Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Nanoscale ; 16(16): 8046-8059, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38563130

RESUMO

The biomedical application of nanotechnology in cancer treatment has demonstrated significant potential for improving treatment efficiencies and ameliorating adverse effects. However, the medical translation of nanotechnology-based nanomedicines faces challenges including hazardous environmental effects, difficulties in large-scale production, and possible excessive costs. In the present study, we extracted and purified natural exosome-like nanoparticles (ELNs) from Phellinus linteus. These nanoparticles (denoted as P-ELNs) had an average particle size of 154.1 nm, displayed a negative zeta potential of -31.3 mV, and maintained stability in the gastrointestinal tract. Furthermore, P-ELNs were found to contain a diverse array of functional components, including lipids and pharmacologically active small-molecule constituents. In vitro investigations suggested that they exhibited high internalization efficiency in liver tumor cells (Hepa 1-6) and exerted significant anti-proliferative, anti-migratory, and anti-invasive effects against Hepa 1-6 cells. Strikingly, the therapeutic outcomes of oral P-ELNs were confirmed in an animal model of metastatic hepatocellular carcinoma by amplifying reactive oxygen species (ROS) and rebalancing the gut microbiome. These findings demonstrate the potential of P-ELNs as a promising oral therapeutic platform for liver cancer treatment.


Assuntos
Carcinoma Hepatocelular , Exossomos , Microbioma Gastrointestinal , Neoplasias Hepáticas , Espécies Reativas de Oxigênio , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/metabolismo , Humanos , Camundongos , Linhagem Celular Tumoral , Exossomos/metabolismo , Exossomos/química , Microbioma Gastrointestinal/efeitos dos fármacos , Basidiomycota/química , Basidiomycota/metabolismo , Nanopartículas/química , Phellinus/química , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Administração Oral
2.
Biomaterials ; 307: 122530, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38493672

RESUMO

The therapeutic efficacy of oral nanotherapeutics against colorectal cancer (CRC) is restricted by inadequate drug accumulation, immunosuppressive microenvironment, and intestinal microbiota imbalance. To overcome these challenges, we elaborately constructed 6-gingerol (Gin)-loaded magnetic mesoporous silicon nanoparticles and functionalized their surface with mulberry leaf-extracted lipids (MLLs) and Pluronic F127 (P127). In vitro experiments revealed that P127 functionalization and alternating magnetic fields (AMFs) promoted internalization of the obtained P127-MLL@Gins by colorectal tumor cells and induced their apoptosis/ferroptosis through Gin/ferrous ion-induced oxidative stress and magneto-thermal effect. After oral administration, P127-MLL@Gins safely passed to the colorectal lumen, infiltrated the mucus barrier, and penetrated into the deep tumors under the influence of AMFs. Subsequently, the P127-MLL@Gin (+ AMF) treatment activated antitumor immunity and suppressed tumor growth. We also found that this therapeutic modality significantly increased the abundance of beneficial bacteria (e.g., Bacillus and unclassified-c-Bacilli), reduced the proportions of harmful bacteria (e.g., Bacteroides and Alloprevotella), and increased lipid oxidation metabolites. Strikingly, checkpoint blockers synergistically improved the therapeutic outcomes of P127-MLL@Gins (+ AMF) against orthotopic and distant colorectal tumors and significantly prolonged mouse life spans. Overall, this oral therapeutic platform is a promising modality for synergistic treatment of CRC.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Lipossomos , Nanopartículas , Camundongos , Animais , Neoplasias Colorretais/tratamento farmacológico , Nanopartículas/uso terapêutico , Administração Oral , Fenômenos Magnéticos , Microambiente Tumoral
3.
J Nanobiotechnology ; 22(1): 4, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38169394

RESUMO

The clinical application of conventional medications for hepatocellular carcinoma treatment has been severely restricted by their adverse effects and unsatisfactory therapeutic effectiveness. Inspired by the concept of 'medicine food homology', we extracted and purified natural exosome-like lipid nanoparticles (LNPs) from black mulberry (Morus nigra L.) leaves. The obtained MLNPs possessed a desirable hydrodynamic particle size (162.1 nm), a uniform size distribution (polydispersity index = 0.025), and a negative surface charge (-26.6 mv). These natural LNPs were rich in glycolipids, functional proteins, and active small molecules (e.g., rutin and quercetin 3-O-glucoside). In vitro experiments revealed that MLNPs were preferentially internalized by liver tumor cell lines via galactose receptor-mediated endocytosis, increased intracellular oxidative stress, and triggered mitochondrial damage, resulting in suppressing the viability, migration, and invasion of these cells. Importantly, in vivo investigations suggested that oral MLNPs entered into the circulatory system mainly through the jejunum and colon, and they exhibited negligible adverse effects and superior anti-liver tumor outcomes through direct tumor killing and intestinal microbiota modulation. These findings collectively demonstrate the potential of MLNPs as a natural, safe, and robust nanomedicine for oral treatment of hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Morus , Nanopartículas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Folhas de Planta
4.
Nanomedicine (Lond) ; 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38275157

RESUMO

Aim: Despite some successful examples of therapeutic nanoparticles reaching clinical stages, there is still a significant need for novel formulations in order to improve the selectivity and efficacy of cancer treatment. Methods: The authors developed two novel dendrimer-gold (Au) complex-based nanoparticles using two different synthesis routes: complexation method (formulation A) and precipitation method (formulation B). Using a biomimetic cancer-on-a-chip model, the authors evaluated the possible cytotoxicity and internalization by colorectal cancer cells of dendrimer-Au complex-based nanoparticles. Results: The results showed promising capabilities of these nanoparticles for selectively targeting cancer cells and delivering drugs, particularly for the formulation A nanoparticles. Conclusion: This work highlights the potential of dendrimer-Au complex-based nanoparticles as a new strategy to improve the targeting of anticancer drugs.

5.
Small ; : e2307247, 2024 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-38243871

RESUMO

Oral treatment of colon diseases with the CRISPR/Cas9 system has been hampered by the lack of a safe and efficient delivery platform. Overexpressed CD98 plays a crucial role in the progression of ulcerative colitis (UC) and colitis-associated colorectal cancer (CAC). In this study, lipid nanoparticles (LNPs) derived from mulberry leaves are functionalized with Pluronic copolymers and optimized to deliver the CRISPR/Cas gene editing machinery for CD98 knockdown. The obtained LNPs possessed a hydrodynamic diameter of 267.2 nm, a narrow size distribution, and a negative surface charge (-25.6 mV). Incorporating Pluronic F127 into LNPs improved their stability in the gastrointestinal tract and facilitated their penetration through the colonic mucus barrier. The galactose end groups promoted endocytosis of the LNPs by macrophages via asialoglycoprotein receptor-mediated endocytosis, with a transfection efficiency of 2.2-fold higher than Lipofectamine 6000. The LNPs significantly decreased CD98 expression, down-regulated pro-inflammatory cytokines (TNF-α and IL-6), up-regulated anti-inflammatory factors (IL-10), and polarized macrophages to M2 phenotype. Oral administration of LNPs mitigated UC and CAC by alleviating inflammation, restoring the colonic barrier, and modulating intestinal microbiota. As the first oral CRISPR/Cas9 delivery LNP, this system offers a precise and efficient platform for the oral treatment of colon diseases.

6.
ACS Nano ; 18(4): 3651-3668, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38241481

RESUMO

Oral administration is the most preferred approach for treating colon diseases, and in situ vaccination has emerged as a promising cancer therapeutic strategy. However, the lack of effective drug delivery platforms hampered the application of in situ vaccination strategy in oral treatment of colorectal cancer (CRC). Here, we construct an oral core-shell nanomedicine by preparing a silk fibroin-based dual sonosensitizer (chlorin e6, Ce6)- and immunoadjuvant (imiquimod, R837)-loaded nanoparticle as the core, with its surface coated with plant-extracted lipids and pluronic F127 (p127). The resultant nanomedicines (Ce6/R837@Lp127NPs) maintain stability during their passage through the gastrointestinal tract and exert improved locomotor activities under ultrasound irradiation, achieving efficient colonic mucus infiltration and specific tumor penetration. Thereafter, Ce6/R837@Lp127NPs induce immunogenic death of colorectal tumor cells by sonodynamic treatment, and the generated neoantigens in the presence of R837 serve as a potent in situ vaccine. By integrating with immune checkpoint blockades, the combined treatment modality inhibits orthotopic tumors, eradicates distant tumors, and modulates intestinal microbiota. As the first oral in situ vaccination, this work spotlights a robust oral nanoplatform for producing a personalized vaccine against CRC.


Assuntos
Neoplasias Colorretais , Nanopartículas , Vacinas , Humanos , Imiquimode , Linhagem Celular Tumoral , Nanomedicina , Neoplasias Colorretais/tratamento farmacológico , Vacinação , Imunoterapia
7.
Molecules ; 28(21)2023 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-37959830

RESUMO

As an emerging technology, microneedles offer advantages such as painless administration, good biocompatibility, and ease of self-administration, so as to effectively treat various diseases, such as diabetes, wound repair, tumor treatment and so on. How to regulate the release behavior of loaded drugs in polymer microneedles is the core element of transdermal drug delivery. As an emerging on-demand drug-delivery technology, intelligent responsive microneedles can achieve local accurate release of drugs according to external stimuli or internal physiological environment changes. This review focuses on the research efforts in smart responsive polymer microneedles at home and abroad in recent years. It summarizes the response mechanisms based on various stimuli and their respective application scenarios. Utilizing innovative, responsive microneedle systems offers a convenient and precise targeted drug delivery method, holding significant research implications in transdermal drug administration. Safety and efficacy will remain the key areas of continuous efforts for research scholars in the future.


Assuntos
Pele , Polímeros Responsivos a Estímulos , Administração Cutânea , Sistemas de Liberação de Medicamentos , Preparações Farmacêuticas , Polímeros/farmacologia
8.
Biomaterials ; 302: 122332, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37801790

RESUMO

The treatment efficacies of conventional medications against colorectal cancer (CRC) are restricted by a low penetrative, hypoxic, and immunosuppressive tumor microenvironment. To address these restrictions, we developed an innovative antitumor platform that employs calcium overload-phototherapy using mitochondrial N770-conjugated mesoporous silica nanoparticles loaded with CaO2 (CaO2-N770@MSNs). A loading level of 14.0 wt% for CaO2-N770@MSNs was measured, constituting an adequate therapeutic dosage. With the combination of oxygen generated from CaO2 and hyperthermia under near-infrared irradiation, CaO2-N770@MSNs penetrated through the dense mucus, accumulated in the colorectal tumor tissues, and inhibited tumor cell growth through endoplasmic reticulum stress and mitochondrial damage. The combination of calcium overload and phototherapy revealed high therapeutic efficacy against orthotopic colorectal tumors, alleviated the immunosuppressive microenvironment, elevated the abundance of beneficial microorganisms (e.g., Lactobacillaceae and Lachnospiraceae), and decreased harmful microorganisms (e.g., Bacteroidaceae and Muribaculaceae). Moreover, together with immune checkpoint blocker (αPD-L1), these nanoparticles showed an ability to eradicate both orthotopic and distant tumors, while potentiating systemic antitumor immunity. This treatment platform (CaO2-N770@MSNs plus αPD-L1) open a new horizon of synergistic treatment against hypoxic CRC with high killing power and safety.


Assuntos
Neoplasias Colorretais , Hipertermia Induzida , Nanopartículas , Humanos , Cálcio , Linhagem Celular Tumoral , Fototerapia , Neoplasias Colorretais/terapia , Imunoterapia , Hipóxia , Microambiente Tumoral
9.
Acta Biomater ; 170: 202-214, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37619895

RESUMO

Current treatments for cancer, such as chemotherapy, radiotherapy, immunotherapy, and surgery, have positive results but are generally ineffective against metastatic tumors. Treatment effectiveness can be improved by employing bioengineered cancer traps, typically utilizing chemoattractant-loaded materials, to attract infiltrating cancer cells preventing their uncontrolled spread and potentially enabling eradication. However, the encapsulated chemical compounds can have adverse effects on other cells causing unwanted responses, and the generated gradients can evolve unpredictably. Here, we report the development of a cancer trap based on mechanical ratchet structures to capture metastatic cells. The traps use an array of asymmetric local features to mechanically attract cancer cells and direct their migration for prolonged periods. The trapping efficiency was found to be greater than isotropic or inverse anisotropic ratchet structures on either disseminating cancer cells and tumor spheroids. Importantly, the traps exhibited a reduced effectiveness when targeting non-metastatic and non-tumorigenic cells, underscoring their particular suitability for capturing highly invasive cancer cells. Overall, this original approach may have therapeutic implications for fighting cancer, and may also be used to control cell motility for other biological processes. STATEMENT OF SIGNIFICANCE: Current cancer treatments have limitations in treating metastatic tumors, where cancer cells can invade distant organs. Biomaterials loaded with chemoattractants can be implanted to attract and capture metastatic cells preventing uncontrolled spread. However, encapsulated chemical compounds can have adverse effects on other cells, and gradients can evolve unpredictably. This paper presents an original concept of "cancer traps" based on using mechanical ratchet-based structures to capture metastatic cancer cells, with greater trapping efficiency and stability than previously studied methods. This innovative approach has significant potential clinical implications for fighting cancer, particularly in treating metastatic tumors. Additionally, it could be applied to control cell motility for other biological processes, opening new possibilities for biomedicine and tissue engineering.

10.
Artigo em Inglês | MEDLINE | ID: mdl-36600447

RESUMO

The alarming increase in the number of breast cancer patients worldwide and the increasing death rate indicate that the traditional and current medicines are insufficient to fight against it. The onset of chemo- and radioresistances and cancer stem cell-based recurrence make this problem harder, and this hour needs a novel treatment approach. Competent nanoparticle-based accurate drug delivery and cancer nanotheranostics like photothermal therapy, photodynamic therapy, chemodynamic therapy, and sonodynamic therapy can be the key to solving this problem due to their unique characteristics. These innovative formulations can be a better cargo with fewer side effects than the standard chemotherapy and can eliminate the stability problems associated with cancer immunotherapy. The nanotheranostic systems can kill the tumor cells and the resistant breast cancer stem cells by novel mechanisms like local hyperthermia and reactive oxygen species and prevent tumor recurrence. These theranostic systems can also combine with chemotherapy or immunotherapy approaches. These combining approaches can be the future of anticancer therapy, especially to overcome the breast cancer stem cells mediated chemo- and radioresistances. This review paper discusses several novel theranostic systems and smart nanoparticles, their mechanism of action, and their modifications with time. It explains their relevance and market scope in the current era. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.


Assuntos
Neoplasias da Mama , Nanopartículas , Humanos , Feminino , Neoplasias da Mama/tratamento farmacológico , Nanomedicina Teranóstica , Sistemas de Liberação de Medicamentos , Nanopartículas/uso terapêutico , Células-Tronco Neoplásicas/patologia , Linhagem Celular Tumoral
11.
Adv Biol (Weinh) ; 7(4): e2200141, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36658719

RESUMO

Breast cancer is still the leading cause of women's death due to relapse and metastasis. In vitro tumor models are considered reliable tools for drug screening and understanding cancer-driving mechanisms due to the possibility of mimicking tumor heterogeneity. Herein, a 3D breast cancer model (3D-BCM) is developed based on enzymatically-crosslinked silk fibroin (eSF) hydrogels. Human MCF7 breast cancer cells are encapsulated into eSF hydrogels, with and without human mammary fibroblasts. The spontaneously occurring conformational change from random coil to ß-sheet is correlated with increased eSF hydrogels' stiffness over time. Moreover, mechanical properties analysis confirms that the cells can modify the stiffness of the hydrogels, mimicking the microenvironment stiffening occurring in vivo. Fibroblasts support cancer cells growth and assembly in the eSF hydrogels up to 14 days of culture. Co-cultured 3D-BCM exhibits an upregulated expression of genes related to extracellular matrix remodeling and fibroblast activation. The 3D-BCM is subjected to doxorubicin and paclitaxel treatments, showing differential drug response. Overall, these results suggest that the co-culture of breast cancer cells and fibroblasts in eSF hydrogels allow the development of a mimetic in vitro platform to study cancer progression. This opens up new research avenues to investigate novel molecular targets for anti-cancer therapy.


Assuntos
Antineoplásicos , Neoplasias da Mama , Humanos , Feminino , Técnicas de Cocultura , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Recidiva Local de Neoplasia , Antineoplásicos/farmacologia , Hidrogéis , Fibroblastos/patologia , Microambiente Tumoral
12.
Mater Today Bio ; 18: 100537, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36659998

RESUMO

Cell migration is essential in numerous living processes, including embryonic development, wound healing, immune responses, and cancer metastasis. From individual cells to collectively migrating epithelial sheets, the locomotion of cells is tightly regulated by multiple structural, chemical, and biological factors. However, the high complexity of this process limits the understanding of the influence of each factor. Recent advances in materials science, tissue engineering, and microtechnology have expanded the toolbox and allowed the development of biomimetic in vitro assays to investigate the mechanisms of cell migration. Particularly, three-dimensional (3D) hydrogels have demonstrated a superior ability to mimic the extracellular environment. They are therefore well suited to studying cell migration in a physiologically relevant and more straightforward manner than in vivo approaches. A myriad of synthetic and naturally derived hydrogels with heterogeneous characteristics and functional properties have been reported. The extensive portfolio of available hydrogels with different mechanical and biological properties can trigger distinct biological responses in cells affecting their locomotion dynamics in 3D. Herein, we describe the most relevant hydrogels and their associated physico-chemical characteristics typically employed to study cell migration, including established cell migration assays and tracking methods. We aim to give the reader insight into existing literature and practical details necessary for performing cell migration studies in 3D environments.

13.
Adv Healthc Mater ; 12(14): e2201442, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35998112

RESUMO

Microfluidic platforms represent a powerful approach to miniaturizing important characteristics of cancers, improving in vitro testing by increasing physiological relevance. Different tools can manipulate cells and materials at the microscale, but few offer the efficiency and versatility of light and optical technologies. Moreover, light-driven technologies englobe a broad toolbox for quantifying critical biological phenomena. Herein, the role of photonics in microfluidic 3D cancer modeling and biosensing from three major perspectives is reviewed. First, optical-driven technologies are looked upon, as these allow biomaterials and living cells to be manipulated with microsized precision and present opportunities to advance 3D microfluidic models by engineering cancer microenvironments' hallmarks, such as their architecture, cellular complexity, and vascularization. Second, the growing field of optofluidics is discussed, exploring how optical tools can directly interface microfluidic chips, enabling the extraction of relevant biological data, from single fluorescent signals to the complete 3D imaging of diseased cells within microchannels. Third, advances in optical cancer biosensing are reviewed, focusing on how light-matter interactions can detect biomarkers, rare circulating tumor cells, and cell-derived structures such as exosomes. Photonic technologies' current challenges and caveats in microfluidic 3D cancer models are overviewed, outlining future research avenues that may catapult the field.


Assuntos
Microfluídica , Células Neoplásicas Circulantes , Humanos , Microfluídica/métodos , Óptica e Fotônica , Materiais Biocompatíveis , Modelos Biológicos , Microambiente Tumoral
14.
Artif Organs ; 47(1): 47-61, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36029128

RESUMO

BACKGROUND: Several factors like three-dimensional microstructure, growth factors, cytokines, cell-cell communication, and coculture with functional cells can affect the stem cells behavior and differentiation. The purpose of this study was to investigate the potential of decellularized placental sponge as adipose-derived mesenchymal stem cells (AD-MSCs) and macrophage coculture systems, and guiding the osteogenic differentiation of stem cells. METHODS: The decellularized placental sponge (DPS) was fabricated, and its mechanical characteristics were evaluated using degradation assay, swelling rate, and pore size determination. Its structure was also investigated using hematoxylin and eosin staining and scanning electron microscopy. Mouse peritoneal macrophages and AD-MSCs were isolated and characterized. The differentiation potential of AD-MSCs co-cultured with macrophages was evaluated by RT-qPCR of osteogenic genes on the surface of DPS. The in vivo biocompatibility of DPS was determined by subcutaneous implantation of scaffold and histological evaluations of the implanted site. RESULTS: The DPS had 67% porosity with an average pore size of 238 µm. The in vitro degradation assay showed around 25% weight loss during 30 days in PBS. The swelling rate was around 50% during 72 h. The coculture of AD-MSCs/macrophages on the DPS showed a significant upregulation of four differentiation osteogenic lineage genes in AD-MSCs on days 14 and 21 and a significantly higher mineralization rate than the groups without DPS. Subcutaneous implantation of DPS showed in vivo biocompatibility of scaffold during 28 days follow-up. CONCLUSIONS: Our findings suggest the decellularized placental sponge as an excellent bone substitute providing a naturally derived matrix substrate with biostructure close to the natural bone that guided differentiation of stem cells toward bone cells and a promising coculture substrate for crosstalk of macrophage and mesenchymal stem cells in vitro.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Gravidez , Feminino , Camundongos , Animais , Osteogênese/fisiologia , Técnicas de Cocultura , Alicerces Teciduais/química , Placenta , Diferenciação Celular/fisiologia , Macrófagos/metabolismo , Células Cultivadas
15.
Adv Exp Med Biol ; 1379: 81-112, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35760989

RESUMO

Despite the significant amount of resources invested, cancer remains a considerable burden in our modern society and a leading cause of death. There is still a lack of knowledge about the mechanistic determinants of the disease, the mechanism of action of drugs, and the process of tumor relapse. Current methodologies to study all these events fail to provide accurate information, threatening the prognosis of cancer patients. This failure is due to the inadequate procedure in how tumorigenesis is studied and how drug discovery and screening are currently made. Traditionally, they both rely on seeding cells on static flat cultures and on the immunolabelling of cellular structures, which are usually limited in their ability to reproduce the complexity of the native cellular habitat and provide quantitative data. Similarly, more complex animal models are employed for-unsuccessfully-mimicking the human physiology and evaluating the etiology of the disease or the efficacy/toxicity of pharmacological compounds. Despite some breakthroughs and success obtained in understanding the disease and developing novel therapeutic approaches, cancer still kills millions of people worldwide, remaining a global healthcare problem with a high social and economic impact. There is a need for novel integrative methodologies and technologies capable of providing valuable readouts. In this regard, the combination of microfluidics technology with miniaturized biosensors offers unprecedented advantages to accelerate the development of drugs. This integrated technology have the potential to unravel the key pathophysiological processes of cancer progression and metastasis, overcoming the existing gap on in vitro predictive platforms and in vivo model systems. Herein, we discuss how this combination may boost the field of cancer theranostics and drug discovery/screening toward more precise devices with clinical relevance.


Assuntos
Técnicas Biossensoriais , Microfluídica , Animais , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Humanos , Microfluídica/métodos , Recidiva Local de Neoplasia
16.
Adv Exp Med Biol ; 1379: 307-318, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35760997

RESUMO

Early cancer detection is still a major clinical challenge. The development of innovative and noninvasive screening approaches for the detection of predictive biomarkers indicating the stage of the disease could save many lives. Traditional in vitro and in vivo models are not adequate to copycat the native tumor microenvironment and for the discovery of new biomarkers. Recent advances in microfluidics, biosensors, and 3D cell biology speed up the development of micro-physiological bioengineered systems that improve the discovery of new potential cancer biomarkers. This can accelerate the individualization of cancer treatments leading to precision medicine-oriented approaches that could improve patient prognosis. For this reason, it is necessary to develop point-of-care diagnostic tools that can be user-friendly, miniaturized, and easily translated into clinical practice. This chapter describes how far this new generation of cutting-edge technologies, such as microfluidics, label-free detection systems, and molecular diagnostics, are from being applied in the current clinical practice.


Assuntos
Técnicas Biossensoriais , Neoplasias , Biomarcadores Tumorais , Detecção Precoce de Câncer , Humanos , Microfluídica , Neoplasias/diagnóstico , Medicina de Precisão
17.
Adv Exp Med Biol ; 1379: 369-387, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35761000

RESUMO

Early cancer screening and effective diagnosis is the most effective form to diminish the number of cancer-related deaths. Liquid biopsy constitutes an attractive alternative to tumor biopsy due to its non-invasive nature and sample accessibility, which permits effective screening and patient monitoring. Within the plethora of biomarkers present in circulation, liquid biopsy has mainly been performed by analyzing circulating tumor cells, and more recently, extracellular vesicles. Tracking these biological particles could provide valuable insights into cancer origin, progression, treatment efficacy, and patient prognosis. Microfluidic devices have emerged as viable solutions for point-of-care cancer screening and monitoring due to their user-friendly operation, low operation costs, and capability of processing, quantifying, and analyzing these bioparticles in a single device. However, the size difference between cells and exosomes (micrometer vs nanometer) requires an adaptation of microfluidic isolation approaches, particularly in label-free methodologies governed by particle and fluid mechanics. This chapter will explore the theory behind particle isolation and sorting in different microfluidic techniques necessary to guide researchers into the design and development of such devices.


Assuntos
Exossomos , Células Neoplásicas Circulantes , Biomarcadores Tumorais/análise , Exossomos/patologia , Humanos , Biópsia Líquida , Microfluídica/métodos , Células Neoplásicas Circulantes/patologia
18.
Adv Exp Med Biol ; 1379: 461-495, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35761004

RESUMO

Microfluidics and biosensors have already demonstrated their potential in cancer research. Typical applications of microfluidic devices include the realistic modeling of the tumor microenvironment for mechanistic investigations or the real-time monitoring/screening of drug efficacy. Similarly, point-of-care biosensing platforms are instrumental for the early detection of predictive biomarkers and their accurate quantification. The combination of both technologies offers unprecedented advantages for the management of the disease, with an enormous potential to contribute to improving patient prognosis. Despite their high performance, these methodologies are still encountering obstacles for being adopted by the healthcare market, such as a lack of standardization, reproducibility, or high technical complexity. Therefore, the cancer research community is demanding better tools capable of boosting the efficiency of cancer diagnosis and therapy. During the last years, innovative microfluidic and biosensing technologies, both individually and combined, have emerged to improve cancer theranostics. In this chapter, we discuss how these emerging-and in some cases unconventional-microfluidics and biosensor technologies, tools, and concepts can enhance the predictive power of point-of-care devices and the development of more efficient cancer therapies.


Assuntos
Técnicas Biossensoriais , Neoplasias , Técnicas Biossensoriais/métodos , Humanos , Dispositivos Lab-On-A-Chip , Microfluídica/métodos , Neoplasias/diagnóstico , Neoplasias/terapia , Medicina de Precisão , Reprodutibilidade dos Testes , Microambiente Tumoral
19.
Bioengineering (Basel) ; 9(4)2022 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-35447726

RESUMO

In vitro cancer models are envisioned as high-throughput screening platforms for potential new therapeutic discovery and/or validation. They also serve as tools to achieve personalized treatment strategies or real-time monitoring of disease propagation, providing effective treatments to patients. To battle the fatality of metastatic cancers, the development and commercialization of predictive and robust preclinical in vitro cancer models are of urgent need. In the past decades, the translation of cancer research from 2D to 3D platforms and the development of diverse in vitro cancer models have been well elaborated in an enormous number of reviews. However, the meagre clinical success rate of cancer therapeutics urges the critical introspection of currently available preclinical platforms, including patents, to hasten the development of precision medicine and commercialization of in vitro cancer models. Hence, the present article critically reflects the difficulty of translating cancer therapeutics from discovery to adoption and commercialization in the light of in vitro cancer models as predictive tools. The state of the art of in vitro cancer models is discussed first, followed by identifying the limitations of bench-to-bedside transition. This review tries to establish compatibility between the current findings and obstacles and indicates future directions to accelerate the market penetration, considering the niche market.

20.
Lab Chip ; 22(6): 1093-1125, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35253032

RESUMO

Extracellular vesicles (EVs) are small lipidic particles packed with proteins, DNA, messenger RNA and microRNAs of their cell of origin that act as critical players in cell-cell communication. These vesicles have been identified as pivotal mediators in cancer progression and the formation of metastatic niches. Hence, their isolation and analysis from circulating biofluids is envisioned as the next big thing in the field of liquid biopsies for early non-invasive diagnosis and patient follow-up. Despite the promise, current benchtop isolation strategies are not compatible with point-of-care testing in a clinical setting. Microfluidic platforms are disruptive technologies capable of recovering, analyzing, and quantifying EVs within clinical samples with limited volume, in a high-throughput manner with elevated sensitivity and multiplexing capabilities. Moreover, they can also be employed for the controlled production of synthetic EVs and effective drug loading to produce EV-based therapies. In this review, we explore the use of microfluidic platforms for the isolation, characterization, and quantification of EVs in cancer, and compare these platforms with the conventional methodologies. We also highlight the state-of-the-art in microfluidic approaches for EV-based cancer therapeutics. Finally, we analyze the currently active or recently completed clinical trials involving EVs for cancer diagnosis, treatment or therapy monitoring and examine the future of EV-based point-of-care testing platforms in the clinic and EV-based therapy production by the industry.


Assuntos
Vesículas Extracelulares , MicroRNAs , Neoplasias , Vesículas Extracelulares/metabolismo , Humanos , Biópsia Líquida , MicroRNAs/metabolismo , Microfluídica , Neoplasias/diagnóstico , Neoplasias/metabolismo , Neoplasias/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA