Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ACS Nano ; 18(34): 23497-23507, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39146387

RESUMEN

Colorectal cancer (CRC) is a major global health concern, and the development of effective treatment strategies is crucial. Enzyme prodrug therapy (EPT) shows promise in combating tumors but faces challenges in achieving sustained expression of therapeutic enzymes and optimal biological distribution. To address these issues, a fungi-triggered in situ chemotherapeutics generator (named as SC@CS@5-FC) was constructed via oral delivery of a prodrug (5-fluorocytosine, 5-FC) for the treatment of orthotopic colorectal tumor. When SC@CS@5-FC targets the tumor through tropism by Saccharomyces cerevisiae (SC), the chemotherapeutic generator could be degraded under abundant hyaluronidase (HAase) in the tumor microenvironment by an enzyme-responsive gate to release prodrug (5-FC). And nontoxic 5-FC was catalyzed to toxic chemotherapy drug 5-fluorouracil (5-FU) by cytosine deaminase (CD) of SC. Meanwhile, SC and zinc-coordinated chitosan nanoparticles could be used as immune adjuvants to activate antigen-presenting cells and further enhance the therapeutic effect. Our results demonstrated that SC@CS@5-FC could effectively inhibit tumor growth and prolong mouse survival in an orthotopic colorectal cancer model. This work utilizes living SC as a dynamotor and positioning system for the chemotherapeutic generator SC@CS@5-FC, providing a strategy for oral enzyme prodrug therapy for the treatment of orthotopic colorectal.


Asunto(s)
Neoplasias Colorrectales , Flucitosina , Fluorouracilo , Inmunoterapia , Profármacos , Saccharomyces cerevisiae , Profármacos/química , Profármacos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Animales , Ratones , Humanos , Flucitosina/farmacología , Flucitosina/química , Administración Oral , Fluorouracilo/farmacología , Fluorouracilo/química , Fluorouracilo/administración & dosificación , Citosina Desaminasa/metabolismo , Quitosano/química , Antineoplásicos/farmacología , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Hialuronoglucosaminidasa/metabolismo , Ratones Endogámicos BALB C , Nanopartículas/química , Ensayos de Selección de Medicamentos Antitumorales
2.
Nat Commun ; 15(1): 7096, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39154092

RESUMEN

The intratumor microbiome imbalance in pancreatic cancer promotes a tolerogenic immune response and triggers immunotherapy resistance. Here we show that Lactobacillus rhamnosus GG probiotics, outfitted with a gallium-polyphenol network (LGG@Ga-poly), bolster immunotherapy in pancreatic cancer by modulating microbiota-immune interactions. Upon oral administration, LGG@Ga-poly targets pancreatic tumors specifically, and selectively eradicates tumor-promoting Proteobacteria and microbiota-derived lipopolysaccharides through a gallium-facilitated disruption of bacterial iron respiration. This elimination of intratumor microbiota impedes the activation of tumoral Toll-like receptors, thus reducing immunosuppressive PD-L1 and interleukin-1ß expression by tumor cells, diminishing immunotolerant myeloid populations, and improving the infiltration of cytotoxic T lymphocytes in tumors. Moreover, LGG@Ga-poly hampers pancreatic tumor growth in both preventive and therapeutic contexts, and amplifies the antitumor efficacy of immune checkpoint blockade in preclinical cancer models in female mice. Overall, we offer evidence that thoughtfully designed biomaterials targeting intratumor microbiota can efficaciously augment immunotherapy for the challenging pancreatic cancer.


Asunto(s)
Galio , Lacticaseibacillus rhamnosus , Microbiota , Neoplasias Pancreáticas , Polifenoles , Probióticos , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/microbiología , Animales , Probióticos/administración & dosificación , Ratones , Femenino , Humanos , Lacticaseibacillus rhamnosus/inmunología , Polifenoles/farmacología , Microbiota/inmunología , Microbiota/efectos de los fármacos , Línea Celular Tumoral , Inmunoterapia/métodos , Ratones Endogámicos C57BL , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Linfocitos T Citotóxicos/inmunología
3.
Adv Mater ; 36(6): e2305384, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37672674

RESUMEN

Adoptive cell therapy has emerged as a promising approach for cancer treatment. However, the transfer of macrophages exhibits limited efficacy against solid tumors due to the dynamic cellular phenotypic shift from antitumor to protumor states within the immunosuppressive tumor microenvironment. In this study, a strategy of attaching bacteria to macrophages (Mø@bac) is reported that endows adoptively infused macrophages with durable stimulation by leveraging the intrinsic immunogenicity of bacteria. These attached bacteria, referred to as backpacks, are encapsulated with adhesive nanocoatings and can sustainably control the cellular phenotypes in vivo. Moreover, Mø@bac can repolarize endogenous tumor-associated macrophages, leading to a more robust immune response and thus reducing the tumor progression in a murine 4T1 cancer model without any side effects. This study utilizing bacteria as cellular backpacks opens a new avenue for the development of cell therapies.


Asunto(s)
Neoplasias , Ratones , Animales , Neoplasias/patología , Macrófagos , Traslado Adoptivo , Bacterias , Microambiente Tumoral , Inmunoterapia
4.
ACS Nano ; 17(24): 24947-24960, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38055727

RESUMEN

Cancer vaccines have been considered to be an alternative therapeutic strategy for tumor therapy in the past decade. However, the popularity and efficacy of cancer vaccines were hampered by tumor antigen heterogeneity and the impaired function of cross-presentation in the tumor-infiltrating dendritic cells (TIDCs). To overcome these challenges, we engineered an in situ nanovaccine (named as TPOP) based on lipid metabolism-regulating and innate immune-stimulated nanoparticles. TPOP could capture tumor antigens and induce specific recognition by TIDCs to be taken up. Meanwhile, TPOP could manipulate TIDC lipid metabolism and inhibit de novo synthesis of fatty acids, thus improving the ability of TIDCs to cross-present by reducing their lipid accumulation. Significantly, intratumoral injection of TPOP combined with pretreatment with doxorubicin showed a considerable therapeutic effect in the subcutaneous mouse colorectal cancer model and melanoma model. Moreover, in combination with immune checkpoint inhibitors, such TPOP could markedly inhibit the growth of distant tumors by systemic antitumor immune responses. This work provides a safe and promising strategy for improving the function of immune cells by manipulating their metabolism and activating the immune system effectively for in situ cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Nanopartículas , Neoplasias , Ratones , Animales , Nanovacunas , Células Dendríticas , Metabolismo de los Lípidos , Inmunoterapia , Neoplasias/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Antígenos de Neoplasias/metabolismo , Modelos Animales de Enfermedad
5.
Adv Mater ; 35(22): e2300977, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37029611

RESUMEN

Despite the recognition that the gut microbiota acts a clinically significant role in cancer chemotherapy, both mechanistic understanding and translational research are still limited. Maximizing drug efficacy requires an in-depth understanding of how the microbiota contributes to therapeutic responses, while microbiota modulation is hindered by the complexity of the human body. To address this issue, a 3D experimental model named engineered microbiota (EM) is reported for bridging microbiota-drug interaction research and therapeutic decision-making. EM can be manipulated in vitro and faithfully recapitulate the human gut microbiota at the genus/species level while allowing co-culture with cells, organoids, and isolated tissues for testing drug responses. Examination of various clinical and experimental drugs by EM reveales that the gut microbiota affects drug efficacy through three pathways: immunological effects, bioaccumulation, and drug metabolism. Guided by discovered mechanisms, custom-tailored strategies are adopted to maximize the therapeutic efficacy of drugs on orthotopic tumor models with patient-derived gut microbiota. These strategies include immune synergy, nanoparticle encapsulation, and host-guest complex formation, respectively. Given the important role of the gut microbiota in influencing drug efficacy, EM will likely become an indispensable tool to guide drug translation and clinical decision-making.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Hidrogeles/farmacología , Interacciones Farmacológicas , Modelos Teóricos
6.
Adv Mater ; 32(45): e2004529, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33006175

RESUMEN

While microbial-based therapy has been considered as an effective strategy for treating diseases such as colon cancer, its safety remains the biggest challenge. Here, probiotics and prebiotics, which possess ideal biocompatibility and are extensively used as additives in food and pharmaceutical products, are combined to construct a safe microbiota-modulating material. Through the host-guest chemistry between commercial Clostridium butyricum and chemically modified prebiotic dextran, prebiotics-encapsulated probiotic spores (spores-dex) are prepared. It is found that spores-dex can specifically enrich in colon cancers after oral administration. In the lesion, dextran is fermented by C. butyricum, and thereby produces anti-cancer short-chain fatty acids (SCFAs). Additionally, spores-dex regulate the gut microbiota, augment the abundance of SCFA-producing bacteria (e.g., Eubacterium and Roseburia), and markedly increase the overall richness of microbiota. In subcutaneous and orthotopic tumor models, drug-loaded spores-dex inhibit tumor growth up to 89% and 65%, respectively. Importantly, no obvious adverse effect is found. The work sheds light on the possibility of using a highly safe strategy to regulate gut microbiota, and provides a promising avenue for treating various gastrointestinal diseases.


Asunto(s)
Neoplasias del Colon/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Prebióticos , Probióticos/farmacología , Esporas/fisiología , Neoplasias del Colon/tratamiento farmacológico , Dextranos/química , Humanos , Probióticos/química , Seguridad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA