Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nano Lett ; 23(18): 8674-8682, 2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37721331

RESUMO

The VEGF-VEGFR2 (VEGF = vascular endothelial growth factor) signaling has been a promising target in cancer therapy. However, because conventional anti-angiogenic therapeutics suffer from drawbacks, particularly severe side effects, novel anti-angiogenic strategies are much needed. Herein, we report the rational engineering of VEGF-targeted molecularly imprinted polymer nanoparticles (nanoMIP) for anti-angiogenic cancer therapy. The anti-VEGF nanomedicine was prepared via a state-of-the-art molecular imprinting approach using the N-terminal epitope of VEGF as the template. The nanoMIP could target the two major pro-angiogenic isoforms (VEGF165 and VEGF121) with high affinity and thereby effectively block the VEGF-VEGFR2 signaling, yielding a potent anti-angiogenic effect of "killing two birds with one stone". In vivo experiments demonstrated that the anti-VEGF nanoMIP effectively suppressed tumor growth via anti-angiogenesis in a xenograft model of human colon carcinoma without apparent side effects. Thus, this study not only proposes an unprecedented anti-angiogenic strategy for cancer therapy but also provides a new paradigm for the rational development of MIPs-based "drug-free" nanomedicines.

2.
ACS Appl Mater Interfaces ; 15(23): 27658-27669, 2023 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-37267068

RESUMO

Reprogramming tumor-associated macrophages (TAMs) has emerged as a promising strategy in cancer immunotherapy. Targeted therapeutics integrating multiple functions to fully leverage the antitumor immune functions of macrophages without affecting systemic or tissue-resident macrophages are crucial for TAM reprogramming. Herein, by integrating molecular imprinting and nanotechnology, we rationally designed and engineered an unprecedented nanocoordinator for targeted remolding of TAMs to fully leverage the antitumor efficacy of macrophages by inducing a cascade effect. The nanocoordinator features a magnetic iron oxide nanoinner core and sialic acid-imprinted shell. Intravenously administered into systemic circulation, the nanocoordinator can rapidly accumulate at the tumor site in response to an external magnet. Then, by specifically binding to sialic acid overexpressed on tumor cells, the nanocoordinator anchors at the tumor site with prolonged retention time. Via binding with the nanocoordinator, tumor cells are tagged with a foreign substance, which promotes the intrinsic phagocytosis of macrophages. Subsequently, the nanocoordinator taken up by macrophages effectively promotes the polarization of macrophages toward the M1 phenotype, thus activating the immunotherapeutic efficacy of macrophages. Synergized by the cascade effect, this nanocoordinator effectively harnesses TAMs for macrophage-mediated immunotherapy. This study offers new TAM-targeted therapeutics that allows us to fully leverage the antitumor immune functions of macrophages without affecting the normal tissue.


Assuntos
Ácido N-Acetilneuramínico , Neoplasias , Humanos , Ácido N-Acetilneuramínico/metabolismo , Macrófagos , Neoplasias/tratamento farmacológico , Fagocitose , Imunoterapia , Microambiente Tumoral
3.
Chem Sci ; 14(10): 2553-2561, 2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36908957

RESUMO

Cis-diol metabolic reprogramming evolves during primary liver cancer (PLC) initiation and progression. However, owing to the low concentrations and highly structural heterogeneity of cis-diols in vivo, severe interference from complex biofluids and limited profiling coverage of existing methods, in-depth profiling of cis-diol metabolites and linking their specific changes with PLC remain challenging. Besides, due to the low specificity of widely used protein biomarkers, accurate classification of PLC from hepatitis still represents an unmet need in clinical diagnostics. Herein, to high-coverage profile cis-diols and explore the translational potential of them as biomarkers, a machine learning-empowered boronate affinity extraction-solvent evaporation assisted enrichment-mass spectrometry (MLE-BESE-MS) was developed. A single analytical platform integrated with multiple complementary functions, including pH-controlled boronate affinity extraction, solvent evaporation-assisted enrichment and nanoelectrospray ionization-based cis-diol identification, was constructed, which significantly improved the metabolite coverage. Meanwhile, by virtue of machine learning (principal components analysis, orthogonal partial least-squares discrimination analysis and random forest), collected cis-diols were statistically screened to extract efficient features for precise PLC diagnosis, and the results outperform the routinely used protein biomarker-based methods both in sensitivity (87.5% vs. less than 70%) and specificity (85.7% vs. ca. 80%). This machine learning-empowered integrated MS platform advanced the targeted metabolic analysis for early cancer diagnosis, rendering great promise for clinical translation.

4.
Angew Chem Int Ed Engl ; 62(17): e202301202, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-36814079

RESUMO

Harnessing innate immunity is an appealing strategy for cancer treatment. Herein, we report a new strategy called molecularly imprinted nanobeacons (MINBs) for redirecting innate immune killing towards triple-negative breast cancer (TNBC). The MINBs were molecularly imprinted nanoparticles with the N-epitope of glycoprotein nonmetastatic B (GPNMB) as the template and grafted with plentiful fluorescein moieties as the hapten. The MINBs could tag the TNBC cells via binding with GPNMB and thereby provide navigation for recruiting hapten-specific antibodies. The gathered antibodies could further trigger effective Fc-domain-mediated immune killing towards the tagged cancer cells. In vivo experiments showed that the TNBC growth was significantly inhibited after MINBs treatment by intravenous injection as compared with control groups. This study not only opens a new access for redirecting innate immunity towards TNBC but also paves the way for innate immunity-based therapy of other diseases.


Assuntos
Nanopartículas , Neoplasias de Mama Triplo Negativas , Humanos , Anticorpos , Linhagem Celular Tumoral , Glicoproteínas de Membrana , Impressão Molecular , Neoplasias de Mama Triplo Negativas/patologia , Imunidade Inata
5.
Chem Sci ; 13(36): 10897-10903, 2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36320712

RESUMO

Blocking the PD-1/PD-L1 immune checkpoint has emerged as a promising strategy in cancer immunotherapy, in which monoclonal antibodies are predominately used as inhibitors. Despite their remarkable success, monoclonal antibody-based therapeutics suffer from drawbacks due to the use of antibodies, such as high cost, low stability and high frequency of immune-related adverse effects. Therefore, novel anti-PD-1/PD-L1 therapeutics that can address these issues are of significant importance. Herein, we report a molecularly imprinted polymer (MIP) based PD-1 nano inhibitor for blocking the PD-1/PD-L1 axis. The anti-PD-1 nanoMIP was rationally designed and engineered by epitope imprinting using the N-terminal epitope of PD-1 as the binding site. The anti-PD-1 nanoMIP showed good specificity and high affinity towards PD-1, yielding a disassociation constant at the 10-8 M level, much better than that between PD-1 and PD-L1. Via steric hindrance, this inhibitor could effectively block PD-1/PD-L1 interaction. Besides, it could effectively reactivate T cells and reverse the chemoresistance of tumor cells. Therefore, this present study not only provides a novel and promising immune checkpoint blockade inhibitor but also boosts further development of MIPs for cancer immunotherapy.

6.
Small ; 18(46): e2201671, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36161701

RESUMO

Exploring new targets and developing novel targeted therapies are urgently needed for neuroblastoma therapy. Polysialic acid (polySia), a linear homopolymer of sialic acid units that correlates well with tumor progression and poor prognosis, has emerged as a potential target for neuroblastoma. However, the lack of polySia-specific binding reagents has severely limited the development of polySia-targeting therapeutics for neuroblastoma. Herein, the construction of polySia-targeting nanomissiles via molecular imprinting for the photothermal therapy of neuroblastoma is reported. Oligosialic acid (oligoSia) containing 3-4 units is considered as a characteristic structure for the recognition of polySia, while oligoSia containing 4-7 units digested from polySia is employed as the template. Via boronate-affinity controllable oriented surface imprinting, oligoSia-imprinted nanoparticles (oSia-MIP) are prepared. The oSia-MIP allows for specifically recognizing polySia and targeting polySia overexpressed neuroblastoma cells in vitro and in vivo. oSia-MIP loaded with indocyanine green is prepared and experimentally demonstrated to be a potent targeted photothermal therapeutic for neuroblastoma. Equipping the core substrate with functional entities, the developed polySia targeting nanoplatform can be accommodated to various therapeutic modalities, holding great promise for neuroblastoma targeted therapy.


Assuntos
Neuroblastoma , Terapia Fototérmica , Humanos , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Neuroblastoma/terapia , Ácido N-Acetilneuramínico
7.
ACS Appl Mater Interfaces ; 14(39): 44098-44110, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36149803

RESUMO

Chemodynamic therapy (CDT) that kills tumor cells by converting low-reactivity H2O2 into highly toxic hydroxyl radicals (•OH) is an emerging tumor therapeutic modality, but its therapeutic efficacy is largely limited by both the lack of tumor targeting and redox homeostasis in tumor cells. Herein, we report Cu2+-encapsulated and GalNAc-imprinted biodegradable silica nanoparticles (nanoMIP) for boosting CDT. In this strategy, the Cu2+ was first encapsulated into disulfide-bridged silica nanoparticles with a high loading capacity of ∼18.3%, followed by in situ functionalization via molecular imprinting using GalNAc as a template. Such a nanovector could specifically target tumor cells overexpressing the Tn antigen to promote the cellular uptake. After internalization into tumor cells, the degradation of nanoMIP occurred in response to the tumor microenvironment, spontaneously releasing Cu2+/Cu+ via redox cycles, which in turn promoted highly potent GSH depletion and triggered •OH generation by a Fenton-like reaction. Notably, we found that the catalase activity could be effectively inhibited by the produced Cu+, which indirectly upregulated the endogenous H2O2 level. As a result, the "maladjusted" tumor cells lost the resistance against •OH damage, finally resulting in the apoptosis of tumor cells. In vitro and in vivo experiments demonstrated that our nanoMIP exhibited excellent cytotoxicity against tumor cells and high efficacy of tumor inhibition in the xenograft tumor model with negligible side effects. Taken together, our study provides not only a promising strategy for maximizing the CDT efficacy but also a new insight for developing MIP-based nanomedicine.


Assuntos
Nanopartículas , Neoplasias , Catalase/metabolismo , Linhagem Celular Tumoral , Dissulfetos/farmacologia , Homeostase , Humanos , Peróxido de Hidrogênio/metabolismo , Nanopartículas/uso terapêutico , Neoplasias/terapia , Oxirredução , Dióxido de Silício/farmacologia , Microambiente Tumoral
8.
Chem Sci ; 13(16): 4589-4597, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35656127

RESUMO

Accurately analysing the particular glycosylation status of protein biomarkers is of significant importance in the precise, early diagnosis of cancer. Existing methods mainly rely on the use of antibodies and lectins. However, due to the macroscopic and microscopic heterogeneity of glycans, precise analysis of glycosylation status still remains a challenge. Molecularly imprinted polymers (MIPs), as a synthetic alternative to antibodies or lectins, may provide new solutions but have not yet been explored. Herein, we report an appealing strategy called triple MIP-based plasmonic immunosandwich assay (triMIP-PISA) for precise cancer diagnosis in terms of the relative glycosylation expression of glycoprotein biomarkers. As proof of the principle, alpha fetoprotein (AFP), which has been used as a clinical biomarker for early detection of hepatocellular carcinoma (HCC), as well as its Lens culinaris agglutinin (LCA)-reactive fraction (AFP-L3), which is mainly composed of core-fucosylated glycans, were used as two target proteoforms to test in this study. Using two MIPs that can specifically recognize the peptide sequence of AFP as well as a fucose-imprinted MIP that can specifically recognize the AFP-L3 fraction, facile simultaneous plasmon-enhanced Raman detection of AFP and AFP-L3 in serum was achieved, which allowed HCC patients to be distinguished from healthy individuals. Due to the excellent recognition properties of the MIPs that are comparable to those of antibodies and superior to those of lectins, our triMIP-PISA method exhibited improved precision as compared with an antibody plus lectin-based immunofluorescence assay. Thus, this strategy opened a new avenue towards the precise diagnosis of cancer.

9.
Anal Chem ; 94(20): 7375-7382, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35544739

RESUMO

The ATP-binding cassette, subfamily G, isoform 2 protein (ABCG2), as an important member of ABC transporters, plays a key role in multidrug resistance (MDR) in cancer and has been widely considered as a marker of cancer stem cells (CSC). Reagents capable of simultaneously targeting ABCG2 and reversing MDR have great clinical application values, but their development is highly challenging. Herein, ABCG2 glycosylated extracellular region-binding aptamers were efficiently screened by a cladded molecularly imprinted polymer (cMIP)-based in vitro screening method and further rationally engineered into cyclic bivalent aptamers. Experiments showed that both the monovalent and cyclic bivalent aptamers could specifically bind ABCG2 and thereby specially target CSC of human colorectal carcinomas (CoCSC), while the latter could effectively reverse MDR in drug-resistant liver cancer cells (HepG2/ADR). Different from currently predominant small molecule inhibitors, the reversal of MDR relied on a different mechanism; the cyclic bivalent aptamers bound the two monomers of ABCG2 dimers simultaneously and thereby blocked the ABCG2-mediated drug-pumping channel, resulting in increased intracellular accumulation of substrate drugs. This study opened a new access to the development of affinity reagents for targeting CSC and reversing MDR, holding great prospects in cancer diagnosis and treatment.


Assuntos
Antineoplásicos , Neoplasias , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Humanos , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo
10.
Angew Chem Int Ed Engl ; 61(21): e202113528, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35194906

RESUMO

The glycosylation pattern of alpha fetoprotein (AFP) paves the basis for precise early diagnosis of hepatocellular carcinoma (HCC). However, existing analytical methods ignore the contribution of terminal sialic acid, which has been reported to be highly connected with HCC. Besides, the development of diagnostic assays is severely hindered by the preparation of anti-glycans antibodies. Molecularly imprinted polymers (MIPs), as synthetic antibody mimics, provide unique strengths to address these issues. Herein, we report a MIPs-based dual-modal ratiometric immunoassay for precise HCC diagnosis. Using a "pit one against ten" MIP to recognize a subset of glycans containing sialic acid and/or core fucose, we demonstrated our assay exhibited improved precision as compared with ELISA. This assay provided not only a glycoform-resolved method for precise HCC diagnosis, but also a new paradigm for developing antibody mimics via molecular imprinting towards challenging biomedical applications.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Impressão Molecular , Anticorpos , Carcinoma Hepatocelular/diagnóstico , Humanos , Imunoensaio/métodos , Neoplasias Hepáticas/diagnóstico , Impressão Molecular/métodos , Polímeros Molecularmente Impressos , Ácido N-Acetilneuramínico
11.
ACS Nano ; 15(11): 18214-18225, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34664930

RESUMO

Although protein therapeutics is of significance in therapeutic intervention of cancers, controlled delivery of therapeutic proteins still faces substantial challenges including susceptibility to degradation and denaturation and poor membrane permeability. Herein, we report a sialic acid (SA)-imprinted biodegradable silica nanoparticles (BS-NPs)-based protein delivery strategy for targeted cancer therapy. Cytotoxic ribonuclease A (RNase A) was effectively caged in the matrix of disulfide-hybridized silica NPs (encapsulation efficiency of ∼64%), which were further functionalized with cancer targeting capability via surface imprinting with SA as imprinting template. Such nanovectors could not only maintain high stability in physiological conditions but also permit redox-triggered biodegradation for both concomitant release of the loaded therapeutic cargo and in vivo clearance. In vitro experiments confirmed that the SA-imprinted RNase A@BS-NPs could selectively target SA-overexpressed tumor cells, promote cells uptake, and subsequently be cleaved by intracellular glutathione (GSH), resulting in rapid release kinetics and enhanced cell cytotoxicity. In vivo experiments further confirmed that the SA-imprinted RNase A@BS-NPs had specific tumor-targeting ability and high therapeutic efficacy of RNase A in xenograft tumor model. Due to the specific targeting and traceless GSH-stimulated intracellular protein release, the SA-imprinted BS-NPs provided a promising platform for the delivery of biomacromolecules in cancer therapy.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Ribonuclease Pancreático/metabolismo , Nanopartículas/uso terapêutico , Dióxido de Silício/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/patologia , Oxirredução , Proteínas/metabolismo , Linhagem Celular Tumoral
12.
J Biomater Sci Polym Ed ; 32(17): 2293-2305, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34429025

RESUMO

For the past few years, organic-inorganic hybrid nanocarriers have been widely explored for effective drug delivery and preferable disease treatments. In this article, hydrothermal method was utilized to prepare fine dispersed layered double hydroxide (Mg-Al LDH) suspension. Polyethylene glycol (PEG) was grafted on the surface of LDH lamella in order to improve the dispersibility of LDH. Besides, the anti-cancer drug gemcitabine was grafted on the surface of LDH lamellas through chemical grafting. Hence a novel new type of organic-inorganic hybrid drug delivery system LDH-mPEG-Gemcitabine was obtained. In addition, the siRNA was intercalated into the LDH interlamination by ion exchange method to realize drug and gene co-delivery. The loading capacity of LDH and LDH-mPEG-Gemcitabine was evaluated by agarose gel electrophoresis. The characterization by laser particle size analyzer, TEM, FT-IR, XRD, in vitro cell viability and in vitro drug release demonstrated that LDH-mPEG-Gemcitabine possessed fine dispersibility, uniform morphology and particle size, fine biocompatibility, ideal drug loading and releasing capacity and held great potential to be used as a desired co-delivery system for drug and gene.


Assuntos
Hidróxidos , Polietilenoglicóis , Desoxicitidina/análogos & derivados , Sistemas de Liberação de Medicamentos , Espectroscopia de Infravermelho com Transformada de Fourier , Gencitabina
13.
Angew Chem Int Ed Engl ; 60(5): 2663-2667, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33078504

RESUMO

Prodrug and drug delivery systems are two effective strategies for improving the selectivity of chemotherapeutics. Molecularly imprinted polymers (MIPs) have emerged as promising carriers in targeted drug delivery for cancer treatment, but they have not yet been integrated with the prodrug strategy. Reported here is an MIP-based smart prodrug delivery system for specific targeting, prolonged retention time, and tumor microenvironment-triggered release. 5'-Deoxy-5-fluorocytidine (DFCR) and sialic acid (SA) were used as a prodrug and a marker for tumor targeting, respectively. Their co-imprinted nanoparticles were prepared as a smart carrier. Prodrug-loaded MIP specifically and sustainably accumulated at the tumor site and then gradually released. Unlike conventional prodrug designs, which often require in-liver bioconversion, this MIP-based prodrug delivery is liver-independent but tumor-dependent. Thus, this study opens new access to the development of smart prodrug delivery nanoplatforms.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Polímeros Molecularmente Impressos/química , Pró-Fármacos/química , Humanos , Microambiente Tumoral
14.
Angew Chem Int Ed Engl ; 60(8): 3858-3869, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32789971

RESUMO

Molecularly imprinted polymers (MIPs) are chemically synthesized affinity materials with tailor-made binding cavities complementary to the template molecules in shape, size, and functionality. Recently, engineering MIP-based nanomedicines to improve cancer therapy has become a rapidly growing field and future research direction. Because of the unique properties and functions of MIPs, MIP-based nanoparticles (nanoMIPs) are not only alternatives to current nanomaterials for cancer therapy, but also hold the potential to fill gaps associated with biological ligand-based nanomedicines, such as immunogenicity, stability, applicability, and economic viability. Here, we survey recent advances in the design and fabrication of nanoMIPs for cancer therapy and highlight their distinct features. In addition, how to use these features to achieve desired performance, including extended circulation, active targeting, controlled drug release and anti-tumor efficacy, is discussed and summarized. We expect that this minireview will inspire more advanced studies in MIP-based nanomedicines for cancer therapy.


Assuntos
Polímeros Molecularmente Impressos/química , Nanopartículas/química , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/química , Humanos , Nanomedicina , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo
15.
Colloids Surf B Biointerfaces ; 165: 235-242, 2018 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29486452

RESUMO

In this study, the copolymer of methoxy poly(ethylene glycol) methacrylate-co-poly(methylacrylic acid) [poly(mPEGMA-co-MAA)] was synthesized via radical polymerization. Based on this copolymer, novel chitosan-modified poly(mPEGMA-co-MAA) nanoparticles (CS/NPs) were developed to improve the bio-availability of ibuprofen (IBU). Fourier transform infrared spectroscopy (FTIR) and 1H nuclear magnetic resonance (1H NMR) spectra were used to confirm the synthesis of the copolymers. The morphology of CS/NPs was investigated with transmission electron microscopy (TEM). Thermogravimetric analysis (TGA) was used to reveal the thermodynamic properties of the CS/NPs. The cytotoxicity of CS/NPs was assessed by the cell viability of 293T cells. FTIR and 1H NMR spectra confirmed the synthesis of the novel copolymer. TEM photographs showed that the CS/NPs had a core-shell structure. High cell viability indicated that the CS/NPs were nontoxic. The in vitro release profiles suggested that the CS/NPs released IBU in pH 7.4 buffer in a continuous manner. Furthermore, the IBU-CS/NPs showed a long antifebrile effect. Animal experiments showed that the IBU-CS/NPs had obvious antifebrile effects. Therefore, CS/NPs could reduce the dosing frequency of IBU, and improve its bio-availability.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Quitosana/química , Portadores de Fármacos , Febre/tratamento farmacológico , Ibuprofeno/farmacologia , Metacrilatos/química , Nanopartículas/química , Polietilenoglicóis/química , Acrilatos/química , Administração Oral , Animais , Anti-Inflamatórios não Esteroides/química , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Febre/induzido quimicamente , Febre/fisiopatologia , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Ibuprofeno/química , Injeções Subcutâneas , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Termodinâmica , Fermento Seco/administração & dosagem
16.
Biomater Sci ; 5(4): 698-706, 2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28184404

RESUMO

A novel injectable and high-solid-content drug-loaded supramolecular hydrogel (PTX-mPECT NP/α-CDgel) was prepared by self-assembly of inclusion complexes based on PTX-loaded mPECT (methoxy poly(ethylene glycol)-b-poly(ε-caprolactone-co-1,4,8-trioxa[4.6]spiro-9-un-decanone)) nanoparticles (PTX-mPECT NPs) and α-cyclodextrin (α-CD). Paclitaxel (PTX) was chosen as a hydrophobic drug encapsulated into mPECT NPs. Then, gelation occurred when the aqueous solution of α-CD was added to the PTX-mPECT NPs aqueous dispersion within several seconds after stirring. Importantly, with the erosion of the hydrogel, PTX-loaded NPs could be released again and then PTX released further. Rheological studies showed that PTX-mPECT NP/α-CDgel with good injectability underwent a shear-induced sol-gel transition. The results of in vitro drug-release studies demonstrated a sustained-release profile, and the cumulative release of PTX was ≈35% after 20 days. The results of cell-uptake studies and in vitro cytotoxicity studies indicated that the PTX-loaded NPs have been efficiently delivered to cells and killed tumor cells. Higher suppression of tumor growth demonstrated the remarkable anticancer effect of PTX-mPECT NP/α-CDgel upon peritumoral injection. These results showed that high-solid-content PTX-mPECT NP/α-CDgel based on in situ systems could be a promising candidate for local and sustained drug delivery.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Caproatos/química , Preparações de Ação Retardada/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Lactonas/química , Nanopartículas/química , Paclitaxel/administração & dosagem , Polietilenoglicóis/química , alfa-Ciclodextrinas/química , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células HeLa , Humanos , Injeções , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Transição de Fase
17.
Nanoscale ; 8(44): 18782-18791, 2016 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-27801924

RESUMO

For in situ thermosensitive hydrogels, it is a big challenge to achieve high drug loading, long-term local retention, and effective drug release simultaneously. To address these issues, we combined the strategy of drug nanocrystals (NCs) and thermosensitive hydrogels with higher gel strength. In particular, we developed paclitaxel NC-based hydrogels using PECT, a thermosensitive polymer synthesized by us (PTX-NC-PECT), and a nanoparticle-based system was used as the control (PTX-NP-PECT). First, high levels of PTX could be loaded in both PECT hydrogels. Moreover, in vivo near infrared fluorescence (NIRF) imaging showed that both hydrogel systems were able to maintain the payloads of 1,1-dioctadecyltetramethyl indotricarbocyanine iodide (DiR) at a peri-tumoural site for at least 21 days, much longer than that achieved with the control hydrogel of Pluronic® F127. Furthermore, we observed that PTX-NCs released free PTX more effectively and homogeneously than PTX-NPs in vitro. It was further verified in vivo that the release of DiR from DiR-NC-PECT was more complete than that from DiR-NP-PECT. Finally, PTX-NC-PECT gel demonstrated the strongest anti-tumour efficacy on MCF-7 breast cancer. In conclusion, PTX-NC-PECT hydrogel might be a high-performance thermosensitive hydrogel for local cancer therapy.

18.
Biomater Sci ; 4(10): 1493-502, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27546028

RESUMO

Thermo-sensitive injectable hydrogels based on poly(ε-caprolactone)/poly(ethylene glycol) (PCL/PEG) block copolymers have attracted considerable attention for sustained drug release and tissue engineering applications. Previously, we have reported a thermo-sensitive hydrogel of P(CL-co-TOSUO)-PEG-P(CL-co-TOSUO) (PECT) triblock copolymers modified by hydrophilic cyclic ether pendant groups 1,4,8-trioxa-[4.6]spiro-9-undecanone (TOSUO). Unfortunately, the low gel modulus of PECT (only 50-70 Pa) may limit its applications. Herein, another kind of thermogelling triblock copolymer of a pendant cyclic ether-modified caprolactonic poloxamer analog, PEG-P(CL-co-TOSUO)-PEG (PECTE), was successfully prepared by control of the hydrophilicity/hydrophobicity balance and chemical compositions of the copolymers. PECTE powder could directly disperse in water to form a stable nanoparticle (NP) aqueous dispersion and underwent sol-gel-sol transition behavior at a higher concentration with the temperature increasing from ambient or lower temperatures. Significantly, the microstructure parameters (e.g., different chemical compositions of the hydrophobic block and topology) played a critical role in the phase transition behavior. Furthermore, comparison studies on PECTE and PEG-PCL-PEG (PECE) showed that the introduction of pendant cyclic ether groups into PCL blocks could avoid unexpected ahead-of-time gelling of the PECE aqueous solution. In addition, the rheological analysis of PECTE and PECT indicated that the storage modulus of the PECTE hydrogel could be 100 times greater than that of the PECT hydrogel under the same mole ratios of TOSUO/CL and lower molecular weight. Consequently, PECTE thermal hydrogel systems are believed to be promising as in situ gel-forming biomaterials for drug delivery and tissue engineering.


Assuntos
Éteres Cíclicos/química , Hidrogéis/química , Nanopartículas/química , Poliésteres/química , Polietilenoglicóis/química , Temperatura
19.
Eur J Pharm Biopharm ; 104: 251-9, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27185379

RESUMO

Intraperitoneal (IP) chemotherapy is a preferable treatment option for peritoneal carcinomatosis of malignancies by delivering chemotherapeutic drugs into the abdominal cavity. A persistent major challenge in IP chemotherapy is the need to provide effective drug concentration in the peritoneal cavity for an extended period of time. In the present work, the thermosensitive hydrogel system (PTX/PECT(gel)) assembled by PTX (paclitaxel)-loaded amphiphilic copolymer (PECT, poly (ε-caprolactone-co-1,4,8-trioxa [4.6]spiro-9-undecanone)-poly(ethylene glycol)-poly (ε-caprolactone-co-1,4,8-trioxa [4.6]spiro-9-undecanone)) nanoparticles was developed for sustained IP chemotherapy of peritoneal carcinomatosis model. Cytotoxicity assay indicated that PECT hydrogel was biocompatible with very low cytotoxicity and PTX/PECT(gel) had enhanced cytotoxicity than free PTX. In vivo toxicity study demonstrated the biocompatibility and biosafety of PECT hydrogel as an IP chemotherapy carrier. The fluorescence imaging method was employed to monitor the intraperitoneal degradation of PECT hydrogel by labeling PECT with rhodamine B. PECT hydrogel with the dose of 200µL showed about 8days' retention time and most of the injected hydrogel was located in the intestine. The anti-tumor efficacy study was carried out in mice bearing CT26 intraperitoneal ascites fluid as colorectal peritoneal carcinomatosis model. The result showed that intraperitoneal administration of PTX/PECT(gel) could effectively suppress growth and metastasis of CT26 peritoneal carcinomatosis in vivo, compared with Taxol® group. The pharmacokinetic studies demonstrated that PTX/PECT(gel) could improve the bioavailability of PTX by being formulated in PECT hydrogel. Overall, sustained drug concentration at peritoneal levels in combination with drug in the form of nanoparticle contributes to the enhanced anti-tumor efficacy. Thus, our results suggested that PTX/PECT(gel) may have great potential applications in IP chemotherapy.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Hidrogéis/química , Nanopartículas/química , Paclitaxel/química , Neoplasias Peritoneais/tratamento farmacológico , Polímeros/química , Animais , Antineoplásicos Fitogênicos/farmacocinética , Linhagem Celular Tumoral , Camundongos , Camundongos Endogâmicos BALB C , Paclitaxel/uso terapêutico , Distribuição Tecidual
20.
Macromol Biosci ; 16(8): 1188-99, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27105589

RESUMO

Injectable and biodegradable supramolecular hydrogel mPECT NP/α-CD(gel) composed of high-concentration nanoparticle dispersion (≤20% W/V) and α-cyclodextrins (α-CD) are prepared by a two-level physical cross-linking using amphiphilic block polymer methoxy poly(ethylene glycol)-b-poly(ε-caprolactone-co-1,4,8-trioxa[4.6]spiro-9-undecanone) (mPECT) and α-CD. The gelation behavior depends on the concentration of nanoparticles and α-CD. The viscoelasticity and shear thinning of mPECT NP/α-CD(gel) are confirmed. In vitro hydrogel erosion is demonstrated to be mainly a concentration-dependent dissociation process with general release of discrete mPECT nanoparticles about 50 nm that can be easily taken up by cells. The in vitro release behavior can be modulated by changing the concentration of nanoparticles or α-CD. In vitro and in vivo cytotoxicity study demonstrates its biocompatibility and biosafety. Gel formation after subcutaneous injection is also confirmed and mPECT NP/α-CD(gel) shows about 2 weeks retention time. This work validates the potential application for this supramolecular hydrogel in local and sustained delivery of nanoparticles.


Assuntos
Preparações de Ação Retardada/farmacologia , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanopartículas/química , alfa-Ciclodextrinas/química , Animais , Varredura Diferencial de Calorimetria , Morte Celular , Linhagem Celular Tumoral , Endocitose , Etilenoglicóis/síntese química , Etilenoglicóis/química , Humanos , Camundongos Endogâmicos BALB C , Poliésteres/síntese química , Poliésteres/química , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA