Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Drug Deliv Rev ; 203: 115135, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37931847

RESUMO

Chimeric Antigen Receptor T cell (CAR-T) therapy has emerged as a transformative therapeutic strategy for hematological malignancies. However, its efficacy in treating solid tumors remains limited. An in-depth and comprehensive understanding of CAR-T cell signaling pathways and the ability to track CAR-T cell biodistribution and activation in real-time within the tumor microenvironment will be instrumental in designing the next generation of CAR-T cells for solid tumor therapy. This review summarizes the signaling network and the cellular and molecular imaging tools and platforms that are utilized in CAR-T cell-based immune therapies, covering both in vitro and in vivo studies. Firstly, we provide an overview of the existing understanding of the activation and cytotoxic mechanisms of CAR-T cells, compared to the mechanism of T cell receptor (TCR) signaling pathways. We further describe the commonly employed tools for live cell imaging, coupled with recent research progress, with a focus on genetically encoded fluorescent proteins (FPs) and biosensors. We then discuss the utility of diverse in vivo imaging modalities, including fluorescence and bioluminescence imaging, Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET), and photoacoustic (PA) imaging, for noninvasive monitoring of CAR-T cell dynamics within tumor tissues, thereby providing critical insights into therapy's strengths and weaknesses. Lastly, we discuss the current challenges and future directions of CAR-T cell therapy from the imaging perspective. We foresee that a comprehensive and integrative approach to CAR-T cell imaging will enable the development of more effective treatments for solid tumors in the future.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Distribuição Tecidual , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Imunoterapia , Linfócitos T , Imagem Molecular , Microambiente Tumoral
2.
Front Cell Dev Biol ; 10: 1064484, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36483679

RESUMO

Immune-engineering is a rapidly emerging field in the past few years, as immunotherapy evolved from a paradigm-shifting therapeutic approach for cancer treatment to promising immuno-oncology models in clinical trials and commercial products. Linking the field of biomedical engineering with immunology, immuno-engineering applies engineering principles and utilizes synthetic biology tools to study and control the immune system for diseases treatments and interventions. Over the past decades, there has been a deeper understanding that mechanical forces play crucial roles in regulating immune cells at different stages from antigen recognition to actual killing, which suggests potential opportunities to design and tailor mechanobiology tools to novel immunotherapy. In this review, we first provide a brief introduction to recent technological and scientific advances in mechanobiology for immune cells. Different strategies for immuno-engineering are then discussed and evaluated. Furthermore, we describe the opportunities and challenges of applying mechanobiology and related technologies to study and engineer immune cells and ultimately modulate their function for immunotherapy. In summary, the synergetic integration of cutting-edge mechanical biology techniques into immune-engineering strategies can provide a powerful platform and allow new directions for the field of immunotherapy.

3.
Clin Transl Med ; 12(12): e1141, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36495108

RESUMO

Cell-based immunotherapy, for example, chimeric antigen receptor T (CAR-T) cell immunotherapy, has revolutionized cancer treatment, particularly for blood cancers. However, factors such as insufficient T cell tracking, tumour heterogeneity, inhibitory tumour microenvironment (TME) and T cell exhaustion limit the broad application of CAR-based immunotherapy for solid tumours. In particular, the TME is a complex and evolving entity, which is composed of cells of different types (e.g., cancer cells, immune cells and stromal cells), vasculature, soluble factors and extracellular matrix (ECM), with each component playing a critical role in CAR-T immunotherapy. Thus, developing approaches to mitigate the inhibitory TME factors is critical for future success in applying CAR-T cells for solid tumour treatment. Accordingly, understanding the bilateral interaction of CAR-T cells with the TME is in pressing need to pave the way for more efficient therapeutics. In the following review, we will discuss TME-associated aspects with an emphasis on T cell trafficking, ECM barriers, abnormal vasculature, solid tumour heterogenicity and immune suppressive microenvironment. We will then summarize current engineering strategies to overcome the challenges posed by the TME-associated factors. Lastly, the future directions for engineering efficient CAR-T cells for solid tumour therapy will be discussed.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Imunoterapia Adotiva , Microambiente Tumoral , Linfócitos T/metabolismo , Linfócitos T/patologia
4.
Front Cell Dev Biol ; 8: 504, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32656213

RESUMO

Focused ultrasound (FUS) is a rapidly developing stimulus technology with the potential to uncover novel mechanosensory dependent cellular processes. Since it is non-invasive, it holds great promise for future therapeutic applications in patients used either alone or as a complement to boost existing treatments. For example, FUS stimulation causes invasive but not non-invasive cancer cell lines to exhibit marked activation of calcium signaling pathways. Here, we identify the membrane channel PANNEXIN1 (PANX1) as a mediator for activation of calcium signaling in invasive cancer cells. Knockdown of PANX1 decreases calcium signaling in invasive cells, while PANX1 overexpression enhances calcium elevations in non-invasive cancer cells. We demonstrate that FUS may directly stimulate mechanosensory PANX1 localized in endoplasmic reticulum to evoke calcium release from internal stores. This process does not depend on mechanosensory stimulus transduction through an intact cytoskeleton and does not depend on plasma membrane localized PANX1. Plasma membrane localized PANX1, however, plays a different role in mediating the spread of intercellular calcium waves via ATP release. Additionally, we show that FUS stimulation evokes cytokine/chemokine release from invasive cancer cells, suggesting that FUS could be an important new adjuvant treatment to improve cancer immunotherapy.

5.
Curr Opin Biotechnol ; 66: 88-94, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32717634

RESUMO

Recent synthetic biology advancements have shown that cells can be engineered to respond to external stimuli such as chemical compounds and light, which significantly improves the specificity and controllability of CAR T therapy. However, the lack of both spatiotemporal and depth control is still the main issue in the clinic of CAR T treatment. At the same time, mechanogenetics, capable of penetrating deep tissues with high spatiotemporal precision, is rapidly evolving and advancing to reveal its potential for cancer immunotherapy. In the past few years, researchers have demonstrated the precise and remote control of engineered cells with mechanical perturbation originated from ultrasound, which may become a new solution to circumvent the limitations of CAR T therapy in the future. This review will discuss mechanobiology and the state-of art designs of controllable CAR T cells. A specific focus of this review will be on the mechanical control of CAR T therapy.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T , Engenharia Celular , Humanos , Imunoterapia , Neoplasias/genética , Neoplasias/terapia , Linfócitos T
6.
Microsyst Nanoeng ; 6: 39, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34567652

RESUMO

Advancements in diagnostic systems for metastatic cancer over the last few decades have played a significant role in providing patients with effective treatment by evaluating the characteristics of cancer cells. Despite the progress made in cancer prognosis, we still rely on the visual analysis of tissues or cells from histopathologists, where the subjectivity of traditional manual interpretation persists. This paper presents the development of a dual diagnosis and treatment tool using an in vitro acoustic tweezers platform with a 50 MHz ultrasonic transducer for label-free trapping and bursting of human breast cancer cells. For cancer cell detection and classification, the mechanical properties of a single cancer cell were quantified by single-beam acoustic tweezers (SBAT), a noncontact assessment tool using a focused acoustic beam. Cell-mimicking phantoms and agarose hydrogel spheres (AHSs) served to standardize the biomechanical characteristics of the cells. Based on the analytical comparison of deformability levels between the cells and the AHSs, the mechanical properties of the cells could be indirectly measured by interpolating the Young's moduli of the AHSs. As a result, the calculated Young's moduli, i.e., 1.527 kPa for MDA-MB-231 (highly invasive breast cancer cells), 2.650 kPa for MCF-7 (weakly invasive breast cancer cells), and 2.772 kPa for SKBR-3 (weakly invasive breast cancer cells), indicate that highly invasive cancer cells exhibited a lower Young's moduli than weakly invasive cells, which indicates a higher deformability of highly invasive cancer cells, leading to a higher metastasis rate. Single-cell treatment may also be carried out by bursting a highly invasive cell with high-intensity, focused ultrasound.

7.
Artigo em Inglês | MEDLINE | ID: mdl-29994523

RESUMO

Ultrasound guided needle biopsy is an important method for collection of breast cancer tissue. In this paper, we report on the design and testing of a high-voltage 1 to 64 Multiplexer/Demultiplexer (MUX/De-MUX) integrated circuit (IC) for ultrasound-guided breast biopsy applications implemented in a high-voltage CMOS process. The IC is intended to be incorporated inside the breast biopsy needle and is designed to fit inside the needle inner diameter of 2.38 mm. The MUX/De-MUX electronics are made up of three parts, including a low-voltage 6 to 64 decoder, a level shifter to convert from low voltage to high voltage, and analog high-voltage switches. Experimental results show a -3-dB bandwidth of over 70 MHz, Rds (on) of , -2.279-dB insertion loss, and -17.5-dB off isolation at 70 MHz with low-voltage input. Finally, we present results obtained via synthetic aperture imaging using the fabricated MUX/De-Mux device and a high-frequency ultrasound array. This device and technique hold promise for high-frequency imaging probes where a limited number of elements are used and the depth of penetration is short such as in breast biopsy and intravascular applications.


Assuntos
Biópsia por Agulha/métodos , Mama/diagnóstico por imagem , Biópsia Guiada por Imagem/métodos , Ultrassonografia/métodos , Neoplasias da Mama/diagnóstico por imagem , Feminino , Humanos , Transdutores
8.
Ann Biomed Eng ; 46(1): 48-59, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29086222

RESUMO

In recent years, ultrasound has gained attention in new biological applications due to its ability to induce specific biological responses at the cellular level. Although the biophysical mechanisms underlying the interaction between ultrasound and cells are not fully understood, many agree on a pivotal role of Ca2+ signaling through mechanotransduction pathways. Because Ca2+ regulates a vast range of downstream cellular processes, a better understanding of how ultrasound influences Ca2+ signaling could lead to new applications for ultrasound. In this study, we investigated the mechanism of ultrasound-induced Ca2+ mobilization in human mesenchymal stem cells using 47 MHz focused ultrasound to stimulate single cells at low intensities (~ 110 mW/cm2). We found that ultrasound exposure triggers opening of connexin 43 hemichannels on the plasma membrane, causing release of ATP into the extracellular space. That ATP then binds to G-protein-coupled P2Y1 purinergic receptors on the membrane, in turn activating phospholipase C, which evokes production of inositol trisphosphate and release of Ca2+ from intracellular stores.


Assuntos
Cálcio/metabolismo , Conexina 43/metabolismo , Células-Tronco Mesenquimais/efeitos da radiação , Ondas Ultrassônicas , Sobrevivência Celular , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/metabolismo
9.
Front Oncol ; 7: 161, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28824873

RESUMO

Cancer cells undergo a number of biophysical changes as they transform from an indolent to an aggressive state. These changes, which include altered mechanical and electrical properties, can reveal important diagnostic information about disease status. Here, we introduce a high-throughput, functional technique for assessing cancer cell invasion potential, which works by probing for the mechanically excitable phenotype exhibited by invasive cancer cells. Cells are labeled with fluorescent calcium dye and imaged during stimulation with low-intensity focused ultrasound, a non-contact mechanical stimulus. We show that cells located at the focus of the stimulus exhibit calcium elevation for invasive prostate (PC-3 and DU-145) and bladder (T24/83) cancer cell lines, but not for non-invasive cell lines (BPH-1, PNT1A, and RT112/84). In invasive cells, ultrasound stimulation initiates a calcium wave that propagates from the cells at the transducer focus to other cells, over distances greater than 1 mm. We demonstrate that this wave is mediated by extracellular signaling molecules and can be abolished through inhibition of transient receptor potential channels and inositol trisphosphate receptors, implicating these proteins in the mechanotransduction process. If validated clinically, our technology could provide a means to assess tumor invasion potential in cytology specimens, which is not currently possible. It may therefore have applications in diseases such as bladder cancer, where cytologic diagnosis of tumor invasion could improve clinical decision-making.

10.
Ultrasonics ; 63: 94-101, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26150401

RESUMO

Extracellular matrix proteins such as fibronectin (FNT) play crucial roles in cell proliferation, adhesion, and migration. For better understanding of these associated cellular activities, various microscopic manipulation tools have been used to study their intracellular signaling pathways. Recently, it has appeared that acoustic tweezers may possess similar capabilities in the study. Therefore, we here demonstrate that our newly developed acoustic tweezers with a high-frequency lithium niobate ultrasonic transducer have potentials to study intracellular calcium signaling by FNT-binding to human breast cancer cells (SKBR-3). It is found that intracellular calcium elevations in SKBR-3 cells, initially occurring on the microbead-contacted spot and then eventually spreading over the entire cell, are elicited by attaching an acoustically trapped FNT-coated microbead. Interestingly, they are suppressed by either extracellular calcium elimination or phospholipase C (PLC) inhibition. Hence, this suggests that our acoustic tweezers may serve as an alternative tool in the study of intracellular signaling by FNT-binding activities.


Assuntos
Micromanipulação/instrumentação , Microesferas , Transdução de Sinais , Ultrassom/métodos , Neoplasias da Mama/metabolismo , Separação Celular/instrumentação , Desenho de Equipamento , Fibronectinas , Humanos , Transdutores , Células Tumorais Cultivadas
11.
ACS Nano ; 7(4): 3061-77, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23557138

RESUMO

Chemotherapy for intracranial gliomas is hampered by limited delivery of therapeutic agents through the blood brain barrier (BBB). An optimal therapeutic agent for brain tumors would selectively cross the BBB, accumulates in the tumor tissue and be activated from an innocuous prodrug within the tumor. Here we show brain tumor-targeted delivery and therapeutic efficacy of a nanometer-sized prodrug (nanoprodrug) of camptothecin (CPT) to treat experimental glioblastoma multiforme (GBM). The CPT nanoprodrug was prepared using spontaneous nanoemulsification of a biodegradable, antioxidant CPT prodrug and α-tocopherol. The oxidized nanoprodrug was activated more efficiently than nonoxidized nanoprodrug, suggesting enhanced therapeutic efficacy in the oxidative tumor microenvironment. The in vitro imaging of U-87 MG glioma cells revealed an efficient intracellular uptake of the nanoprodrug via direct cell membrane penetration rather than via endocytosis. The in vivo study in mice demonstrated that the CPT nanoprodrug passed through the BBB and specifically accumulated in brain tumor tissue, but not in healthy brain tissue and other organs. The accumulation preferably occurred at the periphery of the tumor where cancer cells are most actively proliferating, suggesting optimal therapeutic efficacy of the nanoprodrug. The nanoprodrug was effective in treating subcutaneous and intracranial tumors. The nanoprodrug inhibited subcutaneous tumor growth more than 80% compared with control. The median survival time of mice implanted with an intracranial tumor increased from 40.5 days for control to 72.5 days for CPT nanoprodrug. This nanoprodrug approach is a versatile method for developing therapeutic nanoparticles enabling tumor-specific targeting and treatment. The nontoxic, tumor-specific targeting properties of the nanoprodrug system make it a safe, low cost, and versatile nanocarrier for pharmaceuticals, imaging agents, and diagnostic agents.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Camptotecina/administração & dosagem , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Nanocápsulas/administração & dosagem , Pró-Fármacos/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/administração & dosagem , Neoplasias Encefálicas/patologia , Camptotecina/química , Linhagem Celular Tumoral , Glioblastoma/patologia , Camundongos , Nanocápsulas/química , Nanocápsulas/ultraestrutura , Resultado do Tratamento , alfa-Tocoferol/administração & dosagem
12.
J Drug Deliv ; 2011: 980720, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21603162

RESUMO

We demonstrated that hydrophobic derivatives of the nonsteroidal anti-inflammatory drug (NSAID)flufenamic acid (FA), can be formed into stable nanometer-sized prodrugs (nanoprodrugs) that inhibit the growth of glioma cells, suggesting their potential application as anticancer agent. We synthesized highly hydrophobic monomeric and dimeric prodrugs of FA via esterification and prepared nanoprodrugs using spontaneous emulsification mechanism. The nanoprodrugs were in the size range of 120 to 140 nm and physicochemically stable upon long-term storage as aqueous suspension, which is attributed to the strong hydrophobic interaction between prodrug molecules. Importantly, despite the highly hydrophobic nature and water insolubility, nanoprodrugs could be readily activated into the parent drug by porcine liver esterase, presenting a potential new strategy for novel NSAID prodrug design. The nanoprodrug inhibited the growth of U87-MG glioma cells with IC(50) of 20 µM, whereas FA showed IC(50) of 100 µM, suggesting that more efficient drug delivery was achieved with nanoprodrugs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA