Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 18(24): 15790-15801, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38847355

RESUMO

Targeted drug delivery systems based on metal-organic frameworks (MOFs) have progressed tremendously since inception and are now widely applicable in diverse scientific fields. However, translating MOF agents directly to targeted drug delivery systems remains a challenge due to the biomolecular corona phenomenon. Here, we observed that supramolecular conjugation of antibodies to the surface of MOF particles (MOF-808) via electrostatic interactions and coordination bonding can reduce protein adhesion in biological environments and show stealth shields. Once antibodies are stably conjugated to particles, they were neither easily exchanged with nor covered by biomolecule proteins, which is indicative of the stealth effect. Moreover, upon conjugation of the MOF particle with specific targeted antibodies, namely, anti-CD44, human epidermal growth factor receptor 2 (HER2), and epidermal growth factor receptor (EGFR), the resulting hybrid exhibits an augmented targeting efficacy toward cancer cells overexpressing these receptors, such as HeLa, SK-BR-3, and 4T1, as evidenced by flow cytometry. The therapeutic effectiveness of the antibody-conjugated MOF (anti-M808) was further evaluated through in vivo imaging and the assessment of tumor inhibition effects using IR-780-loaded EGFR-M808 in a 4T1 tumor xenograft model employing nude mice. This study therefore provides insight into the use of supramolecular antibody conjugation as a promising method for developing MOF-based drug delivery systems.


Assuntos
Estruturas Metalorgânicas , Camundongos Nus , Estruturas Metalorgânicas/química , Humanos , Animais , Camundongos , Sistemas de Liberação de Medicamentos , Anticorpos/química , Anticorpos/imunologia , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Linhagem Celular Tumoral , Células HeLa , Camundongos Endogâmicos BALB C , Antineoplásicos/química , Antineoplásicos/farmacologia , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Feminino
2.
Nanoscale ; 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38921728

RESUMO

Targeted drug delivery using metal-organic frameworks (MOFs) has shown significant progress. However, the tumor microenvironment (TME) impedes efficient MOF particle transfer into tumor cells. To tackle this issue, we pre-coated nano-sized MOF-808 particles with multifunctional proteins: glutathione S-transferase (GST)-affibody (Afb) and collagenase, aiming to navigate the TME more effectively. The surface of MOF-808 particles is coated with GST-Afb-a fusion protein of GST and human epidermal growth factor receptor 2 (HER2) Afb or epidermal growth factor receptor (EGFR) Afb which has target affinity. We also added collagenase enzymes capable of breaking down collagen in the extracellular matrix (ECM) through supramolecular conjugation, all without chemical modification. By stabilizing these proteins on the surface, GST-Afb mitigate biomolecule absorption, facilitating specific tumor cell targeting. Simultaneously, collagenase degrades the ECM in the TME, enabling deep tissue penetration of MOF particles. Our resulting system, termed collagenase-GST-Afb-MOF-808 (Col-Afb-M808), minimizes undesired interactions between MOF particles and external biological proteins. It not only induces cell death through Afb-mediated cell-specific targeting, but also showcases advanced cellular internalization in 3D multicellular spheroid cancer models, with effective deep tissue penetration. The therapeutic efficacy of Col-Afb-M808 was further assessed via in vivo imaging and evaluation of tumor inhibition following injection of IR-780 loaded Col-Afb-M808 in 4T1tumor-bearing nude mice. This study offers key insights into the regulation of the multifunctional protein-adhesive surface of MOF particles, paving the way for the designing even more effective targeted drug delivery systems with nano-sized MOF particles.

3.
J Colloid Interface Sci ; 649: 1014-1022, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37392681

RESUMO

Targeted delivery along with controlled drug release is considered crucial in development of a drug delivery system (DDS) for efficient cancer treatment. In this paper, we present a strategy to obtain such a DDS by utilizing disulfide-incorporated mesoporous organosilica nanoparticles (MONs), which were engineered to minimize the surface interactions with proteins for better targeting and therapeutic performance. That is, after MONs were loaded with a chemodrug doxorubicin (DOX) through the inner pores, their outer surface was treated for conjugation to the glutathione-S-transferase (GST)-fused cell-specific affibody (Afb) (GST-Afb). These particles exhibited prompt responsivity to the SS bond-dissociating glutathione (GSH), which resulted in considerable degradation of the initial particle morphology and DOX release. As the protein adsorption to the MON surface appeared largely reduced, their targeting ability with GSH-stimulated therapeutic activities was demonstrated in vitro by employing two kinds of the GST-Afb protein, which target human cancer cells with the surface membrane receptor, HER2 or EGFR. Compared with unmodified control particles, the presented results show that our system can significantly enhance cancer-therapeutic outcomes of the loaded drug, offering a promising way of designing a more efficacious DDS.


Assuntos
Nanopartículas , Neoplasias , Humanos , Portadores de Fármacos/uso terapêutico , Proteínas de Membrana/metabolismo , Proteínas de Membrana/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/metabolismo , Doxorrubicina/uso terapêutico , Glutationa/metabolismo , Neoplasias/tratamento farmacológico , Oxirredução , Dióxido de Silício/uso terapêutico , Porosidade , Liberação Controlada de Fármacos
4.
Small ; 19(22): e2300218, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36864579

RESUMO

Metal-organic framework (MOF) nanoparticles have recently emerged as a promising vehicle for drug delivery with high porosity and feasibility. However, employing a MOF-based drug delivery system remains a challenge due to the difficulty in controlling interfaces of particles in a biological environment. In this paper, protein corona-blocked Zr6 -based MOF (PCN-224) nanoparticles are presented for targeted cancer therapy with high efficiency. The unmodified PCN-224 surface is precoated with glutathione transferase (GST)-fused targetable affibody (GST-Afb) proteins via simple mixing conjugations instead of chemical modifications that can induce the impairment of proteins. GST-Afb proteins are shown to stably protect the surface of PCN-224 particles in a specific orientation with GST adsorbed onto the porous surface and the GST-linked Afb posed outward, minimizing the unwanted interfacial interactions of particles with external biological proteins. The Afb-directed cell-specific targeting ability of particles and consequent induction of cell death is demonstrated both in vitro and in vivo by using two kinds of Afb, which targets the surface membrane receptor, human epidermal growth factor receptor 2 (HER2) or epidermal growth factor receptor (EGFR). This study provides insight into the way of regulating the protein-adhesive surface of MOF nanoparticles and designing a more effective MOF-hosted targeted delivery system.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Humanos , Estruturas Metalorgânicas/metabolismo , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Proteínas de Membrana
5.
Biomater Sci ; 10(6): 1448-1455, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35229845

RESUMO

For efficient drug delivery, stable encapsulation of a large amount of anticancer drugs is crucial, not to mention cell-specific delivery. Among many possible nanocarriers, mesoporous silica nanoparticles are versatile frameworks that satisfy those requirements owing to their characteristic internal pores with a large surface area and a tunable surface composition. By using a noncovalent post-modification strategy, MSN-based drug delivery systems with enhanced therapeutic efficiency can be prepared in a simple one-pot process by loading small anticancer drugs in the unmodified mesopores and by subsequently blocking the drug-loaded pores with a stimuli-responsive polymer gatekeeper. For targeted delivery, drug-loaded MSNs can be functionalized with suitable targeting components such as targeting ligands or artificial protein corona. This mini-review highlights the recent research in which MSN-supported nanocarriers are designed, synthesized, and characterized to possess a high drug loading capacity and encapsulation stability along with targeting capability for more efficient cancer treatment.


Assuntos
Nanopartículas , Dióxido de Silício , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Preparações Farmacêuticas , Porosidade
6.
Nat Commun ; 9(1): 4548, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30382085

RESUMO

Targeted drug delivery using nanoparticles can minimize the side effects of conventional pharmaceutical agents and enhance their efficacy. However, translating nanoparticle-based agents into clinical applications still remains a challenge due to the difficulty in regulating interactions on the interfaces between nanoparticles and biological systems. Here, we present a targeting strategy for nanoparticles incorporated with a supramolecularly pre-coated recombinant fusion protein in which HER2-binding affibody combines with glutathione-S-transferase. Once thermodynamically stabilized in preferred orientations on the nanoparticles, the adsorbed fusion proteins as a corona minimize interactions with serum proteins to prevent the clearance of nanoparticles by macrophages, while ensuring systematic targeting functions in vitro and in vivo. This study provides insight into the use of the supramolecularly built protein corona shield as a targeting agent through regulating the interfaces between nanoparticles and biological systems.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas/química , Coroa de Proteína/química , Animais , Antineoplásicos/farmacologia , Proteínas Sanguíneas/química , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Ligação Proteica , Proteômica , Células RAW 264.7
7.
Biomacromolecules ; 19(7): 3030-3039, 2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29883544

RESUMO

Current drug delivery systems are hampered by poor delivery to tumors, in part reflecting poor encapsulation stability of nanocarriers. Although nanocarriers such as polymeric micelles have high colloidal stability and do not aggregate or precipitate in bulk solution, nanocarriers with low encapsulation stability can lose their cargo during circulation in blood due to interactions with blood cells, cellular membranes, serum proteins, and other biomacromolecules. The resulting premature drug release from carriers limits the therapeutic efficacy at target sites. Herein, we report a simple and robust technique to improve encapsulation stability of drug delivery systems. Specifically, we show that installation of disulfide cross-linked noncovalent polymer gatekeepers onto mesoporous silica nanoparticles with a high loading capacity for hydrophobic drugs enhances in vivo therapeutic efficacy by preventing premature release of cargo. Subsequent release of drug cargos was triggered by cleavage of disulfide cross-linking by glutathione, leading to improved antitumor activity of doxoroubicin in mice. These findings provide novel insights into the development of nanocarriers with high encapsulation stability and improved in vivo therapeutic efficacy.


Assuntos
Nanocápsulas/química , Animais , Antineoplásicos/administração & dosagem , Coloides/química , Reagentes de Ligações Cruzadas/química , Doxorrubicina/administração & dosagem , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Nus , Micelas , Nanocápsulas/efeitos adversos , Nanocápsulas/normas , Dióxido de Silício/química
8.
ACS Biomater Sci Eng ; 4(5): 1716-1722, 2018 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-33445328

RESUMO

Systemic administration of mesoporous silica nanoparticles (MSNs) in biomedical applications has recently been questioned because of poor degradability, which is necessary for the successful development of new drug-delivery systems. Herein, we report the development of colloidal-state-degradable MSNs functionalized with versatile polymer-gatekeepers with a cancer-cell-targeted moiety. The polymer MSNs (PMSNs) were designed with disulfide cross-linking enabling safe encapsulation until cargos are delivered to target cancer cells. Selective targeting was achieved by decoration of CD44-receptor-targeting ligands, hyaluronic acid (HA), with HA-PMSNs. The selective cellular uptake mechanism of the fabricated targeted nanocarrier into CD44-overexpressed cancer cells was demonstrated through the clathrin- and macropinocytosis-mediated pathways. Upon internalization into cancer cells, doxorubicin loaded into the HA-PMSNs can be released by degradation of the polymer shells in the reducing intracellular microenvironment that consequentially induces cell death and further degradation of the MSNs. This study offers a simple technique to fabricate a versatile drug carrier with a high drug loading capacity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...