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











Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 36(19): e2309927, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38387609

RESUMO

Cytokines are powerful in cancer immunotherapy, however, their therapeutic potential is limited by the severe systemic toxicity. Here a potent strategy to reduce the toxicity of systemic cytokine therapy by delivering its denatured form using a finely designed nanochaperone, is described. It is demonstrated that even if the denatured protein cargos are occasionally released under normal physiological conditions they are still misfolded, while can effectively refold into native states and release to function in tumor microenvironment. Consequently, the systemic toxicity of cytokines is nearly completely overcome. Moreover, an immunogenic cell death (ICD)-inducing chemotherapeutic is further loaded and delivered to tumor using this nanochaperone to trigger the release of tumor-associated antigens (TAAs) that are subsequently captured in situ by nanochaperone and then reflows into lymph nodes (LNs) to promote antigen cross-presentation. This optimized personalized nanochaperone-vaccine demonstrates unprecedented suppressive effects against large, advanced tumors, and in combination with immune checkpoint blockade (ICB) therapy results in a significant abscopal effect and inhibition of postoperative tumor recurrence and metastasis. Hence, this approach provides a simple and universal delivery strategy to reduce the systemic toxicities of cytokines, as well as provides a robust personalized cancer vaccination platform, which may find wide applications in cancer immunotherapy.


Assuntos
Antígenos de Neoplasias , Imunoterapia , Interleucina-12 , Nanoestruturas , Animais , Humanos , Camundongos , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Vacinas Anticâncer/química , Linhagem Celular Tumoral , Morte Celular Imunogênica/efeitos dos fármacos , Imunoterapia/métodos , Interleucina-12/química , Interleucina-12/metabolismo , Interleucina-12/toxicidade , Nanopartículas/química , Nanoestruturas/química , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Dobramento de Proteína , Microambiente Tumoral/efeitos dos fármacos
2.
Bioact Mater ; 27: 288-302, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37113688

RESUMO

New antimicrobial strategies are urgently needed to meet the challenges posed by the emergence of drug-resistant bacteria and bacterial biofilms. This work reports the facile synthesis of antimicrobial dynamic covalent nano-networks (aDCNs) composing antibiotics bearing multiple primary amines, polyphenols, and a cross-linker acylphenylboronic acid. Mechanistically, the iminoboronate bond drives the formation of aDCNs, facilitates their stability, and renders them highly responsive to stimuli, such as low pH and high H2O2 levels. Besides, the representative A1B1C1 networks, composed of polymyxin B1(A1), 2-formylphenylboronic acid (B1), and quercetin (C1), inhibit biofilm formation of drug-resistant Escherichia coli, eliminate the mature biofilms, alleviate macrophage inflammation, and minimize the side effects of free polymyxins. Excellent bacterial eradication and inflammation amelioration efficiency of A1B1C1 networks are also observed in a peritoneal infection model. The facile synthesis, excellent antimicrobial performance, and biocompatibility of these aDCNs potentiate them as a much-needed alternative in current antimicrobial pipelines.

3.
Small Methods ; 7(5): e2201051, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36228110

RESUMO

Despite unprecedented successes of antibody-based cancer immunotherapy, the serious side effects and rapid clearance following systemic administration remain big challenges to realize its full potential. At the same time, combination immunotherapy using multiple antibodies has shown particularly promising in cancer treatment. It is noticed that the working mechanisms of natural holdase and foldase chaperone are desirable to overcome the limitations of therapeutic antibodies. Holdase chaperone stabilizes unfolded client and prevents it from activation and degradation, while foldase chaperone assists unfolded client to its native state to function. Here a holdase/foldase mimetic nanochaperone (H/F-nChap) to co-delivery two types of monoclonal antibodies (mAbs), αCD16 and αPDL1, and resiquimod (R848) is developed, which significantly improves cancer immunotherapy. The H/F-nChap presents holdase activity in blood and normal tissues that hides and protects mAbs from unnecessary targeted activation and degradation, thereby prolonging blood circulation and reducing immunotoxicity in vivo. Furthermore, H/F-nChap switches to foldase activity in the tumor microenvironment that exposes mAbs and releases R848 to enhance the engagement between NK cells and tumor cells and promote immune activation, respectively. The H/F-nChap represents a strategy for safe and spatiotemporal delivery of multiple mAbs, providing a promising platform for improved cancer immunotherapy.


Assuntos
Anticorpos Monoclonais , Neoplasias , Humanos , Anticorpos Monoclonais/uso terapêutico , Imunoterapia , Neoplasias/terapia
4.
ACS Appl Mater Interfaces ; 14(39): 44211-44221, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36153949

RESUMO

Exendin-4 (Ex-4) is a promising therapeutic peptide for the clinical treatment of type 2 diabetes, but its instability and immunogenicity result in a short circulating half-life and low bioavailability, which severely limit its clinical application. Here, complex micelles with 4-carboxy-3-fluorophenylboronic acid (FPBA)-modified and positively charged hydrophobic domains on the surface were devised as nanochaperones to mediate the delivery of Ex-4. The nanochaperones can bind Ex-4 on the surface via the synergy of electrostatic and hydrophobic interactions, leading to efficient loading and stabilization of Ex-4. More importantly, the immunogenic site of Ex-4 was shielded by the nanochaperones, thereby effectively reducing the immune clearance and prolonging the half-life. Hyperglycemia-triggered release of Ex-4 was achieved by the hydrophobic to the hydrophilic transformation of the FPBA-modified domains and the introduced negative charge because of the binding of glucose by FPBA. The Ex-4-loaded nanochaperones exhibited an enhanced therapeutic effect on type 2 diabetic mice.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Exenatida/farmacologia , Exenatida/uso terapêutico , Glucose/metabolismo , Camundongos , Micelas , Peptídeos/farmacologia , Peptídeos/uso terapêutico
5.
Biomater Sci ; 10(13): 3575-3584, 2022 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-35635244

RESUMO

Protein therapeutics have been viewed as powerful candidates for cancer treatment by virtue of highly specific bioactivity and minimized adverse effects. However, the intracellular delivery of protein drugs remains enormously challenging due to multiple successive biological barriers in vivo. Herein, a bioinspired nanochaperone is developed to assist proteins in vanquishing the sequential physiological barriers in a holistic manner and enhance synergistic tumor therapy. By concurrently mimicking the N-terminal-binding domain and C-terminal-stabilizing domain of natural chaperones, this nanochaperone can efficiently capture the protein by multiple interactions and hide them in the confined spaces on the surface, serving as a shield to resist enzymatic degradation and avoid immune clearance during blood circulation. Upon reaching the tumor site, the nanochaperone rapidly responds to the acidic tumor microenvironment and turns into partial protonation, acting as a spear to facilitate tumor cellular internalization. More importantly, further protonation of nanochaperone in the lysosome of tumor cells enables it to blast the lysosome and achieve cytosolic protein delivery with reserved bioactivities. Furthermore, this nanochaperone-based protein transduction strategy is demonstrated to combine with small-molecule drugs to synergistically amplify the anti-tumor therapeutic effect in vitro and in vivo, providing a potential platform for the exploitation of diverse combinations of anti-tumor therapies.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/química , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Microambiente Tumoral
6.
ACS Macro Lett ; 10(6): 662-670, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35549098

RESUMO

Human islet amyloid polypeptide (hIAPP) aggregation is closely associated with dysfunction and apoptosis of pancreatic ß-cells in type 2 diabetes (T2D). Accordingly, hIAPP amyloid inhibitors have shown promise against T2D. Here, by mimicking the function of natural molecular chaperones, nanochaperones (nChaps) based on self-assembled polymeric micelles with tunable surface microdomains for T2D treatment are reported. By capturing the aggregation-prone species of hIAPP onto the hydrophobic microdomains and segregating them by hydrophilic PEG chains, this kind of nChaps could effectively prevent hIAPP aggregation, block cell adhesion of hIAPP, facilitate hIAPP aggregates degradation and reduce hIAPP-related cytotoxicity. Therefore, our work will provide useful insights to develop a biomimetic strategy for the treatment of T2D.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Apoptose , Diabetes Mellitus Tipo 2/tratamento farmacológico , Humanos , Interações Hidrofóbicas e Hidrofílicas , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química
7.
Nano Lett ; 20(6): 4454-4463, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32401534

RESUMO

Inspired by heat shock proteins (HSPs), a self-assembly nanochaperone (nChap) is developed as a novel nanovaccine for boosting antitumor immune responses. Taking advantage of HSP-like microdomains and surface-decorated mannose, this nChap can efficiently capture antigens and ferry them into the dendritic cells (DCs). Subsequently, the nChap can blast lysosomes by transforming the structure and property of surface microdomains, thereby promoting antigen escape and enhancing their cross-presentation in cytoplasm. As a result, the nChap-based nanovaccine can elicit both CD4+ and CD8+ T cell-based immune responses and shows an excellent preventive effect on melanoma. Further combination of the nanovaccine with antiprogrammed death-1 (anti-PD-1) checkpoint blockade offers effective inhibition on the growth of already-established melanoma. Therefore, this nC ap-based nanovaccine provides a simple and robust strategy in mimicking HSPs to realize structure-assisted antigen capture, surface-receptor-mediated DC internalization, and both activation of humoral immunity and cellular immunity, promising for efficient cancer immunotherapy.


Assuntos
Vacinas Anticâncer , Proteínas de Choque Térmico , Imunoterapia , Melanoma , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Humanos , Imunidade , Melanoma/tratamento farmacológico
8.
Front Chem ; 7: 861, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31970146

RESUMO

Cascade reactions integrate two or more reactions, of which each subsequent reaction can only start when the previous reaction step is completed. Employing natural substrates in the human body such as glucose and oxygen, cascade reactions can generate reactive oxygen species (ROS) to kill tumor cells, but cascade reactions may also have potential as a direly needed, novel bacterial infection-control strategy. ROS can disintegrate the EPS matrix of infectious biofilm, disrupt bacterial cell membranes, and damage intra-cellular DNA. Application of cascade reactions producing ROS as a new infection-control strategy is still in its infancy. The main advantages for infection-control cascade reactions include the fact that they are non-antibiotic based and induction of ROS resistance is unlikely. However, the amount of ROS generated is generally low and antimicrobial efficacies reported are still far <3-4 log units necessary for clinical efficacy. Increasing the amounts of ROS generated by adding more substrate bears the risk of collateral damage to tissue surrounding an infection site. Collateral tissue damage upon increasing substrate concentrations may be prevented by locally increasing substrate concentrations, for instance, using smart nanocarriers. Smart, pH-responsive nanocarriers can self-target and accumulate in infectious biofilms from the blood circulation to confine ROS production inside the biofilm to yield long-term presence of ROS, despite the short lifetime (nanoseconds) of individual ROS molecules. Increasing bacterial killing efficacies using cascade reaction components containing nanocarriers constitutes a first, major challenge in the development of infection-control cascade reactions. Nevertheless, their use in combination with clinical antibiotic treatment may already yield synergistic effects, but this remains to be established for cascade reactions. Furthermore, specific patient groups possessing elevated levels of endogenous substrate (for instance, diabetic or cancer patients) may benefit from the use of cascade reaction components containing nanocarriers.

9.
Langmuir ; 34(40): 12116-12125, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30212220

RESUMO

Insulin plays a significant role in diabetes treatment. Although a huge number of insulin-loaded, glucose-responsive nanocarriers have been developed in past decades, most of them showed a lower loading capacity and efficiency due to the weak interaction between insulin and nanocarriers. In this work, a novel insulin-encapsulated glucose-responsive polymeric complex micelle (CM) is devised, showing (i) enhanced insulin-loading efficiency owing to the zinc ions' chelation by nitrilotriacetic acid (NTA) groups of NTA-functioned glycopolymer and the histidine imidazole of insulin, (ii) the glucose-triggered pulse release of insulin, and (iii) long stability under physiological conditions. This CM was fabricated by the self-assembly of block copolymer PEG- b-P(Asp- co-AspPBA) and glycopolymer P(Asp- co-AspGA- co-AspNTA), resulting in complex micelles with a PEG shell and a cross-linked core composed of phenylboronic acid (PBA)/glucose complexations. Notably, the modified nitrilotriacetic acid (NTA) groups of CM could specifically bind insulin via chelated zinc ions, thus enhancing the loading efficacy of insulin compared to that of nonmodified CM. The dynamic PBA/glucose complexation core of CM dissociates under the trigger of high glucose concentration (>2 g/L) while being quite stable in low glucose concentrations (<2 g/L), as demonstrated by the pulse release of insulin in vitro. Finally, in a murine model of type 1 diabetes, NTA-modified complex micelles loading an insulin (NTA-CM-INS) group exhibited a long hypoglycemic effect which is superior to that of free insulin in the PBS (PBS-INS) group and insulin-loaded complex micelles without an NTA modification (CM-INS) group. This long-term effect benefited from Zn(II) chelation by NTA-modified complex micelles and could avoid hypoglycemia caused by the burst release of insulin. Taken together, this constitutes a highly effective way to encapsulate insulin and release insulin via an on-demand manner for blood glucose control in diabetes.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Portadores de Fármacos/química , Glucose/química , Insulina/uso terapêutico , Micelas , Ácido Nitrilotriacético/química , Animais , Ácidos Borônicos/síntese química , Ácidos Borônicos/química , Ácidos Borônicos/toxicidade , Quelantes/síntese química , Quelantes/química , Quelantes/toxicidade , Portadores de Fármacos/síntese química , Portadores de Fármacos/toxicidade , Liberação Controlada de Fármacos , Camundongos , Células NIH 3T3 , Ácido Nitrilotriacético/síntese química , Ácido Nitrilotriacético/toxicidade , Peptídeos/síntese química , Peptídeos/química , Peptídeos/toxicidade , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polietilenoglicóis/toxicidade , Zinco/química
10.
ACS Biomater Sci Eng ; 4(6): 2007-2017, 2018 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-33445271

RESUMO

Protein drugs play a significant role in the treatment of many diseases such as diabetes, cancers, and immune system diseases. Though polymeric nanocarriers have been designed to deliver protein drugs for prolonging circulation lifetime and providing stimuli-triggered release, problems are still often encountered including lower loading efficiency and capacity as well as poor circulation stability because of the weak interaction between protein drugs and nanocarriers. Herein, we described a new kind of bifunctional polymeric nanogels for efficient loading and glucose-triggered release of insulin. Biodegradable poly(N-isopropylacrylamide) (PNIPAM)-based nanogels was synthesized with nitrilotriacetic acid (NTA) and phenylboronic acid (PBA) as functional groups and ethylene glycol dimethacrylate (EGDMA) as cross-linker. The NTA groups could specifically bind imidazole-containing protein drugs such as insulin via chelated zinc ions, leading an efficient loading of insulin. The structure, morphology, and drug-loading properties of the nanogels were well-characterized, and glucose-triggered insulin release was achieved based on the glucose-responsiveness of PBA groups. MTT assay and enzymatic degradation revealed good biocompatibility and biodegradability for the nanogels. This kind of bifunctional nanogels would be promising candidates for glucose-responsive delivery of insulin in the future.

11.
Theranostics ; 6(9): 1277-92, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27375779

RESUMO

Chemotherapy for cancer treatment has been demonstrated to cause some side effects on healthy tissues and multidrug resistance of the tumor cells, which greatly limits therapeutic efficacy. To address these limitations and achieve better therapeutic efficacy, combination therapy based on nanoparticle platforms provides a promising approach through delivering different agents simultaneously to the same destination with synergistic effect. In this study, a novel green tea catechin-based polyion complex (PIC) micelle loaded with doxorubicin (DOX) and (-)-Epigallocatechin-3-O-gallate (EGCG) was constructed through electrostatic interaction and phenylboronic acid-catechol interaction between poly(ethylene glycol)-block-poly(lysine-co-lysine-phenylboronic acid) (PEG-PLys/PBA) and EGCG. DOX was co-loaded in the PIC micelles through π-π stacking interaction with EGCG. The phenylboronic acid-catechol interaction endowed the PIC micelles with high stability under physiological condition. Moreover, acid cleavability of phenylboronic acid-catechol interaction in the micelle core has significant benefits for delivering EGCG and DOX to same destination with synergistic effects. In addition, benefiting from the oxygen free radicals scavenging activity of EGCG, combination therapy with EGCG and DOX in the micelle core could protect the cardiomyocytes from DOX-mediated cardiotoxicity according to the histopathologic analysis of hearts. Attributed to modulation of EGCG on P-glycoprotein (P-gp) activity, this kind of PIC micelles could effectively reverse multidrug resistance of cancer cells. These results suggested that EGCG based PIC micelles could effectively overcome DOX induced cardiotoxicity and multidrug resistance.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Catequina/administração & dosagem , Doxorrubicina/farmacologia , Portadores de Fármacos/administração & dosagem , Micelas , Nanoestruturas/administração & dosagem , Chá/química , Antibióticos Antineoplásicos/administração & dosagem , Cardiotoxicidade/prevenção & controle , Catequina/análogos & derivados , Catequina/isolamento & purificação , Catequina/farmacologia , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Portadores de Fármacos/química , Resistência a Múltiplos Medicamentos , Humanos , Nanoestruturas/química
13.
Biomacromolecules ; 15(10): 3634-42, 2014 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25308336

RESUMO

Exploring ideal nanocarriers for drug delivery systems has encountered unavoidable hurdles, especially the conflict between enhanced cellular uptake and prolonged blood circulation, which have determined the final efficacy of cancer therapy. Here, based on controlled self-assembly, surface structure variation in response to external environment was constructed toward overcoming the conflict. A novel micelle with mixed shell of hydrophilic poly(ethylene glycol) PEG and pH responsive hydrophobic poly(ß-amino ester) (PAE) was designed through the self-assembly of diblock amphiphilic copolymers. To avoid the accelerated clearance from blood circulation caused by the surface exposed targeting group c(RGDfK), here c(RGDfK) was conjugated to the hydrophobic PAE and hidden in the shell of PEG at pH 7.4. At tumor pH, charge conversion occurred, and c(RGDfK) stretched out of the shell, leading to facilitated cellular internalization according to the HepG2 cell uptake experiments. Meanwhile, the heterogeneous surface structure endowed the micelle with prolonged blood circulation. With the self-regulated multifunctional collaborated properties of enhanced cellular uptake and prolonged blood circulation, successful inhibition of tumor growth was achieved from the demonstration in a tumor-bearing mice model. This novel nanocarrier could be a promising candidate in future clinical experiments.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Feminino , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Micelas , Polietilenoglicóis/química , Polímeros/química
14.
Langmuir ; 30(16): 4797-805, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24697573

RESUMO

Complexation between 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrin (TPPS) and poly(ethylene glycol)-block-poly(L-lysine) (PEG-b-PLL) was performed via electrostatic interaction. Two kinds of primary arrays of TPPS with different supramolecular chirality induced by PLL were obtained in the resultant complex by inverting the mixing procedure of the two components. These arrays could be displaced by poly(sodium-p-styrenesulfonate) (PSS) from the chiral PLL template through competitive electrostatic complexation, and then PSS formed a polyion complex micelle with PEG-b-PLL. The template-removed TPPS arrays preserved their induced chirality and served as primary subunits for the secondary aggregation of TPPS. The morphology of the secondary aggregates was strongly dependent upon the asymmetric primary supramolecular arrangement of TPPS. The rodlike nanostructure that was ∼200 nm in length was composed of the primary arrays that showed opposite exciton chirality between the J- and H-bands. In contrast, the micrometer-sized fibrils observed were composed of the arrays with the same exciton chirality at the J- and H-bands.


Assuntos
Polietilenoglicóis/química , Porfirinas/química , Estereoisomerismo
15.
Biomacromolecules ; 14(10): 3434-43, 2013 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-24063314

RESUMO

Herein, a series of biocompatible, robust, pH/sugar-sensitive, core-cross-linked, polyion complex (PIC) micelles based on phenylboronic acid-catechol interaction were developed for protein intracellular delivery. The rationally designed poly(ethylene glycol)-b-poly(glutamic acid-co-glutamicamidophenylboronic acid) (PEG-b-P(Glu-co-GluPBA)) and poly(ethylene glycol)-b-poly(l-lysine-co-ε-3,4-dihydroxyphenylcarboxyl-L-lysine) (PEG-b-P(Lys-co-LysCA)) copolymers were successfully synthesized and self-assembled under neutral aqueous condition to form uniform micelles. These micelles possessed a distinct core-cross-linked core-shell structure comprised of the PEG outer shell and the PGlu/PLys polyion complex core bearing boronate ester cross-linking bonds. The cross-linked micelles displayed superior physiological stabilities compared with their non-cross-linked counterparts while swelling and disassembling in the presence of excess fructose or at endosomal pH. Notably, either negatively or positively charged proteins can be encapsulated into the micelles efficiently under mild conditions. The in vitro release studies showed that the release of protein cargoes under physiological conditions was minimized, while a burst release occurred in response to excess fructose or endosomal pH. The cytotoxicity of micelles was determined by cck-8 assay in HepG2 cells. The cytochrome C loaded micelles could efficiently delivery proteins into HepG2 cells and exhibited enhanced apoptosis ability. Hence, this type of core-cross-linked PIC micelles has opened a new avenue to intracellular protein delivery.


Assuntos
Antineoplásicos/farmacologia , Reagentes de Ligações Cruzadas/química , Citocromos c/metabolismo , Frutose/farmacologia , Micelas , Polímeros/química , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Ácidos Borônicos/química , Ácidos Borônicos/metabolismo , Catecóis/química , Catecóis/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Íons/química , Íons/farmacologia , Estrutura Molecular , Relação Estrutura-Atividade
16.
Biomacromolecules ; 14(2): 460-7, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23281663

RESUMO

The miserable targeting performance of nanocarriers for cancer therapy arises largely from the rapid clearance from blood circulation and the major accumulation in the organs of the reticuloendothelial system (RES), leading to inefficient enhanced permeability and retention (EPR) effect after intravenous injection (i.v.). Herein, we reported an efficient method to prolong the blood circulation of nanoparticles and decrease their deposition in liver and spleen. In this work, we fabricated a series of mixed shell micelles (MSMs) with approximately the same size, charge and core composition but with varied hydrophilic/hydrophobic ratios in the shell through spontaneously self-assembly of block copolymers poly(ethylene glycol)-block-poly(l-lysine) (PEG-b-PLys) and poly(N-isopropylacrylamide)-block-poly(aspartic acid) (PNIPAM-b-PAsp) in aqueous medium. The effect of the surface heterogeneity on the in vivo biodistribution was systematically investigated through in vivo tracking of the (125)I-labeled MSMs determined by Gamma counter. Compared with single PEGylated micelles, some MSMs were proved to be significantly efficient with more than 3 times lower accumulation in liver and spleen and about 6 times higher concentration in blood at 1 h after i.v.. The results provide us a novel strategy for future development of long-circulating nanocarriers for efficient cancer therapy.


Assuntos
Portadores de Fármacos , Nanopartículas , Polímeros/farmacologia , Distribuição Tecidual/efeitos dos fármacos , Transporte Biológico , Sobrevivência Celular/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Fígado/metabolismo , Micelas , Sistema Fagocitário Mononuclear/metabolismo , Neoplasias/tratamento farmacológico , Baço/metabolismo , Propriedades de Superfície
17.
Macromol Biosci ; 12(2): 251-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22076739

RESUMO

A multifunctional drug delivery system based on MCM-41-type mesoporous silica nanoparticles is described that behaves as if nanogates were covalently attached to the outlets of the mesopores through a highly acid-sensitive benzoic-imine linker. Tumor-specific uptake and intracellular delivery results from the pH-dependent progressive hydrolysis of the benzoic-imine linkage that starts at tumor extracellular pH = 6.8 and increases with decreasing pH. The cleavage of the benzoic-imine bond leads to the removal of the polypseudorotaxane caps and subsequent release of the payload drugs at tumor sites. At the same time, the carrier surface becomes positively charged, which further facilitates cellular uptake of the nanocarriers, thus offering a tremendous potential for targeted tumor therapy.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Portadores de Fármacos/síntese química , Nanopartículas/química , Dióxido de Silício/química , Antibióticos Antineoplásicos/química , Benzaldeídos/química , Benzoatos/química , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Citometria de Fluxo , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Iminas/química , Polietilenoglicóis/química , Porosidade
19.
Langmuir ; 25(21): 12522-8, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19810675

RESUMO

Poly(ethylene glycol)-block-poly(acrylic acid-co-acrylamidophenylboronic acid) [PEG(114)-b-(PAA(63)-co-PAAPBA(107))] was synthesized by the modification of poly(ethylene glycol)-block-poly(acrylic acid) (PEG(114)-b-PAA(170)) with 3-aminophenylboronic acid (APBA). Glucose-responsive PEG(114)-b-(PAA(63)-co-PAAPBA(107)) self-assembled into core-shell micelles with the hydrophobic core composed of PAAPBA and hydrophilic shell composed of PEG in aqueous solution. The swelling and disaggregating behaviors of micelles responding to glucose were investigated by using light scattering in aqueous solution at pH 7.4. Characterization of insulin-loaded micelles and their drug release in solutions with various glucose concentrations were further studied. The results demonstrated that the drug release rate can be controlled by variation of glucose concentration.

20.
Langmuir ; 23(14): 7498-504, 2007 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-17547432

RESUMO

The pyranine-induced micellization of poly(ethylene glycol)-block-poly(4-vinylpyridine) (PEG114-b-P4VP61) in aqueous solutions and pH-triggered release of pyranine from the complex micelles were studied by dynamic and static light scattering, transmission electron microscopy, 1H NMR spectroscopy, and UV-vis spectroscopy. At pH 2, the ionized pyranine can ionically cross-link the protonated P4VP block and result in well-defined spherical complex micelles with a P4VP/pyranine core surrounded by a PEG corona. The ratio of pyranine to pyridyl units can influence the structure and the properties of the resultant complex micelles. The complex micelles are stable upon dilution and heating but are sensitive to pH changes. pH-triggered release of the incorporated pyranine from the complex micelles demonstrates that the release behavior is pH-tunable and displays good controlled-release characteristics at pH approximately 4.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA