Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 36(16): e2309920, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38213134

RESUMEN

Advancements in gene delivery systems are pivotal for gene-based therapeutics in oncological, inflammatory, and infectious diseases. This study delineates the design of a self-assembled oligopeptoplex (SA-OP) optimized for shRNA delivery to adipocytes, targeting obesity and associated metabolic syndromes. Conventional systems face challenges, including instability due to electrostatic interactions between genetic materials and cationic oligopeptides. Additionally, repeated injections induce discomfort and compromise patient well-being. To circumvent these issues, a dissolvable hyaluronic acid-based, self-locking microneedle (LMN) patch is developed, with improved micro-dose efficiency, for precise SA-OP delivery. This platform offers pain-free administration and improved SA-OP storage stability. In vitro studies in 3T3-L1 cells demonstrated improvements in SA-OP preservation and gene silencing efficacy. In vivo evaluation in a mice model of diet-induced type 2 diabetes yielded significant gene silencing in adipose tissue and a 21.92 ± 2.51% reduction in body weight with minimum relapse risk at 6-weeks post-treatment, representing a superior therapeutic efficacy in a truncated timeframe relative to the GLP-1 analogues currently available on the market. Additionally, SA-OP (LMN) mitigated insulin resistance, inflammation, and hepatic steatosis. These findings establish SA-OP (LMN) as a robust, minimally invasive transdermal gene delivery platform with prolonged storage stability for treating obesity and its metabolic comorbidities.


Asunto(s)
Diabetes Mellitus Tipo 2 , Humanos , Ratones , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Adipocitos , Administración Cutánea , Terapia Genética , Sistemas de Liberación de Medicamentos
2.
Biomater Res ; 27(1): 136, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38111068

RESUMEN

BACKGROUND: The emergence of cancer immunotherapies, notably immune checkpoint inhibitors, has revolutionized anti-cancer treatments. These treatments, however, have been reported to be effective in a limited range of cancers and cause immune-related adverse effects. Thus, for a broader applicability and enhanced responsiveness to solid tumor immunotherapy, immunomodulation of the tumor microenvironment is crucial. Transforming growth factor-ß (TGF-ß) has been implicated in reducing immunotherapy responsiveness by promoting M2-type differentiation of macrophages and facilitating cancer cell metastasis. METHODS: In this study, we developed macrophage membrane-coated nanoparticles loaded with a TGF-ßR1 kinase inhibitor, SD-208 (M[Formula: see text]-SDNP). Inhibitions of M2 macrophage polarization and epithelial-to-mesenchymal transition (EMT) of cancer cells were comprehensively evaluated through in vitro and in vivo experiments. Bio-distribution study and in vivo therapeutic effects of M[Formula: see text]-SDNP were investigated in orthotopic breast cancer model and intraveneously injected metastasis model. RESULTS: M[Formula: see text]-SDNPs effectively inhibited cancer metastasis and converted the immunosuppressive tumor microenvironment (cold tumor) into an immunostimulatory tumor microenvironment (hot tumor), through specific tumor targeting and blockade of M2-type macrophage differentiation. Administration of M[Formula: see text]-SDNPs considerably augmented the population of cytotoxic T lymphocytes (CTLs) in the tumor tissue, thereby significantly enhancing responsiveness to immune checkpoint inhibitors, which demonstrates a robust anti-cancer effect in conjunction with anti-PD-1 antibodies. CONCLUSION: Collectively, responsiveness to immune checkpoint inhibitors was considerably enhanced and a robust anti-cancer effect was demonstrated with the combination treatment of M[Formula: see text]-SDNPs and anti-PD-1 antibody. This suggests a promising direction for future therapeutic strategies, utilizing bio-inspired nanotechnology to improve the efficacy of cancer immunotherapy.

3.
Adv Mater ; 35(10): e2209966, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36528846

RESUMEN

Advancements in micro-resolution 3D printers have significantly facilitated the development of highly complex mass-producible drug delivery platforms. Conventionally, due to the limitations of micro-milling machineries, dissolvable microneedles (MNs) are mainly fabricated in cone-shaped geometry with limited drug delivery accuracy. Herein, to overcome the limitations of conventional MNs, a novel projection micro-stereolithography 3D printer-based self-locking MN for precise skin insertion, adhesion, and transcutaneous microdose drug delivery is presented. The geometry of self-locking MN consists of a sharp skin-penetrating tip, a wide skin interlocking body, and a narrow base with mechanical supports fabricated over a flexible hydrocolloid patch to improve the accuracy of skin penetration into irregular surfaces. Melanoma, a type of skin cancer, is selected as the model for the investigation of self-locking MNs due to its irregular and uneven surface. In vivo immunotherapy efficacy is evaluated by integrating SD-208, a novel transforming growth factor-ß (TGF-ß) inhibitor that suppresses the proliferation and metastasis of tumors, and anti-PD-L1 (aPD-L1 Ab), an immune checkpoint inhibitor that induces T cell-mediated tumor cell death, into self-locking MNs and comparing them with intratumoral injection. Evaluation of (aPD-L1 Ab)/SD-208 delivery effectiveness in B16F10 melanoma-bearing mice model confirms significantly improved dose efficacy of self-locking MNs compared with intratumoral injection.


Asunto(s)
Melanoma , Piel , Ratones , Animales , Piel/metabolismo , Sistemas de Liberación de Medicamentos , Adyuvantes Inmunológicos , Inmunoterapia
4.
Adv Sci (Weinh) ; 9(33): e2203286, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36209391

RESUMEN

Persistent uptake of high-calorie diets induces the storage of excessive lipid in visceral adipose tissue. Lipids secreted from obese adipose tissue are accumulated in peripheral tissues such as the liver, pancreas, and muscle, and impair insulin sensitivity causing type 2 diabetes mellitus (T2DM). Furthermore, the accumulation of inflammatory cytokines and lipids in the liver induces apoptosis and fibrogenesis, and ultimately causes nonalcoholic steatohepatitis (NASH). To modulate obese tissue environments, it is challenged to selectively deliver inducers of heme oxygenase-1 (HO-1) to adipose tissue with the aid of a prohibitin targeting drug delivery system. Prohibitin binding peptide (PBP), an oligopeptide targeting prohibitin rich in adipose tissue, is conjugated on the surface of Hemin- or CoPP-loaded poly(lactide-co-glycolide) nanoparticles (PBP-NPs). PBP-NPs efficiently differentiate lipid storing white adipocytes into energy-generating brown adipocytes in T2DM and NASH models. In addition, PBP-NPs are found to target prohibitin overexpressed fatty liver in the NASH model and inhibit hepatic uptake of circulating lipids. Furthermore, PBP-NPs switch phenotypes of inflammatory macrophages in damaged organs and lower inflammation. Taken together, dual-targeted induction of HO-1 in fatty adipose and liver tissues is proven to be a promising therapeutic strategy to ameliorate obesity, insulin resistance, and steatohepatitis by lowering lipids and cytokines.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Nanopartículas , Enfermedad del Hígado Graso no Alcohólico , Humanos , Hemo-Oxigenasa 1/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Tejido Adiposo/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Resistencia a la Insulina/fisiología , Citocinas/metabolismo , Lípidos
5.
Biomaterials ; 279: 121209, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34700224

RESUMEN

Obesity is a serious health problem with tremendous economic and social consequences, which is associated with metabolic diseases and cancer. Currently available anti-obesity drugs acting in the gastrointestinal tract, or the central nervous system have shown limited efficacy in the reduction of obesity, accompanied by severe side effects. Therefore, a novel therapeutic delivery targeting adipocytes and normalizing excess fat transport and accumulation is necessary to maximize efficacy and reduce side effects for long-term treatment. Fatty acid binding protein 4 (FABP4) is an adipokine that coordinates lipid transport in mature adipocyte and its inhibition in obesity model showed weight loss and normalized insulin response. Reduction of FABP4 level in adipocytes was compensated by fatty acid binding protein 5 (FABP5), which resulted in reduction of recovery of obesity and co-morbidities related to obesity by FABP4 knock-down alone. In this study, we developed a non-viral gene delivery system, sh (FABP4/5)/ATS9R, that silences FABP4 and FABP5 simultaneously with oligopeptide (ATS9R) that can selectively target mature adipocyte. For future clinical application to increase patient compliance, sh (FABP4/5)/ATS9R was administered subcutaneously and intraperitoneally to obese animal model and both routes demonstrated startling dual gene efficacy in visceral adipose tissues. Furthermore, dual gene silencing efficiently alleviated obesity, improved insulin sensitivity and restored hepatic metabolism in high fat diet-induced type 2 diabetes mouse model. Targeted-dual gene silencing of sh (FABP4/5)/ATS9R in adipose tissues demonstrated synergistic effects to overcome obesity and obesity-induced metabolic diseases and beneficial effects against liraglutide, providing a great potential for future translational research.


Asunto(s)
Diabetes Mellitus Tipo 2 , Proteínas de Unión a Ácidos Grasos , Resistencia a la Insulina , Obesidad , Adipocitos Blancos/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/terapia , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Silenciador del Gen , Ratones , Proteínas de Neoplasias , Obesidad/genética , Obesidad/terapia
6.
J Control Release ; 336: 344-353, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34147573

RESUMEN

Sepsis is a systemic inflammatory response syndrome caused by bacterial infection. The sepsis therapy has involved prescription of adequate antibiotics, requiring several days to determine the proper type without reducing the inflammatory response. Thus, it is necessary to rapidly decrease fundamental inflammation, which can induce serious organ damage. In the inflammatory mechanism, tumor necrosis factor-alpha (TNF-α) produced by macrophages has an important role in infiltration of macrophages into infected sites and as a trigger for secretion of pro-inflammatory cytokines. However, commercialized TNF-α antibody medicines have limits such as fibrosis, cytokine storms, and high production costs. There is a growing need for anti-inflammatory sepsis treatment free from side effects. For this reason, TNF-α converting enzyme (TACE) could be an innovative target to break the positive feedback loop of inflammatory mediators (TNF-α) since it converts the inactive TNF-α membrane bound form to the activated soluble form in macrophages. A non-viral gene delivery system was developed in this study to deliver siRNA into inflammation-mediated macrophages without toxicity. The peptide-based gene carrier created by conjugating positively-charged nine arginine (9R) and the TKPR (Thr-Lys-Pro-Arg) sequence from the Fc region of Immunoglobulin G (IgG) specifically binds to the neuropilin-1 (NRP-1) receptor on the macrophage surface. Our results demonstrated that siTACE/TKPR-9R complexes were internalized in macrophages and successfully down-regulated TACE mRNA level. Finally, RNA interference with cell-targeted peptide carriers indicates a fundamental therapy for acute inflammatory sepsis free of off-target effects.


Asunto(s)
Sepsis , Factor de Necrosis Tumoral alfa , Regulación hacia Abajo , Humanos , Macrófagos/metabolismo , Interferencia de ARN , Sepsis/terapia , Factor de Necrosis Tumoral alfa/metabolismo
7.
J Control Release ; 332: 109-126, 2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33571549

RESUMEN

Cancer immunotherapy has revolutionized the standard of care for solid tumors in multiple disease sites. In light of this, immune checkpoint blockade, directly interfering with various immunosuppressive mechanisms in tumor sites, has been actively studied. Inhibitors of cytotoxic T-lymphocyte-associated protein 4 (CTLA 4) and programmed cell death 1 receptor (PD-1) / programmed cell death receptor ligand 1 (PD-L1) could successfully increase survival rate in patients with advanced cancers including melanoma and non-small cell lung cancer, leading to increased survival and different patterns of response including durable response and pseudo-progression. Despite continued development of the immune checkpoint blockades, however, fairly low overall response rate (ORR) levels have been reported for patients with various types of cancer. Fewer than 13% of patients with cancer were reported to respond to immune checkpoint blockades and some patients were diagnosed with severe immune-related adverse events (irAEs). The tumor microenvironment, controlled via various components and mediators, is regarded as the primary cause responsible for failure of immune checkpoint blockades in clinical investigations. In fact, there has been a clinical report that epidermal growth factor receptor (EGFR) inhibition in tumor microenvironment enhanced ORR of PD-1 inhibitors for 29.7% in EGFR-mutated non-small-cell lung carcinoma (NSCLC) patients. Therefore, to enhance the effectiveness and reduce adverse effects of immune checkpoint blockades, the majority of studies have focused on targeting and suppressing the immunosuppressive characteristics of the tumor microenvironment. Herein, we review the components and mediators of tumor microenvironment responsible for failure of immune checkpoint blockades and introduce the recent approaches of tumor microenvironment component-targeted nanomedicine delivery capable of enhancing the efficacy of immune checkpoint blockades. Understanding the active targeting candidates of tumor microenvironment components and the associated treatment strategies could offer insights into the development of combination therapeutics boosting immune checkpoint blockades for clinical applications.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Humanos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Nanomedicina , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...