Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Control Release ; 373: 201-215, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39004104

RESUMEN

Traditional bolus vaccines typically require multiple doses, which complicates the vaccination process and may cause missed shots, leading to sub-optimal immunity and reduced vaccine effectiveness. Herein, a gel-based long-acting vaccine system with self-adjuvant properties based on laponite was constructed to simplify vaccination procedures and improve vaccine effectiveness. Firstly, the gel system could recruit multiple types of immune cells to form immune niches. Secondly, it could achieve sustained delivery of antigens to lymph nodes by active transport and passive drainage. Then, the gel system triggered the formation of a large number of germinal centers, which elicited enhanced and durable humoral immune responses, as well as strong cellular immune responses. As a result, it eventually showed good prophylactic and therapeutic effects in a variety of tumor models including melanoma, colorectal cancer and peritoneal metastasis models. By further combining the immunoadjuvant CpG ODN and cytokine IL-12, the effect of the gel-vaccine could be further enhanced. In a murine peritoneal metastasis model of colorectal carcinoma, a single administration of the gel-vaccine resulted in complete tumor eradication in 8/9 mice. In summary, this study developed an immunologically active gel-vaccine system. And as a robust and versatile vaccine platform, by loading different antigens and adjuvants, this gel-vaccine system is expected to realize its better therapeutic potential.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra el Cáncer , Geles , Ratones Endogámicos C57BL , Silicatos , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Silicatos/química , Silicatos/administración & dosificación , Femenino , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/química , Línea Celular Tumoral , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/química , Sistemas de Liberación de Medicamentos , Ratones Endogámicos BALB C , Interleucina-12/inmunología , Ratones , Neoplasias Colorrectales/inmunología
2.
Adv Healthc Mater ; : e2401723, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39049538

RESUMEN

Pyroptosis, a highly inflammatory form of programmed cell death, has emerged as a promising target for cancer immunotherapy. However, in the context of pyroptosis execution, while both caspase-3 and GSDME are essential, it is noteworthy that GSDME is frequently under-expressed in cold tumors. To overcome this limitation, engineered cellular nanovesicles (NVs) presenting TRAIL on their membranes (NVTRAIL) are developed to trigger the upregulation of cleaved caspase-3. When strategically combined with the chemotherapeutic agent mitoxantrone (MTO), known for its ability to enhance GSDME expression, MTO@NVTRAIL can convert cancer cells from apoptosis into pyroptosis, inhibit the tumor growth and metastasis successfully in primary tumor. The microparticles released by pyroptotic tumor cells also exhibited certain cytotoxicity against other tumor cells. In addition, tumor cells exposed to the combination treatment of MTO@NVTRAIL in vitro have also demonstrated potential utility as a novel form of vaccine for cancer immunotherapy. Flow analysis of the tumor microenvironment and draining lymph nodes reveals an increased proportion of matured dendritic cells and activation of T cells. In summary, the research provided a reference and alternative approach to induce cancer pyroptosis for clinical antitumor therapy based on engineered cellular nanovesicles and chemotherapy.

3.
Acta Pharm Sin B ; 14(5): 2247-2262, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38799631

RESUMEN

Immunogenic dying tumor cells hold promising prospects as cancer vaccines to activate systemic immunity against both primary and metastatic tumors. Especially, X-ray- induced dying tumor cells are rich in highly immunogenic tumor-associated antigens and self-generated dsDNA as potent adjuvants. However, we found that the X-ray induction process can result in the excessive exposure of phosphatidylserine in cancer vaccines, which can specifically bind with the MerTK receptor on macrophages, acting as a "checkpoint" to facilitate immune silence in the tumor microenvironment. Therefore, we developed a novel strategy combining X-ray-induced cancer vaccines with UNC2250, a macrophage MerTK "checkpoint inhibitor," for treating peritoneal carcinomatosis in colon cancer. By incorporating UNC2250 into the treatment regimen, immunosuppressive efferocytosis of macrophages, which relies on MerTK-directed recognition of phosphatidylserine on vaccines, was effectively blocked. Consequently, the immune analysis revealed that this combination strategy promoted the maturation of dendritic cells and M1-like repolarization of macrophages, thereby simultaneously eliciting robust adaptive and innate immunity. This innovative approach utilizing X-ray-induced vaccines combined with a checkpoint inhibitor may provide valuable insights for developing effective cancer vaccines and immunotherapies targeting colon cancer.

4.
J Control Release ; 369: 746-764, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38599547

RESUMEN

Acute respiratory distress syndrome (ARDS) is a critical illness characterized by severe lung inflammation. Improving the delivery efficiency and achieving the controlled release of anti-inflammatory drugs at the lung inflammatory site are major challenges in ARDS therapy. Taking advantage of the increased pulmonary vascular permeability and a slightly acidic-inflammatory microenvironment, pH-responsive mineralized nanoparticles based on dexamethasone sodium phosphate (DSP) and Ca2+ were constructed. By further biomimetic modification with M2 macrophage membranes, hybrid mineralized nanovesicles (MM@LCaP) were designed to possess immunomodulatory ability from the membranes and preserve the pH-sensitivity from core nanoparticles for responsive drug release under acidic inflammatory conditions. Compared with healthy mice, the lung/liver accumulation of MM@LCaP in inflammatory mice was increased by around 5.5 times at 48 h after intravenous injection. MM@LCaP promoted the polarization of anti-inflammatory macrophages, calmed inflammatory cytokines, and exhibited a comprehensive therapeutic outcome. Moreover, MM@LCaP improved the safety profile of glucocorticoids. Taken together, the hybrid mineralized nanovesicles-based drug delivery strategy may offer promising ideas for enhancing the efficacy and reducing the toxicity of clinical drugs.


Asunto(s)
Antiinflamatorios , Dexametasona , Glucocorticoides , Pulmón , Nanopartículas , Síndrome de Dificultad Respiratoria , Animales , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacocinética , Glucocorticoides/uso terapéutico , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Dexametasona/uso terapéutico , Dexametasona/análogos & derivados , Distribución Tisular , Nanopartículas/química , Ratones , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Liberación de Fármacos , Neumonía/tratamiento farmacológico , Neumonía/inducido químicamente , Células RAW 264.7 , Sistemas de Liberación de Medicamentos , Calcio/metabolismo , Citocinas/metabolismo
5.
J Control Release ; 369: 215-230, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38508529

RESUMEN

In the progression of acute inflammation, the activation and recruitment of macrophages and neutrophils are mutually reinforcing, leading to amplified inflammatory response and severe tissue damage. Therefore, to regulate the axis of neutrophils and macrophages is essential to avoid tissue damage induced from acute inflammatory. Apoptotic neutrophils can regulate the anti-inflammatory activity of macrophages through the efferocytosis. The strategy of in situ targeting and inducing neutrophil apoptosis has the potential to modulate macrophage activity and transfer anti-inflammatory drugs. Herein, a natural glycyrrhiza protein nanoparticle loaded with dexamethasone (Dex@GNPs) was constructed, which could simultaneously regulate neutrophil and macrophage function during acute inflammation treatment by combining in situ neutrophil apoptosis and macrophage efferocytosis. Dex@GNPs can be rapidly and selectively internalized by neutrophils and subsequently induce neutrophils apoptosis through a ROS-dependent mechanism. The efferocytosis of apoptotic neutrophils not only promoted the polarization of macrophages into anti-inflammatory state, but also facilitated the transfer of Dex@GNPs to macrophages. This enabled dexamethasone to further modulate macrophage function. In mouse models of acute respiratory distress syndrome and sepsis, Dex@GNPs significantly ameliorated the disordered immune microenvironment and alleviated tissue injury. This study presents a novel strategy for drug delivery and inflammation regulation to effectively treat acute inflammatory diseases.


Asunto(s)
Antiinflamatorios , Apoptosis , Dexametasona , Glycyrrhiza , Inflamación , Macrófagos , Nanopartículas , Neutrófilos , Animales , Dexametasona/administración & dosificación , Dexametasona/farmacología , Apoptosis/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Nanopartículas/química , Macrófagos/efectos de los fármacos , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Inflamación/tratamiento farmacológico , Glycyrrhiza/química , Ratones Endogámicos C57BL , Masculino , Ratones , Fagocitosis/efectos de los fármacos , Humanos , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Células RAW 264.7 , Eferocitosis
6.
ACS Nano ; 18(2): 1658-1677, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38166370

RESUMEN

Acute Respiratory Distress Syndrome (ARDS) is a clinically severe respiratory disease that causes severe medical and economic burden. To improve therapeutic efficacy, effectively targeting delivery to the inflamed lungs and inflamed cells remains an ongoing challenge. Herein, we designed engineered biomimetic nanovesicles (DHA@ANeu-DDAB) by fusion of lung-targeting functional lipid, neutrophil membrane containing activated ß2 integrins, and the therapeutic lipid, docosahexaenoic acid (DHA). By the advantage of lung targeting lipid and ß2 integrin targeting adhesion, DHA@ANeu-DDAB can first target lung tissue and further target inflammatory vascular endothelial cells, to achieve "tissue first, cell second" hierarchical delivery. In addition, the ß2 integrins in DHA@ANeu-DDAB could bind to the intercellular cell adhesion molecule-1/2 (ICAM-1/2) ligand on the endothelium in the inflamed blood vessels, thus inhibiting neutrophils' infiltration in the blood circulation. DHA administration to inflamed lungs could effectively regulate macrophage phenotype and promote its anti-inflammatory activity via enhanced biosynthesis of specialized pro-resolving mediators. In the lipopolysaccharide-induced ARDS mouse model, DHA@ANeu-DDAB afforded a comprehensive and efficient inhibition of lung inflammation and promoted acute lung damage repair. Through mimicking physiological processes, these engineered biomimetic vesicles as a delivery system possess good potential in targeting therapy for ARDS.


Asunto(s)
Neutrófilos , Compuestos de Amonio Cuaternario , Síndrome de Dificultad Respiratoria , Animales , Ratones , Humanos , Neutrófilos/metabolismo , Células Endoteliales/metabolismo , Biomimética , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Pulmón/metabolismo , Integrinas , Lípidos
7.
Acta Pharm Sin B ; 13(10): 4318-4336, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37799395

RESUMEN

Delayed diabetic wound healing has placed an enormous burden on society. The key factors limiting wound healing include unresolved inflammation and impaired angiogenesis. Platelet-rich plasma (PRP) gel, a popular biomaterial in the field of regeneration, has limited applications due to its non-injectable properties and rapid release and degradation of growth factors. Here, we prepared an injectable hydrogel (DPLG) based on PRP and laponite by a simple one-step mixing method. Taking advantages of the non-covalent interactions, DPLG could overcome the limitations of PRP gels, which is injectable to fill irregular injures and could serve as a local drug reservoir to achieve the sustained release of growth factors in PRP and deferoxamine (an angiogenesis promoter). DPLG has an excellent ability in accelerating wound healing by promoting macrophage polarization and angiogenesis in a full-thickness skin defect model in type I diabetic rats and normal rats. Taken together, this study may provide the ingenious and simple bioactive wound dressing with a superior ability to promote wound healing.

8.
ACS Appl Mater Interfaces ; 15(36): 42378-42394, 2023 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-37658814

RESUMEN

The interaction between nanoparticles and cells is closely associated with the therapeutic effects of nanomedicine. Nanoparticles could be transported among cells, but the process-related mechanism remains to be further explored. In this study, it was found that endocytosed cationic polymer nanoparticles (cNPs) could be excreted in an extracellular vesicle (EV)-coated form (cNP@EVs). It was deduced that cNPs may pass through early endosomes, multivesicular bodies (MVBs), and autophagic MVBs within cells. Moreover, a high level of autophagy facilitated the exocytosis process. Since EVs were the effective vehicles for conveying biological information and substances, cNP@EVs were proved to be efficient forms for the intercellular transportation of nanoparticles and have the potential as efficient biomimetic drug delivery systems. These properties endowed cNP@EVs with deep penetration and enhanced antitumor activity. Our findings provided a proof-of-concept for understanding the transfer process of nanoparticles among cells and may help us to further utilize EV-mediated transportation of nanoparticles, therefore, expanding its clinical application.


Asunto(s)
Vesículas Extracelulares , Nanopartículas , Neoplasias , Humanos , Autofagia , Vesículas Transportadoras , Polímeros , Neoplasias/tratamiento farmacológico
9.
J Control Release ; 359: 359-372, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37290722

RESUMEN

Whole tumor cells can act as effective antigen depots and have been regarded as prospective candidate for cancer vaccines. However, the clinical outcomes of whole tumor cell vaccine were restricted by the poor immunogenicity and potential in vivo oncogenicity risks. Herein, a simple and effective cancer vaccine named frozen dying tumor cells (FDT) was constructed to initiate a cascade of immune attacks against cancer. By introducing immunogenic dying tumor cells and integrating cryogenic freezing technology, FDT was endowed with high immunogenicity, good in vivo safety, and long-time storage superiority. In syngeneic mice with malignant melanoma, FDT primed the polarization of follicular helper T cells and the differentiation of germinal center B cells in lymph nodes, and promoted the infiltration of cytotoxic CD8+ T cells in the tumor microenvironment, triggering the dual activation of humoral and cellular immunity. Of note, when combined with cytokines and immune checkpoint inhibitors, the FDT vaccine achieved 100% eradication of pre-existing tumors in mice, as demonstrated in the peritoneal metastasis model of colorectal carcinoma. Taken together, our work suggests an efficient cancer vaccine inspired by dying tumor cells and provides an alternative treatment candidate for cancer.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Ratones , Animales , Linfocitos T CD8-positivos , Inmunidad Humoral , Inmunidad Celular , Vacunas contra el Cáncer/uso terapéutico , Melanoma/tratamiento farmacológico , Microambiente Tumoral
10.
ACS Appl Mater Interfaces ; 15(20): 24134-24148, 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37163695

RESUMEN

In recent years, microbiota-based tumor immunotherapy has become a hotspot in cancer research. However, the use of microorganisms alone to activate the immune response for antitumor therapy was unsatisfactory. In this study, we biosynthesized gold nanoparticles (AuNPs) and platinum nanoparticles (PtNPs) based on yeast microcapsules to activate the immune response for antitumor treatment in synergy with chemodynamic therapy (CDT) and photothermal therapy (PTT). We generated AuNPs and PtNPs on yeast microcapsules (YAP) and fabricated nanoscale particles (Bre-YAP) by ultrasonic fragmentation and differential centrifugation. Bre-YAP retained the glucan component of yeast as an adjuvant; in the meantime, these two kinds of metal nanoparticles contained were excellent CDT and PTT mediators. By inspection, they could reach a high level of distribution in tumors and tumor-draining lymph nodes (TDLNs). Under the laser irradiation of tumors, this immunological nanomaterial significantly remodeled the microenvironments of tumors and TDLNs. The primary tumors were effectively inhibited or even eradicated, and the overall survival of mice was significantly improved as well. Therefore, yeast microcapsule-based Bre-YAP with immune properties could be used as an effective cancer treatment modality.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias , Animales , Ratones , Fototerapia , Nanopartículas del Metal/química , Oro/química , Saccharomyces cerevisiae , Cápsulas , Línea Celular Tumoral , Platino (Metal)/química , Nanopartículas/química , Neoplasias/patología , Inmunoterapia , Microambiente Tumoral
11.
Adv Healthc Mater ; 12(6): e2202209, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36401821

RESUMEN

Cell-derived nanovesicles are widely utilized as therapeutic agents for cancer therapy. Current research mostly focuses on their ability to activate antitumor cellular immunity. However, whether they can activate and participate in antitumor humoral immunity is rarely studied. Here, doxorubicin-loaded hybrid cell nanovesicles (DNVs) are designed for boosting antitumor humoral and cellular immunity. The hybrid cell nanovesicles are generated through fusion of nanovesicles derived from M1-type macrophages and 4T1 tumor cells. It is found that DNVs can accumulate at tumor tissues and draining lymph nodes effectively, which results in the activation of antitumor immune response and significant inhibition of tumor progression. During this process, dendritic cells are effectively activated, subsequently inducing cytotoxicity T lymphocytes-mediated cellular immunity. Furthermore, DNVs elicit the antitumor humoral immunity through boosting T follicular helper cells and germinal center B cells. By analyzing the mechanism behind humoral immunity activation, it is found that M1-type macrophages repolarized by DNVs play an important role. In general, besides antitumor cellular immunity, the proposed hybrid nanovesicles provide a promising strategy for enhancing antitumor humoral immunity by macrophages repolarization and germinal center B cells activation.


Asunto(s)
Inmunidad Humoral , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Macrófagos , Centro Germinal , Doxorrubicina/farmacología
12.
Adv Sci (Weinh) ; 10(2): e2204178, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36424135

RESUMEN

Biological vesicles, containing genetic materials and proteins of the original cells, are usually used for local or systemic communications among cells. Currently, studies on biological vesicles as therapeutic strategies or drug delivery carriers mainly focus on exogenously generated biological vesicles. However, the limitations of yield and purity caused by the complex purification process still hinder their clinical transformation. Recently, it has been reported that living organisms, including cells and bacteria, can produce functional/therapeutic biological vesicles within body automatically. Therefore, using organisms to produce endogenous biological vesicles in body as drug/bio-information delivery carriers has become a potential therapeutic strategy. In this review, the current development status and application prospects of in situ organism-produced biological vesicles are introduced. The advantages and effects of this endogenous biological vesicles-based strategy in drug delivery and disease treatments are analyzed. According to the type of endogenous biological vesicles, they are divided into four categories: exosomes, platelet-derived microparticles, apoptotic bodies, and bacteria-released outer membrane vesicles. And finally, the shortcomings of current research and future development are analyzed. This review is believed to open up the application of endogenous biological vesicles in the field of biomedicine and shed light on current research.


Asunto(s)
Micropartículas Derivadas de Células , Exosomas , Vesículas Extracelulares , Sistemas de Liberación de Medicamentos , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Portadores de Fármacos/metabolismo
13.
J Mater Chem B ; 11(2): 244-260, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36512384

RESUMEN

Inflammatory bowel disease (IBD) is a type of recurrent intestinal diseases. Natural product molecules have been gradually developed into an important source of anti-inflammatory drugs for treating IBD owing to their high anti-inflammatory activity, well known safety, structural specificity and therapeutic mechanism diversity. However, most of the natural products are restricted by poor solubility in actual application. How to achieve satisfactory bioavailability during the treatment of IBD is one of the urgent problems to be solved in the current research. Micro/nano drug delivery systems could improve the solubility of drugs with targeted delivery of anti-inflammatory drugs to the colon with responsive release property. Therefore, using micro/nano drug delivery systems, the problems mentioned above involving natural product molecules in the treatment of IBD could be solved. According to the compositions of the intestinal tract and inflammatory characteristics of IBD, the strategies of using micro/nano drug delivery systems for natural products could be summarized in two steps: targeted delivery and responsive release. In this review, the targeted and responsive release strategies of the micro/nano drug delivery systems combined with their anti-inflammatory effects in IBD animal models to illustrate that the proposed strategies could be potential treatments for symptomatic IBD are described.


Asunto(s)
Productos Biológicos , Enfermedades Inflamatorias del Intestino , Animales , Sistema de Administración de Fármacos con Nanopartículas , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Preparaciones Farmacéuticas , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
14.
ACS Nano ; 16(9): 15124-15140, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36037505

RESUMEN

Acute respiratory distress syndrome (ARDS) has been a life threat for patients in ICUs. Vast efforts have been devoted, while no medication has proved viable, which may be ascribed to inadequate drug delivery to damaged tissues and insufficient control of lung inflammation. Given the anti-inflammatory role of M2-type macrophages, M2 macrophage-derived nanovesicles and lung-targeting liposomes are cofused to fabricate hybrid liposomes-nanovesicles (LNVs). Benefiting from the incorporated lung-homing moiety, LNVs demonstrate high pulmonary accumulation with a lung/liver ratio of 14.9, which is approximately 53.3-fold of free nanovesicles. Thus, M2 macrophage-derived nanovesicles can be delivered to lung tissues for executing immunoregulatory functions. LNVs display phagocytosis by the infiltrated neutrophils and macrophages, exhibiting sustained release of preloaded IKK-2 inhibitor (TPCA-1). The integrated nanosystems demonstrate multidimensional suppression of the overwhelming inflammation, such as decreasing infiltration of inflammatory cells, achieving restraint on cytokine storms and alleviating oxidative stress. Therefore, the improved therapeutic outcome in ARDS mice is obtained. Altogether, the hybrid nanoplatform provides a versatile drug delivery paradigm for integrating biological nanovesicles and therapeutic molecules by cofusion of nanovesicles with liposomes, improving lung biodistribution and accomplishing a boosted anti-inflammatory response for ARDS therapy.


Asunto(s)
Síndrome de Liberación de Citoquinas , Síndrome de Dificultad Respiratoria , Animales , Antiinflamatorios/farmacología , Biomimética , Preparaciones de Acción Retardada , Liposomas , Pulmón , Ratones , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Distribución Tisular
15.
Acta Pharm Sin B ; 12(5): 2550-2567, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35646526

RESUMEN

In the development of chemo-immunotherapy, many efforts have been focusing on designing suitable carriers to realize the co-delivery of chemotherapeutic and immunotherapeutic with different physicochemical properties and mechanisms of action. Besides, rapid drug release at the tumor site with minimal drug degradation is also essential to facilitate the antitumor effect in a short time. Here, we reported a cancer cell membrane-coated pH-responsive nanogel (NG@M) to co-deliver chemotherapeutic paclitaxel (PTX) and immunotherapeutic agent interleukin-2 (IL-2) under mild conditions for combinational treatment of triple-negative breast cancer. In the designed nanogels, the synthetic copolymer PDEA-co-HP-ß-cyclodextrin-co-Pluronic F127 and charge reversible polymer dimethylmaleic anhydride-modified polyethyleneimine endowed nanogels with excellent drug-loading capacity and rapid responsive drug-releasing behavior under acidic tumor microenvironment. Benefited from tumor homologous targeting capacity, NG@M exhibited 4.59-fold higher accumulation at the homologous tumor site than heterologous cancer cell membrane-coated NG. Rapidly released PTX and IL-2 enhanced the maturation of dendritic cells and quickly activated the antitumor immune response in situ, followed by prompted infiltration of immune effector cells. By the combined chemo-immunotherapy, enhanced antitumor effect and efficient pulmonary metastasis inhibition were achieved with a prolonged median survival rate (39 days).

16.
J Nanobiotechnology ; 20(1): 226, 2022 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-35549947

RESUMEN

BACKGROUND: Conventional chemotherapy has poor efficacy in triple-negative breast cancer (TNBC) which is highly heterogeneous and aggressive. Imaging-guided therapy is usually combined with diverse treatment modalities, could realize the integration of diagnosis and treatments. Therefore, the primary challenge for combinational therapy is designing proper delivery systems to accomplish multiple synergistic effects. RESULTS: Herein, a facile nanoplatform was manufactured to fulfill the all-in-one approaches for TNBC combinational therapy. Fe3+-based metal-phenolic networks (MPNs) with bovine serum albumin (BSA) modification served as drug delivery carriers to encapsulate bleomycin (BLM), forming BFE@BSA NPs. The self-assembly mechanism, pH-responsive drug release behavior, and other physicochemical properties of this system were characterized. The potential of BFE@BSA NPs as photothermal transduction agents and magnetic resonance imaging (MRI) contrast agents was explored. The synergistic anti-tumor effects consisting of BLM-induced chemotherapy, Fenton reactions-mediated chemodynamic therapy, and photothermal therapy-induced apoptosis were studied both in vitro and in vivo. Once internalized into tumor cells, released BLM could cause DNA damage, while Fenton reactions were initiated to produce highly toxic •OH. Upon laser irradiation, BFE@BSA NPs could convert light into heat to achieve synergistic effects. After intravenous administration, BFE@BSA NPs exhibited great therapeutic effects in 4T1 tumor xenograft model. Moreover, as T1-weighted MRI contrast agents, BFE@BSA NPs could provide diagnosis and treatment monitoring for individualized precise therapy. CONCLUSIONS: A nano-system that integrated imaging and combinational therapy (chemotherapy, chemodynamic therapy and photothermal therapy) were developed to kill the tumor and monitor therapeutic efficacy. This strategy provided an all-in-one theranostic nanoplatform for MRI-guided combinational therapy against TNBC.


Asunto(s)
Nanopartículas , Neoplasias , Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Medios de Contraste , Portadores de Fármacos/uso terapéutico , Humanos , Imagen por Resonancia Magnética , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Fototerapia/métodos , Terapia Fototérmica , Albúmina Sérica Bovina/uso terapéutico , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
17.
J Nanobiotechnology ; 19(1): 415, 2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34895243

RESUMEN

BACKGROUND: Immunochemotherapy is a potent anti-tumor strategy, however, how to select therapeutic drugs to enhance the combined therapeutic effect still needs to be explored. METHODS AND RESULTS: Herein, a magnetic resonance nanoprobe (MnP@Lip) with STING (Stimulator of INterferon Genes) activation character was synthesized and co-administered with platinum-based chemotherapeutics for enhanced immunochemotherapy. MnP@Lip nanoparticles was prepared by simple fabrication process with good reproducibility, pH-sensitive drug release behavior and biocompatibility. In vitro experiments elucidated that Mn2+ can promote the polarization of M0 and/or M2 macrophages to M1 phenotype, and promote the maturation of BMDC cells. Upon Mn2+ treatment, the STING pathway was activated in tumor cells, mouse lung epithelial cells, and immune cells. More importantly, anti-tumor experiments in vivo proved that MnP@Lip combined with platinum-based chemotherapeutics increased T cells infiltration in the tumor microenvironment, and inhibited tumor growth in the orthotopic therapeutic and postoperative tumor models. CONCLUSIONS: This kind of therapeutic strategy that combined MnP@Lip nanoparticles with platinum-based chemotherapeutics may provide a novel insight for immunochemotherapy.


Asunto(s)
Antineoplásicos , Sondas Moleculares , Nanopartículas , Platino (Metal) , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Células Cultivadas , Femenino , Inmunoterapia , Macrófagos/efectos de los fármacos , Imagen por Resonancia Magnética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Sondas Moleculares/química , Sondas Moleculares/farmacocinética , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias Experimentales , Platino (Metal)/química , Platino (Metal)/farmacocinética , Platino (Metal)/farmacología , Microambiente Tumoral/efectos de los fármacos
18.
Small ; 17(50): e2103984, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34723421

RESUMEN

The biosynthesis of nanomedicine has gained enormous attention and exhibited promising prospects, while the underlying mechanism and advantage remain not fully understood. Here, a cell-reactor based on tumor cells is developed to obtain biogenetic gold nanoparticles (Au@MC38) for sensitizing radiotherapy and boosting immune responses. It demonstrates that the intracellular biomineralization and exocytosis process of Au@MC38 can be regulated by the cellular metabolites level and other factors, such as glutathione and reactive oxygen species (ROS), autophagy, and UV irradiation. The elucidation of mechanisms may promote the understanding of interaction principles between nanoparticles and biosystems in the process of biosynthesis. Combined with radiotherapy, Au@MC38 strengthens the radiation-induced DNA damage and ROS generation, thus aggravating cell apoptosis and necrosis. Benefiting from homologous targeting and transcytosis effect, Au@MC38 demonstrates good tumor distribution. Local radiation-induced immunogenic cell death initiates an effective immune response. Especially, CD8a+ dendritic cells are significantly increased in mice that received combinatorial treatment. This radio-sensitization strategy has demonstrated the effective inhibition on primary and metastatic tumors, and achieved satisfactory survival benefit in combinatorial with immune checkpoint blockade. Thus, this bio-inspired synthetic strategy may impulse the development of biosynthesis and its therapeutic applications, contributing to a non-invasive and efficient modality for nanomedicine exploitation.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias , Animales , Línea Celular Tumoral , Oro , Inmunidad , Ratones , Nanomedicina , Neoplasias/terapia
19.
ACS Appl Mater Interfaces ; 13(33): 39003-39017, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433253

RESUMEN

Improving tumor immunogenicity is critical for increasing the responsiveness of triple-negative breast cancer (TNBC) to anti-PD-(L)1 treatment. Here, we verified that chidamide (CHI), an epigenetic modulator, could elicit immunogenic cell death within TNBC to enhance cancer immunogenicity and elicit an antitumor immune response. Additionally, CHI increased the expression level of PD-L1, MHC I, and MHC II on cancer cells, which contributed to T-cell recognition and PD-1/PD-L1 blockade therapy response. The synergistic antitumor efficacy of CHI and PD-L1 blockade therapy was further explored through liposomes co-delivering CHI and BMS-202 (a small-molecule PD-L1 inhibitor). The liposomes possessed good biocompatibility, security, and controllable drug release and endowed therapeutics drugs with favorable tumor accumulation. Furthermore, the drug-loaded liposomes could obviously boost the antitumor immunity of TNBC through CHI-enhanced tumor immunogenicity and BMS-202-mediated PD-L1 blockade, thereby effectively inhibiting the growth of primary and metastatic tumors with an inhibitory rate of metastasis of up to 96%. In summary, this work provided a referable and optional approach for clinical antitumor therapy based on the combination of an epigenetic modulator and PD-1/PD-L1 blockade therapy.


Asunto(s)
Acetamidas/química , Aminopiridinas/química , Antineoplásicos/farmacología , Benzamidas/química , Portadores de Fármacos/química , Inhibidores de Puntos de Control Inmunológico/química , Piridinas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/inmunología , Acetamidas/farmacología , Aminopiridinas/farmacología , Animales , Benzamidas/farmacología , Materiales Biocompatibles/química , Línea Celular Tumoral , Terapia Combinada/métodos , Liberación de Fármacos , Epigénesis Genética/efectos de los fármacos , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Distribución Tisular , Resultado del Tratamiento
20.
Acta Pharm Sin B ; 11(10): 3060-3091, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33977080

RESUMEN

Acute respiratory distress syndrome (ARDS) is characterized by the severe inflammation and destruction of the lung air-blood barrier, leading to irreversible and substantial respiratory function damage. Patients with coronavirus disease 2019 (COVID-19) have been encountered with a high risk of ARDS, underscoring the urgency for exploiting effective therapy. However, proper medications for ARDS are still lacking due to poor pharmacokinetics, non-specific side effects, inability to surmount pulmonary barrier, and inadequate management of heterogeneity. The increased lung permeability in the pathological environment of ARDS may contribute to nanoparticle-mediated passive targeting delivery. Nanomedicine has demonstrated unique advantages in solving the dilemma of ARDS drug therapy, which can address the shortcomings and limitations of traditional anti-inflammatory or antioxidant drug treatment. Through passive, active, or physicochemical targeting, nanocarriers can interact with lung epithelium/endothelium and inflammatory cells to reverse abnormal changes and restore homeostasis of the pulmonary environment, thereby showing good therapeutic activity and reduced toxicity. This article reviews the latest applications of nanomedicine in pre-clinical ARDS therapy, highlights the strategies for targeted treatment of lung inflammation, presents the innovative drug delivery systems, and provides inspiration for strengthening the therapeutic effect of nanomedicine-based treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA