Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Lab Chip ; 20(23): 4357-4372, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-32955072

RESUMO

Traditional drug screening models are often unable to faithfully recapitulate human physiology in health and disease, motivating the development of microfluidic organs-on-a-chip (OOC) platforms that can mimic many aspects of human physiology and in the process alleviate many of the discrepancies between preclinical studies and clinical trials outcomes. Linsitinib, a novel anti-cancer drug, showed promising results in pre-clinical models of Ewing Sarcoma (ES), where it suppressed tumor growth. However, a Phase II clinical trial in several European centers with patients showed relapsed and/or refractory ES. We report an integrated, open setting, imaging and sampling accessible, polysulfone-based platform, featuring minimal hydrophobic compound binding. Two bioengineered human tissues - bone ES tumor and heart muscle - were cultured either in isolation or in the integrated platform and subjected to a clinically used linsitinib dosage. The measured anti-tumor efficacy and cardiotoxicity were compared with the results observed in the clinical trial. Only the engineered tumor tissues, and not monolayers, recapitulated the bone microenvironment pathways targeted by linsitinib, and the clinically-relevant differences in drug responses between non-metastatic and metastatic ES tumors. The responses of non-metastatic ES tumor tissues and heart muscle to linsitinib were much closer to those observed in the clinical trial for tissues cultured in an integrated setting than for tissues cultured in isolation. Drug treatment of isolated tissues resulted in significant decreases in tumor viability and cardiac function. Meanwhile, drug treatment in an integrated setting showed poor tumor response and less cardiotoxicity, which matched the results of the clinical trial. Overall, the integration of engineered human tumor and cardiac tissues in the integrated platform improved the predictive accuracy for both the direct and off-target effects of linsitinib. The proposed approach could be readily extended to other drugs and tissue systems.


Assuntos
Antineoplásicos , Sarcoma de Ewing , Antineoplásicos/uso terapêutico , Coração , Humanos , Dispositivos Lab-On-A-Chip , Sarcoma de Ewing/tratamento farmacológico , Engenharia Tecidual , Microambiente Tumoral
2.
Proc Natl Acad Sci U S A ; 115(6): 1256-1261, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29363599

RESUMO

Eight out of 10 breast cancer patients die within 5 years after the primary tumor has spread to the bones. Tumor cells disseminated from the breast roam the vasculature, colonizing perivascular niches around blood capillaries. Slow flows support the niche maintenance by driving the oxygen, nutrients, and signaling factors from the blood into the interstitial tissue, while extracellular matrix, endothelial cells, and mesenchymal stem cells regulate metastatic homing. Here, we show the feasibility of developing a perfused bone perivascular niche-on-a-chip to investigate the progression and drug resistance of breast cancer cells colonizing the bone. The model is a functional human triculture with stable vascular networks within a 3D native bone matrix cultured on a microfluidic chip. Providing the niche-on-a-chip with controlled flow velocities, shear stresses, and oxygen gradients, we established a long-lasting, self-assembled vascular network without supplementation of angiogenic factors. We further show that human bone marrow-derived mesenchymal stem cells, which have undergone phenotypical transition toward perivascular cell lineages, support the formation of capillary-like structures lining the vascular lumen. Finally, breast cancer cells exposed to interstitial flow within the bone perivascular niche-on-a-chip persist in a slow-proliferative state associated with increased drug resistance. We propose that the bone perivascular niche-on-a-chip with interstitial flow promotes the formation of stable vasculature and mediates cancer cell colonization.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Técnicas de Cocultura/instrumentação , Dispositivos Lab-On-A-Chip , Matriz Óssea/patologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Técnicas de Cocultura/métodos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Oxigênio , Perfusão , Alicerces Teciduais
3.
Tissue Eng Part C Methods ; 23(2): 98-107, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28068876

RESUMO

Ewing's sarcoma (ES) is a poorly differentiated pediatric tumor of aggressive behavior characterized by propensity to metastasize to bone. Interactions between the tumor and bone cells orchestrate a vicious cycle in which tumor cells induce osteoclast differentiation and activation to cause osteolytic lesions, broken bones, pain, and hypercalcemia. The lack of controllable models that can recapitulate osteolysis in ES impedes the development of new therapies and limits our understanding of how tumor cells invade bone. In response to this need, tissue-engineered models are now being developed to enable quantitative, predictive studies of human tumors. In this study, we report a novel bioengineered model of ES that incorporates the osteolytic process. Our strategy is based on engineering human bone containing both osteoclasts and osteoblasts within three-dimensional mineralized bone matrix. We show that the bone matrix is resorbed by mature osteoclasts while the new bone matrix is formed by osteoblasts, leading to calcium release and bone remodeling. Introduction of ES cell aggregates into the bone niche induced decreases in bone density, connectivity, and matrix deposition. Additionally, therapeutic reagents, such as zoledronic acid, which have demonstrated efficacy in ES treatment, inhibited bone resorption mediated by osteoclasts in the tumor model.


Assuntos
Neoplasias Ósseas/patologia , Modelos Biológicos , Osteoblastos/citologia , Osteoclastos/citologia , Osteólise/fisiopatologia , Sarcoma de Ewing/patologia , Engenharia Tecidual/métodos , Conservadores da Densidade Óssea/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Diferenciação Celular , Células Cultivadas , Difosfonatos/farmacologia , Humanos , Imidazóis/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Sarcoma de Ewing/tratamento farmacológico , Sarcoma de Ewing/metabolismo , Ácido Zoledrônico
5.
Theranostics ; 6(8): 1119-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27279906

RESUMO

There is a growing interest in the pivotal role of exosomes in cancer and in their use as biomarkers. However, despite the importance of the microenvironment for cancer initiation and progression, monolayer cultures of tumor cells still represent the main in vitro source of exosomes. As a result, their environmental regulation remains largely unknown. Here, we report a three-dimensional tumor model for studying exosomes, using Ewing's sarcoma type 1 as a clinically relevant example. The bioengineered model was designed based on the hypothesis that the 3-dimensionality, composition and stiffness of the tumor matrix are the critical determinants of the size and cargo of exosomes released by the cancer cells. We analyzed the effects of the tumor microenvironment on exosomes, and the effects of exosomes on the non-cancer cells from the bone niche. Exosomes from the tissue-engineered tumor had similar size distribution as those in the patients' plasma, and were markedly smaller than those in monolayer cultures. Bioengineered tumors and the patients' plasma contained high levels of the Polycomb histone methyltransferase EZH2 mRNA relatively to their monolayer counterparts. Notably, EZH2 mRNA, a potential tumor biomarker detectable in blood plasma, could be transferred to the surrounding mesenchymal stem cells. This study provides the first evidence that an in vitro culture environment can recapitulate some properties of tumor exosomes.


Assuntos
Exossomos/química , Exossomos/ultraestrutura , Sarcoma de Ewing/patologia , Biomarcadores/análise , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Modelos Biológicos , RNA Mensageiro/análise , Técnicas de Cultura de Tecidos
6.
Methods Mol Biol ; 1502: 203-11, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27115504

RESUMO

The lack of controllable in vitro models that can recapitulate the features of solid tumors such as Ewing's sarcoma limits our understanding of the tumor initiation and progression and impedes the development of new therapies. Cancer research still relies of the use of simple cell culture, tumor spheroids, and small animals. Tissue-engineered tumor models are now being grown in vitro to mimic the actual tumors in patients. Recently, we have established a new protocol for bioengineering the Ewing's sarcoma, by infusing tumor cell aggregates into the human bone engineered from the patient's mesenchymal stem cells. The bone niche allows crosstalk between the tumor cells, osteoblasts and supporting cells of the bone, extracellular matrix, and the tissue microenvironment. The bioreactor platform used in these experiments also allows the implementation of physiologically relevant mechanical signals. Here, we describe a method to build an in vitro model of Ewing's sarcoma that mimics the key properties of the native tumor and provides the tissue context and physical regulatory signals.


Assuntos
Pesquisa Biomédica/métodos , Neoplasias Ósseas/patologia , Osso e Ossos/patologia , Sarcoma de Ewing/patologia , Engenharia Tecidual/métodos , Animais , Fenômenos Biomecânicos , Pesquisa Biomédica/instrumentação , Reatores Biológicos , Osso e Ossos/citologia , Agregação Celular , Linhagem Celular , Linhagem Celular Tumoral , Desenho de Equipamento , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/patologia , Esferoides Celulares/citologia , Esferoides Celulares/patologia , Estresse Mecânico , Engenharia Tecidual/instrumentação , Células Tumorais Cultivadas
7.
Biomaterials ; 35(22): 5785-94, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24746967

RESUMO

Monolayer cultures of tumor cells and animal studies have tremendously advanced our understanding of cancer biology. However, we often lack animal models for human tumors, and cultured lines of human cells quickly lose their cancer signatures. In recent years, simple 3D models for cancer research have emerged, including cell culture in spheroids and on biomaterial scaffolds. Here we describe a bioengineered model of human Ewing's sarcoma that mimics the native bone tumor niche with high biological fidelity. In this model, cancer cells that have lost their transcriptional profiles after monolayer culture re-express genes related to focal adhesion and cancer pathways. The bioengineered model recovers the original hypoxic and glycolytic tumor phenotype, and enables re-expression of angiogenic and vasculogenic mimicry features that favor tumor adaptation. We propose that differentially expressed genes between the monolayer cell culture and native tumor environment are potential therapeutic targets that can be explored using the bioengineered tumor model.


Assuntos
Neoplasias Ósseas/genética , Sarcoma de Ewing/genética , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Engenharia Genética , Células HEK293 , Humanos , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patologia , Esferoides Celulares , Transcrição Gênica , Células Tumorais Cultivadas , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA