer Treat Rev ; 68: 38-46, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29793113

RESUMO

Angiogenesis plays a critical role in the growth, progression, and metastasis of numerous solid tumor types, and thus, antiangiogenic agents have been studied for many years as potential therapeutic agents. Many different antiangiogenic agents, including monoclonal antibodies and multi-targeted tyrosine kinase inhibitors (TKIs), have been approved for various oncology indications, and promising clinical activity has been demonstrated. However, some of these agents have also been associated with serious safety concerns. Because angiogenesis is an important step in the wound healing process, agents targeting the angiogenesis pathway may interfere with wound healing, thus increasing the risk of surgical wound complications, such as dehiscence, surgical site bleeding, and wound infection. Nevertheless, antiangiogenic agents can be safely used in the perioperative setting if oncologists and surgeons are educated on the biology and pharmacokinetics of these agents. This review discusses the available published literature regarding surgical complications associated with the use of antiangiogenic agents and provides updated clinical recommendations on the optimal timing between surgery and antiangiogenic therapy. Due to the paucity of data surrounding this topic, current and future clinical trials need to evaluate prospectively the potential risks for surgical complications associated with antiangiogenic therapies to establish specific guidelines for their safe and effective use within the surgical oncology community.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Neoplasias/tratamento farmacológico , Neoplasias/cirurgia , Quimioterapia Adjuvante , Ensaios Clínicos como Assunto , Humanos , Terapia Neoadjuvante , Neoplasias/irrigação sanguínea , Neovascularização Patológica/tratamento farmacológico , Complicações Pós-Operatórias/induzido quimicamente , Complicações Pós-Operatórias/etiologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Medição de Risco , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Procedimentos Cirúrgicos Operatórios/métodos
4.
Mol Imaging Biol ; 19(2): 194-202, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27519522

RESUMO

PURPOSE: The purposes of the present study is to evaluate a new ultrasound molecular imaging approach in its ability to image a preclinical tumor model and to investigate the capacity to visualize and quantify co-registered microvascular and molecular imaging volumes. PROCEDURES: Molecular imaging using the new technique was compared with a conventional ultrasound molecular imaging technique (multi-pulse imaging) by varying the injected microbubble dose and scanning each animal using both techniques. Each of the 14 animals was randomly assigned one of three doses; bolus dose was varied, and the animals were imaged for three consecutive days so that each animal received every dose. A microvascular scan was also acquired for each animal by administering an infusion of nontargeted microbubbles. These scans were paired with co-registered molecular images (VEGFR2-targeted microbubbles), the vessels were segmented, and the spatial relationships between vessels and VEGFR2 targeting locations were analyzed. In five animals, an additional scan was performed in which the animal received a bolus of microbubbles targeted to E- and P-selectins. Vessel tortuosity as a function of distance from VEGF and selectin targeting was analyzed in these animals. RESULTS: Although resulting differences in image intensity due to varying microbubble dose were not significant between the two lowest doses, superharmonic imaging had significantly higher contrast-to-tissue ratio (CTR) than multi-pulse imaging (mean across all doses 13.98 dB for molecular acoustic angiography vs. 0.53 dB for multi-pulse imaging; p = 4.9 × 10-10). Analysis of registered microvascular and molecular imaging volumes indicated that vessel tortuosity decreases with increasing distance from both VEGFR2- and selectin-targeting sites. CONCLUSIONS: Molecular acoustic angiography (superharmonic molecular imaging) exhibited a significant increase in CTR at all doses tested due to superior rejection of tissue artifact signals. Due to the high resolution of acoustic angiography molecular imaging, it is possible to analyze spatial relationships in aligned microvascular and molecular superharmonic imaging volumes. Future studies are required to separate the effects of biomarker expression and blood flow kinetics in comparing local tortuosity differences between different endothelial markers such as VEGFR2, E-selectin, and P-selectin.


Assuntos
Acústica , Angiografia/métodos , Imagem Molecular/métodos , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico por imagem , Animais , Meios de Contraste/química , Modelos Animais de Doenças , Processamento de Imagem Assistida por Computador , Microvasos/patologia , Ratos Endogâmicos F344
5.
Int J Nanomedicine ; 11: 4735-4741, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27695329

RESUMO

Analytical and Monte Carlo simulations have been used to predict dose enhancement factors in nanoparticle-enhanced X-ray radiation therapy. Both simulations predict an increase in dose enhancement in the presence of nanoparticles, but the two methods predict different levels of enhancement over the studied energy, nanoparticle materials, and concentration regime for several reasons. The Monte Carlo simulation calculates energy deposited by electrons and photons, while the analytical one only calculates energy deposited by source photons and photoelectrons; the Monte Carlo simulation accounts for electron-hole recombination, while the analytical one does not; and the Monte Carlo simulation randomly samples photon or electron path and accounts for particle interactions, while the analytical simulation assumes a linear trajectory. This study demonstrates that the Monte Carlo simulation will be a better choice to evaluate dose enhancement with nanoparticles in radiation therapy.


Assuntos
Nanopartículas Metálicas/uso terapêutico , Nanomedicina/métodos , Radioterapia/métodos , Linhagem Celular Tumoral , Simulação por Computador , Elétrons , Humanos , Método de Monte Carlo , Neoplasias/irrigação sanguínea , Fótons , Raios X
6.
IEEE Trans Biomed Eng ; 63(5): 1082-6, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26394410

RESUMO

GOAL: Abnormalities of microvascular morphology have been associated with tumor angiogenesis for more than a decade, and are believed to be intimately related to both tumor malignancy and response to treatment. However, the study of these vascular changes in-vivo has been challenged due to the lack of imaging approaches which can assess the microvasculature in 3-D volumes noninvasively. Here, we use contrast-enhanced "acoustic angiography" ultrasound imaging to observe and quantify heterogeneity in vascular morphology around solid tumors. METHODS: Acoustic angiography, a recent advance in contrast-enhanced ultrasound imaging, generates high-resolution microvascular images unlike anything possible with standard ultrasound imaging techniques. Acoustic angiography images of a genetically engineered mouse breast cancer model were acquired to develop an image acquisition and processing routine that isolated radially expanding regions of a 3-D image from the tumor boundary to the edge of the imaging field for assessment of vascular morphology of tumor and surrounding vessels. RESULTS: Quantitative analysis of vessel tortuosity for the tissue surrounding tumors 3 to 7 mm in diameter revealed that tortuosity decreased in a region 6 to 10 mm from the tumor boundary, but was still significantly elevated when compared to control vasculature. CONCLUSION: Our analysis of angiogenesis-induced changes in the vasculature outside the tumor margin reveals that the extent of abnormal tortuosity extends significantly beyond the primary tumor mass. SIGNIFICANCE: Visualization of abnormal vascular tortuosity may make acoustic angiography an invaluable tool for early tumor detection based on quantifying the vascular footprint of small tumors and a sensitive method for understanding changes in the vascular microenvironment during tumor progression.


Assuntos
Interpretação de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Neoplasias/diagnóstico por imagem , Neovascularização Patológica/diagnóstico por imagem , Ultrassonografia/métodos , Angiografia , Animais , Camundongos , Microvasos/diagnóstico por imagem , Neoplasias/irrigação sanguínea
7.
Int J Radiat Oncol Biol Phys ; 94(1): 189-205, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26700713

RESUMO

The past decade has seen a dramatic increase in interest in the use of gold nanoparticles (GNPs) as radiation sensitizers for radiation therapy. This interest was initially driven by their strong absorption of ionizing radiation and the resulting ability to increase dose deposited within target volumes even at relatively low concentrations. These early observations are supported by extensive experimental validation, showing GNPs' efficacy at sensitizing tumors in both in vitro and in vivo systems to a range of types of ionizing radiation, including kilovoltage and megavoltage X rays as well as charged particles. Despite this experimental validation, there has been limited translation of GNP-mediated radiation sensitization to a clinical setting. One of the key challenges in this area is the wide range of experimental systems that have been investigated, spanning a range of particle sizes, shapes, and preparations. As a result, mechanisms of uptake and radiation sensitization have remained difficult to clearly identify. This has proven a significant impediment to the identification of optimal GNP formulations which strike a balance among their radiation sensitizing properties, their specificity to the tumors, their biocompatibility, and their imageability in vivo. This white paper reviews the current state of knowledge in each of the areas concerning the use of GNPs as radiosensitizers, and outlines the steps which will be required to advance GNP-enhanced radiation therapy from their current pre-clinical setting to clinical trials and eventual routine usage.


Assuntos
Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Neoplasias/radioterapia , Radiossensibilizantes/uso terapêutico , Animais , Tempo de Circulação Sanguínea , Química Farmacêutica/métodos , Ouro/química , Ouro/farmacocinética , Humanos , Método de Monte Carlo , Neoplasias/irrigação sanguínea , Tamanho da Partícula , Fótons/uso terapêutico , Terapia com Prótons , Tolerância a Radiação , Radiossensibilizantes/química , Radiossensibilizantes/farmacocinética , Propriedades de Superfície
8.
Biomaterials ; 58: 82-92, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25941785

RESUMO

The assessment of the fractional vascular volume (vV) in the tumor area is of great interest in the characterization of tumor and it can be useful to monitor the outcome of anti-angiogenetic therapies. The high spatial and temporal resolution of Magnetic Resonance Imaging makes it the election imaging modality to monitor in vivo the vascular volume changes. Commonly used MRI methods to obtain this information rely on the administration of contrast agents that modify the bulk water relaxation times but, unfortunately, they can provide only an estimate of vV since they are not fully retained in the vascular space. Herein, Gd-loaded Red Blood Cells (Gd-RBCs) are proposed as a contrast agent able to provide quantitative information on tumor vascularization. Being Gd-RBCs fully retained in the vascular space, the proposed method does not suffer for the limitations associated to the use of extracellular Gd-agents that quickly extravasate in the leaky tumor vasculature. Furthermore, the long half-life and biocompatibility of Gd-RBCs allows repeating the measurement many times upon their administration; this ensures the possibility to in vivo evaluate the change of vascular volume during tumor growth. For these reasons, Gd-RBCs may represent a highly biocompatible imaging reporter of vasculature, able to quantitatively assess changes in the vascular volume in the ROI.


Assuntos
Meios de Contraste/química , Eritrócitos/efeitos dos fármacos , Gadolínio/química , Imageamento por Ressonância Magnética , Neoplasias/irrigação sanguínea , Animais , Materiais Biocompatíveis , Encéfalo/patologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Músculos/patologia , Neovascularização Patológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Carga Tumoral
9.
Phys Med ; 31(5): 536-41, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25979209

RESUMO

The small-scale dosimetry of radionuclides in solid-tumours is directly related to the intra-tumoral distribution of the administered radiopharmaceutical, which is affected by its egress from the vasculature and dispersion within the tumour. The aim of the present study was to evaluate the combined dosimetric effects of radiopharmaceutical distribution and range of the emitted radiation in a model of tumour microvasculature. We developed a computational model of solid-tumour microenvironment around a blood capillary vessel, and we simulated the transport of radiation emitted by (223)Ra, (111)In, (131)I and (177)Lu using the GEANT4 Monte Carlo. For each nuclide, several models of radiopharmaceutical dispersion throughout the capillary vessel were considered. Radial dose profiles around the capillary vessel, the Initial Radioactivity (IR) necessary to deposit 100 Gy of dose at the edge of the viable tumour-cell region, the Endothelial Cell Mean Dose (ECMD) and the Tumour Edge Mean Dose (TEMD), i.e. the mean dose imparted at the 250-µm layer of tissue, were computed. The results for beta and Auger emitters demonstrate that the photon dose is about three to four orders of magnitude lower than that deposited by electrons. For (223)Ra, the beta emissions of its progeny deliver a dose about three orders of magnitude lower than that delivered by the alpha emissions. Such results may help to characterize the dose inhomogeneities in solid tumour therapies with radiopharmaceuticals, taking into account the interplay between drug distribution from vasculature and range of ionizing radiations.


Assuntos
Capilares/efeitos da radiação , Método de Monte Carlo , Neoplasias/irrigação sanguínea , Neoplasias/radioterapia , Medicina Nuclear , Radiometria/métodos , Compostos Radiofarmacêuticos/uso terapêutico , Radioisótopos de Índio/uso terapêutico , Radioisótopos do Iodo/uso terapêutico , Marcação por Isótopo , Lutécio/uso terapêutico , Dosagem Radioterapêutica , Rádio (Elemento)/uso terapêutico
10.
Antioxid Redox Signal ; 21(8): 1145-55, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-24597714

RESUMO

AIMS: The tumor microenvironment is characterized by a highly reducing redox status, a low pH, and hypoxia. Anti-angiogenic therapies for solid tumors frequently function in two steps: the transient normalization of structurally and functionally aberrant tumor blood vessels with increased blood perfusion, followed by the pruning of tumor blood vessels and the resultant cessation of nutrients and oxygen delivery required for tumor growth. Conventional anatomic or vascular imaging is impractical or insufficient to distinguish between the two steps of tumor response to anti-angiogenic therapies. Here, we investigated whether the noninvasive imaging of the tumor redox state and energy metabolism could be used to characterize anti-angiogenic drug-induced transient vascular normalization. RESULTS: Daily treatment of squamous cell carcinoma (SCCVII) tumor-bearing mice with the multi-tyrosine kinase inhibitor sunitinib resulted in a rapid decrease in tumor microvessel density and the suppression of tumor growth. Tumor pO2 imaging by electron paramagnetic resonance imaging showed a transient increase in tumor oxygenation after 2-4 days of sunitinib treatment, implying improved tumor perfusion. During this window of vascular normalization, magnetic resonance imaging of the redox status using an exogenously administered nitroxide probe and hyperpolarized (13)C MRI of the metabolic flux of pyruvate/lactate couple revealed an oxidative shift in tumor redox status. INNOVATION: Redox-sensitive metabolic couples can serve as noninvasive surrogate markers to identify the vascular normalization window in tumors with imaging techniques. CONCLUSION: A multimodal imaging approach to characterize physiological, metabolic, and redox changes in tumors is useful to distinguish between the different stages of anti-angiogenic treatment.


Assuntos
Inibidores da Angiogênese/farmacologia , Indóis/farmacologia , Neoplasias/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Pirróis/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Meios de Contraste/metabolismo , Óxidos N-Cíclicos/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Humanos , Indóis/uso terapêutico , Imageamento por Ressonância Magnética , Camundongos Endogâmicos C3H , Camundongos Nus , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Oxirredução , Oxigênio/metabolismo , Pirróis/uso terapêutico , Ácido Pirúvico/metabolismo , Sunitinibe , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Angiogenesis ; 15(3): 433-42, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22535383

RESUMO

PURPOSE: To develop and test a real-time motion compensation algorithm for contrast-enhanced ultrasound imaging of tumor angiogenesis on a clinical ultrasound system. MATERIALS AND METHODS: The Administrative Institutional Panel on Laboratory Animal Care approved all experiments. A new motion correction algorithm measuring the sum of absolute differences in pixel displacements within a designated tracking box was implemented in a clinical ultrasound machine. In vivo angiogenesis measurements (expressed as percent contrast area) with and without motion compensated maximum intensity persistence (MIP) ultrasound imaging were analyzed in human colon cancer xenografts (n = 64) in mice. Differences in MIP ultrasound imaging signal with and without motion compensation were compared and correlated with displacements in x- and y-directions. The algorithm was tested in an additional twelve colon cancer xenograft-bearing mice with (n = 6) and without (n = 6) anti-vascular therapy (ASA-404). In vivo MIP percent contrast area measurements were quantitatively correlated with ex vivo microvessel density (MVD) analysis. RESULTS: MIP percent contrast area was significantly different (P < 0.001) with and without motion compensation. Differences in percent contrast area correlated significantly (P < 0.001) with x- and y-displacements. MIP percent contrast area measurements were more reproducible with motion compensation (ICC = 0.69) than without (ICC = 0.51) on two consecutive ultrasound scans. Following anti-vascular therapy, motion-compensated MIP percent contrast area significantly (P = 0.03) decreased by 39.4 ± 14.6 % compared to non-treated mice and correlated well with ex vivo MVD analysis (Rho = 0.70; P = 0.05). CONCLUSION: Real-time motion-compensated MIP ultrasound imaging allows reliable and accurate quantification and monitoring of angiogenesis in tumors exposed to breathing-induced motion artifacts.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica/diagnóstico por imagem , Ultrassom , Algoritmos , Animais , Feminino , Imunofluorescência , Humanos , Camundongos , Camundongos Nus , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Transplante Heterólogo , Ultrassonografia
12.
Eur Radiol ; 22(7): 1442-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22302501

RESUMO

Imaging is key in the accurate monitoring of response to cancer therapies targeting tumour vascularity to inhibit its growth and dissemination. Dynamic contrast enhanced ultrasound (DCE ultrasound) is a quantitative method with the advantage of being non-invasive, widely available, portable, cost effective, highly sensitive and reproducible using agents that are truly intravascular. Under the auspices of the initiative of the Experimental Cancer Medicine Centre Imaging Network, bringing together experts from the UK, Europe and North America for a 2-day workshop in May 2010, this consensus paper aims to provide guidance on the use of DCE ultrasound in the measurement of tumour vascular support in clinical trials. Key Points • DCE ultrasound can quantify and extract specific blood flow parameters, such as flow velocity, relative vascular volume and relative blood flow rate. • DCE ultrasound can be performed repeatedly and is therefore ideally suited for pharmacokinetic and pharmacodynamic studies evaluating vascular-targeted drugs. • DCE ultrasound provides a reproducible method of assessing the vascular effects of therapy in pre-clinical and early clinical trials, which is easily translatable into routine clinical practice.


Assuntos
Ensaios Clínicos como Assunto/normas , Meios de Contraste/normas , Neoplasias/diagnóstico por imagem , Neovascularização Patológica/diagnóstico por imagem , Guias de Prática Clínica como Assunto , Ultrassonografia/normas , Europa (Continente) , Humanos , Neoplasias/irrigação sanguínea , América do Norte , Padrões de Referência
13.
Int J Radiat Oncol Biol Phys ; 82(5): e725-31, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22330998

RESUMO

PURPOSE: [(18)F]-fluorodeoxyglucose-positron emission tomography (FDG-PET) images are usually quantitatively analyzed in "whole-tumor" volumes of interest. Also parameters determined with dynamic PET acquisitions, such as the Patlak glucose metabolic rate (MR(glc)) and pharmacokinetic rate constants of two-tissue compartment modeling, are most often derived per lesion. We propose segmentation of tumors to determine tumor heterogeneity, potentially useful for dose-painting in radiotherapy and elucidating mechanisms of FDG uptake. METHODS AND MATERIALS: In 41 patients with 104 lesions, dynamic FDG-PET was performed. On MR(glc) images, tumors were segmented in quartiles of background subtracted maximum MR(glc) (0%-25%, 25%-50%, 50%-75%, and 75%-100%). Pharmacokinetic analysis was performed using an irreversible two-tissue compartment model in the three segments with highest MR(glc) to determine the rate constants of FDG metabolism. RESULTS: From the highest to the lowest quartile, significant decreases of uptake (K(1)), washout (k(2)), and phosphorylation (k(3)) rate constants were seen with significant increases in tissue blood volume fraction (V(b)). CONCLUSIONS: Tumor regions with highest MR(glc) are characterized by high cellular uptake and phosphorylation rate constants with relatively low blood volume fractions. In regions with less metabolic activity, the blood volume fraction increases and cellular uptake, washout, and phosphorylation rate constants decrease. These results support the hypothesis that regional tumor glucose phosphorylation rate is not dependent on the transport of nutrients (i.e., FDG) to the tumor.


Assuntos
Fluordesoxiglucose F18/farmacocinética , Glucose/metabolismo , Neoplasias/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Adulto , Idoso , Volume Sanguíneo , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/metabolismo , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/metabolismo , Masculino , Pessoa de Meia-Idade , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico por imagem , Fosforilação
14.
Eur Radiol ; 22(7): 1430-41, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22367468

RESUMO

Dynamic contrast-enhanced computed tomography (DCE-CT) assesses the vascular support of tumours through analysis of temporal changes in attenuation in blood vessels and tissues during a rapid series of images acquired with intravenous administration of iodinated contrast material. Commercial software for DCE-CT analysis allows pixel-by-pixel calculation of a range of validated physiological parameters and depiction as parametric maps. Clinical studies support the use of DCE-CT parameters as surrogates for physiological and molecular processes underlying tumour angiogenesis. DCE-CT has been used to provide biomarkers of drug action in early phase trials for the treatment of a range of cancers. DCE-CT can be appended to current imaging assessments of tumour response with the benefits of wide availability and low cost. This paper sets out guidelines for the use of DCE-CT in assessing tumour vascular support that were developed using a Delphi process. Recommendations encompass CT system requirements and quality assurance, radiation dosimetry, patient preparation, administration of contrast material, CT acquisition parameters, terminology and units, data processing and reporting. DCE-CT has reached technical maturity for use in therapeutic trials in oncology. The development of these consensus guidelines may promote broader application of DCE-CT for the evaluation of tumour vascularity. Key Points • DCE-CT can robustly assess tumour vascular support • DCE-CT has reached technical maturity for use in therapeutic trials in oncology • This paper presents consensus guidelines for using DCE-CT in assessing tumour vascularity.


Assuntos
Meios de Contraste/normas , Previsões , Neoplasias/diagnóstico por imagem , Neovascularização Patológica/diagnóstico por imagem , Guias de Prática Clínica como Assunto , Tomografia Computadorizada por Raios X/normas , Tomografia Computadorizada por Raios X/tendências , Humanos , Neoplasias/irrigação sanguínea , Padrões de Referência
15.
World J Gastroenterol ; 18(47): 7026-32, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23323004

RESUMO

AIM: To quantitatively assess the ability of double contrast-enhanced ultrasound (DCUS) to detect tumor early response to pre-operative chemotherapy. METHODS: Forty-three patients with gastric cancer treated with neoadjuvant chemotherapy followed by curative resection between September 2011 and February 2012 were analyzed. Pre-operative chemotherapy regimens of fluorouracil + oxaliplatin or S-1 + oxaliplatin were administered in 2-4 cycles over 6-12 wk periods. All patients underwent contrast-enhanced computed tomography (CT) scan and DCUS before and after two courses of pre-operative chemotherapy. The therapeutic response was assessed by CT using the response evaluation criteria in solid tumors (RECIST 1.1) criteria. Tumor area was assessed by DCUS as enhanced appearance of gastric carcinoma due to tumor vascularity during the contrast phase as compared to the normal gastric wall. Histopathologic analysis was carried out according to the Mandard tumor regression grade criteria and used as the reference standard. Receiver operating characteristic (ROC) analysis was used to evaluate the efficacy of DCUS parameters in differentiating histopathological responders from non-responders. RESULTS: The study population consisted of 32 men and 11 women, with mean age of 59.7 ± 11.4 years. Neither age, sex, histologic type, tumor site, T stage, nor N stage was associated with pathological response. The responders had significantly smaller mean tumor size than the non-responders (15.7 ± 7.4 cm vs 33.3 ± 14.1 cm, P < 0.01). According to Mandard's criteria, 27 patients were classified as responders, with 11 (40.7%) showing decreased tumor size by DCUS. In contrast, only three (18.8%) of the 16 non-responders showed decreased tumor size by DCUS (P < 0.01). The area under the ROC curve was 0.64, with a 95%CI of 0.46-0.81. The effects of several cut-off points on diagnostic parameters were calculated in the ROC curve analysis. By maximizing Youden's index (sensitivity + specificity - 1), the best cut-off point for distinguishing responders from non-responders was determined, which had optimal sensitivity of 62.9% and specificity of 56.3%. Using this cut-off point, the positive and negative predictive values of DCUS for distinguishing responders from non-responders were 70.8% and 47.4%, respectively. The overall accuracy of DCUS for therapeutic response assessment was 60.5%, slightly higher than the 53.5% for CT response assessment with RECIST criteria (P = 0.663). Although the advantage was not statistically significant, likely due to the small number of cases assessed. DCUS was able to identify decreased perfusion in responders who showed no morphological change by CT imaging, which can be occluded by such treatment effects as fibrosis and edema. CONCLUSION: DCUS may represent an innovative tool for more accurately predicting histopathological response to neoadjuvant chemotherapy before surgical resection in patients with locally-advanced gastric cancer.


Assuntos
Terapia Neoadjuvante , Neoplasias Gástricas/diagnóstico por imagem , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/tratamento farmacológico , Adulto , Idoso , Meios de Contraste/farmacologia , Feminino , Fluoruracila/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/irrigação sanguínea , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Curva ROC , Fatores de Tempo , Tomografia Computadorizada por Raios X , Resultado do Tratamento , Ultrassonografia
16.
NMR Biomed ; 24(6): 612-35, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21567513

RESUMO

MRI and MRS techniques are being applied to the characterisation of various aspects of the tumour microenvironment and to the assessment of tumour response to therapy. For example, kinetic parameters describing tumour blood vessel flow and permeability can be derived from dynamic contrast-enhanced MRI data and have been correlated with a positive tumour response to antivascular therapies. The ongoing development and validation of noninvasive, high-resolution anatomical/molecular MR techniques will equip us with the means to detect specific tumour biomarkers early on, and then to monitor the efficacy of cancer treatments efficiently and reliably, all within a clinically relevant time frame. Reliable tumour microenvironment imaging biomarkers will provide obvious advantages by enabling tumour-specific treatment tailoring and potentially improving patient outcome. However, for routine clinical application across many disease types, such imaging biomarkers must be quantitative, robust, reproducible, sufficiently sensitive and cost-effective. These characteristics are all difficult to achieve in practice, but image biomarker development and validation have been greatly facilitated by an increasing number of pertinent preclinical in vivo cancer models. Emphasis must now be placed on discovering whether the preclinical results translate into an improvement in patient care and, therefore, overall survival.


Assuntos
Imageamento por Ressonância Magnética , Neoplasias/patologia , Neoplasias/terapia , Microambiente Tumoral , Meios de Contraste , Matriz Extracelular/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Neoplasias/irrigação sanguínea
17.
Strahlenther Onkol ; 187(4): 260-6, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21437770

RESUMO

BACKGROUND AND PURPOSE: Hypoxia is a characteristic of tumors, is known to increase aggressiveness, and causes treatment resistance. Traditional classification suggests two types of hypoxia: chronic and acute. Acute hypoxia is mostly caused by transient disruptions in perfusion, while chronic hypoxia is caused by diffusion limitations. This classification may be insufficient in terms of pathogenetic and pathophysiological mechanisms. Therefore, we quantified hypoxia subtypes in tumors based on (immuno-)fluorescent marker distribution patterns in microcirculatory supply units (MCSUs). MATERIAL AND METHODS: Cryosections from hSCC lines (SAS, FaDu, UT-SCC-5, UT-SCC-14, UT-SCC-15) were analyzed. Hypoxia was identified by pimonidazole, perfusion by Hoechst 33342, and endothelial cells by CD31. The following patterns were identified in vital tumor tissue: (1) normoxia: Hoechst 33342 fluorescence around microvessels, no pimonidazole, (2) chronic hypoxia: Hoechst 33342 fluorescence around microvessels, pimonidazole distant from microvessels, (3) acute hypoxia: no Hoechst 33342 fluorescence around microvessels, pimonidazole in immediate vicinity of microvessels, and (4) hypoxemic hypoxia: Hoechst 33342 fluorescence and pimonidazole directly around microvessels. RESULTS: Quantitative assessment of MCSUs show predominance for normoxia in 4 out of 5 tumor lines (50.1-72.8%). Total hypoxia slightly prevails in UT-SCC-15 (56.9%). Chronic hypoxia is the dominant subtype (65.4-85.9% of total hypoxia). Acute hypoxia only accounts for 12.9-29.8% and hypoxemic hypoxia for 1.2-6.4% of total hypoxia. The fraction of perfused microvessels ranged from 82.5-96.6%. CONCLUSION: Chronic hypoxia is the prevailing subtype in MCSUs. Acute hypoxia and hypoxemic hypoxia account for only a small fraction. This approach enables assessment and recognition of different hypoxia subtypes including hypoxemic hypoxia and may facilitate methods to (clinically) identify and eliminate hypoxia.


Assuntos
Hipóxia Celular/fisiologia , Microcirculação/fisiologia , Microscopia de Fluorescência , Neoplasias/irrigação sanguínea , Células Tumorais Cultivadas/classificação , Células Tumorais Cultivadas/patologia , Benzimidazóis , Linhagem Celular Tumoral , Difusão , Humanos , Microvasos , Nitroimidazóis , Imagem de Perfusão , Prognóstico , Radiossensibilizantes
18.
Integr Biol (Camb) ; 3(4): 375-87, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21321746

RESUMO

The tumor microenvironment provides a rich source of potential targets for selective therapeutic intervention with properly designed anticancer agents. Significant physiological differences exist between the microvessels that nourish tumors and those that supply healthy tissue. Selective drug-mediated damage of these tortuous and chaotic microvessels starves a tumor of necessary nutrients and oxygen and eventually leads to massive tumor necrosis. Vascular targeting strategies in oncology are divided into two separate groups: angiogenesis inhibiting agents (AIAs) and vascular disrupting agents (VDAs). The mechanisms of action between these two classes of compounds are profoundly distinct. The AIAs inhibit the actual formation of new vessels, while the VDAs damage and/or destroy existing tumor vasculature. One subset of small-molecule VDAs functions by inhibiting the assembly of tubulin into microtubules, thus causing morphology changes to the endothelial cells lining the tumor vasculature, triggered by a cascade of cell signaling events. Ultimately this results in catastrophic damage to the vessels feeding the tumor. The rapid emergence and subsequent development of the VDA field over the past decade has led to the establishment of a synergistic combination of preclinical state-of-the-art tumor imaging and biological evaluation strategies that are often indicative of future clinical efficacy for a given VDA. This review focuses on an integration of the appropriate biochemical and biological tools necessary to assess (preclinically) new small-molecule, tubulin active VDAs for their potential to be clinically effective anticancer agents.


Assuntos
Diagnóstico por Imagem/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Células Endoteliais/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Moduladores de Tubulina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Hipóxia/induzido quimicamente , Hipóxia/patologia , Imageamento por Ressonância Magnética/métodos , Camundongos , Microscopia de Fluorescência/métodos , Microvasos/patologia , Estrutura Molecular , Neoplasias/patologia , Ratos , Tubulina (Proteína)/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/uso terapêutico , Ultrassonografia Doppler/métodos
19.
Angiogenesis ; 14(2): 143-53, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21225337

RESUMO

Inflammation plays a prominent role in tumor growth. Anti-inflammatory drugs have therefore been proposed as anti-cancer therapeutics. In this study, we determined the anti-angiogenic activity of a single dose of liposomal prednisolone phosphate (PLP-L), by monitoring tumor vascular function and viability over a period of one week. C57BL/6 mice were inoculated subcutaneously with B16F10 melanoma cells. Six animals were PLP-L-treated and six served as control. Tumor tissue and vascular function were probed using MRI before and at three timepoints after treatment. DCE-MRI was used to determine K(trans), v(e), time-to-peak, initial slope and the fraction of non-enhancing pixels, complemented with immunohistochemistry. The apparent diffusion coefficient (ADC), T(2) and tumor size were assessed with MRI as well. PLP-L treatment resulted in smaller tumors and caused a significant drop in K(trans) 48 h post-treatment, which was maintained until one week after drug administration. However, this effect was not sufficient to significantly distinguish treated from non-treated animals. The therapy did not affect tumor tissue viability but did prevent the ADC decrease observed in the control group. No evidence for PLP-L-induced tumor vessel normalization was found on histology. Treatment with PLP-L altered tumor vascular function. This effect did not fully explain the tumor growth inhibition, suggesting a broader spectrum of PLP-L activities.


Assuntos
Inibidores da Angiogênese/farmacologia , Glucocorticoides/farmacologia , Lipossomos/química , Prednisolona/análogos & derivados , Inibidores da Angiogênese/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Contraste , Imagem de Difusão por Ressonância Magnética , Glucocorticoides/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Microvasos/efeitos dos fármacos , Microvasos/patologia , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Prednisolona/farmacologia , Prednisolona/uso terapêutico
20.
Int J Radiat Oncol Biol Phys ; 81(1): 270-6, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21163591

RESUMO

PURPOSE: Tumor endothelial cell damage during radiation therapy may contribute significantly to tumor eradication and treatment efficacy. Gold nanoparticles (AuNPs) delivered preferentially to the walls of tumor blood vessels produce low-energy, short-range photoelectrons during external beam radiotherapy, boosting dose to the tumor microvasculature. In this study dosimetry at the single-cell level is used to estimate the anticipated AuNP-mediated dose enhancement to tumor endothelial cells during 6-MV X-ray irradiation. METHODS AND MATERIALS: Endothelial cells are modeled as thin slabs with 100-nm-diameter AuNPs attached within the blood vessel. The number of photoelectrons emitted per AuNP per gray of X-rays is computed at multiple points along the external beam central axis by use of a Monte Carlo-generated energy fluence spectrum. The energy deposited from AuNP emissions to the endothelium is calculated based on an analytic method incorporating the energy-loss formula of Cole. The endothelial dose enhancement factor (EDEF) is the ratio of the overall (externally plus internally generated) dose to endothelial cells in the presence of AuNPs to the dose without AuNPs (from the external beam only). RESULTS: At 20-cm depth, the EDEF is 1.7 (70% dose increase) for an intravascular AuNP concentration of 30 mg/g. Most of this dose enhancement arises from the low-energy (approximately 100 keV) portion of the linear accelerator X-ray spectrum. Furthermore, for AuNP concentrations ranging from 7 to 140 mg/g, EDEF values of 1.2 to 4.4 (20-340% dose increase) are calculated. CONCLUSIONS: In contrast to calculations assuming that AuNPs distributed homogeneously throughout the target volume (macrodosimetry), our cellular microdosimetry calculations predict a major dose enhancement to tumor microvasculature from conventional linear accelerator X-rays. This effect may enable the delivery of ablative therapeutic doses to these sensitive microstructures while maintaining established dose constraints for the organs at risk.


Assuntos
Células Endoteliais/efeitos da radiação , Endotélio Vascular/citologia , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Neoplasias/irrigação sanguínea , Neoplasias/radioterapia , Algoritmos , Microcirculação/efeitos da radiação , Método de Monte Carlo , Aceleradores de Partículas , Fótons/uso terapêutico , Radioterapia/métodos , Dosagem Radioterapêutica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA