Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Bio Mater ; 7(5): 3306-3315, 2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38634490

RESUMEN

Photodynamic therapy (PDT) and ferroptosis show significant potential in tumor treatment. However, their therapeutic efficacy is often hindered by the oxygen-deficient tumor microenvironment and the challenges associated with efficient intracellular drug delivery into tumor cells. Toward this end, this work synthesized perfluorocarbon (PFC)-modified Pluronic F127 (PFC-F127), and then exploits it as a carrier for codelivery of photosensitizer Chlorin e6 (Ce6) and the ferroptosis promoter sorafenib (Sor), yielding an oxygen self-supplying nanoplatform denoted as Ce6-Sor@PFC-F127. The PFCs on the surface of the micelle play a crucial role in efficiently solubilizing and delivering oxygen as well as increasing the hydrophobicity of the micelle surface, giving rise to enhanced endocytosis by cancer cells. The incorporation of an oxygen-carrying moiety into the micelles enhances the therapeutic impact of PDT and ferroptosis, leading to amplified endocytosis and cytotoxicity of tumor cells. Hypotonic saline technology was developed to enhance the cargo encapsulation efficiency. Notably, in a murine tumor model, Ce6-Sor@PFC-F127 effectively inhibited tumor growth through the combined use of oxygen-enhanced PDT and ferroptosis. Taken together, this work underscores the promising potential of Ce6-Sor@PFC-F127 as a multifunctional therapeutic nanoplatform for the codelivery of multiple cargos such as oxygen, photosensitizers, and ferroptosis inducers.


Asunto(s)
Antineoplásicos , Clorofilidas , Ensayos de Selección de Medicamentos Antitumorales , Ferroptosis , Fluorocarburos , Micelas , Oxígeno , Fotoquimioterapia , Fármacos Fotosensibilizantes , Ferroptosis/efectos de los fármacos , Fluorocarburos/química , Fluorocarburos/farmacología , Animales , Ratones , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/síntesis química , Humanos , Oxígeno/química , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Ensayo de Materiales , Tamaño de la Partícula , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/síntesis química , Porfirinas/química , Porfirinas/farmacología , Supervivencia Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratones Endogámicos BALB C , Sorafenib/química , Sorafenib/farmacología , Sorafenib/administración & dosificación , Poloxámero/química , Línea Celular Tumoral , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Experimentales/metabolismo , Estructura Molecular
2.
Adv Healthc Mater ; 13(9): e2303305, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38277491

RESUMEN

Nanomedicine in combination with immunotherapy has shown great potential in the cancer treatment, but phototherapeutic nanomaterials that specifically activate the immunopharmacological effects in deep tumors have rarely been developed due to limited laser penetration depth and tumor immune microenvironment. Herein, this work reports a newly synthesized semiconducting polymer (SP) grafted with imiquimod R837 and indoxmid encapsulated micelle (SPRIN-micelle) with strong absorption in the second near infrared window (NIR-II) that can relieve tumor immunosuppression and enhance the photothermal immunotherapy and catabolic modulation on tumors. Immune agonists (Imiquimod R837) and immunometabolic modulators (indoxmid) are covalently attached to NIR-II SP sensors via a glutathione (GSH) responsive self-immolation linker and then loaded into Pluronic F127 (F127) micelles by a temperature-sensitive critical micelle concentration (CMC)-switching method. Using this method, photothermal effect of SPRIN-micelles in deep-seated tumors can be activated, leading to effective tumor ablation and immunogenic cell death (ICD). Meanwhile, imiquimod and indoxmid are tracelessly released in response to the tumor microenvironment, resulting in dendritic cell (DC) maturation by imiquimod R837 and inhibition of both indoleamine 2,3-dioxygenase (IDO) activity and Treg cell expression by indoxmid. Ultimately, cytotoxic T-lymphocyte infiltration and tumor metastasis inhibition in deep solid tumors (9 mm) are achieved. In summary, this work demonstrates a new strategy for the combination of photothermal immunotherapy and metabolic modulation by developing a dual functional polymer system including activable SP and temperature-sensitive F127 for the treatment of deep solid tumors.


Asunto(s)
Nanopartículas , Neoplasias , Polietilenos , Polipropilenos , Humanos , Imiquimod/farmacología , Polímeros/farmacología , Micelas , Fototerapia/métodos , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Línea Celular Tumoral , Microambiente Tumoral
3.
J Control Release ; 357: 210-221, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36972864

RESUMEN

CRISPR-Cas9 is a central focus of the emerging field of gene editing and photodynamic therapy (PDT) is a clinical-stage ablation modality combining photosensitizers with light irradiation. But metal coordination biomaterials for the applications of both have rarely been investigated. Herein, Chlorin-e6 (Ce6) Manganese (Mn) coordination micelles loaded with Cas9, termed Ce6-Mn-Cas9, were developed for augmented combination anti-cancer treatment. Manganese played multiple roles to facilitate Cas9 and single guide RNA (sgRNA) ribonucleoprotein (RNP) delivery, Fenton-like effect, and enhanced endonuclease activity of RNP. Histidine (His)-tagged RNP could be coordinated to Ce6 encapsulated in Pluronic F127 (F127) micelles by simple admixture. Triggered by ATP and endolysosomal acidic pH, Ce6-Mn-Cas9 released Cas9 without altering protein structure or function. Dual guide RNAs were designed to target the antioxidant regulator MTH1 and the DNA repair protein APE1, resulting in increased oxygen and enhanced PDT effect. In a murine tumor model, Ce6-Mn-Cas9 inhibited tumor growth with the combination therapy of PDT and gene editing. Taken together, Ce6-Mn-Cas9 represents a new biomaterial with a high degree of versatility to enable photo- and gene-therapy approaches.


Asunto(s)
Neoplasias , Fotoquimioterapia , Porfirinas , Humanos , Animales , Ratones , Fotoquimioterapia/métodos , Micelas , Manganeso , Edición Génica , Fototerapia , Fármacos Fotosensibilizantes/química , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Porfirinas/química , Línea Celular Tumoral
4.
Small ; 19(17): e2206981, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36693779

RESUMEN

CRISPR/Cas9-based gene therapy and photodynamic therapy both show promise for cancer treatment but still have their drawbacks limited by tumor microenvironment and long treatment duration. Herein, CRISPR/Cas9 genome editing and photodynamic strategy for a synergistic anti-tumor therapeutic modality is merged. Chlorophyll (Chl) extracted from natural green vegetables is encapsulated in Pluronic F127 (F127) micelles and Histidine-tagged Cas9 can be effectively chelated onto micelles via metal coordination by simple incubation, affording Cas9-Chl@F127 micelles. Mg2+ acts as an enzyme cofactor to correlatively enhance Cas9 gene-editing activity. Upon laser irradiation, Chl as an effective photosensitizer generates reactive oxygen species (ROS) to kill tumor cells. Meanwhile, CRISPR/Cas9, mediated by dual deliberately designed gRNAs of APE1 and NRF2, can reprogram the tumor microenvironment by increasing the intracellular oxygen accumulation and impairing the oxidative defense system of tumor cells. Cas9-Chl@F127 micelles can responsively release Cas9 in the presence of abundant ATP or low pH in tumor cells. In a murine tumor model, Cas9-Chl@F127 complexed with dual gRNAs including APE1 and NRF2 significantly inhibits the tumor growth. Taken together, Cas9-Chl@F127 micelles, representing the first Chl-based green biomaterial for the delivery of Cas9, show great promise for the synergistic anti-tumor treatment by PDT and gene editing.


Asunto(s)
Neoplasias , Fotoquimioterapia , Ratones , Animales , Micelas , Edición Génica , Clorofila , Sistemas CRISPR-Cas/genética , Factor 2 Relacionado con NF-E2 , Neoplasias/genética , Neoplasias/terapia
5.
Bioconjug Chem ; 33(10): 1944-1952, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36191256

RESUMEN

In the tumor microenvironment, there exist microorganisms that metabolize anticancer drugs, leading to chemotherapy failure. To solve this problem, herein, we develop antibiotic and anticancer drug co-delivery micelles, termed colistin crosslinked gemcitabine micelle (CCGM). A self-immolative linker enables colistin and gemcitabine to be released on demand without affecting their antibacterial and anticancer effects. Once CCGM is delivered to the tumor microenvironment, intracellular glutathione triggers the release of colistin and gemcitabine, inhibiting the growth of microbes in the tumor, thus eliminating the microbe-induced drug resistance of tumor.


Asunto(s)
Antineoplásicos , Micelas , Colistina/farmacología , Resistencia a Antineoplásicos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Glutatión , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Línea Celular Tumoral , Gemcitabina
6.
J Biomed Mater Res A ; 110(9): 1590-1598, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35593460

RESUMEN

Colistin is a potent antibiotic but its severe side effects including nephrotoxicity and neurotoxicity are the roadblock for their wide use in clinics. To solve this problem, we synthesized a new prodrug, mannose-maltose-colistin conjugate, termed MMCC that can reversibly mask the five amines of colistin that are primarily responsible for the toxicity. The deliberated design of disulfide-based self-immolative linker warranted the reversibly release of the pristine amines of colistin on demand without sacrificing antimicrobial efficacy. Once MMCC was delivered in cells, reducing agents cleaves the disulfide bond and release the pristine amines. The targeting ligands of maltose and mannose were grafted on colistin conjugate for targeting delivery of colistin to bacteria and macrophages, respectively. Taken together, MMCC as a new class of antimicrobial biomaterials, demonstrates its great potential for the treatment of intracellular bacterial infections.


Asunto(s)
Infecciones Bacterianas , Profármacos , Aminas , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones Bacterianas/tratamiento farmacológico , Colistina/química , Colistina/uso terapéutico , Disulfuros , Humanos , Maltosa , Manosa , Profármacos/farmacología , Profármacos/uso terapéutico
7.
Adv Healthc Mater ; 11(10): e2102365, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34989166

RESUMEN

CRISPR-Cas9 as a powerful gene-editing tool has tremendous potential for the treatment of genetic diseases. Herein, a new mesoporous nanoflower (NF)-like delivery nanoplatform termed Cas9-NF is reported by crosslinking Cas9 and polymeric micelles that enables efficient intracellular delivery and controlled release of Cas9 in response to reductive microenvironment in tumor cells. The flower morphology is flexibly tunable by the protein concentration and different types of crosslinkers. Cas9 protein, embedded between polymeric micelles and protected by Cas9-NF, remains stable even under extreme pH conditions. Responsive cleavage of crosslinkers in tumor cells, leads to the traceless release of Cas9 for efficient gene knockout in nucleus. This crosslinked nanoparticle exhibits excellent capability of downregulating oncogene expression and inhibiting tumor growth in a murine tumor model. Taken together, these findings pave a new pathway toward the application of the protein-micelle crosslinked nanoflower for protein delivery, which warrants further investigations for gene regulation and cancer treatment.


Asunto(s)
Edición Génica , Nanopartículas , Animales , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Ratones , Micelas , Polímeros/metabolismo
8.
ACS Appl Mater Interfaces ; 14(2): 2510-2521, 2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-34986639

RESUMEN

The development of nanocarriers capable of codelivering antigens and immune-activating adjuvants is an emerging area of research and is relevant for cancer vaccines that target induction of antigen-specific CD8+ T-cell responses. Here, we report a system for delivery of short peptide antigens to dendritic cells for strong cellular immune responses, based on block copolymers chemically modified with a hydrophobic and self-immolative linker. After modification, micelles effectively and reversibly capture antigens and adjuvants via a covalent bond within several minutes in an aqueous solution. After uptake in antigen presenting cells, the polymer disulfide bond is cleaved by intracellular glutathione, leading to release of pristine antigens, along with the upregulated expression of costimulatory molecules. The induced antigen-specific CD8+ T cells have strong tumor cell killing efficacy in the murine B16OVA and human papilloma virus-E6/E7 subcutaneous and lung metastasis tumor models. In addition, delivery to lymph nodes can be imaged to visualize vaccine trafficking. Taken together, multifunctional self-immolative micelles represent a versatile class of a vaccine delivery system for the generation of a cellular immune response that warrants further exploration as a component of cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Inmunoterapia , Neoplasias Pulmonares/terapia , Adyuvantes Inmunológicos , Animales , Humanos , Neoplasias Pulmonares/inmunología , Ensayo de Materiales , Ratones , Micelas , Vacunación
9.
J Control Release ; 341: 329-340, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34843813

RESUMEN

Effective delivery of antimicrobial agents to intracellular pathogens represents a major bottleneck for a wide variety of infectious diseases. To address this, we developed SIR-micelles(+), as a new delivery vehicle comprising antibiotic-loaded micelles with rapid self-immolation within cells for targeted delivery to macrophages, where most intracellular bacterial reside. After phagocytosis, SIR-micelles(+) rapidly release the pristine antibiotic after the cleavage of the disulfide bonds by intracellular reducing agents such as glutathione (GSH). Colistin, a hydrophilic and potent "last-resort" antibiotic used for the treatment of drug-resistant bacterial infection, was encapsulated in SIR-micelles with 40% yield and good short-term storage stability. Hydrophobic moieties and mannose ligands in SIR-micelles(+) enhanced the delivery of colistin into macrophages. The traceless and thiol-responsive release of colistin effectively eliminated intracellular Escherichia coli within twenty minutes. In a murine pneumonia model, SIR-micelles(+) significantly reduced bacterial lung burden of multidrug-resistant Klebsiella pneumoniae. Furthermore, SIR-micelles(+) improved the survival rate and reduced the bacterial burden of organs infected by intracellular bacteria transferred from donor mice. Using this formulation approach, the nephrotoxicity and neurotoxicity induced by antibiotic were reduced by about 5- 15 fold. Thus, SIR-micelles(+) represent a new class of material that can be used for targeting treatment of intracellular and drug-resistant pathogens.


Asunto(s)
Antibacterianos , Micelas , Animales , Colistina , Escherichia coli , Glutatión/química , Ratones
10.
Small ; 18(6): e2104132, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34850550

RESUMEN

Photoacoustic imaging (PA) in the second near infrared (NIR-II) window presents key advantages for deep tissue imaging owing to reduced light scattering and low background signal from biological structures. Here, a thiadiazoloquinoxaline-based semiconducting polymer (SP) with strong absorption in the NIR-II region is reported. After encapsulation of SP in Pluronic F127 (F127) followed by removal of excess surfactant, a dual functional polymer system named surfactant-stripped semiconductor polymeric micelles (SSS-micelles) are generated with water solubility, storage stability, and high photothermal conversion efficiency, permitting tumor theranostics in a mouse model. SSS-micelles have a wideband absorption in the NIR-II window, allowing for the PA imaging at both 1064 and 1300 nm wavelengths. The PA signal of the SSS-micelles can be detected through 6.5 cm of chicken breast tissue in vitro. In mice or rats, SSS-micelles can be visualized in bladder and intestine overlaid 5 cm (signal to noise ratio, SNR ≈ 17 dB) and 5.8 cm (SNR over 10 dB) chicken breast tissue, respectively. This work demonstrates the SSS-micelles as a nanoplatform for deep tissue theranostics.


Asunto(s)
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Animales , Ratones , Micelas , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Técnicas Fotoacústicas/métodos , Fototerapia , Polímeros/química , Medicina de Precisión , Ratas , Tensoactivos/química
11.
ACS Appl Mater Interfaces ; 13(8): 9630-9642, 2021 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-33616382

RESUMEN

One potential approach to address the rising threat of antibiotic resistance is through novel formulations of established drugs. We designed antibiotic cross-linked micelles (ABC-micelles) by cross-linking the Pluronic F127 block copolymers with an antibiotic itself, via a novel one-pot synthesis in aqueous solution. ABC-micelles enhanced antibiotic encapsulation while also reducing systemic toxicity in mice. Using colistin, a hydrophilic, potent ″last-resort" antibiotic, ABC-micelle encapsulation yield was 80%, with good storage stability. ABC-micelles exhibited an improved safety profile, with a maximum tolerated dose of over 100 mg/kg colistin in mice, at least 16 times higher than the free drug. Colistin-induced nephrotoxicity and neurotoxicity were reduced in ABC-micelles by 10-50-fold. Despite reduced toxicity, ABC-micelles preserved bactericidal activity, and the clinically relevant combination of colistin and rifampicin (co-loaded in the micelles) showed a synergistic antimicrobial effect against antibiotic-resistant strains of Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumannii. In a mouse model of sepsis, colistin ABC-micelles showed equivalent efficacy as free colistin but with a substantially higher therapeutic index. Microscopic single-cell imaging of bacteria revealed that ABC-micelles could kill bacteria in a more rapid manner with distinct cell membrane disruption, possibly reflecting a different antimicrobial mechanism from free colistin. This work shows the potential of drug cross-linked micelles as a new class of biomaterials formed from existing antibiotics and represents a new and generalized approach for formulating amine-containing drugs.


Asunto(s)
Antibacterianos/uso terapéutico , Colistina/uso terapéutico , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Micelas , Sepsis/tratamiento farmacológico , Animales , Antibacterianos/síntesis química , Antibacterianos/toxicidad , Bacterias/efectos de los fármacos , Colistina/síntesis química , Colistina/toxicidad , Ciclofosfamida , Femenino , Ratones , Pruebas de Sensibilidad Microbiana , Síndromes de Neurotoxicidad/prevención & control , Poloxámero/síntesis química , Poloxámero/química , Poloxámero/toxicidad , Sepsis/inducido químicamente
12.
ACS Appl Bio Mater ; 4(6): 4982-4989, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35007046

RESUMEN

Prodrugs can be formed by chemical modification of the existing active pharmaceutical ingredients (APIs); however, this often sacrifices their functional efficacy. Self-immolative linkers have recently attracted attention, as they can be designed to release pristine APIs. Herein, we report a strategy to generate a self-immolative prodrug (SIP) that can release pristine doxorubicin (DOX). Compared to conventional linkers, the key SIP DOX (KSIP-DOX) developed here can rapidly and quantitatively release the API due to its strong leaving group after reduction by thiol groups, which are present in tumors. KSIP-DOX has enhanced cellular uptake and improved anticancer efficacy, demonstrating its utility for cancer treatment.


Asunto(s)
Antibióticos Antineoplásicos , Doxorrubicina , Profármacos , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/toxicidad , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/toxicidad , Diseño de Fármacos , Liberación de Fármacos , Femenino , Glutatión/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Profármacos/administración & dosificación , Profármacos/química , Profármacos/farmacocinética , Profármacos/toxicidad
13.
Int J Mol Sci ; 21(17)2020 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-32887466

RESUMEN

Stimulus-responsive drug delivery systems generally aim to release the active pharmaceutical ingredient (API) in response to specific conditions and have recently been explored for disease treatments. These approaches can also be extended to molecular imaging to report on disease diagnosis and management. The stimuli used for activation are based on differences between the environment of the diseased or targeted sites, and normal tissues. Endogenous stimuli include pH, redox reactions, enzymatic activity, temperature and others. Exogenous site-specific stimuli include the use of magnetic fields, light, ultrasound and others. These endogenous or exogenous stimuli lead to structural changes or cleavage of the cargo carrier, leading to release of the API. A wide variety of stimulus-responsive systems have been developed-responsive to both a single stimulus or multiple stimuli-and represent a theranostic tool for disease treatment. In this review, stimuli commonly used in the development of theranostic nanoplatforms are enumerated. An emphasis on chemical structure and property relationships is provided, aiming to focus on insights for the design of stimulus-responsive delivery systems. Several examples of theranostic applications of these stimulus-responsive nanomedicines are discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanomedicina , Nanopartículas/administración & dosificación , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Polímeros/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...