Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Macromol Rapid Commun ; 45(11): e2400036, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38453138

RESUMO

Preparation of materials that possess highly strong and tough properties simultaneously is a great challenge. Thermosetting resins as a type of widely used polymeric materials without synergistic strength and toughness limit their applications in some special fields. In this report, an effective strategy to prepare thermosetting resins with synergistic strength and toughness, is presented. In this method, the soft and rigid microspheres with dynamic hemiaminal bonds are fabricated first, followed by hot-pressing to crosslink at the interfaces. Specifically, the rigid or soft microspheres are prepared via precipitation polymerization. After hot-pressing, the resulting rigid-soft blending materials exhibit superior strength and toughness, simultaneously. As compared with the precursor rigid or soft materials, the toughness of the rigid-soft blending films (RSBFs) is improved to 240% and 2100%, respectively, while the strength is comparable to the rigid precursor. As compared with the traditional crushing, blending, and hot-pressing of rigid or soft materials to get the nonuniform materials, the strength and toughness of the RSBFs are improved to 168% and 255%, respectively. This approach holds significant promise for the fabrication of polymer thermosets with a unique combination of strength and toughness.


Assuntos
Polimerização , Resinas Sintéticas/química , Microesferas , Polímeros/química , Temperatura , Teste de Materiais , Propriedades de Superfície , Tamanho da Partícula
2.
Biomacromolecules ; 24(11): 4622-4645, 2023 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-37870458

RESUMO

Polymersomes have been extensively investigated for drug delivery as nanocarriers for two decades due to a series of advantages including high stability under physiological conditions, simultaneous encapsulation of hydrophilic and hydrophobic drugs inside inner cavities and membranes, respectively, and facile adjustment of membrane and surface properties, as well as controlled drug release through incorporation of stimuli-responsive components. Despite these features, polymersome nanocarriers frequently suffer from nontargeting delivery and poor membrane permeability. In recent years, polymersomes have been functionalized for more efficient drug delivery. The surface shells were explored to be modified with diverse active targeting groups to improve disease-targeting delivery. The membrane permeability of the polymersomes was adjusted by incorporation of the stimuli-responsive components for smart controlled transportation of the encapsulated drugs. Therefore, being the polymersome-biointerface, tailorable properties can be introduced by its carefully modulated engineering. This review elaborates on the role of polymersome membranes as a platform to incorporate versatile features. First, we discuss how surface functionalization facilitates the directional journey to the targeting sites toward specific diseases, cells, or intracellular organelles via active targeting. Moreover, recent advances in the past decade related to membrane permeability to control drug release are also summarized. We finally discuss future development to promote polymersomes as in vivo drug delivery nanocarriers.


Assuntos
Sistemas de Liberação de Medicamentos , Polímeros , Polímeros/química , Liberação Controlada de Fármacos , Permeabilidade
3.
Chem Commun (Camb) ; 59(54): 8323-8331, 2023 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-37318285

RESUMO

Radiotherapy as one of the most important cancer treatment modalities has been widely used in the therapy of various cancers. The clinically used radiation (e.g. X-ray) for radiotherapy has the advantages of precise spatiotemporal controllability and deep tissue penetration. However, traditional radiotherapy is frequently limited by the high side effects and tumor hypoxia. The combination of radiotherapy and other cancer treatment modalities may overcome the disadvantages of radiotherapy and improve the final therapeutic efficacy. In recent years, X-ray-activable prodrugs and polymeric nanocarriers have been extensively explored to introduce other treatment modalities in the precise position during radiotherapy, which can reduce the side toxicity of the drugs and improve the combination therapeutic efficacy. In this review, we focus on recent advances in X-ray-activable prodrugs and polymeric nanocarriers to boost X-ray-based multimodal synergistic therapy with reduced toxicity. The design strategies of prodrugs and polymeric nanocarriers are highlighted. Finally, challenges and outlooks of X-ray-activable prodrugs and polymeric nanocarriers are discussed.


Assuntos
Nanopartículas , Neoplasias , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Raios X , Neoplasias/patologia , Hipóxia Tumoral , Terapia Combinada , Portadores de Fármacos/farmacologia
4.
ACS Nano ; 17(10): 9374-9387, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37141569

RESUMO

Stimulator of interferon genes (STING) activation by STING agonists has been recognized as one of the potent and promising immunotherapy strategies. However, the immunosuppressive tumor microenvironment always hinders the therapeutic efficacy of cancer immunotherapy. In this report, we present polymeric metal-organic framework (PMOF) nanoparticles (NPs) for the combination of photodynamic therapy (PDT) and enhanced STING activation to improve the immunotherapeutic efficacy. The PMOF NPs with poly(ethylene glycol) (PEG) shells were obtained via coordination between the block copolymer ligand PEG-b-PABDA consisting of 1,4-bezenedicarboxylic acid-bearing polyacrylamide (PABDA), meso-tetra(carboxyphenyl)porphyrin (TCPP), thioketal diacetic acid, and zirconyl chloride. Subsequently, the STING agonist SR-717 was loaded into the porous structure of PMOF to obtain SR@PMOF NPs which show excellent stability under the physiological conditions. After intravenous injection and tumor accumulation, light irradiation on the tumor sites results in efficient singlet oxygen (1O2) production from TCPP and cellular apoptosis to release fragmented DNA and tumor-associated antigens. Simultaneously, thioketal bonds can be broken by 1O2 to destroy the PMOF structure and rapidly release SR717. SR-717 and PDT synergistically enhance the antitumor immunity via combination photodynamic-immunotherapy due to reversal of the immunosuppressive tumor microenvironment and enhanced endogenous STING activation, which can suppress the growth of the primary and distant tumors efficiently. The oxidation-responsive SR@PMOF NPs represent a promising delivery system of STING agonists and efficient PDT NPs for simultaneous suppression of the primary and metastatic tumors via the rational combination of PDT and enhanced STING activation.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/química , Linhagem Celular Tumoral , Neoplasias/terapia , Nanopartículas/química , Polímeros , Imunoterapia , Microambiente Tumoral
5.
Bioconjug Chem ; 34(2): 377-391, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36716444

RESUMO

The multi-drug resistance (MDR) of cancers is one of the main barriers for the success of diverse chemotherapeutic methods and is responsible for most cancer deaths. Developing efficient approaches to overcome MDR is still highly desirable for efficient chemotherapy of cancers. The delivery of targeted anticancer drugs that can interact with mitochondrial DNA is recognized as an effective strategy to reverse the MDR of cancers due to the relatively weak DNA-repairing capability in the mitochondria. Herein, we report on a polyprodrug that can sequentially target cancer cells and mitochondria using folic acid (FA) and tetraphenylphosphonium (TPP) targeting moieties, respectively. They were conjugated to the terminal groups of the amphiphilic block copolymer prodrugs composed of poly[oligo(ethylene glycol) methyl ether methacrylate] (POEGMA) and copolymerized monomers containing cinnamaldehyde (CNM) and doxorubicin (DOX). After self-assembly into micelles with the suitable size (∼30 nm), which were termed as TF@CNM + DOX, and upon intravenous administration, the micelles can accumulate in tumor tissues. After FA-mediated endocytosis, the endosomal acidity (∼pH 5) can trigger the release of CNM from TF@CNM + DOX micelles, followed by enhanced accumulation into the mitochondria via the TPP target. This promotes the overproduction of reactive oxygen species (ROS), which can subsequently enhance the intracellular oxidative stress and trigger ROS-responsive release of DOX into the mitochondria. TF@CNM + DOX shows great potential to inhibit the growth of DOX-resistant MCF-7 ADR tumors without observable side effects. Therefore, the tumor and mitochondria dual-targeting polyprodrug design represents an ideal strategy to treat MDR tumors through improvement of the intracellular oxidative level and ROS-responsive drug release.


Assuntos
Micelas , Neoplasias , Humanos , Liberação Controlada de Fármacos , Espécies Reativas de Oxigênio/metabolismo , Células MCF-7 , Doxorrubicina , Resistência a Múltiplos Medicamentos , Mitocôndrias/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
6.
ACS Appl Mater Interfaces ; 14(45): 50601-50615, 2022 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-36335599

RESUMO

The unique tumor microenvironment (TME) characteristic of severe hypoxia, overexpressed intracellular glutathione (GSH), and elevated hydrogen peroxide (H2O2) concentration limit the anticancer effect by monotherapy. In this report, glucose oxidase (GOx)-encapsulated mesoporous hollow Co9S8 nanoreactors are constructed with the coverage of polyphenol diblock polymers containing poly(oligo(ethylene glycol) methacrylate) and dopamine moieties containing methacrylate polymeric block, which are termed as GOx@PCoS. After intravenous injection, tumor accumulation, and cellular uptake, GOx@PCoS deplete GSH by Co3+ ions. GOx inside the nanoreactors produce H2O2 via oxidation of glucose to enhance •OH-based chemodynamic therapy (CDT) through the Fenton-like reaction under the catalysis of Co2+. Moreover, Co3+ ions possess catalase activity to catalyze production of O2 from H2O2 to relieve tumor hypoxia. Upon 808 nm laser irradiation, GOx@PCoS exhibit photothermal and photodynamic effects with a high photothermal conversion efficiency (45.06%) and generation capacity of the toxic superoxide anion (•O2-) for photothermal therapy (PTT) and photodynamic therapy (PDT). The synergetic antitumor effects can be realized by GSH depletion, starvation, and combined CDT, PTT, and PDT with enhanced efficacy. Notably, GOx@PCoS can also be used as a magnetic resonance imaging (MRI) contrast agent to monitor the antitumor performance. Thus, GOx@PCoS show great potentials to effectively modulate TME and perform synergistic multimodal therapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Terapia Fototérmica , Peróxido de Hidrogênio , Linhagem Celular Tumoral , Microambiente Tumoral , Glutationa , Glucose Oxidase , Polímeros , Metacrilatos , Nanotecnologia
7.
ACS Nano ; 16(9): 14982-14999, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36017992

RESUMO

Immunotherapeutic efficacy of tumors based on immune checkpoint blockade (ICB) therapy is frequently limited by an immunosuppressive tumor microenvironment and cross-reactivity with normal tissues. Herein, we develop reactive oxygen species (ROS)-responsive nanocomplexes with the function of ROS production for delivery and triggered release of anti-mouse programmed death ligand 1 antibody (αPDL1) and glucose oxidase (GOx). GOx and αPDL1 were complexed with oligomerized (-)-epigallocatechin-3-O-gallate (OEGCG), which was followed by chelation with Fe3+ and coverage of the ROS-responsive block copolymer, POEGMA-b-PTKDOPA, consisting of poly(oligo(ethylene glycol)methacrylate) (POEGMA) and the block with thioketal bond-linked dopamine moieties (PTKDOPA) as the side chains. After intravenous injection, the nanocomplexes show prolonged circulation in the bloodstream with a half-life of 8.72 h and efficient tumor accumulation. At the tumor sites, GOx inside the nanocomplexes can produce H2O2 via oxidation of glucose for Fenton reaction to generate hydroxyl radicals (•OH) which further trigger the release of the protein cargos through ROS-responsive cleavage of thioketal bonds. The released GOx improves the production efficiency of •OH to kill cancer cells for release of tumor-associated antigens via chemodynamic therapy (CDT). The enhanced immunogenic cell death (ICD) can activate the immunosuppressive tumor microenvironment and improve the immunotherapy effect of the released αPDL1, which significantly suppresses primary and metastatic tumors. Thus, the nanocomplexes with Fenton reaction-triggered protein release show great potentials to improve the immunotherapeutic efficacy of ICB via combination with CDT.


Assuntos
Glucose Oxidase , Neoplasias , Antígeno B7-H1 , Linhagem Celular Tumoral , Dopamina , Glucose , Humanos , Peróxido de Hidrogênio , Inibidores de Checkpoint Imunológico , Imunoterapia , Neoplasias/tratamento farmacológico , Polietilenoglicóis , Espécies Reativas de Oxigênio , Microambiente Tumoral
8.
Small ; 18(37): e2202369, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35971160

RESUMO

Photodynamic therapy (PDT) of cancers is seriously restricted by tumor hypoxia. In addition to the intrinsic hypoxic microenvironment, continuous photoirradiation further aggravates intratumoral hypoxia, thereby reducing the PDT effect significantly. Oxygen-independent PDT is recognized as an efficient approach to overcome this issue. Herein, singlet oxygen (1 O2 )-stored covalent organic framework (COF) nanoparticles loading the near-infrared (NIR) dye cypate, which realize oxygen-independent 1 O2 production for concurrent photothermal therapy (PTT) and PDT under NIR irradiation, are presented. The cypate-loading COF nanoparticles are prepared by using the photosensitizers and 1 O2 -stored molecules via formation of Schiff base bonds, followed by coverage of poly(vinyl pyrrolidone). The COF nanoparticles significantly improve the photostability and photothermal conversion efficiency of cypate by protecting them from photodegradation under NIR irradiation. Upon 660 nm laser irradiation, 1 O2 is produced by the photosensitizer motifs and is successfully stored by the 1 O2 -stored moieties. After intravenous injection and tumor accumulation, the COF nanoparticles can generate heat quickly upon 808 nm irradiation which induces the efficient release of the stored 1 O2 to ablate tumors via O2 -independent concurrent PTT/PDT. Accordingly, the COF nanocarriers of 1 O2 provide a paradigm to develop O2 -independent concurrent PTT/PDT for precise cancer treatment upon NIR irradiation.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Fotoquimioterapia , Linhagem Celular Tumoral , Humanos , Hipóxia/tratamento farmacológico , Indóis , Estruturas Metalorgânicas/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Oxigênio/química , Fármacos Fotossensibilizantes/química , Terapia Fototérmica , Propionatos , Oxigênio Singlete , Microambiente Tumoral
9.
ACS Macro Lett ; 11(4): 543-548, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35575322

RESUMO

Nanofibers self-assembled from peptides have attracted much attention to inhibit cancer cells. However, there are still some disadvantages, including high concentration for self-assembly and incapability to load drugs, which limit their applications. In this report, we rationally integrate self-assembled peptides, glutathione-sensitive disulfide bonds, and mitochondrial targeting moieties into the amphiphilic block copolymer to construct the nanocarriers, which can be used to load anticancer drug doxorubicin (DOX). After cellular internalization, the nanocarriers can reassemble from micelles to nanofibers under the trigger by glutathione and locate in mitochondria. The released DOX and nanofibers induce mitochondrial dysfunction and activate the apoptosis pathway to synergistically inhibit tumor cells. This organelle-specific drug delivery system with reassembly capability from micelles to nanofibers shows great potential for effectively killing cancer cells.


Assuntos
Micelas , Nanofibras , Doxorrubicina/farmacologia , Glutationa/farmacologia , Mitocôndrias , Peptídeos , Polímeros/química
10.
J Control Release ; 339: 418-429, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34662586

RESUMO

Combination chemo-immunotherapy of cancers has attracted great attention due to its significant synergistic antitumor effect. The response rates and therapeutic efficacy of immunotherapy can be enhanced significantly after proper combination with chemotherapy. However, chemo-immunotherapy is frequently limited by severe immune-related adverse events and systemic side toxicity. In this report, efficient nanofactory-directed enzyme prodrug chemo-immunotherapy is demonstrated based on enzyme-loaded tumor-dilatable polymersomes with optimized membrane cross-linking density. Upon intravenous injection of the nanofactories, they can passively accumulate at the tumor site. The tumor pH-responsive nanofactories can swell from ~100 nm to ~200 nm under the trigger of tumor acidity, leading to prolonged retention of up to one week inside tumor tissues. Simultaneously, the membrane permeability of the nanofactories has improved significantly, which allows hydrophilic small molecules to pass across the membranes while keeping the enzymes in the inner cavities. Subsequently, the non-toxic prodrug mixtures of chemo-immunotherapy are administrated three times within 6 days, which are in situ activated by the nanofactories selectively at tumor sites. Activated chemotherapeutic drugs kill cancer cells and generate tumor-associated antigens to promote the maturation of dendritic cells. Activated indoleamine 2, 3-dioxygenase 1 inhibitors reverse the immunosuppressive tumor microenvironment. Finally, primary tumors can be effectively suppressed while causing minimal systemic toxicity. The distant tumors that are established after treatment can also be inhibited completely via activation of antitumor immunity in mice. Thus, the tumor-dilatable polymersome nanofactories with long-term intratumoral retention offer a promising paradigm for enhanced enzyme prodrug chemo-immunotherapy.


Assuntos
Neoplasias , Pró-Fármacos , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/uso terapêutico , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Microambiente Tumoral
11.
Biomacromolecules ; 22(11): 4857-4870, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34689560

RESUMO

The hypoxia environment inside tumors is tightly associated with tumor growth, metastasis, and drug resistance. However, the heterogonous distribution of hypoxic areas limits the efficacy of hypoxia-activatable drug delivery systems. Herein, we report the hypoxia-activable block copolymer polyprodrugs, which are composed of poly(ethylene glycol) (PEG) and copolymerized segments of ortho-nitrobenzyl-linked camptothecin (CPT) methacrylate and 2-(piperidin-1-yl)ethyl methacrylate (PEMA) monomers. After self-assembly in aqueous solution, indocyanine green (ICG) photosensitizers were encapsulated to formulate ICG-loaded micellar nanoparticles (ICG@CPTNB) for near-infrared (NIR) light-boosted photodynamic therapy (PDT), tumor hypoxia aggravation, and responsive drug activation. Through intravenous injection and prolonged blood circulation, the nanoparticles can accumulate into tumor efficiently. Tumor acidity-triggered charge transition of PEMA units remarkably promotes cellular internalization of the nanoparticles. Upon exposure to NIR laser irradiation, ICG inside the nanoparticles produced reactive oxygen species (ROS) along with local hypothermia. Simultaneously, the oxygen consumption during ROS production aggravated the intratumoral hypoxia, which amplified hypoxia-responsive self-immolative CPT release from the nanoparticles. The combined photodynamic chemotherapy using hypoxia-responsive polyprodrug nanoparticles, ICG@CPTNB, overcomes the limitations of single therapy of hypoxia-activable prodrugs or PDT, which remarkably improves the efficiency of tumor growth suppression.


Assuntos
Fotoquimioterapia , Sistemas de Liberação de Medicamentos , Humanos , Hipóxia/tratamento farmacológico , Raios Infravermelhos , Fármacos Fotossensibilizantes/uso terapêutico
12.
J Control Release ; 339: 130-142, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34560158

RESUMO

The inherent hypoxic microenvironment of solid tumors has an important influence on tumor growth, distant metastasis, and invasiveness. The heterogeneous distribution of hypoxic regions inside tumors limits the therapeutic efficacy of O2-assisted therapeutic strategy (e.g. photodynamic therapy (PDT)). On the other hand, the hypoxia-activable prodrugs cannot work effectively in the regions with enough O2 concentration. To address the issues, we prepare a block copolymer polyprodrug consisting of polyethylene glycol (PEG) and copolymerized segments of nitroimidazole-linked camptothecin (CPT) methacrylate and 5,10,15,20-tetraphenylporphyrin (TPP)-containing methacrylate monomers for complementary photodynamic-chemotherapy. The polyprodrug can self-assemble into polymeric micelles in aqueous solution with suitable size and high stability. After intravenous injection, the polyprodrug micelles show tumor accumulation. Followed by light irradiation (650 nm) at tumor sites, TPP moieties induce singlet oxygen (1O2) production in the oxygen-rich area to exert PDT and cause transformation of the oxygen-rich areas into hypoxia. Simultaneously, in the hypoxic areas, the hypoxia-responsive polyprodrugs can be activated to release free CPT due to the cleavage of nitroimidazole linkages. The polyprodrug micelles with the segments for PDT and hypoxia-activable CPT efficiently suppress the growth of HeLa tumors. The well-defined polyprodrug amphiphiles offer an effective strategy to overcome the disadvantages of single treatment of PDT or hypoxia-responsive prodrugs for complementary photodynamic-chemotherapy of cancers.


Assuntos
Fotoquimioterapia , Pró-Fármacos , Linhagem Celular Tumoral , Humanos , Hipóxia/tratamento farmacológico , Micelas , Fármacos Fotossensibilizantes/uso terapêutico
13.
Macromol Biosci ; 21(8): e2100133, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34117839

RESUMO

Stimulator of interferon genes (STING) are located in the endoplasmic reticulum of cells, which have been demonstrated to show considerable potentials to achieve efficient antitumor immunity by inducing various pro-inflammatory cytokines and chemokines, such as type I interferons. A variety of STING agonists have been prepared for STING activation, and many of them have been promoted to preclinical trials or clinical applications for the immunotherapy of cancers. However, the intrinsic disadvantages of the small molecule STING agonists can limit the in vivo application and final therapeutic efficacy due to low bioavailability of targeting tissues. Moreover, a cascade of physiological barriers for in vivo STING activation also limit the accumulation of STING agonists in targeting tissues. Drug delivery systems play an important role to improve the STING activation efficiency. In recent years, a variety of nanoparticle-mediated STING agonist delivery systems have been engineered and exploited to address the challenges related to the in vivo STING activation, including liposomes, polymeric micelles, polymersomes, and so on. In this review article, the progresses concerning STING agonists and related delivery systems in recent years will be summarized and discussed.


Assuntos
Nanopartículas , Neoplasias , Sistemas de Liberação de Medicamentos , Humanos , Imunoterapia , Proteínas de Membrana , Neoplasias/tratamento farmacológico
14.
J Control Release ; 333: 500-510, 2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33848558

RESUMO

Chemodynamic therapy (CDT) has been proposed to convert tumoral H2O2 into toxic hydroxyl radicals (OH) via Fenton or Fenton-like reactions for antitumor efficacy, which is frequently limited by low H2O2 concentrations or lack of enough metal ions inside tumor tissues. In this report, we present ferrocene-containing responsive polymersome nanoreactors via loading glucose oxidase (GOD) and hypoxia-activable prodrug tirapazamine (TPZ) in the inner aqueous cavities. After intravenous injection, the polymersome nanoreactors with the optimized nanoparticle size of ~100 nm and poly(ethylene glycol) corona facilitate tumor accumulation. The tumor acidic microenvironment can trigger the permeability of the polymersome membranes to activate the nanoreactors and release the loaded TPZ prodrugs. Tumor oxygen and glucose can enter the polymersome nanoreactors and are transformed into H2O2 under the catalysis of GOD, which are further converted into OH via Fenton reaction under catalysis of ferrocene moieties. The oxygen consumption can aggravate tumor hypoxia to activate hypoxia-responsive TPZ prodrugs which can produce benzotriazinyl (BTZ) radicals and OH. All the produced radicals synergistically kill tumor cells via the amplified CDT and suppress the tumor growth efficiently. Thus, the ferrocene-containing responsive polymersome nanoreactors loading GOD and TPZ represent a potent nanoplatform to exert amplified CDT for improved anticancer efficacy.


Assuntos
Neoplasias , Pró-Fármacos , Linhagem Celular Tumoral , Humanos , Peróxido de Hidrogênio , Metalocenos , Nanotecnologia , Microambiente Tumoral
15.
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
16.
Chemosphere ; 268: 129391, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33360138

RESUMO

The anaerobic fermentation (AF) of waste activated sludge (WAS) with an electrochemical pretreatment (EPT) was investigated to determine its correlation with the release of phosphorus and the disintegration of WAS. The sludge was pretreated by holding under 4.5 V for 60 min, followed by AF for 9 days. Untreated sludge was used as the control group (no-EPT). Results showed that, with pretreatment, the total dissolved P (TDP), orthophosphate (PO43--P) and organic P (OP) reached the maximum values of 7.30 mg/L, 4.77 mg/L and 2.35 mg/L on day 8, respectively, which were approximately 5.3, 9.2 and 2.7 times greater than that in the control group. The analysis of soluble chemical oxygen demand (SCOD), protein and polysaccharides showed that the EPT promoted the disintegration of sludge, thereby enhancing the P release. The SCOD reached 1625 mg/L on day 6 in pretreatment experiment, which was about 9.8 times greater than that in control group. Additionally, the EPT contributed to fewer metal ions in sludge supernatant. This mechanism might have been due to the anions accumulating in the supernatant from the greater degree of sludge collapse after EPT, which caused the released metal ions to combine with anions to form insoluble compounds. In conclusion, EPT could be a promising method for the dissolution of sludge and the recovery of phosphorus from WAS under AF. Besides, the economic benefit evaluation showed the potential value of EPT for P recovery.


Assuntos
Fósforo , Esgotos , Anaerobiose , Análise da Demanda Biológica de Oxigênio , Fermentação , Esgotos/análise , Eliminação de Resíduos Líquidos
17.
Chemosphere ; 259: 127403, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32603963

RESUMO

The ability of thiol-modified humic acids (HAs) to release arsenic in tailings soil after being modified with different sulfur-containing reagents were significantly improved. The structure and physicochemical properties of humic acid (HA) before and after thiol-modification were characterized. The 3-MPTS-HA treated with 3-mercaptopropyltrimethoxysilane (3-MPTS) effectively improved the mobility of arsenic, and its reducing ability was increased from 2 mmol g-1 to 3.54 mmol g-1. The S content of humic acids were also significantly increased after treatment with sulfur-containing reagents, in which the oxygen-containing functional group (e.g., C = O, C-O) on the surface of HA may be the active sites for binding with sulfur-containing reagents. It was found in the XPS spectrum that because the thiol group is easily oxidized, there are many S forms in thiol-modified HA. The -SH content in Na2S·9H2O-HA, l (+)-Cysteine-HA (Cys-HA), thioglycolic acid (TGA-HA) and 3-MPTS-HA was determined by fluorescence method to be 13.9, 78.45, 90.34, and 192.29 µmol g-1, respectively. The study demonstrated that surface thiol modification can increase the abundance of thiol in HA and enhance reactivity, which will further promote the application of HA in the treatment of heavy metal contaminated tailing soil.


Assuntos
Arsênio/química , Poluentes do Solo/química , Substâncias Húmicas/análise , Metais Pesados/química , Solo/química , Poluentes do Solo/análise , Compostos de Sulfidrila
18.
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
19.
Biomaterials ; 195: 63-74, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30612064

RESUMO

In tumor tissues, reactive oxygen species (ROS) level is significantly higher than that in normal tissues, which has been frequently explored as the specific stimulus to trigger drug release. However, the low intrinsic ROS concentration and heterogeneous distribution in tumor tissues hinder the applications as the stimulus for drug delivery. Herein, we developed integrated nanoparticles to remold tumor microenvironment via specific amplification of the tumor oxidative stress and simultaneously realize ROS-responsive drug release. The amphiphilic block copolymer prodrugs composed of poly(ethylene glycol) and polymerized methacrylate monomer containing thioketal-linked camptothecin (CPT) were synthesized and self-assembled to form core-shell micelles for encapsulation of ß-lapachone (Lapa@NPs). After tumor accumulation and internalization into tumor cells post systemic administration of Lapa@NPs, Lapa can selectively induce remarkable ROS level increase via the catalysis of NAD(P)H: quinone oxidoreductase-1 (NQO1) enzyme overexpressed in cancer cells. Subsequently, enhanced ROS concentration would trigger the cleavage of thioketal linkers to release drug. The released CPT together with high ROS level achieved a synergistic therapy to suppress tumor growth. Moreover, Lapa@NPs exhibited superior biosafety due to the tumor-specific activation of the cascade reaction. Accordingly, Lapa@NPs represent a novel polymer prodrug design and drug release strategy via tumor-specific oxidative stress amplification and subsequent ROS-responsive drug release.


Assuntos
Nanopartículas/química , Polímeros/química , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Espécies Reativas de Oxigênio/metabolismo , Animais , Liberação Controlada de Fármacos , Humanos , Estresse Oxidativo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...