Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Acc Chem Res ; 57(12): 1722-1735, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38819691

RESUMO

ConspectusIn human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient's own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering.Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies.We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).


Assuntos
Engenharia Celular , Humanos , Engenharia Celular/métodos , Receptores de Antígenos Quiméricos/metabolismo , Nanotecnologia/métodos , Linfócitos T/citologia , Linfócitos T/metabolismo , Eletroporação/métodos , Injeções
2.
ACS Biomater Sci Eng ; 10(5): 2894-2910, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38556768

RESUMO

Over the past decades, evidence has consistently shown that treatment of central nervous system (CNS)-related disorders, including Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis, and brain cancer, is limited due to the presence of the blood-brain barrier (BBB). To assist with the development of new therapeutics, it is crucial to engineer a drug delivery system that can cross the BBB efficiently and reach target cells within the brain. In this study, we present a potentially efficient strategy for targeted brain delivery through utilization of folic acid (FA)-conjugated brush polymers, that specifically target the reduced folate carrier (RFC, SLC19A1) expressed on brain endothelial cells. Here, azide (N3)-decorated brush polymers were prepared in a straightforward manner coupling a heterotelechelic α-NH2, ω-N3-poly(2-ethyl-2-oxazoline) (NH2-PEtOx-N3) to N-acylated poly(amino ester) (NPAE)-based brushes. Strain-promoted azide-alkyne cycloaddition (SPAAC) 'click chemistry' with DBCO-folic acid (FA) yielded FA-brush polymers. Interestingly, while azide functionalization of the brush polymers dramatically reduced their association to brain microvascular endothelial cells (hCMEC/D3), the introduction of FA to azide led to a substantial accumulation of the brush polymers in hCMEC/D3 cells. The ability of the polymeric brush polymers to traverse the BBB was quantitatively assessed using different in vitro BBB models including static Transwell and microfluidic platforms. FA-brush polymers showed efficient transport across hCMEC/D3 cells in a manner dependent on FA composition, whereas nonfunctionalized brush polymers exhibited limited trafficking under the same conditions. Further, cellular uptake inhibition studies suggested that the interaction and transport pathway of FA-brush polymers across BBB relies on the RFC-mediated pathways. The potential application of the developed FA-brush polymers in brain cancer delivery was also investigated in a microfluidic model of BBB-glioblastoma. Brush polymers with more FA units successfully presented an enhanced accumulation into U-87 MG glioma cells following its BBB crossing, compared to controls. These results demonstrate that FA-modified brush polymers hold a great potential for more efficient delivery of future brain therapeutics.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Ácido Fólico , Polímeros , Ácido Fólico/química , Ácido Fólico/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Humanos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Polímeros/química , Sistemas de Liberação de Medicamentos/métodos , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Portadores de Fármacos/química
3.
ACS Appl Mater Interfaces ; 16(10): 12161-12174, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38416873

RESUMO

Glioblastoma multiforme (GBM) is an aggressive brain cancer with high malignancy and resistance to conventional treatments, resulting in a bleak prognosis. Nanoparticles offer a way to cross the blood-brain barrier (BBB) and deliver precise therapies to tumor sites with reduced side effects. In this study, we developed angiopep-2 (Ang2)-functionalized lipid cubosomes loaded with cisplatin (CDDP) and temozolomide (TMZ) for crossing the BBB and providing targeted glioblastoma therapy. Developed lipid cubosomes showed a particle size of around 300 nm and possessed an internal ordered inverse primitive cubic phase, a high conjugation efficiency of Ang2 to the particle surface, and an encapsulation efficiency of more than 70% of CDDP and TMZ. In vitro models, including BBB hCMEC/D3 cell tight monolayer, 3D BBB cell spheroid, and microfluidic BBB/GBM-on-a-chip models with cocultured BBB and glioblastoma cells, were employed to study the efficiency of the developed cubosomes to cross the BBB and showed that Ang2-functionalized cubosomes can penetrate the BBB more effectively. Furthermore, Ang2-functionalized cubosomes showed significantly higher uptake by U87 glioblastoma cells, with a 3-fold increase observed in the BBB/GBM-on-a-chip model as compared to that of the bare cubosomes. Additionally, the in vivo biodistribution showed that Ang2 modification could significantly enhance the brain accumulation of cubosomes in comparison to that of non-functionalized particles. Moreover, CDDP-loaded Ang2-functionalized cubosomes presented an enhanced toxic effect on U87 spheroids. These findings suggest that the developed Ang2-cubosomes are prospective for improved BBB crossing and enhanced delivery of therapeutics to glioblastoma and are worth pursuing further as a potential application of nanomedicine for GBM treatment.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Peptídeos , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Barreira Hematoencefálica/patologia , Distribuição Tecidual , Estudos Prospectivos , Linhagem Celular Tumoral , Temozolomida , Neoplasias Encefálicas/patologia , Nanopartículas/uso terapêutico , Lipídeos/uso terapêutico
4.
Adv Healthc Mater ; 13(12): e2302902, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38199238

RESUMO

Brain cancers, especially glioblastoma multiforme, are associated with poor prognosis due to the limited efficacy of current therapies. Nanomedicine has emerged as a versatile technology to treat various diseases, including cancers, and has played an indispensable role in combatting the COVID-19 pandemic as evidenced by the role that lipid nanocarrier-based vaccines have played. The tunability of nanocarrier physicochemical properties -including size, shape, surface chemistry, and drug release kinetics- has resulted in the development of a wide range of nanocarriers for brain cancer treatment. These nanocarriers can improve the pharmacokinetics of drugs, increase blood-brain barrier transfer efficiency, and specifically target brain cancer cells. These unique features would potentially allow for more efficient treatment of brain cancer with fewer side effects and better therapeutic outcomes. This review provides an overview of brain cancers, current therapeutic options, and challenges to efficient brain cancer treatment. The latest advances in nanomedicine strategies are investigated with an emphasis on targeted and stimulus-responsive nanocarriers and their potential for clinical translation.


Assuntos
Neoplasias Encefálicas , Portadores de Fármacos , Nanopartículas , Humanos , Neoplasias Encefálicas/tratamento farmacológico , Portadores de Fármacos/química , Nanopartículas/química , Nanopartículas/uso terapêutico , Nanomedicina/métodos , Barreira Hematoencefálica/metabolismo , COVID-19 , Animais , Sistemas de Liberação de Medicamentos/métodos , SARS-CoV-2 , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia
5.
Biosensors (Basel) ; 13(8)2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37622865

RESUMO

Microfluidic technology is applied across various research areas including organ-on-chip (OOC) systems. The main material used for microfluidics is polydimethylsiloxane (PDMS), a silicone elastomer material that is biocompatible, transparent, and easy to use for OOC systems with well-defined microstructures. However, PDMS-based OOC systems can absorb hydrophobic and small molecules, making it difficult and erroneous to make quantitative analytical assessments for such compounds. In this paper, we explore the use of a synthetic fluoropolymer, poly(4,5-difluoro-2,2-bis(trifluoromethyl)-1,3-dioxole-co-tetrafluoroethylene) (Teflon™ AF 2400), with excellent "non-stick" properties to functionalize OOC systems. Cannabinoids, including cannabidiol (CBD), are classes of hydrophobic compounds with a great potential for the treatment of anxiety, depression, pain, and cancer. By using CBD as a testing compound, we examined and systematically quantified CBD absorption into PDMS by means of an LC-MS/MS analysis. In comparison to the unmodified PDMS microchannels, an increase of approximately 30× in the CBD signal was detected with the fluoropolymer surface modification after 3 h of static incubation. Under perfusion conditions, we observed an increase of nearly 15× in the CBD signals from the surface-modified microchannels than from the unmodified microchannels. Furthermore, we also demonstrated that fluoropolymer-modified microchannels are compatible for culturing hCMEC/D3 endothelial cells and for CBD perfusion experiments.


Assuntos
Canabidiol , Canabinoides , Polímeros de Fluorcarboneto , Cromatografia Líquida , Células Endoteliais , Espectrometria de Massas em Tandem
6.
Adv Mater ; 35(44): e2304122, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37434421

RESUMO

Chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising cell-based immunotherapy approach for treating blood disorders and cancers, but genetically engineering CAR-T cells is challenging due to primary T cells' sensitivity to conventional gene delivery approaches. The current viral-based method can typically involve significant operating costs and biosafety hurdles, while bulk electroporation (BEP) can lead to poor cell viability and functionality. Here, a non-viral electroactive nanoinjection (ENI) platform is developed to efficiently negotiate the plasma membrane of primary human T cells via vertically configured electroactive nanotubes, enabling efficient delivery (68.7%) and expression (43.3%) of CAR genes in the T cells, with minimal cellular perturbation (>90% cell viability). Compared to conventional BEP, the ENI platform achieves an almost threefold higher CAR transfection efficiency, indicated by the significantly higher reporter GFP expression (43.3% compared to 16.3%). By co-culturing with target lymphoma Raji cells, the ENI-transfected CAR-T cells' ability to effectively suppress lymphoma cell growth (86.9% cytotoxicity) is proved. Taken together, the results demonstrate the platform's remarkable capacity to generate functional and effective anti-lymphoma CAR-T cells. Given the growing potential of cell-based immunotherapies, such a platform holds great promise for ex vivo cell engineering, especially in CAR-T cell therapy.


Assuntos
Linfoma , Receptores de Antígenos de Linfócitos T , Humanos , Linfócitos T , Transfecção , Eletroporação , Linfoma/metabolismo
7.
Angew Chem Int Ed Engl ; 62(39): e202306100, 2023 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-37278399

RESUMO

Drug delivery systems (DDSs) are designed to deliver therapeutic agents to specific target sites while minimizing systemic toxicity. Recent developments in drug-loaded DDSs have demonstrated promising characteristics and paved new pathways for cancer treatment. Light, a prevalent external stimulus, is widely utilized to trigger drug release. However, conventional light sources primarily concentrate on the ultraviolet (UV) and visible light regions, which suffer from limited biological tissue penetration. This limitation hinders applications for deep-tissue tumor drug release. Given their deep tissue penetration and well-established application technology, X-rays have recently received attention for the pursuit of controlled drug release. With precise spatiotemporal and dosage controllability, X-rays stand as an ideal stimulus for achieving controlled drug release in deep-tissue cancer therapy. This article explores the recent advancements in using X-rays for stimulus-triggered drug release in DDSs and delves into their action mechanisms.


Assuntos
Nanopartículas , Neoplasias , Humanos , Raios X , Liberação Controlada de Fármacos , Sistemas de Liberação de Medicamentos , Luz , Preparações Farmacêuticas , Neoplasias/tratamento farmacológico
8.
ACS Appl Bio Mater ; 6(9): 3532-3554, 2023 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-37294445

RESUMO

Recent preclinical and clinical studies have focused on the active area of therapeutic peptides due to their high potency, selectivity, and specificity in treating a broad range of diseases. However, therapeutic peptides suffer from multiple disadvantages, such as limited oral bioavailability, short half-life, rapid clearance from the body, and susceptibility to physiological conditions (e.g., acidic pH and enzymolysis). Therefore, high peptide dosages and dose frequencies are required for effective patient treatment. Recent innovations in pharmaceutical formulations have substantially improved therapeutic peptide administration by providing the following advantages: long-acting delivery, precise dose administration, retention of biological activity, and improvement of patient compliance. This review discusses therapeutic peptides and challenges in their delivery and explores recent peptide delivery formulations, including micro/nanoparticles (based on lipids, polymers, porous silicon, silica, and stimuli-responsive materials), (stimuli-responsive) hydrogels, particle/hydrogel composites, and (natural or synthetic) scaffolds. This review further covers the applications of these formulations for prolonged delivery and sustained release of therapeutic peptides and their impact on peptide bioactivity, loading efficiency, and (in vitro/in vivo) release parameters.


Assuntos
Hidrogéis , Peptídeos , Humanos , Polímeros
9.
Pharmaceutics ; 15(5)2023 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-37242631

RESUMO

Despite the clinical benefits that chemotherapeutics has had on the treatment of breast cancer, drug resistance remains one of the main obstacles to curative cancer therapy. Nanomedicines allow therapeutics to be more targeted and effective, resulting in enhanced treatment success, reduced side effects, and the possibility of minimising drug resistance by the co-delivery of therapeutic agents. Porous silicon nanoparticles (pSiNPs) have been established as efficient vectors for drug delivery. Their high surface area makes them an ideal carrier for the administration of multiple therapeutics, providing the means to apply multiple attacks to the tumour. Moreover, immobilising targeting ligands on the pSiNP surface helps direct them selectively to cancer cells, thereby reducing harm to normal tissues. Here, we engineered breast cancer-targeted pSiNPs co-loaded with an anticancer drug and gold nanoclusters (AuNCs). AuNCs have the capacity to induce hyperthermia when exposed to a radiofrequency field. Using monolayer and 3D cell cultures, we demonstrate that the cell-killing efficacy of combined hyperthermia and chemotherapy via targeted pSiNPs is 1.5-fold higher than applying monotherapy and 3.5-fold higher compared to using a nontargeted system with combined therapeutics. The results not only demonstrate targeted pSiNPs as a successful nanocarrier for combination therapy but also confirm it as a versatile platform with the potential to be used for personalised medicine.

10.
Bioeng Transl Med ; 8(1): e10325, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36684100

RESUMO

Green chemistry has been a growing multidisciplinary field in recent years showing great promise in biomedical applications, especially for cancer therapy. Chitosan (CS) is an abundant biopolymer derived from chitin and is present in insects and fungi. This polysaccharide has favorable characteristics, including biocompatibility, biodegradability, and ease of modification by enzymes and chemicals. CS-based nanoparticles (CS-NPs) have shown potential in the treatment of cancer and other diseases, affording targeted delivery and overcoming drug resistance. The current review emphasizes on the application of CS-NPs for the delivery of a chemotherapeutic agent, doxorubicin (DOX), in cancer therapy as they promote internalization of DOX in cancer cells and prevent the activity of P-glycoprotein (P-gp) to reverse drug resistance. These nanoarchitectures can provide co-delivery of DOX with antitumor agents such as curcumin and cisplatin to induce synergistic cancer therapy. Furthermore, co-loading of DOX with siRNA, shRNA, and miRNA can suppress tumor progression and provide chemosensitivity. Various nanostructures, including lipid-, carbon-, polymeric- and metal-based nanoparticles, are modifiable with CS for DOX delivery, while functionalization of CS-NPs with ligands such as hyaluronic acid promotes selectivity toward tumor cells and prevents DOX resistance. The CS-NPs demonstrate high encapsulation efficiency and due to protonation of amine groups of CS, pH-sensitive release of DOX can occur. Furthermore, redox- and light-responsive CS-NPs have been prepared for DOX delivery in cancer treatment. Leveraging these characteristics and in view of the biocompatibility of CS-NPs, we expect to soon see significant progress towards clinical translation.

11.
J Drug Target ; 31(3): 310-319, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36440540

RESUMO

BACKGROUND: The drug delivery for treatment of glioblastoma multiforme (GBM) has been unsatisfactory mainly due to the drug resistance and low targeting efficiency. The selective targeting of GBM cells and using a cocktail of therapeutic agents to synergistically induce apoptosis may help enhance the drug delivery. METHODS: In this study, mesenchymal stem cells (MSCs) were engineered to produce exosomes, i.e. nanosized natural vesicles presenting anti-EGFRvIII (ab139) antibody on their surface while encapsulating two apoptosis-inducing gene therapy agents, i.e. cytosine deaminase (CDA) and miR-34a. Exosomes were characterised for their size, morphology, protein content and markers using dynamic light scattering and nanoparticle tracking analysis, cryo-TEM, Western blotting, respectively. miR-34a overexpression and Lamp2-ab139 protein expression were analysed using real-time PCR and flow cytometry, respectively. The armed exosomes were delivered to EGFRvIII positive GBM cells (U87EGFRvIII) as well as wild type cell line (U87), which was EGFRvIII negative. Apoptosis was quantified using flow cytometry in both EGFRvIII negative and positive U87 cells, receiving one gene therapy agent (either CDA or miR-34a) or a combination of them (CDAmiR). RESULTS: Spherical shape exosomes with an average diameter of 110 nm and a membrane thickness of 6.5 nm were isolated from MSCs. Lamp2-ab139 was successfully expressed on the surface of transfected cells and their secreted exosomes. Induced apoptosis rates was significantly higher in U87EGFRvIII cells than for U87 cells, indicating selectivity. The cell death rate was 6%, 9% and 12% in U87, 13%, 21% and 40% in U87EGFRvIII cells for CDA, miR-34a and CDAmiR treatment respectively, showing a higher apoptosis rate in the cells receiving both drugs compared to when single therapy was applied. CONCLUSION: The experimental findings clearly show the improved apoptosis rate of GBM cells when treated by engineered exosomes armed with two gene therapy agents and targeted towards EGFRvIII antigen.


Assuntos
Exossomos , Glioblastoma , MicroRNAs , Humanos , Glioblastoma/tratamento farmacológico , Exossomos/metabolismo , Linhagem Celular Tumoral , Apoptose , MicroRNAs/genética , MicroRNAs/metabolismo
12.
Mol Pharm ; 20(1): 545-560, 2023 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-36484477

RESUMO

Clinical treatment of glioblastoma (GBM) remains a major challenge because of the blood-brain barrier, chemotherapeutic resistance, and aggressive tumor metastasis. The development of advanced nanoplatforms that can efficiently deliver drugs and gene therapies across the BBB to the brain tumors is urgently needed. The protein "downregulated in renal cell carcinoma" (DRR) is one of the key drivers of GBM invasion. Here, we engineered porous silicon nanoparticles (pSiNPs) with antisense oligonucleotide (AON) for DRR gene knockdown as a targeted gene and drug delivery platform for GBM treatment. These AON-modified pSiNPs (AON@pSiNPs) were selectively internalized by GBM and human cerebral microvascular endothelial cells (hCMEC/D3) cells expressing Class A scavenger receptors (SR-A). AON was released from AON@pSiNPs, knocked down DRR and inhibited GBM cell migration. Additionally, a penetration study in a microfluidic-based BBB model and a biodistribution study in a glioma mice model showed that AON@pSiNPs could specifically cross the BBB and enter the brain. We further demonstrated that AON@pSiNPs could carry a large payload of the chemotherapy drug temozolomide (TMZ, 1.3 mg of TMZ per mg of NPs) and induce a significant cytotoxicity in GBM cells. On the basis of these results, the nanocarrier and its multifunctional strategy provide a strong potential for clinical treatment of GBM and research for targeted drug and gene delivery.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Animais , Camundongos , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Silício , Porosidade , Células Endoteliais , Distribuição Tecidual , Linhagem Celular Tumoral , Temozolomida/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Resistencia a Medicamentos Antineoplásicos/genética
13.
ACS Appl Mater Interfaces ; 14(49): 54539-54549, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36469497

RESUMO

An approach to differentially modify the internal surface of porous silicon nanoparticles (pSiNPs) with hydrophobic dodecene and the external surface with antifouling poly-N-(2-hydroxypropyl) acrylamide (polyHPAm) as well as a cell-targeting peptide was developed. Specifically, to generate these core-shell pSiNPs, the interior surface of a porous silicon (pSi) film was hydrosilylated with 1-dodecene, followed by ultrasonication to create pSiNPs. The new external surfaces were modified by silanization with a polymerization initiator, and surface-initiated atom transfer radical polymerization was performed to introduce polyHPAm brushes. Afterward, a fraction of the polymer side chain hydroxyl groups was activated to conjugate cRGDfK─a peptide with a high affinity and selectivity for the ανß3 integrin receptor that is overexpressed in prostate and melanoma cancers. Finally, camptothecin, a hydrophobic anti-cancer drug, was successfully loaded into the pores. This drug delivery system showed excellent colloidal stability in a cell culture medium, and the in vitro drug release kinetics could be fine-tuned by the combination of internal and external surface modifications. In vitro studies by confocal microscopy and flow cytometry revealed improved cellular association attributed to cRGDfK. Furthermore, the cell viability results showed that the drug-loaded and peptide-functionalized nanoparticles had enhanced cytotoxicity toward a C4-2B prostate carcinoma cell line in both 2D cell culture and a 3D spheroid model.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Silício/química , Porosidade , Nanopartículas/química , Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Linhagem Celular Tumoral
14.
Lab Chip ; 22(19): 3579-3602, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36004771

RESUMO

The blood-brain barrier (BBB) represents a key challenge in developing brain-penetrating therapeutic molecules. BBB dysfunction is also associated with the onset and progression of various brain diseases. The BBB-on-a-chip (µBBB), an organ-on-chip technology, has emerged as a powerful in vitro platform that closely mimics the human BBB microenvironments. While the µBBB technology has seen wide application in the study of brain cancer, its utility in other brain disease models ("µBBB+") is less appreciated. Based on the advances of the µBBB technology and the evolution of in vitro models for brain diseases over the last decade, we propose the concept of a "µBBB+" system and summarize its major promising applications in pathological studies, personalized medical research, drug development, and multi-organ-on-chip approaches. We believe that such a sophisticated "µBBB+" system is a highly tunable and promising in vitro platform for further advancement of the understanding of brain diseases.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Transporte Biológico , Encéfalo , Neoplasias Encefálicas/patologia , Humanos , Dispositivos Lab-On-A-Chip , Microambiente Tumoral
15.
Adv Biol (Weinh) ; 6(12): e2200152, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35999436

RESUMO

A continuous, sealed endothelial membrane is essential for the blood-brain barrier (BBB) to protect neurons from toxins present in systemic circulation. Endothelial cells are critical sensors of the capillary environment, where factors like fluid shear stress (FSS) and systemic signaling molecules activate intracellular pathways that either promote or disrupt the BBB. The brain vasculature exhibits complex heterogeneity across the bed, which is challenging to recapitulate in BBB microfluidic models with fixed dimensions and rectangular cross-section microchannels. Here, a Cayley-tree pattern, fabricated using lithography-less, fluid shaping technique in a modified Hele-Shaw cell is used to emulate the brain vasculature in a microfluidic chip. This geometry generates an inherent distribution of heterogeneous FSS, due to smooth variations in branch height and width. hCMEC/D3 endothelial cells cultured in the Cayley-tree designed chip generate a 3D monolayer of brain endothelium with branching hierarchy, enabling the study of the effect of heterogeneous FSS on the brain endothelium. The model is employed to study neuroinflammatory conditions by stimulating the brain endothelium with tumor necrosis factor-α under heterogeneous FSS conditions. The model has immense potential for studies involving drug transport across the BBB, which can be misrepresented in fixed dimension models.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Células Endoteliais/metabolismo , Encéfalo , Estresse Mecânico , Microfluídica
16.
Adv Sci (Weinh) ; 9(26): e2201740, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35851766

RESUMO

Central Nervous System (CNS) diseases, such as Alzheimer's diseases (AD), Parkinson's Diseases (PD), brain tumors, Huntington's disease (HD), and stroke, still remain difficult to treat by the conventional molecular drugs. In recent years, various gene therapies have come into the spotlight as versatile therapeutics providing the potential to prevent and treat these diseases. Despite the significant progress that has undoubtedly been achieved in terms of the design and modification of genetic modulators with desired potency and minimized unwanted immune responses, the efficient and safe in vivo delivery of gene therapies still poses major translational challenges. Various non-viral nanomedicines have been recently explored to circumvent this limitation. In this review, an overview of gene therapies for CNS diseases is provided and describes recent advances in the development of nanomedicines, including their unique characteristics, chemical modifications, bioconjugations, and the specific applications that those nanomedicines are harnessed to deliver gene therapies.


Assuntos
Doenças do Sistema Nervoso Central , Nanomedicina , Barreira Hematoencefálica , Doenças do Sistema Nervoso Central/tratamento farmacológico , Doenças do Sistema Nervoso Central/terapia , Sistemas de Liberação de Medicamentos , Terapia Genética , Humanos
17.
Adv Healthc Mater ; 11(12): e2200076, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35306736

RESUMO

Porous silicon nanoparticles (pSiNPs) are widely utilized as drug carriers due to their excellent biocompatibility, large surface area, and versatile surface chemistry. However, the dispersion in pore size and biodegradability of pSiNPs arguably have hindered the application of pSiNPs for controlled drug release. Here, a step-changing solution to this problem is described involving the design, synthesis, and application of three different linker-drug conjugates comprising anticancer drug doxorubicin (DOX) and different stimulus-cleavable linkers (SCLs) including the photocleavable linker (ortho-nitrobenzyl), pH-cleavable linker (hydrazone), and enzyme-cleavable linker (ß-glucuronide). These SCL-DOX conjugates are covalently attached to the surface of pSiNP via copper (I)-catalyzed alkyne-azide cycloaddition (CuAAC, i.e., click reaction) to afford pSiNP-SCL-DOXs. The mass loading of the covalent conjugation approach for pSiNP-SCL-DOX reaches over 250 µg of DOX per mg of pSiNPs, which is notably twice the mass loading achieved by noncovalent loading. Moreover, the covalent conjugation between SCL-DOX and pSiNPs endows the pSiNPs with excellent stability and highly controlled release behavior. When tested in both in vitro and in vivo tumor models, the pSiNP-SCL-DOXs induces excellent tumor growth inhibition.


Assuntos
Nanopartículas , Neoplasias , Doxorrubicina/farmacologia , Portadores de Fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Porosidade , Silício
18.
Anal Chem ; 94(3): 1726-1732, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35014786

RESUMO

It is well known that hydrogen peroxide (H2O2) is a signaling molecule essential for vital physiological reactions in mammalian cells, such as cell survival, intercellular communication, and cancer metabolism. However, to fully understand the function of H2O2, it is critical to monitor its intracellular and/or extracellular concentrations. Current techniques implemented to address this need require large sample volumes, expensive instrumentation, and long sample preparation and analysis times, inapplicable to inline or online monitoring. In this paper, a new integrated microfluidic device capable of overcoming these limitations is demonstrated for the colorimetric detection of extracellular hydrogen peroxide H2O2. The device contains an optical waveguide to determine absorbance changes and micromixers to enable complete mixing of reagents using a passive approach. This novel H2O2-sensing device has allowed the detection of H2O2 in the range of 0.5-60 µM with a detection limit of 167 ± 5.8 nM and a sensitivity of 13.5 ± 0.1 AU/mM. Proof of concept of the device was demonstrated by quantifying H2O2 release from benign prostatic epithelial (BPH-1) cells upon stimulation with phorbol 12-myristate 13-acetate (PMA). Results show that this integrated device can be potentially utilized to continuously monitor cell-released metabolites autonomously without constant human supervision during the process. Furthermore, this can be achieved without interfering with the cell culture conditions, as only a very small volume of conditioned media (less than 0.4 µL), and not the cells, is required.


Assuntos
Colorimetria , Peróxido de Hidrogênio , Animais , Humanos , Peróxido de Hidrogênio/análise , Dispositivos Lab-On-A-Chip , Acetato de Tetradecanoilforbol
19.
Carbohydr Polym ; 272: 118491, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34420747

RESUMO

An important motivation for the use of nanomaterials and nanoarchitectures in cancer therapy emanates from the widespread emergence of drug resistance. Although doxorubicin (DOX) induces cell cycle arrest and DNA damage by suppressing topoisomerase activity, resistance to DOX has severely restricted its anti-cancer potential. Hyaluronic acid (HA) has been extensively utilized for synthesizing nanoparticles as it interacts with CD44 expressed on the surface of cancer cells. Cancer cells can take up HA-modified nanoparticles through receptor-mediated endocytosis. Various types of nanostructures such as carbon nanomaterials, lipid nanoparticles and polymeric nanocarriers have been modified with HA to enhance the delivery of DOX to cancer cells. Hyaluronic acid-based advanced materials provide a platform for the co-delivery of genes and drugs along with DOX to enhance the efficacy of anti-cancer therapy and overcome chemoresistance. In the present review, the potential methods and application of HA-modified nanostructures for DOX delivery in anti-cancer therapy are discussed.


Assuntos
Doxorrubicina , Ácido Hialurônico , Animais , Portadores de Fármacos , Humanos
20.
Biosens Bioelectron ; 192: 113496, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34274623

RESUMO

Electrochemical devices for transdermal monitoring of key biomarkers are the potential next frontier of wearable technologies for point-of-care disease diagnosis, including Cancer in which Cancer is the leading cause of death worldwide with estimated 10 million deaths in 2018 according to the World Health Organization and breast cancer is one of the five most common causes of cancer death with over two million cases recorded in 2018. Early diagnosis and prognosis based on monitoring of breast cancer biomarkers is of high importance. In this work, high-density gold coated silicon microneedle arrays (Au-Si-MNA) were simultaneously used as biomarker extraction platform and electrochemical transducer, enabling the selective immunocapture of epidermal growth factor receptor 2 (ErbB2), a key breast cancer biomarker, and its subsequent quantification. The analytical performance of the device was tested in artificial interstitial fluid exhibiting a linear response over a wide concentration range from 10 to 250 ng/mL, with a detection limit of 4.8 ng/mL below the biomarker levels expected in breast cancer patients. As a proof of concept, the immunosensor demonstrated its ability to successfully extract ErbB2 from a phantom gel mimicking the epidermis and dermis layers, and subsequently quantify it showing a linear range from 50 to 250 ng/mL and a detection limit of 25 ng/mL. The uniqueness of this sensing platform combining direct transdermal biomarker extraction and quantification opens up new avenues towards the development of high performing wearable point-of-care devices.


Assuntos
Técnicas Biossensoriais , Neoplasias da Mama , Nanopartículas Metálicas , Biomarcadores Tumorais , Neoplasias da Mama/diagnóstico , Técnicas Eletroquímicas , Feminino , Ouro , Humanos , Imunoensaio , Limite de Detecção , Silício
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA