Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Clin Oral Investig ; 28(5): 259, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38639763

RESUMEN

OBJECTIVES: Natural enzymes mouthwash has been proposed as salivary substitutes to treat xerostomia. This study aims to evaluate the efficacy of the mouthwash to treat xerostomia. MATERIALS AND METHODS: A double-blind, parallel group randomised control clinical trial involving N = 49 adult participants with xerostomia was carried out. Intervention group received natural enzymes moisturising mouthwash (with active ingredients lactoferrin, lysozyme, lactoperoxidase and glucose oxidase); while control group received benzydamine mouthwash. Mouthwashes were repacked, labelled with specific code, and were given to participants by third-party. Subjects were instructed to rinse with the mouthwash 4 times per day at a specific period, for 2 weeks. Symptoms of xerostomia were assessed using Xerostomia Inventory at day 0 and 14; together with the assessment of Clinical Oral Dryness Score (CODS), and measurement of resting and stimulated salivary flow rate. RESULTS: 48 participants completed the clinical follow-up, and n = 1 had lost of follow-up. From the 48 participants, n = 23 received natural enzymes mouthwash, while n = 25 received benzydamine mouthwash. Intervention group achieved reduction in symptoms of xerostomia from baseline. Intervention group also showed significantly better improvements in the cognitive perception of dry mouth and oromotor function such as chewing, swallowing and speech of the participants; and reduction in waking up at night to drink water (p < 0.05). The CODS and resting salivary flow rate were also significantly improved in intervention group (p < 0.05). CONCLUSION: Use of natural enzymes mouthwash improved signs and symptoms of xerostomia. CLINICAL RELEVANCE: Natural enzymes mouthwash is potentially effective to treat xerostomia, well-tolerated and safe to be used by xerostomia patients. CLINICAL TRIAL REGISTRATION NUMBER: This study was retrospectively registered in ClinicalTrials.gov ID NCT05640362 on 7 December 2022.


Asunto(s)
Bencidamina , Xerostomía , Adulto , Humanos , Antisépticos Bucales/uso terapéutico , Bencidamina/uso terapéutico , Xerostomía/tratamiento farmacológico , Glucosa Oxidasa/uso terapéutico , Deglución
2.
J Mater Chem B ; 11(41): 9798-9839, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37842806

RESUMEN

Nanomaterial-based cancer therapy faces significant limitations due to the complex nature of the tumor microenvironment (TME). Starvation therapy is an emerging therapeutic approach that targets tumor cell metabolism using glucose oxidase (GOx). Importantly, it can provide a material or environmental foundation for other diverse therapeutic methods by manipulating the properties of the TME, such as acidity, hydrogen peroxide (H2O2) levels, and hypoxia degree. In recent years, this cascade strategy has been extensively applied in nanoplatforms for ongoing synergetic therapy and still holds undeniable potential. However, only a few review articles comprehensively elucidate the rational designs of nanoplatforms for synergetic therapeutic regimens revolving around the conception of the cascade strategy. Therefore, this review focuses on innovative cascade strategies for GOx-based synergetic therapy from representative paradigms to state-of-the-art reports to provide an instructive, comprehensive, and insightful reference for readers. Thereafter, we discuss the remaining challenges and offer a critical perspective on the further advancement of GOx-facilitated cancer treatment toward clinical translation.


Asunto(s)
Nanopartículas , Nanoestructuras , Neoplasias , Humanos , Glucosa Oxidasa/uso terapéutico , Glucosa Oxidasa/metabolismo , Peróxido de Hidrógeno/uso terapéutico , Neoplasias/terapia , Nanoestructuras/uso terapéutico , Microambiente Tumoral
3.
Nano Lett ; 23(14): 6610-6618, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37458704

RESUMEN

Diabetic ulcers have received much attention in recent years due to their high incidence and mortality, motivating the scientific community to develop various strategies for such chronic disease treatments. However, the therapeutic outcome of these approaches is highly compromised by invasive bacteria and a severe inflammatory microenvironment. To overcome these dilemmas, microenvironment-responsive self-delivery glucose oxidase@manganese sulfide (GOx@MnS) nanoparticles (NPs) are developed by one-step biomineralization. When they encounter the high glucose level in the ulcer site, GOx particles catalyze glucose to decrease the local pH and trigger the steady release of both manganese ions (Mn2+) and hydrogen sulfide (H2S). Mn2+ reacts with hydrogen peroxide to generate hydroxyl radicals for the elimination of bacterial infection; meanwhile, H2S is able to suppress the inflammatory response and accelerate diabetic wound healing through macrophage polarization. The excellent biocompatibility, strong bactericidal activity, and considerable immunomodulatory effect promise GOx@MnS NPs have great therapeutic potential for diabetic wound treatment.


Asunto(s)
Diabetes Mellitus , Sulfuro de Hidrógeno , Nanopartículas , Humanos , Glucosa Oxidasa/uso terapéutico , Glucosa , Peróxido de Hidrógeno
4.
Adv Mater ; 35(46): e2305512, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37487702

RESUMEN

Simultaneous lactate metabolism inhibition and intracellular acidification (LIIA) is a promising approach for inducing tumor regression by depleting ATP. However, given the limited efficacy of individual metabolic modulators, a combination of various modulators is required for highly efficient LIIA. Herein, a co-delivery system that combines lactate transporter inhibitor, glucose oxidase, and O2 -evolving nanoparticles is proposed. As a vehicle, a facile room-temperature synthetic method for large-pore mesoporous silica nanoparticles (L-MSNs) is developed. O2 -evolving nanoparticles are then conjugated onto L-MSNs, followed by immobilizing the lactate transporter inhibitor and glucose oxidase inside the pores of L-MSNs. To load the lactate transporter inhibitor, which is too small to be directly loaded into the large pores, it is encapsulated in albumin by controlling the albumin conformation before being loaded into L-MSNs. Notably, inhibiting lactate efflux shifts the glucose consumption mechanism from lactate metabolism to glucose oxidase reaction, which eliminates glucose and produces acid. This leads to synergistic LIIA and subsequent ATP depletion in cancer cells. Consequently, L-MSN-based co-delivery of modulators for LIIA shows high anticancer efficacy in several mouse tumor models without toxicity in normal tissues. This study provides new insights into co-delivery of small-molecule drugs, proteins, and nanoparticles for synergistic metabolic modulation in tumors.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Ratones , Glucosa Oxidasa/uso terapéutico , Transportadores de Ácidos Monocarboxílicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Nanopartículas/uso terapéutico , Glucosa , Concentración de Iones de Hidrógeno , Adenosina Trifosfato , Albúminas , Dióxido de Silicio , Porosidad , Sistemas de Liberación de Medicamentos , Portadores de Fármacos/uso terapéutico
5.
Adv Healthc Mater ; 12(27): e2300516, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37285596

RESUMEN

Reactive oxygen species (ROS)-involved tumor therapeutic strategy, chemodynamic therapy (CDT), has attracted extensive research interest in the scientific community. However, the therapeutic effect of CDT is insufficient and unsustainable owing to the limited endogenous H2 O2 level in the tumor microenvironment. Here, peroxidase (POD)-like RuTe2 nanozyme with the immobilization of glucose oxidase (GOx) and allochroic 3,3',5,5'-tetramethylbenzidine (TMB) molecule have been synthesized to construct RuTe2 -GOx-TMB nanoreactors (RGT NRs) as cascade reaction systems for tumor-specific and self-replenishing cancer therapy. GOx in sequential nanocatalysts can effectively deplete glucose in tumor cells. Meanwhile, a sustainable supply of H2 O2 for subsequent Fenton-like reactions catalyzed by RuTe2 nanozyme is achieved in response to the mild acidic tumor microenvironment. Through this cascade reaction, highly toxic hydroxyl radicals (·OH) are produced, which can further oxidize TMB to trigger tumor-specific "turn-on" photothermal therapy (PTT). In addition, PTT and massive ROS can stimulate the tumor immune microenvironment and activate the systematic anti-tumor immune responses, exerting a notable effect on hindering tumor recurrence and metastasis. This study paves a promising paradigm for synergistic starvation therapy, PTT, and CDT cancer therapy with high efficiency.


Asunto(s)
Neoplasias , Humanos , Especies Reactivas de Oxígeno , Neoplasias/tratamiento farmacológico , Glucosa , Glucosa Oxidasa/uso terapéutico , Peroxidasa , Microambiente Tumoral , Peróxido de Hidrógeno , Línea Celular Tumoral
6.
ACS Appl Mater Interfaces ; 14(45): 50583-50591, 2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36322919

RESUMEN

Protein therapy has been considered to be one of the most direct and safe ways to regulate cell function and treat tumors. However, safe and effective intracellular delivery of protein drugs is still a key challenge. Herein, we developed a tannic acid-assisted biomineralization strategy for the encapsulation and intracellular delivery of protein drugs. RNase A and glucose oxidase (GOD) were choose as the protein drug model. RNase A, GOD, TA, and Mn2+ are mixed in one pot to attain RG@MT, and CaCO3 coating is subsequently carried out to construct RG@MT@C through biomineralization. Once RG@MT@C is endocytosed, the acidic environment of the lysosome will dissolve the protective layer of CaCO3 and produce plenty of CO2 to cause lysosome bursting, ensuring the lysosome escape of the RG@MT@C and thus releasing the generated TA-Mn2+, RNase A, and GOD into the cytoplasm. The released substances would activate starvation therapy, chemodynamic therapy, and protein therapy pathways to ensure a high performance of cancer therapy. Due to simple preparation, low toxicity, and controlled release in the tumor microenvironment, we expect it can realize efficient and nondestructive delivery of protein drugs and meet the needs for precise, high performance of synergistically antitumor therapy in biomedical applications.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Taninos/farmacología , Taninos/uso terapéutico , Ribonucleasa Pancreática/uso terapéutico , Preparaciones Farmacéuticas , Biomineralización , Neoplasias/tratamiento farmacológico , Glucosa Oxidasa/uso terapéutico , Línea Celular Tumoral , Microambiente Tumoral
7.
J Control Release ; 352: 1-14, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36241091

RESUMEN

Bacterial biofilms, especially ones caused by multi-drug resistant strains, are increasingly posing a significant threat to human health. Inspired by nature, we report the fabrication of glucose oxidase-loaded iron-phenolic networks that can power the cascade reaction to generate free radicals to eradicate bacterial biofilms. A soft template, sodium deoxycholate, is employed to guarantee glucose oxidase activity during encapsulation, yielding the porous nanocomplexes after removing the template. The porous nature of nanocomplexes, characterized via transmission electron microscopy, N2 adsorption isotherms, and thermogravimetric analysis, facilitates the diffusion of substrates and products during the cascade reaction and protects glucose oxidase from protease attack. Our optimized nanocomplexes (Fe-GA/GOx) could efficiently kill drug-resistant ESKAPE pathogens, including the clinically isolated strains and eradicate their biofilms. In this regard, Fe-GA/GOx could induce over 90% of the biomass of Klebsiella pneumoniae and Staphylococcus aureus biofilms. In the murine peritonitis infection model induced by Staphylococcus aureus and pneumonia model induced by Klebsiella pneumoniae, our Fe-GA/GOx nanocomplexes could efficiently eradicate the bacteria (over 3-log reduction in colony-forming units) and alleviate the inflammatory response without notable side effects on normal tissues. Therefore, our strategy may provide an efficient alternative treatment to combat bacterial biofilms and address the emergence of drug resistance.


Asunto(s)
Hierro , Infecciones Estafilocócicas , Animales , Ratones , Antibacterianos/química , Bacterias , Biopelículas , Glucosa Oxidasa/uso terapéutico , Klebsiella pneumoniae , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus
8.
ACS Appl Mater Interfaces ; 14(37): 42541-42557, 2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36094305

RESUMEN

Nanotechnology has shown a revolution in cancer treatments, including breast cancers. However, there remain some challenges and translational hurdles. Surgery, radiotherapy, and chemotherapy are the primary treatment methods for breast cancer, although drug combinations showed promising results in preclinical studies. Herein we report the development of a smart drug delivery system (DDS) to efficiently treat breast cancer by pyroptosis-starvation-chemotherapeutic combination. Cancer-starvation agent glucose oxidase was chemically attached to synthesized iron oxide nanoparticles which were entrapped inside poly(lactic-co-glycolic acid) along with apoptosis-associated speck-like protein containing a caspase recruitment domain plasmid and paclitaxel (PTX). An emulsion solvent evaporation method was used to prepare the DDS. The surface of the DDS was modified with chitosan to which aptamer was attached to achieve site-specific targeting. Hence, the prepared DDS could be targeted to a tumor site by both external magnet and aptamer to obtain an enhanced accumulation of drugs at the tumor site. The final size of the aptamer-decorated DDS was less than 200 nm, and the encapsulation efficiency of PTX was 76.5 ± 2.5%. Drug release from the developed DDS was much higher at pH 5.5 than at pH 7.4, ensuring the pH sensitivity of the DDS. Due to efficient dual targeting of the DDS, in vitro viability of 4T1 cells was reduced to 12.1 ± 1.6%, whereas the nontargeted group and free PTX group could reduce the viability of cells to 29.2 ± 2.4 and 46.2 ± 1.6%, respectively. Our DDS showed a synergistic effect in vitro and no severe side effects in vivo. This DDS has strong potential to treat various cancers.


Asunto(s)
Neoplasias de la Mama , Quitosano , Nanopartículas , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Quitosano/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Emulsiones , Femenino , Glucosa Oxidasa/uso terapéutico , Humanos , Fenómenos Magnéticos , Nanopartículas/química , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico , Polímeros/química , Piroptosis , Solventes
9.
ACS Nano ; 16(10): 17062-17079, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36153988

RESUMEN

Activated fibroblast-like synovial (FLS) cells are regarded as an important target for rheumatoid arthritis (RA) treatment via starvation therapy mediated by glucose oxidase (GOx). However, the hypoxic RA-FLS environment greatly reduces the oxidation process of glucose and leads to a poor therapeutic effect of the GOx-based starvation therapy. In this work, we designed a hollow mesoporous copper sulfide nanoparticles (CuS NPs)-based smart GOx/atovaquone (ATO) codelivery system (named as V-HAGC) targeting RA-FLS cells to realize a O2-economized dual energy inhibition strategy to solve the limitation of GOx-based starvation therapy. V-HAGC armed with dual multi-stimuli-responsive "doorkeepers" can guard drugs intelligently. Once under the stimulation of photothermal and acidic conditions at the targeted area, the dual intelligent responsive "doors" would orderly open to realize the controllable release of drugs. Besides, the efficacy of V-HAGC would be much improved by the additional chemodynamic therapy (CDT) and photothermal therapy (PTT) stimulated by CuS NPs. Meanwhile, the upregulated H2O2 and acid levels by starvation therapy would promote the Fenton-like reaction of CuS NPs under O2-economized dual energy inhibition, which could enhance the PTT and CDT efficacy as well. In vitro and in vivo evaluations revealed V-HAGC with much improved efficacy of this combination therapy for RA. In general, the smart V-HAGC based on the O2-economized dual energy inhibition strategy combined with enhanced CDT and PTT has the potential to be an alternative methodology in the treatment of RA.


Asunto(s)
Artritis Reumatoide , Nanopartículas , Neoplasias , Humanos , Cobre/farmacología , Cobre/uso terapéutico , Terapia Fototérmica , Glucosa Oxidasa/uso terapéutico , Atovacuona/uso terapéutico , Peróxido de Hidrógeno , Nanopartículas/uso terapéutico , Sulfuros/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Glucosa , Nanotecnología , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral
10.
Adv Sci (Weinh) ; 9(31): e2202332, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36156451

RESUMEN

Notwithstanding immune checkpoint blocking (ICB) therapy has made eminent clinical breakthroughs, overcoming immunologically "cold" tumors remains challenging. Here, a cascade potentiated nanomodulator AuPtAg-GOx is engineered for boosting immune responsiveness. Upon 1064 nm laser irradiation, AuPtAg-mediated mild photothermal therapy (PTT) activates cytotoxic T lymphocytes and reverses the immunogenic "cold" tumor microenvironment. Further, to amplify the thermal sensitivity of tumor cells, glucose oxidase (GOx) is introduced to suppress the production of heat shock proteins, thereby promoting mild photothermal therapy. Complementarily, AuPtAg nanozymes with catalase-like activity can ameliorate tumor hypoxia, significantly improving the GOx activity. As a result, the combination of AuPtAg-GOx with self-augmented photothermal ability and PD-L1 antibody can further escalate the antitumor efficacy. The AuPtAg-GOx-based synergistic starvation therapy, mild PTT, and immunotherapy cascade enhancement therapy strategy can be a favorable tool to effectively kill cancer cells.


Asunto(s)
Glucosa Oxidasa , Terapia Fototérmica , Glucosa Oxidasa/uso terapéutico , Glucosa Oxidasa/metabolismo , Línea Celular Tumoral , Inmunoterapia , Fototerapia
11.
Acta Biomater ; 152: 484-494, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36028197

RESUMEN

Poor tumor specificity is one of the key obstacles for clinical applications of nanotheranostic agents, consequently leading to serious side effects and unsatisfactory therapeutic efficacy. Herein, biomolecule-based nanohybrids (named as Hb-PDA-GOx) with multiple stimuli-responsiveness were designed and fabricated to enhance tumor-specific therapy. The nanohybrids embodied two proteins, i.e., hemoglobin (Hb) and glucose oxidase (GOx), which exhibited cascade catalytic activity selectively within the tumor microenvironment (TME). Specifically, GOx catalyzes the overexpressed glucose into gluconic acid and hydrogen peroxide (H2O2), which not only initiated starvation therapy (ST) through cutting off the nutrition supply for carcinoma cells, but also provided H2O2 for sequential Fenton reaction induced by Hb that generating biotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT). Moreover, localized heat generation from polydopamine (PDA) in the nanohybrids can implement photothermal therapy (PTT) and reinforce the CDT efficacy. Excitingly, effective eradication of solid tumors and significant suppression of metastatic tumors growth were achieved by utilizing Hb-PDA-GOx as a versatile theranostic agent. All these results had been verified by in vitro and/or in vivo experiments. In light of the superior anticancer effects and insignificant systemic toxicity, the as-fabricated biomolecule-based nanohybrids could be employed as a promising agent for tumor-specific therapy. More importantly, the high biocompatibility and biodegradability of the selected biomolecules would facilitate subsequent clinical translation. STATEMENT OF SIGNIFICANCE: (1) A facile one-pot synthesis strategy was proposed to fabricate biomolecule-based tumor theranostic agent with high biocompatibility and biodegradability, which would facilitate subsequent clinical translation; (2) The as-developed theranostic agent was endowed with multiple stimuli-responsiveness for achieving tumor-specific and cascade-enhanced synergistic therapy; (3) The in vivo experiments demonstrated that the as-developed theranostic agent can not only effectively eradicate solid tumors, but also significantly suppress metastatic tumors growth.


Asunto(s)
Nanopartículas , Neoplasias , Glucosa/metabolismo , Glucosa Oxidasa/uso terapéutico , Humanos , Peróxido de Hidrógeno/metabolismo , Nanopartículas/uso terapéutico , Neoplasias/patología , Microambiente Tumoral
12.
J Control Release ; 349: 617-633, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35868357

RESUMEN

A hyaluronic acid (HA)-based one-pot hydrogel reactor with single syringe injection and immediate gelation was developed for starvation therapy (ST), chemodynamic therapy (CDT), ferroptosis, and photothermal therapy (PTT) against breast cancer. A rheologically tuned hydrogel network, composed of HA-phenylboronic acid (HP) and HA-dopamine (HD), was designed by introducing a boronate ester linkage (phenylboronic acid-dopamine interaction) and polydopamine bond (pH control). Ferrocene (Fc)-conjugated HP (Fc-HP) was synthesized to achieve ferroptosis, Fenton reaction-involved toxic hydroxyl radical (•OH) generation, and photothermal ablation in cancer therapy. Glucose oxidase (GOx) was entrapped in the pH-modulated Fc-HP (Fc-HP°)/HD hydrogel network for converting intracellular glucose to H2O2 to enable its own supply. The GOx/Fc combination-installed hydrogel reactor system can provide sustained ST/CDT/PTT functions along with ferroptosis. Injection of Fc-HP°/HD/GOx hydrogel with single-syringe injectability, shear-thinning feature, and self-healing capability offered a slow biodegradation rate and high safety profiles. Peritumorally injected Fc-HP°/HD/GOx hydrogel also efficiently suppressed the growth of breast cancer based on multifunctional therapeutic approaches with reduced dosing frequency. Hyperthermia induced by near-infrared (NIR) laser absorption may amplify the therapeutic effects of free radicals. It is expected that this Fc-HP°/HD/GOx hydrogel system can be applied to local cancer therapy with high efficacy and safety profiles.


Asunto(s)
Neoplasias de la Mama , Hipertermia Inducida , Neoplasias , Ácidos Borónicos , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Dopamina/uso terapéutico , Ésteres/uso terapéutico , Femenino , Compuestos Ferrosos , Glucosa/metabolismo , Glucosa Oxidasa/química , Glucosa Oxidasa/uso terapéutico , Humanos , Ácido Hialurónico/química , Hidrogeles/química , Peróxido de Hidrógeno/metabolismo , Radical Hidroxilo/uso terapéutico , Metalocenos/uso terapéutico , Neoplasias/tratamiento farmacológico
13.
Nat Commun ; 13(1): 2688, 2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35577812

RESUMEN

Inhibited immune response and low levels of delivery restrict starvation cancer therapy efficacy. Here, we report on the co-delivery of glucose oxidase (GOx) and indoleamine 2,3-dioxygenase (IDO) inhibitor 1-methyltryptophan using a metal-organic framework (MOF)-based nanoreactor, showing an amplified release for tumor starvation/oxidation immunotherapy. The nanosystem significantly overcomes the biobarriers associated with tumor penetration and improves the cargo bioavailability owing to the weakly acidic tumor microenvironment-activated charge reversal and size reduction strategy. The nanosystem rapidly disassembles and releases cargoes in response to the intracellular reactive oxygen species (ROS). GOx competitively consumes glucose and generates ROS, further inducing the self-amplifiable MOF disassembly and drug release. The starvation/oxidation combined IDO-blockade immunotherapy not only strengthens the immune response and stimulates the immune memory through the GOx-activated tumor starvation and recruitment of effector T cells, but also effectively relieves the immune tolerance by IDO blocking, remarkably inhibiting the tumor growth and metastasis in vivo.


Asunto(s)
Inmunoterapia , Indolamina-Pirrol 2,3,-Dioxigenasa , Estructuras Metalorgánicas , Nanopartículas , Neoplasias , Línea Celular Tumoral , Glucosa Oxidasa/uso terapéutico , Humanos , Inmunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Estructuras Metalorgánicas/uso terapéutico , Nanotecnología , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno , Microambiente Tumoral
14.
Acta Biomater ; 145: 222-234, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35460908

RESUMEN

Cancer cells rely on glycolysis to support a high proliferation rate. Metformin (Met) is a promising drug for tumor treatment that targets hexokinase 2 (HK2) to block the glycolytic process, thereby further disrupting the metabolism of cancer cells. Herein, an intelligent nanomedicine based on glucose deprivation and glycolysis inhibition is creatively constructed for enhanced cancer synergistic treatment. In brief, Met and glucose oxidase (GOx) was encapsulated into histidine/zeolitic imidazolate framework-8 (His/ZIF-8), which was followed by coating with Arg-Gly-Asp (RGD) peptides to obtain the desired nanomedicine (Met/GOx@His/ZIF-8∼RGD). This smart nanomedicine presents the controllable Met and GOx release behavior in an acidic responsive manner. The liberated Met blocks the glycolysis process via suppressing the activity of HK2 and impairing ATP production, which activates the AMP-activated protein kinase (AMPK) pathway and p53 pathway and damages the Warburg effect, eventually leading to cells apoptosis. And the GOx boosts the glucose shortage for starvation therapy by depleting accumulated glucose. According to in vitro and in vivo assays, the combination of glycolysis inhibition and starvation therapy demonstrates efficient cancer cells growth suppression and superior antitumor properties compared to the Met based or GOx-mediated monotherapy. This work provides an advanced therapeutic strategy via disrupting cellular metabolism against cancer. STATEMENT OF SIGNIFICANCE: The obtained nanomedicine (Met/GOx@His/ZIF-8∼RGD) presents the controllable Met and glucose oxidase (GOx) release behavior in an acidic responsive manner. The liberated Met blocks the glycolysis process via suppressing the activity of HK2 and impairing ATP production, which activates the AMP-activated protein kinase (AMPK) pathway and p53 pathway and damages the Warburg effect, eventually leading to cells apoptosis. And the GOx boosts the glucose shortage for starvation therapy by depleting accumulated glucose. The combination of glycolysis inhibition and starvation therapy demonstrate the efficient suppression of cancer cells growth and the superior antitumor properties when compared to the Met based or GOx-mediated monotherapy.


Asunto(s)
Glucosa Oxidasa , Metformina , Neoplasias , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Quimioterapia/métodos , Glucosa , Glucosa Oxidasa/farmacología , Glucosa Oxidasa/uso terapéutico , Glucólisis/efectos de los fármacos , Humanos , Metformina/farmacología , Metformina/uso terapéutico , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo
15.
ACS Biomater Sci Eng ; 8(5): 1892-1906, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35404565

RESUMEN

Organic near-infrared fluorescent dye mediated photothermal therapy (PTT) and photodynamic therapy (PDT) suffer from heat shock response, since, heat shock proteins (HSPs) are overexpressed and can repair the proteins damaged by PTT and PDT. Starvation therapy by glucose oxide (GOx) can inhibit the heat shock response by limiting the energy supply. However, the delivery of sufficient and active GOx remains a challenge. To solve this problem, we utilize liposomes as drug carriers and prepare GOx loaded liposome (GOx@Lipo) with a high drug loading content (12.0%) and high enzymatic activity. The successful delivery of GOx shows excellent inhibition of HSPs and enhances PTT and PDT. Additionally, we apply the same liposome formulation to load near-infrared dye 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbo cyanine iodide (DiR) and prepare DiR contained liposomes (DiR@Lipo) for PTT and PDT. The liposomal formulation substantially enhances the PTT and PDT properties of DiR as well as the cellular uptake and tumor accumulation. Finally, the combination therapy shows excellent tumor inhibition on 4T1 tumor-bearing mice. Interestingly, we also find that the starvation therapy can efficiently inhibit tumor metastasis, which is probably due to the immunogenic effect. Our work presents a biocompatible and effective carrier for the combination of starvation therapy and phototherapy, emphasizing the importance of auxiliary starvation therapy against tumor metastasis and offering important guidance for clinical PTT and PDT.


Asunto(s)
Neoplasias , Fotoquimioterapia , Animales , Glucosa Oxidasa/uso terapéutico , Liposomas/uso terapéutico , Ratones , Neoplasias/tratamiento farmacológico , Terapia Fototérmica
16.
J Mater Chem B ; 10(18): 3462-3473, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35403639

RESUMEN

Sequential nano-catalytic therapy has emerged as a novel therapeutic modality for cancer treatment as it utilizes the unique tumor microenvironment for selective tumor treatment. This study reports a magnetic nanoparticle to achieve Fenton-like reaction and dual-imaging guidance/monitoring. Natural glucose oxidase (GOx) and superparamagnetic Fe3O4 nanoparticles have been integrated into poly(lactic-co-glycolic acid) (PLGA) to fabricate a sequential nanocatalyst (designated as GOx@PLGA-Fe3O4). This nanocatalyst can functionally deplete glucose in tumor tissues, producing a considerable amount of highly cytotoxic hydroxyl radicals via the sequential Fenton-like reaction, and meanwhile maximizing the potential imaging capability as a contrast agent for magnetic resonance imaging and photoacoustic imaging. By ribonucleic acid sequencing (RNA-seq) technology, GOx@PLGA-Fe3O4 nanoparticles are demonstrated to induce tumor cell death by inhibiting multiple gene regulation pathways involving tumor growth and recurrence. Therefore, this finding provides a novel strategy to achieve promising therapeutic efficacy by the rational design of multifunctional nanoparticles with various features, including magnetic targeting, sequential nano-catalytic therapy, and dual-imaging guidance/monitoring.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Glucosa Oxidasa/uso terapéutico , Humanos , Magnetismo , Nanopartículas/uso terapéutico , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Microambiente Tumoral
17.
ACS Biomater Sci Eng ; 8(4): 1522-1531, 2022 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-35274927

RESUMEN

The emergence of peroxidase (POD)-like nanozyme-derived catalytic therapy has provided a promising choice for reactive oxygen species (ROS)-mediated broad-spectrum antibacterials to replace antibiotics, but it still suffers from limitations of low therapeutic efficiency and unusual addition of unstable H2O2. Considering that the higher blood glucose in diabetic wounds provides much more numerous nutrients for bacterial growth, a cascade nanoenzymatic active material was developed by coating glucose oxidase (GOx) onto POD-like Fe2(MoO4)3 [Fe2(MoO4)3@GOx]. GOx could consume the nutrient of glucose to produce gluconic acid (weakly acidic) and H2O2, which could be subsequently converted into highly oxidative •OH via the catalysis of POD-like Fe2(MoO4)3. Accordingly, the synergistic effect of starvation and ROS-mediated therapy showed significantly efficient antibacterial effect while avoiding the external addition of H2O2 that affects the stability and efficacy of the therapy system. Compared with the bactericidal rates of 46.2-59.404% of GOx or Fe2(MoO4)3 alone on extended-spectrum ß-lactamases producing Escherichia coli and methicillin-resistant Staphylococcus aureus, those of the Fe2(MoO4)3@GOx group are 98.396 and 98.776%, respectively. Animal experiments showed that the as-synthesized Fe2(MoO4)3@GOx could much efficiently promote the recovery of infected wounds in type 2 diabetic mice while showing low cytotoxicity in vivo.


Asunto(s)
Diabetes Mellitus Experimental , Staphylococcus aureus Resistente a Meticilina , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Escherichia coli , Glucosa Oxidasa/farmacología , Glucosa Oxidasa/uso terapéutico , Peróxido de Hidrógeno/farmacología , Ratones , Especies Reactivas de Oxígeno/farmacología
18.
Adv Mater ; 34(18): e2200062, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35243699

RESUMEN

Companion diagnostics (CDx) provides critical information for precision medicine. However, current CDx is mostly limited to in vitro tests, which cannot accurately evaluate the disease progression and treatment response in real time. To overcome this challenge, herein a glucose oxidase (GOx)-engineered conjugated polymer (polyaniline, PANI) nanoplatform (denoted as PANITG) is reported for activatable imaging-based CDx and multistage augmented photothermal/starvation synergistic therapy. PANITG comprises a pH-activatable conjugated polymer as a photothermal convertor and photoacoustic (PA) emitter, a GOx as a cancer starvation inducer as well as a H2 O2 and acid producer, and a H2 O2 -cleavable linker as a "switch" for GOx activity. The in vivo PA imaging and photothermal therapy abilities are activated by acidic tumor microenvironment and self-augmented by the reaction between GOx and glucose. Meanwhile, the photothermal effect will enhance the GOx activity in turn. Such multistage augmentation of the therapeutic effects will facilitate effective cancer management. In addition, the in vivo PA imaging with PANITG reveals the tumor pH level which is correlated to the efficiency of the photothermal therapy and to the catalytic activity of GOx at each stage, enabling real-time activatable CDx.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Glucosa Oxidasa/uso terapéutico , Humanos , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Terapia Fototérmica , Polímeros/uso terapéutico , Microambiente Tumoral
19.
ACS Nano ; 16(2): 2319-2329, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35129953

RESUMEN

A nanoscale therapeutic system with good biocompatibility was facilely fabricated by the coassembly of human serum albumin and glucose oxidase (GOD), where the former was pretreated with metal ions through a chelating agent or the chemotherapeutic prodrug oxaliplatin (Oxa(IV)). Among different chelating metal ions used, Mn2+ ion was selected to produce hydroxyl radical (•OH) efficiently through Fenton-like reaction, while GOD loaded in the system was able to generate a large amount of hydrogen peroxide for promoting efficient conversion into highly toxic •OH. In the meanwhile, the conversion of the Oxa(IV) prodrug into chemotherapeutic Oxa(II) was beneficial for the consumption of glutathione, thereby enhancing the chemodynamic therapy (CDT) efficacy. Based on the combined chemotherapy and CDT, the treatment with this system leads to superior antitumor outcome.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Albúminas , Línea Celular Tumoral , Glucosa Oxidasa/uso terapéutico , Glutatión , Humanos , Neoplasias/tratamiento farmacológico
20.
Biomaterials ; 282: 121384, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35078004

RESUMEN

Biomimetic drug delivery systems offer new opportunities to mimic biological components for enhancing tumor targeting efficiency and promoting therapeutic efficacy. Therefore, in this study, the platelet exosomes and photothermal sensitive liposomes were coalesced and incorporated glucose oxidase (GOx, G) and ferric ammonium (FAC, F) to constitute a laser controlled nanosystem (FG@PEL). Results confirmed that FG@PEL possessed two cascade therapeutic strategies. As the first cascade therapeutic strategy, FG@PEL had achieved cascade targeting effect due to the inheritance of biological property of platelet exosomes to adhere cancer cells and respond on the vascular damage triggered by photothermal effect. Subsequently, GOx was used to oxidize glucose to produce hydrogen peroxide which could be catalyzed by FAC to realize cascade reaction for enhancing chemodynamic therapy (CDT). Meanwhile, photothermal effect could accelerate the reaction rate to further improve the therapeutic effect by producing more hydroxyl radicals. So significant therapeutic effect could be acquired through the synergistic modality of FG@PEL, and the mechanism of mutual promotion among different treatment was also clarified. Hence, this research may provide a promising strategy for curative tumor therapy.


Asunto(s)
Exosomas , Nanopartículas , Neoplasias , Línea Celular Tumoral , Glucosa Oxidasa/uso terapéutico , Humanos , Peróxido de Hidrógeno/uso terapéutico , Neoplasias/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...