Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Biomacromolecules ; 21(2): 921-929, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-31961134

RESUMO

A remarkable hallmark of cancer cells is the heterogeneous coexistence of overproduced intracellular glutathione (GSH) and a high level of reactive oxygen species (ROS) compared with those in normal cells, which have been frequently used as the stimuli to trigger drug release from the nanocarriers. Most of the stimuli-responsive delivery vehicles have been designed to respond to only one redox stimulus (e.g., GSH or ROS). Herein, we develop a GSH and ROS dual-responsive amphiphilic diblock copolymer prodrug (BCP) (GR-BCP) consisting of poly(ethylene glycol) (PEG)- and camptothecin (CPT)-conjugated poly(methacrylate) in the side chains via thioether bonds. In comparison, GSH or ROS single-responsive BCPs (G-BCPs or R-BCPs) were prepared, where CPT drugs were linked by disulfide or thioketal bonds, respectively. The three BCPs can form well-defined spherical micellar nanoparticles in an aqueous solution with a diameter of ∼50 nm. Compared with G-BCP and R-BCP, GR-BCP realized the highest cytotoxicity against HeLa cells with the half-inhibitory concentration (IC50) of 6.3 µM, which is much lower than 17.8 µM for G-BCP and 28.9 µM for R-BCP. Moreover, for in vivo antitumor performance, G-BCP, R-BCP, and GR-BCP showed similar efficiencies in blood circulation and tumor accumulation after intravenous injection. However, GR-BCP realized the most efficient tumor suppression with few side effects. Our findings demonstrate that intracellular GSH and ROS dual-responsive BCPs show a more efficient responsive drug release inside tumor cells for boosting the antitumor efficacy as compared with GSH or ROS single-responsive BCPs, which provides novel strategies for designing redox-responsive BCPs.


Assuntos
Antineoplásicos/metabolismo , Liberação Controlada de Fármacos/fisiologia , Glutationa/metabolismo , Polímeros/metabolismo , Pró-Fármacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Polietilenoglicóis/metabolismo , Polímeros/administração & dosagem , Polímeros/química , Polimetil Metacrilato/administração & dosagem , Polimetil Metacrilato/química , Polimetil Metacrilato/metabolismo , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Resultado do Tratamento
2.
Bioconjug Chem ; 29(1): 23-28, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29232506

RESUMO

Hydrophobic segments and amino moieties in polymeric nonviral gene vectors play important roles in overcoming a cascade of barriers for efficient gene delivery. However, it remains a great challenge to facilely construct well-defined multifunctional polymers through optimization of the amino and hydrophobic groups. Herein, we choose thiolactone chemistry to perform the ring opening reaction of varying hydrophobic groups-modified thiolactones by various amines to generate mercapto groups for further Michael addition reaction with poly[2-(acryloyloxy)ethyl methacrylate] (PAOEMA). Based on the combinatorial methodology, a series of multifunctional polymers were prepared and screened. The polymer (P3D) from tetraethylenepentamine and heptafluorobutyric acid-functionalized thiolactone is the most efficacious one with significantly higher gene transfection efficiency and lower cytotoxicity compared with polyethylenimine (PEI) (branched average Mw ∼ 25 000 Da) and Lipofectamine 2000. Cellular uptake and intracellular distribution studies indicate that P3D complexes show high-efficiency endocytosis and excellent endosomal escape. Accordingly, thiolactone chemistry-based combinatorial methodology allows for facile integration of multifunctional groups to prepare simultaneous efficacious and low-cytotoxic gene delivery vectors.


Assuntos
Técnicas de Química Combinatória/métodos , Lactonas/química , Polímeros/química , Compostos de Sulfidrila/química , Transfecção , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Células HeLa , Humanos , Lactonas/síntese química , Polímeros/síntese química , Compostos de Sulfidrila/síntese química , Transfecção/métodos
3.
Biomacromolecules ; 19(6): 1990-1998, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29420880

RESUMO

Relative to normal cells, tumor cells lack adequate capability of reactive oxygen scavenging. Thus, tumor cells can be selectively killed by increasing the concentration of reactive oxygen species in tumor tissue. In this report, we construct an integrated multifunctional polymeric nanoparticle which can selectively improve hydrogen peroxide (H2O2) levels in tumor tissue and convert them into more active hydroxyl radicals by Fenton reaction. First, the diblock copolymers containing polyethylene glycol (PEG) and poly(glutamic acid) modified by ß-cyclodextrin (ß-CD) were synthesized. The block copolymer, ferrocenecarboxylic acid hexadecyl ester (DFc), and ascorbyl palmitate (PA) were coassembled in aqueous solution to obtain stable core-shell micelles through the inclusion complexation between ß-CD moieties in the block copolymer and ferrocene (Fc) groups from DFc. After intravenous injection, the particles achieved significant accumulation in tumor tissue where ascorbic acid at the pharmacological concentration promotes the production of H2O2, and subsequently Fenton reaction was catalyzed by Fc groups to produce hydroxyl radicals to efficiently kill cancer cells and suppress tumor growth. The micellar systems possess great potentials toward cancer therapy through synergistic H2O2 production and conversion into hydroxyl radicals specifically in tumor tissue.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Nanopartículas/química , Polímeros/química , Animais , Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/química , Ácido Ascórbico/farmacocinética , Ácido Ascórbico/farmacologia , Feminino , Compostos Ferrosos/química , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Radical Hidroxila/química , Radical Hidroxila/metabolismo , Injeções Intravenosas , Ferro/química , Células MCF-7 , Metalocenos , Camundongos Endogâmicos BALB C , Micelas , Nanopartículas/administração & dosagem , Polietilenoglicóis/química , Ácido Poliglutâmico/química , Polímeros/farmacocinética , Polímeros/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , beta-Ciclodextrinas/química
4.
Nano Lett ; 17(11): 6983-6990, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-28977746

RESUMO

Therapeutic nanoreactors have been proposed to treat cancers through in situ transformation of low-toxicity prodrugs into toxic therapeutics in the body. However, the in vivo applications are limited by low tissue-specificity and different tissue distributions between sequentially injected nanoreactors and prodrugs. Herein, we construct a block copolymer prodrug-based polymersome nanoreactor that can achieve novel orchestrated oxidation/chemotherapy of cancer via specific activation at tumor sites. The block copolymers composed of poly(ethylene glycol) (PEG) and copolymerized monomers of camptothecin (CPT) and piperidine-modified methacrylate [P(CPTMA-co-PEMA)] were optimized to self-assemble into polymersomes in aqueous solution for encapsulation of glucose oxidase (GOD) to obtain GOD-loaded polymersome nanoreactors (GOD@PCPT-NR). GOD@PCPT-NR maintained inactive in normal tissues upon systemic administration. After deposition in tumor tissues, tumor acidity-triggered protonation of PPEMA segments resulted in high permeability of the polymersome membranes and oxidation reaction of diffused glucose and O2 under the catalysis of GOD. The activation of the reaction generated H2O2, improving the oxidative stress in tumors. Simultaneously, a high level of H2O2 further activated PCPTMA prodrugs, releasing active CPT drugs. High tumor oxidative stress and released CPT drugs synergistically killed cancer cells and suppressed tumor growth via oxidation/chemotherapy. Our study provides a new strategy for engineering therapeutic nanoreactors in an orchestrated fashion for cancer therapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Glucose Oxidase/farmacologia , Peróxido de Hidrogênio/metabolismo , Metacrilatos/farmacologia , Polietilenoglicóis/farmacologia , Pró-Fármacos/farmacologia , Células A549 , Antineoplásicos Fitogênicos/química , Camptotecina/química , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Glucose Oxidase/química , Humanos , Concentração de Íons de Hidrogênio , Metacrilatos/química , Nanoestruturas/química , Neoplasias/tratamento farmacológico , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Polietilenoglicóis/química , Pró-Fármacos/química
5.
Bioconjug Chem ; 28(8): 2190-2198, 2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28661654

RESUMO

The amphiphilic block copolymer anticancer drug nanocarriers clinically used or in the progress of clinical trials frequently suffer from modest final therapeutic efficacy due to a lack of intelligent features. For example, the biodegradable amphiphilic block copolymer, poly(ethylene glycol)-b-poly(d,l-lactide) (PEG-PDLLA) has been approved for clinical applications as a paclitaxel (PTX) nanocarrier (Genexol-PM) due to the optimized pharmacokinetics and biodistribution; however, a lack of intelligent features limits the intracellular delivery in tumor tissue. To endow the mediocre polymer with smart properties via a safe and facile method, we introduced a matrix metalloproteinase (MMP)-responsive peptide GPLGVRGDG into the block copolymer via efficient click chemistry and ring-opening polymerization to prepare PEG-GPLGVRGDG-PDLLA (P1). P1 was further self-assembled into micellar nanoparticles (NPs) to load PTX, which show MMP-2-triggered dePEGylation due to cleavage of the peptide linkage. Moreover, the residual VRGDG sequences are retained on the surface of the NPs after dePEGylation, which can serve as ligands to facilitate the cellular uptake. The cytotoxicity of PTX loaded in P1 NPs against 4T1 cells is significantly enhanced as compared with free PTX or PTX-loaded PEG-GPLGVRG-PDLLA (P2) and PEG-PDLLA (P3) NPs. In vivo studies confirmed that PTX-loaded P1 NPs show prolonged blood circulation, which are similar to P2 and P3 NPs but exhibit more-efficient accumulation in the tumor site. Ultimately, PTX-loaded P1 NPs display statistically significant improvement of antitumor activity against tumor-bearing mice via systemic administration. Therefore, the strategy by facile incorporation of a responsive peptide linkage between PEG and PDLLA is a promising approach to improving the therapeutic efficacy of anticancer-drug-loaded amphiphilic block copolymer micelles.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Interações Hidrofóbicas e Hidrofílicas , Metaloproteinases da Matriz/metabolismo , Peptídeos/química , Poliésteres/química , Polietilenoglicóis/química , Sequência de Aminoácidos , Animais , Antineoplásicos/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Portadores de Fármacos/farmacocinética , Humanos , Camundongos , Distribuição Tecidual
6.
Angew Chem Int Ed Engl ; 56(45): 14025-14030, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28940903

RESUMO

Polymeric nanoreactors (NRs) have distinct advantages to improve chemical reaction efficiency, but the in vivo applications are limited by lack of tissue-specificity. Herein, novel glucose oxidase (GOD)-loaded therapeutic vesicular NRs (theraNR) are constructed based on a diblock copolymer containing poly(ethylene glycol) (PEG) and copolymerized phenylboronic ester or piperidine-functionalized methacrylate (P(PBEM-co-PEM)). Upon systemic injection, theraNR are inactive in normal tissues. At a tumor site, theraNR are specifically activated by the tumor acidity via improved permeability of the membranes. Hydrogen peroxide (H2 O2 ) production by the catalysis of GOD in theraNR increases tumor oxidative stress significantly. Meanwhile, high levels of H2 O2 induce self-destruction of theraNR releasing quinone methide (QM) to deplete glutathione and suppress the antioxidant ability of cancer cells. Finally, theraNR efficiently kill cancer cells and ablate tumors via the synergistic effect.


Assuntos
Antineoplásicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Portadores de Fármacos , Nanoestruturas , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polietilenoglicóis/química , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antioxidantes/metabolismo , Ácidos Borônicos/química , Permeabilidade da Membrana Celular , Ésteres , Glucose Oxidase/metabolismo , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Indolquinonas/química , Metacrilatos/química , Microscopia Eletrônica de Transmissão , Neoplasias/metabolismo , Piperidinas/química , Estudo de Prova de Conceito
7.
Biomacromolecules ; 17(10): 3268-3276, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27564064

RESUMO

Construction of efficient doxorubicin (DOX) delivery systems addressing a cascade of physiological barriers remains a great challenge for better therapeutic efficacy of tumors. Herein, we design well-defined enzyme-responsive peptide-linked block copolymer, PEG-GPLGVRGDG-P(BLA-co-Asp) [PEG and P(BLA-co-Asp) are poly(ethylene glycol) and partially hydrolyzed poly(ß-benzyl l-aspartate) (PBLA), respectively] (P3), with modular functionality for efficient delivery of DOX. The block copolymers were successfully obtained via click reaction to introduce peptide (alkynyl-GPLGVRGDG) into the end of PEG for initiating ring-opening polymerization of ß-benzyl l-aspartate N-carboxyanhydride (BLA-NCA) by terminal amino groups followed by partial hydrolysis of PBLA segments. P3 micelle was demonstrated to encapsulate DOX efficiently through synergistic effect of benzyl group-based hydrophobic and carboxyl moiety-based electrostatic interactions. Effective matrix metalloproteinase-2 (MMP-2)-triggered cleavage of peptide for dePEGylation of P3 micelles was confirmed and residual RGD ligands were retained on the surfaces. Against HT1080 cells overexpressing MMP-2, DOX-loaded P3 micelles showed approximately 4-fold increase of the cellular internalization amount as compared with free DOX and half maximal inhibitory concentration (IC50) value of DOX-loaded P3 micelles was determined to be 0.38 µg/mL compared with 0.66 µg/mL of free DOX due to MMP-triggered dePEGylation, RGD-mediated cellular uptake, and rapid drug release inside cells. Binding and penetration evaluation toward HT1080 multicellular tumor spheroids (MCTs) confirmed high affinity and deep penetration of P3 micelles in tumor tissues. This modular design of enzyme-responsive block copolymers represents an effective strategy to construct intelligent drug delivery vehicles for addressing a cascade of delivery barriers.


Assuntos
Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Peptídeos/administração & dosagem , Linhagem Celular Tumoral , Doxorrubicina/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/genética , Neoplasias/genética , Tamanho da Partícula , Peptídeos/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química
8.
Sci Total Environ ; 919: 170739, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38340854

RESUMO

Nanoplastics (NPs) are unavoidable hazardous materials that result from the human production and use of plastics. While there is evidence that NPs can bioaccumulate in the brain, no enough research regarding the pathways by which NPs reach the brain was conducted, and it is also urgently needed to evaluate the health threat to the nervous system. Here, we observed accumulation of polystyrene nanoplastics (PS-NPs) with different surface modifications (PS, PS-COOH, and PS-NH2) in mouse brains. Further studies showed that PS-NPs disrupted the tight junctions between endothelial cells and transport into endothelial cells via the endocytosis and macropinocytosis pathways. Additionally, NPs exposure induced a series of alternations in behavioral tests, including anxiety- and depression-like changes and impaired social interaction performance. Further results identified that NPs could be internalized into neurons and localized in the mitochondria, bringing about mitochondrial dysfunction and a concurrent decline of ATP production, which might be associated with abnormal animal behaviors. The findings provide novel insights into the neurotoxicity of NPs and provide a basis for the formulation of policy on plastic production and usage by relevant government agencies.


Assuntos
Nanopartículas , Poluentes Químicos da Água , Humanos , Animais , Camundongos , Poliestirenos/toxicidade , Poliestirenos/metabolismo , Microplásticos , Depressão/induzido quimicamente , Células Endoteliais/metabolismo , Poluentes Químicos da Água/toxicidade , Ansiedade/induzido quimicamente , Nanopartículas/toxicidade , Nanopartículas/metabolismo , Neurônios/metabolismo , Plásticos
9.
Int J Biol Macromol ; 258(Pt 1): 128520, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38040150

RESUMO

In this study, we developed an enhanced heterogeneous interface intelligent conductive hydrogel NH3 sensor for individualized treatment of infected wounds. The sensor achieved monitoring, self-diagnosis, and adaptive gear adjustment functions. The PPY@PDA/PANI(3/6) sensor had a minimum NH3 detection concentration of 50 ppb and a response value of 2.94 %. It also had a theoretical detection limit of 49 ppt for infected wound gas. The sensor exhibited a fast response time of 23.2 s and a recovery time of 42.9 s. Tobramycin (TOB) was encapsulated in a self-healing QCS/OD hydrogel formed by quaternized chitosan (QCS) and oxidized dextran (OD), followed by the addition of polydopamine-coated polypyrrole nanowires (PPY@PDA) and polyaniline (PANI) to prepare electrically conductive drug-loaded PPY@PDA/PANI hydrogels. The drug-loaded PPY@PDA/PANI hydrogel was combined with a PANI/PVDF membrane to form an enhanced heterogeneous interfacial PPY@PDA/PANI/PVDF-based sensor, which could adaptively learn the individual wound ammonia response and adjust the speed of drug release from the PPY@PDA/PANI hydrogel with electrical stimulation. Drug release and animal studies demonstrated the efficacy of the PPY@PDA/PANI hydrogel in inhibiting infection and accelerating wound healing. In conclusion, the gas-sensitive conductive hydrogel sensing system is expected to enable intelligent drug delivery and provide personalized treatment for complex wound management.


Assuntos
Quitosana , Polímeros de Fluorcarboneto , Polímeros , Polivinil , Animais , Hidrogéis/farmacologia , Pirróis
10.
Environ Int ; 176: 107968, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37201399

RESUMO

Nanoplastics (NPs), regarded as the emerging contaminants, can enter and be mostly accumulated in the digest tract, which pose the potential threat to intestinal health. In this study, mice were orally exposed to polystyrene (PS), PS-COOH and PS-NH2 NPs with the size of ∼100 nm at a human equivalent dose for 28 consecutive days. All three kinds of PS-NPs triggered Crohn's ileitis-like features, such as ileum structure impairment, increased proinflammatory cytokines and intestinal epithelial cell (IEC) necroptosis, and PS-COOH/PS-NH2 NPs exhibited higher adverse effects on ileum tissues. Furthermore, we found PS-NPs induced necroptosis rather than apoptosis via activating RIPK3/MLKL pathway in IECs. Mechanistically, we found that PS-NPs accumulated in the mitochondria and subsequently caused mitochondrial stress, which initiated PINK1/Parkin-mediated mitophagy. However, mitophagic flux was blocked due to lysosomal deacidification caused by PS-NPs, and thus led to IEC necroptosis. We further found that mitophagic flux recovery by rapamycin can alleviate NP-induced IEC necroptosis. Our findings revealed the underlying mechanisms concerning NP-triggered Crohn's ileitis-like features and might provide new insights for the further safety assessment of NPs.


Assuntos
Doença de Crohn , Ileíte , Nanopartículas , Poluentes Químicos da Água , Animais , Camundongos , Humanos , Poliestirenos/toxicidade , Poliestirenos/química , Microplásticos , Necroptose , Doença de Crohn/metabolismo , Células Epiteliais , Ileíte/metabolismo , Nanopartículas/toxicidade
11.
Biomater Sci ; 9(13): 4613-4629, 2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34190224

RESUMO

Prodrug strategy especially in the field of chemotherapy of cancers possesses significant advantages reducing the side toxicity of anticancer drugs. However, high-efficiency delivery and in situ activation of prodrugs for tumor growth suppression are still a great challenge. Herein, we report rationally engineered pH-responsive endosomolytic polymeric micelles for the delivery of an oxidation-activable prodrug into the cytoplasm of cancer cells and amplification of intracellular oxidative stress for further prodrug activation. The prepared block copolymers consist of a poly(ethylene glycol) (PEG) block and a segment grafted by endosomolytic moieties and acetal linkage-connected cinnamaldehyde groups. The amphiphilic diblock copolymers can self-assemble to form micelles in water for loading the oxidation-activable phenylboronic pinacol ester-caged camptothecin prodrug (ProCPT). The obtained micelles can release free cinnamaldehyde under acidic conditions in tumor tissues and endo/lysosomes followed by efficient endosomal escape, which further induces enhancement of intracellular reactive oxygen species (ROS) to activate the prodrugs. Simultaneously, intracellular glutathione (GSH) can be reduced by quinone methide that was produced during prodrug activation. The ProCPT-loaded micelles can finally achieve efficient tumor accumulation and retention as well as effective tumor growth inhibition. More importantly, hematological and pathological analysis of toxicity reveals that the ProCPT-loaded micelles do not cause obvious toxic side effects toward important organs of mice. A positive immunomodulatory microenvironment in tumor tissue and serum can be detected after treatment with ProCPT-loaded micelles. Therefore, the endosomolytic ProCPT-loaded micelles exert synergistic therapeutic effects toward tumors through amplification of intracellular oxidative stress and activation of the prodrugs.


Assuntos
Antineoplásicos , Nanopartículas , Pró-Fármacos , Animais , Camundongos , Micelas , Estresse Oxidativo , Polietilenoglicóis , Polímeros
12.
J Mater Chem B ; 9(13): 3055-3067, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33885667

RESUMO

Drug resistance of cisplatin significantly limits its therapeutic efficacy in clinical applications against different cancers. Herein, we develop a novel strategy to overcome cisplatin drug resistance through sensitizing cisplatin-resistant human lung cancer cells (A549R) under amplified oxidative stress using a vesicular nanoreactor for simultaneous cisplatin delivery and H2O2 generation. We engineer the nanoreactor by the self-assembly of the amphiphilic diblock copolymers to co-deliver glucose oxidase (GOD) and cisplatin (Cis) (Cis/GOD@Bz-V). Cis/GOD@Bz-V was rationally designed to stay impermeable during blood circulation while mild acidity (pH 6.5-6.8) can activate its molecular-weight selective membrane permeability and release cisplatin locally. Diffusion of small molecules such as oxygen and glucose across the membranes can induce the in situ generation of superfluous H2O2 to promote cellular oxidative stress and sensitize A549R cells via activation of pro-apoptotic pathways. Cis/GOD@Bz-V nanoreactors could effectively kill A549R at pH 6.8 in the presence of glucose by the combination of H2O2 generation and cisplatin release. Growth of A549R xenograft tumors can be inhibited efficiently without the obvious toxic side effects via the systemic administration of Cis/GOD@Bz-V. Accordingly, the tumor acidity-activable cisplatin-loaded nanoreactors show great potential to enhance the therapeutic efficacy against cisplatin-resistant cancers.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Nanotecnologia , Polímeros/farmacologia , Células A549 , Animais , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Nanotecnologia/instrumentação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Estresse Oxidativo/efeitos dos fármacos , Tamanho da Partícula , Polímeros/síntese química , Polímeros/química , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo
13.
J Control Release ; 318: 67-77, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31837355

RESUMO

Filomicelles possess some unique properties for improved in vivo drug delivery efficiency relative to commonly used spherical nanocarriers, which have attracted great interests. However, the length effect of the block copolymer prodrug-based filomicelles with a comparable cross-section diameter on the drug delivery efficiency and antitumor efficacy still need to be systematically studied. In this report, we prepare three optimized nanoparticles with a comparable cross-section diameter of ~40 nm, including long filomicelles (LFMs) with the length of ~2.5 µm, short filomicelles (SFMs) with the length of ~180 nm, and spherical micelles (SMs) with a diameter of ~40 nm. All of them are self-assembled from the pH and oxidation dual-responsive block copolymer prodrug, PEG-b-P(CPTKMA-co-PEMA), consisting of poly(ethylene glycol) (PEG) and a copolymerized block of thioketal-linked camptothecin methacrylate (CPTKMA) and 2-(pentamethyleneimino) ethyl methacrylate (PEMA). At pH 6.5, the nanoparticles are positively charged due to the protonation of PPEMA segments. Among them, SFMs are demonstrated to be internalized into cells most efficiently at pH 6.5 due to larger interaction areas with cell membranes relative to SMs. Moreover, SFMs show prolonged blood circulation similar to SMs as well as deepest tumor penetration and best antitumor efficacy among the three nanoparticles. LFMs show worst in vivo performance because their too long structure limits the cellular uptake and tumor accumulation. Therefore, the responsive polymer prodrug filomicelles with an optimized length show great potentials to overcome the physiological barriers and improve the drug delivery efficiency.


Assuntos
Pró-Fármacos , Sistemas de Liberação de Medicamentos , Concentração de Íons de Hidrogênio , Micelas , Polietilenoglicóis , Polímeros
14.
J Control Release ; 303: 209-222, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31026547

RESUMO

Therapeutic nanoreactors are currently emerging as promising nanoplatforms to in situ transform inert prodrugs into active drugs. Nevertheless, it is still challenging to engineer a nanoreactor with balanced key features of tunable selective membrane permeability and structural stability for prodrug delivery and activation in diseased tissues. Herein, we present a facile strategy to engineer a polymersome nanoreactor with tumor-specific tunable membrane permeability to load both hydrophobic phenylboronic ester-caged anticancer prodrugs (e.g., camptothecin or paclitaxel prodrug) and hydrophilic glucose oxidase (GOD) in the membranes and cavities, respectively. The nanoreactors maintain inactive during blood circulation and in normal tissues. Upon accumulation in tumors, the mild acidic microenvironment triggers selective membrane permeability to allow small molecules (glucose and O2) to diffuse across the membrane and react under the catalysis of GOD. The massively generated H2O2 triggers in situ transformation of innocuous prodrugs into toxic parental drugs through cleavage of the self-immolative degradable caging groups. The developed system showed significantly enhanced antitumor efficacy by H2O2 production and prodrug activation via combined oxidation-chemotherapy. The well-devised polymersome nanoreactors with tumor-pH-tunable membrane permeability to coload H2O2-responsive prodrug and GOD represent a novel strategy to realize prodrug delivery and activation for enhanced therapeutic efficacy with low side toxicity.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Camptotecina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Glucose Oxidase/administração & dosagem , Paclitaxel/administração & dosagem , Polímeros/administração & dosagem , Pró-Fármacos/administração & dosagem , Animais , Antineoplásicos Fitogênicos/química , Camptotecina/química , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Portadores de Fármacos/química , Feminino , Glucose Oxidase/química , Humanos , Peróxido de Hidrogênio/química , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos ICR , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Oxirredução , Paclitaxel/química , Polímeros/química , Pró-Fármacos/química
15.
ACS Nano ; 13(2): 2357-2369, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30699292

RESUMO

Therapeutic nanoreactors are of increasing interest in precise cancer therapy, which have been explored to in situ produce therapeutic compounds from inert prodrugs or intrinsic molecules at the target sites. However, engineering a nanoreactor with tumor activable cascade reactions for efficient cooperative cancer therapy remains a great challenge. Herein, we demonstrate a polymersome nanoreactor with tumor acidity-responsive membrane permeability to activate cascade reactions for orchestrated cooperative cancer treatment. The nanoreactors are constructed from responsive polyprodrug polymersomes incorporating ultrasmall iron oxide nanoparticles and glucose oxidase in the membranes and inner aqueous cavities, respectively. The cascade reactions including glucose consumption to generate H2O2, accelerated iron ion release, Fenton reaction between H2O2 and iron ion to produce hydroxyl radicals (•OH), and •OH-triggered rapid release of parent drugs can be specifically activated by the tumor acidity-responsive membrane permeability. During this process, the orchestrated cooperative cancer therapy including starving therapy, chemodynamic therapy, and chemotherapy is realized for high-efficiency tumor suppression by the in situ consumed and produced compounds. The nanoreactor design with tumor-activable cascade reactions represents an insightful paradigm for precise cooperative cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Radical Hidroxila/farmacologia , Nanopartículas/química , Neoplasias/tratamento farmacológico , Polímeros/farmacologia , Pró-Fármacos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Liberação Controlada de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Corantes Fluorescentes/química , Humanos , Concentração de Íons de Hidrogênio , Radical Hidroxila/síntese química , Radical Hidroxila/química , Estrutura Molecular , Neoplasias/patologia , Polímeros/síntese química , Polímeros/química , Pró-Fármacos/síntese química , Pró-Fármacos/química , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo
16.
Biomaterials ; 154: 261-274, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29149720

RESUMO

Block copolymer prodrugs (BCPs) have attracted considerable attentions in clinical translation of nanomedicine owing to their self-assembly into well-defined core-shell nanoparticles for improved pharmacokinetics, stability in blood circulation without drug leakage, and optimized biodistribution. However, a cascade of physiological barriers against specific delivery of drugs into tumor cells limit the final therapeutic efficacy. Herein, we report a robust and facile strategy based on thiolactone chemistry to fabricate well-defined BCPs with sequential tumor pH-promoted cellular internalization and intracellular stimuli-responsive drug release. A series of BCPs were prepared through one-pot synthesis from clinically used small molecule anticancer drugs. The ring-opening reaction of drug-conjugated thiolactones releases mercapto groups via aminolysis by N-(3-aminopropyl)-imidazole, which further react with poly(ethylene glycol)-block-poly(pyridyldisulfide ethylmethacrylate) (PEG-PDSEMA) to produce imidazole and disulfide bonds-incorporated BCPs. Taking paclitaxel (PTX) for example, PTX BCPs exhibited high drug-loading content (>50%) and low critical micellization concentration (5 × 10-3 g/L), which can self-assemble into micellar nanoparticles in aqueous solution with a small size (∼40 nm). The nanoparticles showed high tumor accumulation and uniform distribution in hypopermeable tumors via systemic administration. Meanwhile, imidazole moieties endow nanoparticles tumor pH-sensitive charge transition from nearly neutral to positive, which promoted cellular internalization. Disulfide bonds can be cleaved by intracellular glutathione (GSH) of cancer cells, which accelerate the release of active PTX drug inside cells. Finally, highly aggressive murine breast cancer 4T1 tumor and hypopermeable human pancreatic adenocarcinoma BxPC3 tumor were completely ablated after treatment by PTX BCP nanoparticles. Consequently, the robust and facile preparation strategy based on thiolactone chemistry represents an efficient approach to construct multifunctional BCPs for better therapeutic efficacy via addressing multiple physiological barriers.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Lactonas/química , Polímeros/química , Pró-Fármacos/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Difusão Dinâmica da Luz , Feminino , Camundongos Endogâmicos BALB C , Nanopartículas/química , Polímeros/síntese química , Espectroscopia de Prótons por Ressonância Magnética , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
17.
J Control Release ; 273: 30-39, 2018 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-29371047

RESUMO

The intrinsic or acquired cisplatin resistance of cancer cells frequently limits the final therapeutic efficacy. Detoxification by the high level of intracellular glutathione (GSH) plays critical roles in the majority of cisplatin-resistant cancers. In this report, we designed an amphiphilic diblock copolymer composed of poly(ethylene glycol) (PEG) and polymerized phenylboronic ester-functionalized methacrylate (PBEMA), PEG-b-PBEMA, which can self-assemble into micelles in aqueous solutions to load hydrophobic cisplatin prodrug (Pt(IV)). Pt(IV)-loaded PEG-b-PBEMA micelles (PtBE-Micelle) reverse cisplatin-resistance of cancer cells through improving cellular uptake efficiency and reducing intracellular GSH level. We found that the cellular uptake amount of platinum from PtBE-Micelle was 6.1 times higher than that of free cisplatin in cisplatin-resistant human lung cancer cells (A549R). Meanwhile, GSH concentration of A549R cells was decreased to 32% upon treatment by PEG-b-PBEMA micelle at the phenyl borate-equivalent concentration of 100µM. PtBE-Micelle displayed significantly higher cytotoxicity toward A549R cells with half maximal inhibitory concentration (IC50) of cisplatin-equivalent 0.20µM compared with free cisplatin of 33.15µM and Pt(IV)-loaded PEG-b-poly(ε-caprolactone) micelles of cisplatin-equivalent 0.75µM. PtBE-Micelle can inhibit the growth of A549R xenograft tumors effectively. Accordingly, PEG-b-PBEMA micelles show great potentials as drug delivery nanocarriers for platinum-based chemotherapy toward cisplatin-resistant cancers.


Assuntos
Antineoplásicos/administração & dosagem , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glutationa/metabolismo , Micelas , Polímeros/administração & dosagem , Pró-Fármacos/administração & dosagem , Células A549 , Animais , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Control Release ; 284: 15-25, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-29894709

RESUMO

Tumor hypoxia strikingly restricts photodynamic therapy (PDT) efficacy and limits its clinical applications in cancer therapy. The ideal strategy to address this issue is to develop oxygen-independent PDT systems. Herein, the rationally designed tumor pH-responsive polymeric micelles are devised to realize oxygen-independent combined PDT and photothermal therapy (PTT) under near-infrared light (NIR) irradiation. The triblock copolymer, poly(ethylene glycol)-b-poly(ε-caprolactone)-b-poly(2-(piperidin-1-yl)ethyl methacrylate) (PEG-b-PCL-b- PPEMA), was prepared to co-encapsulate cypate and singlet oxygen donor (diphenylanthracene endoperoxide, DPAE) via self-assembly to obtain the micellar delivery system (C/O@N-Micelle). C/O@N-Micelle showed remarkable tumor accumulation and improved cellular internalization (2.1 times) as the pH value was changed from 7.4 during blood circulation to 6.8 in tumor tissues. The micelles could produce a potent hyperthermia for PTT of cypate under 808 nm NIR irradiation, which simultaneously induced thermal cycloreversion of DPAE generating abundant singlet oxygen for PDT without participation of tumor oxygen. Finally, the photothermally triggered PDT and PTT combination achieved efficient tumor ablation without remarkable systemic toxicity in an oxygen-independent manner. This work represents an efficient strategy for oxygen-independent combined PDT and PTT of cancers under NIR irradiation through co-encapsulation of cypate and DPAE into tumor pH-responsive polymeric micelles.


Assuntos
Antracenos/administração & dosagem , Preparações de Ação Retardada/química , Indóis/administração & dosagem , Lactonas/química , Neoplasias/terapia , Fármacos Fotossensibilizantes/administração & dosagem , Polietilenoglicóis/química , Ácidos Polimetacrílicos/química , Propionatos/administração & dosagem , Animais , Antracenos/uso terapêutico , Linhagem Celular Tumoral , Terapia Combinada/métodos , Sistemas de Liberação de Medicamentos , Feminino , Concentração de Íons de Hidrogênio , Hipertermia Induzida/métodos , Indóis/uso terapêutico , Camundongos Endogâmicos BALB C , Micelas , Neoplasias/metabolismo , Neoplasias/patologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia/métodos , Propionatos/uso terapêutico , Oxigênio Singlete/metabolismo , Hipóxia Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA