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

Métodos Terapêuticos e Terapias MTCI
Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Asian Pac J Cancer Prev ; 23(2): 561-571, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35225469

RESUMO

OBJECTIVE: The formation of three-dimensional spheroid tumor model using the scaffold-based platforms has been demonstrated over many years now. 3D tumor models are generated mainly in non-scalable culture systems, using synthetic and biological scaffolds. Many of these models fail to reflect the complex tumor microenvironment and do not allow long-term monitoring of tumor progression. This has resulted in inconsistent data in drug testing assays during preclinical and clinical studies. METHODS: To overcome these limitations, we have developed 3D tissueoids model by using novel AXTEX-4D platform. RESULTS: Cancer 3D tissueoids demonstrated the basic features of 3D cell culture with rapid attachment, proliferation, and longevity with contiguous cytoskeleton and hypoxic core. This study also demonstrated greater drug resistance in 3D-MCF-7 tissueoids in comparison to 2D monolayer cell culture. CONCLUSION: In conclusion, 3D-tissueoids are more responsive than 2D-cultured cells in simulating important tumor characteristics, anti-apoptotic features, and their resulting drug resistance.


Assuntos
Antineoplásicos/farmacologia , Técnicas de Cultura de Células em Três Dimensões/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Neoplasias/tratamento farmacológico , Esferoides Celulares/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos
2.
Asian Pac J Cancer Prev ; 23(2): 731-741, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35225487

RESUMO

BACKGROUND: Breast Cancer (BC) is a malignancy with high mortality among women. Recently, scaffold-based three-dimensional (3D) models have been developed for anti-cancer drug research. The present study aimed to investigate the anti-proliferative effects of Astragalus hamosus (A. hamosus) in 3D fibrin gel against MCF-7 cell line. We have also evaluated anti-proliferative effect of A. hamosus differences between 3D and 2D cultures. METHODS: The fibrin gel formulation was first optimized by testing the structural and mechanical properties. Then the cytotoxic effect of A. hamosus extract was assessed on MCF-7 cells by MTT assay. Cell apoptosis was evaluated using TUNEL method and flow cytometry. Cell cycle and proliferation were analyzed by flow cytometry. Apoptosis-related gene expression such as Bcl-2, caspase-3, -8 and -9 were quantified by real time-PCR. RESULTS: TUNEL staining showed a significant damage accompanied with cell apoptosis. Flow cytometry analysis revealed that apoptosis increased after treatment with A. hamosus extract in 3D culture model compared to 2D culture. The A. hamosus extract arrested cell cycle in the S and G2/M phases in 3D model while in the 2D culture G0/G1 phase was affected. Treatment with A. hamosus extract led to upregulation of the caspase-3, -8 and -9 genes and downregulation of the Ki-67 in the 3D-culture compared with the 2D culture. CONCLUSION: These results indicated that A. hamosus extract could be used as a therapeutic candidate for BC due to its anti-proliferative effects. Furthermore, 3D fibrin gel could be better than 2D-cultured cells in simulating important tumor characteristics in vivo, namely, anti-proliferative and anti-apoptotic features.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Astrágalo/química , Neoplasias da Mama/tratamento farmacológico , Técnicas de Cultura de Células em Três Dimensões/métodos , Extratos Vegetais/farmacologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Géis , Humanos , Células MCF-7
3.
Front Immunol ; 12: 781337, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925361

RESUMO

Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.


Assuntos
Bioensaio/métodos , Técnicas de Cultura de Células em Três Dimensões/métodos , Imunoterapia Adotiva , Células Matadoras Naturais/transplante , Neoplasias/terapia , Bioensaio/instrumentação , Técnicas de Cultura de Células em Três Dimensões/instrumentação , Linhagem Celular , Separação Celular , Feminino , Sangue Fetal , Voluntários Saudáveis , Humanos , Células-Tronco Pluripotentes Induzidas , Microscopia Intravital , Células Matadoras Naturais/imunologia , Dispositivos Lab-On-A-Chip , Masculino , Miócitos Cardíacos , Neoplasias/imunologia , Neoplasias/patologia , Cultura Primária de Células , Estudo de Prova de Conceito
5.
Int J Mol Sci ; 22(21)2021 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-34769400

RESUMO

INTRODUCTION: Introducing new drugs for clinical application is a very difficult, long, drawn-out, and costly process, which is why drug repositioning is increasingly gaining in importance. The aim of this study was to analyze the cytotoxic properties of ciprofloxacin and levofloxacin on bladder and prostate cell lines in vitro. METHODS: Bladder and prostate cancer cell lines together with their non-malignant counterparts were used in this study. In order to evaluate the cytotoxic effect of both drugs on tested cell lines, MTT assay, real-time cell growth analysis, apoptosis detection, cell cycle changes, molecular analysis, and 3D cultures were examined. RESULTS: Both fluoroquinolones exhibited a toxic effect on all of the tested cell lines. In the case of non-malignant cell lines, the cytotoxic effect was weaker, which was especially pronounced in the bladder cell line. A comparison of both fluoroquinolones showed the advantage of ciprofloxacin (lower doses of drug caused a stronger cytotoxic effect). Both fluoroquinolones led to an increase in late apoptotic cells and an inhibition of cell cycle mainly in the S phase. Molecular analysis showed changes in BAX, BCL2, TP53, and CDKN1 expression in tested cell lines following incubation with ciprofloxacin and levofloxacin. The downregulation of topoisomerase II genes (TOP2A and TOP2B) was noticed. Three-dimensional (3D) cell culture analysis confirmed the higher cytotoxic effect of tested fluoroquinolone against cancer cell lines. CONCLUSIONS: Our results suggest that both ciprofloxacin and levofloxacin may have great potential, especially in the supportive therapy of bladder cancer treatment. Taking into account the low costs of such therapy, fluoroquinolones seem to be ideal candidates for repositioning into bladder cancer therapeutics.


Assuntos
Biomarcadores Tumorais/metabolismo , Técnicas de Cultura de Células em Três Dimensões/métodos , Ciprofloxacina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Levofloxacino/farmacologia , Neoplasias Urogenitais/tratamento farmacológico , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Ciclo Celular , Proliferação de Células , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Perfilação da Expressão Gênica , Humanos , Inibidores da Topoisomerase II/farmacologia , Células Tumorais Cultivadas , Neoplasias Urogenitais/genética , Neoplasias Urogenitais/metabolismo , Neoplasias Urogenitais/patologia
6.
Stem Cell Reports ; 16(9): 2257-2273, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525385

RESUMO

Hair cell degeneration is a major cause of sensorineural hearing loss. Hair cells in mammalian cochlea do not spontaneously regenerate, posing a great challenge for restoration of hearing. Here, we establish a robust, high-throughput cochlear organoid platform that facilitates 3D expansion of cochlear progenitor cells and differentiation of hair cells in a temporally regulated manner. High-throughput screening of the FDA-approved drug library identified regorafenib, a VEGFR inhibitor, as a potent small molecule for hair cell differentiation. Regorafenib also promotes reprogramming and maturation of hair cells in both normal and neomycin-damaged cochlear explants. Mechanistically, inhibition of VEGFR suppresses TGFB1 expression via the MEK pathway and TGFB1 downregulation directly mediates the effect of regorafenib on hair cell reprogramming. Our study not only demonstrates the power of a cochlear organoid platform in high-throughput analyses of hair cell physiology but also highlights VEGFR-MEK-TGFB1 signaling crosstalk as a potential target for hair cell regeneration and hearing restoration.


Assuntos
Reprogramação Celular , Cóclea/metabolismo , Ensaios de Triagem em Larga Escala , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Organoides/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Técnicas de Cultura de Células em Três Dimensões/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Reprogramação Celular/genética , Cóclea/citologia , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Células Ciliadas Auditivas/citologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Camundongos , Camundongos Transgênicos , Organoides/citologia , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Mol Cancer ; 20(1): 125, 2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34587953

RESUMO

With advances in the discovery of the clinical and molecular landscapes of prostate cancer (PCa), implementation of precision medicine-guided therapeutic testing in the clinic has become a priority. Patient derived organoids (PDOs) are three-dimensional (3D) tissue cultures that promise to enable the validation of preclinical drug testing in precision medicine and coclinical trials by modeling PCa for predicting therapeutic responses with a reliable efficacy. We evaluate the advances in 3D culture and PDO use to model clonal heterogeneity and screen for effective targeted therapies, with a focus on the technological advances in generating PDOs. Recent innovations include the utilization of PDOs both in original research and/or correlative studies in clinical trials to examine drug effects within the PCa tumor microenvironment (TME). There has also been a significant improvement with the utilization of various extracellular matrices and single cell assays for the generation and long-term propagation of PDOs. Single cell derived PDOs could faithfully recapitulate the original tumor and reflect the heterogeneity features. While most PDO use for precision medicine understandably involved tissues derived from metastatic patients, we envision that the generation of PDOs from localized PCa along with the incorporation of cells of the TME in tissue models would fulfill the great potential of PDOs in predicting drug clinical benefits. We conclude that single cell derived PDOs reiterate the molecular features of the original tumor and represent a reliable pre-clinical PCa model to understand individual tumors and design tailored targeted therapies.


Assuntos
Antineoplásicos/farmacologia , Organoides/efeitos dos fármacos , Medicina de Precisão/métodos , Neoplasias da Próstata/patologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais , Técnicas de Cultura de Células em Três Dimensões/métodos , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Imunofluorescência/métodos , Heterogeneidade Genética , Genômica/métodos , Humanos , Imuno-Histoquímica , Masculino , Organoides/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/etiologia , Microambiente Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA