Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 16(7): 6588-6612, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38604156

RESUMEN

BACKGROUND: Liver progenitor cells (LPCs) are a subpopulation of cells that contribute to liver regeneration, fibrosis and liver cancer initiation under different circumstances. RESULTS: By performing adenoviral-mediated transfection, CCK-8 analyses, F-actin staining, transwell analyses, luciferase reporter analyses and Western blotting, we observed that TGF-ß promoted cytostasis and partial epithelial-mesenchymal transition (EMT) in LPCs. In addition, we confirmed that TGF-ß activated the Smad and MAPK pathways, including the Erk, JNK and p38 MAPK signaling pathways, and revealed that TGFß-Smad signaling induced growth inhibition and partial EMT, whereas TGFß-MAPK signaling had the opposite effects on LPCs. We further found that the activity of Smad and MAPK signaling downstream of TGF-ß was mutually restricted in LPCs. Mechanistically, we found that TGF-ß activated Smad signaling through serine phosphorylation of both the C-terminal and linker regions of Smad2 and 3 in LPCs. Additionally, TGFß-MAPK signaling inhibited the phosphorylation of Smad3 but not Smad2 at the C-terminus, and it reinforced the linker phosphorylation of Smad3 at T179 and S213. We then found that overexpression of mutated Smad3 at linker phosphorylation sites intensifies TGF-ß-induced cytostasis and EMT, mimicking the effects of MAPK inhibition in LPCs, whereas mutation of Smad3 at the C-terminus caused LPCs to blunt TGF-ß-induced cytostasis and partial EMT. CONCLUSION: These results suggested that TGF-ß downstream of Smad3 and MAPK signaling were mutually antagonistic in regulating the viability and partial EMT of LPCs. This antagonism may help LPCs overcome the cytostatic effect of TGF-ß under fibrotic conditions and maintain partial EMT and progenitor phenotypes.


Asunto(s)
Transición Epitelial-Mesenquimal , Hígado , Sistema de Señalización de MAP Quinasas , Proteína smad3 , Células Madre , Factor de Crecimiento Transformador beta , Proteína smad3/metabolismo , Células Madre/metabolismo , Animales , Factor de Crecimiento Transformador beta/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Hígado/metabolismo , Supervivencia Celular/efectos de los fármacos , Fosforilación , Ratones , Transducción de Señal
2.
Cancer Gene Ther ; 31(4): 586-598, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38267623

RESUMEN

Glutamate-NMDAR receptors (GRINs) have been reported to influence cancer immunogenicity; however, the relationship between GRIN alterations and the response to immune checkpoint inhibitors (ICIs) has not been determined. This study combined clinical characteristics and mutational profiles from multiple cohorts to form a discovery cohort (n = 901). The aim of this study was to investigate the correlation between the mutation status of the GRIN gene and the response to ICI therapy. Additionally, an independent ICI-treated cohort from the Memorial Sloan Kettering Cancer Center (MSKCC, N = 1513) was used for validation. Furthermore, this study explored the associations between GRIN2A mutations and intrinsic and extrinsic immunity using multiomics analysis. In the discovery cohort, patients with GRIN2A-MUTs had improved clinical outcomes, as indicated by a higher objective response rate (ORR: 36.8% vs 25.8%, P = 0.020), durable clinical benefit (DCB: 55.2% vs 38.7%, P = 0.005), prolonged progression-free survival (PFS: HR = 0.65; 95% CI 0.49 to 0.87; P = 0.003), and increased overall survival (OS: HR = 0.67; 95% CI 0.50 to 0.89; P = 0.006). Similar results were observed in the validation cohort, in which GRIN2A-MUT patients exhibited a significant improvement in overall survival (HR = 0.66; 95% CI = 0.49 to 0.88; P = 0.005; adjusted P = 0.045). Moreover, patients with GRIN2A-MUTs exhibited an increase in tumor mutational burden, high expression of costimulatory molecules, increased activity of antigen-processing machinery, and infiltration of various immune cells. Additionally, gene sets associated with cell cycle regulation and the interferon response were enriched in GRIN2A-mutated tumors. In conclusion, GRIN2A mutation is a novel biomarker associated with a favorable response to ICIs in multiple cancers.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Interferones , Mutación , Biomarcadores de Tumor/genética
3.
Adv Sci (Weinh) ; 11(13): e2307242, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38247171

RESUMEN

N6-methyladenosine (m6A) modification orchestrates cancer formation and progression by affecting the tumor microenvironment (TME). For hepatocellular carcinoma (HCC), immune evasion and angiogenesis are characteristic features of its TME. The role of YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), as an m6A reader, in regulating HCC TME are not fully understood. Herein, it is discovered that trimethylated histone H3 lysine 4 and H3 lysine 27 acetylation modification in the promoter region of YTHDF2 enhanced its expression in HCC, and upregulated YTHDF2 in HCC predicted a worse prognosis. Animal experiments demonstrated that Ythdf2 depletion inhibited spontaneous HCC formation, while its overexpression promoted xenografted HCC progression. Mechanistically, YTHDF2 recognized the m6A modification in the 5'-untranslational region of ETS variant transcription factor 5 (ETV5) mRNA and recruited eukaryotic translation initiation factor 3 subunit B to facilitate its translation. Elevated ETV5 expression induced the transcription of programmed death ligand-1 and vascular endothelial growth factor A, thereby promoting HCC immune evasion and angiogenesis. Targeting YTHDF2 via small interference RNA-containing aptamer/liposomes successfully both inhibited HCC immune evasion and angiogenesis. Together, this findings reveal the potential application of YTHDF2 in HCC prognosis and targeted treatment.


Asunto(s)
Aptámeros de Nucleótidos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas de Unión al ARN , Animales , Angiogénesis , Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Evasión Inmune , Neoplasias Hepáticas/genética , Lisina , Factores de Transcripción/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ADN/metabolismo
4.
Adv Sci (Weinh) ; 11(3): e2305081, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38009498

RESUMEN

Cancer vaccines hold great potential for clinical cancer treatment by eliciting T cell-mediated immunity. However, the limited numbers of antigen-presenting cells (APCs) at the injection sites, the insufficient tumor antigen phagocytosis by APCs, and the presence of a strong tumor immunosuppressive microenvironment severely compromise the efficacy of cancer vaccines. Trained innate immunity may promote tumor antigen-specific adaptive immunity. Here, a personalized cancer vaccine is developed by engineering the inactivated probiotic Escherichia coli Nissle 1917 to load tumor antigens and ß-glucan, a trained immunity inducer. After subcutaneous injection, the cancer vaccine delivering model antigen OVA (BG/OVA@EcN) is highly accumulated and phagocytosed by macrophages at the injection sites to induce trained immunity. The trained macrophages may recruit dendritic cells (DCs) to facilitate BG/OVA@EcN phagocytosis and the subsequent DC maturation and T cell activation. In addition, BG/OVA@EcN remarkably enhances the circulating trained monocytes/macrophages, promoting differentiation into M1-like macrophages in tumor tissues. BG/OVA@EcN generates strong prophylactic and therapeutic efficacy to inhibit tumor growth by inducing potent adaptive antitumor immunity and long-term immune memory. Importantly, the cancer vaccine delivering autologous tumor antigens efficiently prevents postoperative tumor recurrence. This platform offers a facile translatable strategy to efficiently integrate trained immunity and adaptive immunity for personalized cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Probióticos , Humanos , Inmunidad Entrenada , Células Dendríticas , Neoplasias/terapia , Antígenos de Neoplasias , Activación de Linfocitos , Probióticos/uso terapéutico , Microambiente Tumoral
5.
Signal Transduct Target Ther ; 8(1): 408, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37875473

RESUMEN

Immune checkpoint blockade (ICB) therapy, particularly antibodies targeting the programmed death receptor 1 (PD-1) and its ligand (PD-L1), has revolutionized cancer treatment. However, its efficacy as a standalone therapy remains limited. Although ICB therapy in combination with chemotherapy shows promising therapeutic responses, the challenge lies in amplifying chemotherapy-induced antitumor immunity effectively. This relies on efficient drug delivery to tumor cells and robust antigen presentation by dendritic cells (DCs). Here, we developed tumor-repopulating cell (TRC)-derived microparticles with exceptional tumor targeting to deliver doxorubicin (DOX@3D-MPs) for improve anti-PD-1 therapy. DOX@3D-MPs effectively elicit immunogenic tumor cell death to release sufficient tumor antigens. Heat shock protein 70 (HSP70) overexpressed in DOX@3D-MPs contributes to capturing tumor antigens, promoting their phagocytosis by DCs, and facilitating DCs maturation, leading to the activation of CD8+ T cells. DOX@3D-MPs significantly enhance the curative response of anti-PD-1 treatment in large subcutaneous H22 hepatoma, orthotopic 4T1 breast tumor and Panc02 pancreatic tumor models. These results demonstrate that DOX@3D-MPs hold promise as agents to improve the response rate to ICB therapy and generate long-lasting immune memory to prevent tumor relapse.


Asunto(s)
Antineoplásicos , Micropartículas Derivadas de Células , Neoplasias Pancreáticas , Humanos , Linfocitos T CD8-positivos , Doxorrubicina/farmacología , Neoplasias Pancreáticas/terapia , Antígenos de Neoplasias/genética , Antineoplásicos/uso terapéutico
6.
Adv Mater ; 35(45): e2211980, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37755231

RESUMEN

Tumor-cell-derived microparticles (MPs) can function as anticancer drug-delivery carriers. However, short blood circulation time, large-size-induced insufficient tumor accumulation and penetration into tumor parenchyma, as well as limited cellular internalization by tumor cells and cancer stem cells (CSCs), and difficult intracellular drug release restrict the anticancer activity of tumor-cell-derived MP-based drug-delivery systems. In this work, hydrophobicity-adaptive polymers based on poly(N-isopropylacrylamide) are anchored to tumor-cell-derived MPs for enhanced delivery of the anticancer drug doxorubicin (DOX). The polymers are hydrophilic in blood to prolong the circulation time of DOX-loaded MPs (DOX@MPs), while rapidly switching to hydrophobic at the tumor acidic microenvironment. The hydrophobicity of polymers drives the fission of tumor-cell-derived MPs to form small vesicles, facilitating tumor accumulation, deep tumor penetration, and efficient internalization of DOX@MPs into tumor cells and CSCs. Subsequently, the hydrophobicity of polymers in acidic lysosomes further promotes DOX release to nuclei for strong cytotoxicity against tumor cells and CSCs. The work provides a facile and simple strategy for improved anticancer drug delivery of tumor-cell-derived MPs.


Asunto(s)
Antineoplásicos , Micropartículas Derivadas de Células , Neoplasias , Humanos , Polímeros/química , Antineoplásicos/química , Doxorrubicina/química , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Interacciones Hidrofóbicas e Hidrofílicas , Portadores de Fármacos/química , Línea Celular Tumoral , Concentración de Iones de Hidrógeno , Microambiente Tumoral
7.
Nat Commun ; 14(1): 5653, 2023 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-37704614

RESUMEN

The durable response rate to immune checkpoint blockade such as anti-programmed cell death-1 (PD-1) antibody remains relatively low in hepatocellular carcinoma (HCC), mainly depending on an immunosuppressive microenvironment with limited number of CD8+ T cells, especially stem-like CD8+ T cells, in tumor tissues. Here we develop engineered microparticles (MPs) derived from alpha-fetoprotein (AFP)-overexpressing macrophages to load resiquimod (R848@M2pep-MPsAFP) for enhanced anti-PD-1 therapy in HCC. R848@M2pep-MPsAFP target and reprogram immunosuppressive M2-like tumor-associated macrophages (TAMs) into M1-like phenotype. Meanwhile, R848@M2pep-MPsAFP-reprogrammed TAMs act as antigen-presenting cells, not only presenting AFP antigen to activate CD8+ T cell-mediated antitumor immunity, but also providing an intra-tumoral niche to maintain and differentiate stem-like CD8+ T cells. Combination immunotherapy with anti-PD-1 antibody generates strong antitumor immune memory and induces abundant stem-like CD8+ T cell proliferation and differentiation to terminally exhausted CD8+ T cells for long-term immune surveillance in orthotopic and autochthonous HCC preclinical models in male mice. We also show that the R848-loaded engineered MPs derived from macrophages overexpressing a model antigen ovalbumin (OVA) can improve anti-PD-1 therapy in melanoma B16-OVA tumor-bearing mice. Our work presents a facile and generic strategy for personalized cancer immunotherapy to boost anti-PD-1 therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Animales , Ratones , Macrófagos Asociados a Tumores , alfa-Fetoproteínas , Carcinoma Hepatocelular/tratamiento farmacológico , Linfocitos T CD8-positivos , Neoplasias Hepáticas/terapia , Inmunosupresores , Antígenos de Neoplasias , Microambiente Tumoral
8.
Cell Death Dis ; 14(5): 303, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37142578

RESUMEN

Despite past extensive studies, the pathoetiologies underlying tumor metastasis remain poorly understood, which renders its treatment largely unsuccessful. The methyl-CpG-binding domain 2 (MBD2), a "reader" to interpret DNA methylome-encoded information, has been noted to be involved in the development of certain types of tumors, while its exact impact on tumor metastasis remains elusive. Herein we demonstrated that patients with LUAD metastasis were highly correlated with enhanced MBD2 expression. Therefore, knockdown of MBD2 significantly attenuated the migration and invasion of LUAD cells (A549 and H1975 cell lines) coupled with attenuated epithelial-mesenchymal transition (EMT). Moreover, similar results were observed in other types of tumor cells (B16F10). Mechanistically, MBD2 selectively bound to the methylated CpG DNA within the DDB2 promoter, by which MBD2 repressed DDB2 expression to promote tumor metastasis. As a result, administration of MBD2 siRNA-loaded liposomes remarkably suppressed EMT along with attenuated tumor metastasis in the B16F10 tumor-bearing mice. Collectively, our study indicates that MBD2 could be a promising prognostic marker for tumor metastasis, while administration of MBD2 siRNA-loaded liposomes could be a viable therapeutic approach against tumor metastasis in clinical settings.


Asunto(s)
Proteínas de Unión al ADN , Neoplasias , Animales , Ratones , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Metilación de ADN/genética , Liposomas , Línea Celular , ARN Interferente Pequeño/metabolismo , Neoplasias/genética
9.
Adv Sci (Weinh) ; 10(17): e2207080, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37096833

RESUMEN

Bone is the second leading metastatic site for hepatocellular carcinoma (HCC). Patients with HCC and bone metastasis suffer poor quality of life and reduced survival time. Extracellular vesicles (EVs) are widely involved in HCC formation and metastasis. However, the communication between primary HCC and bone lesions mediated by EVs remains unclear and the possible effect of bone metastasis on the progression of HCC remains largely unknown. Here, bone-metastasized HCC-derived EVs (BM-EVs) are found to localize to orthotropic HCC cells and promote HCC progression. Mechanistically, miR-3190-5p (miR-3190) is upregulated in intracellular HCC cells isolated from bone lesions as well as in their derived EVs. miR-3190 in BM-EVs is transferred into orthotopic tumor cells and enhances their metastatic capacity by downregulating AlkB homolog 5 (ALKBH5) expression. Decreased level of ALKBH5 exacerbates the prometastatic characteristics of HCC by modulating gene expression in N6-methyladenosine-dependent and -independent ways. Finally, antagomir-miR-3190-loaded liposomes with HCC affinity successfully suppress HCC progression in mice treated with BM-EVs. These findings reveal that BM-EVs initiate prometastatic cascades in orthotopic HCC by transferring ALKBH5-targeting miR-3190 and miR-3190 is serving as a promising therapeutic target for inhibiting the progression of HCC in patients with bone metastasis.


Asunto(s)
Neoplasias Óseas , Carcinoma Hepatocelular , Vesículas Extracelulares , Neoplasias Hepáticas , MicroARNs , Animales , Ratones , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Calidad de Vida , Vesículas Extracelulares/metabolismo , Línea Celular Tumoral , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo
10.
Adv Drug Deliv Rev ; 188: 114450, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35841955

RESUMEN

Extracellular vesicles (EVs), including microparticles and exosomes, have emerged as potential tools for tumor targeting delivery during the past years. Recently, mass of strategies are applied to assist EVs to accumulate and penetrate into deep tumor sites. In this review, EVs from different cells with unique innate characters and engineered approaches (e.g. chemical engineering, genetical engineering and biomimetic engineering) as drug delivery systems to enhance tumor accumulation and penetration are summarized. Meanwhile, efficient biological function modulation (e.g. extracellular matrix degradation, mechanical property regulation and transcytosis) is introduced to facilitate tumor accumulation and penetration of EVs. Finally, the prospects and challenges on further clinical applications of EVs are discussed.


Asunto(s)
Exosomas , Vesículas Extracelulares , Neoplasias , Sistemas de Liberación de Medicamentos , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Transcitosis
11.
Adv Mater ; 34(52): e2201054, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35726204

RESUMEN

Although immunotherapy harnessing activity of the immune system against tumors has made great progress, the treatment efficacy remains limited in most cancers. Current anticancer immunotherapy is primarily based on T-cell-mediated cellular immunity, which highly relies on efficiency of triggering the cancer-immunity cycle, namely, tumor antigen release, antigen presentation by antigen presenting cells, T cell activation, recruitment and infiltration of T cells into tumors, and recognition and killing of tumor cells by T cells. Unfortunately, these immunotherapies are restricted by inefficient drug delivery and acting on only a single step of the cancer-immunity cycle. Due to high biocompatibility, low immunogenicity, intrinsic cell targeting, and easy chemical and genetic manipulation, extracellular vesicle (EV)-based drug delivery systems are widely used to amplify anticancer immune responses by serving as an integrated platform for multiple drugs or therapeutic strategies to synergistically activate several steps of cancer-immunity cycle. This review summarizes various mechanisms related to affecting cancer-immunity cycle disorders. Meanwhile, preparation and application of EV-based drug delivery systems in modulating cancer-immunity cycle are introduced, especially in the improvement of T cell recruitment and infiltration into tumors. Finally, opportunities and challenges of EV-based drug delivery systems in translational clinical applications are briefly discussed.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Antígenos de Neoplasias , Inmunoterapia
12.
Theranostics ; 12(7): 3503-3517, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35547751

RESUMEN

The perioperative trauma-related platelet recruitment and activation severely affect tumor recurrence and metastasis. Therefore, efficiently killing residual tumor cells and simultaneously inhibiting platelet activation to block platelet-cancer cell interaction might be a promising strategy to prevent postoperative tumor recurrence and metastasis. Methods: Biodegradable PLGA electrospun nanofibrous films co-delivering doxorubicin-loaded tumor repopulating cell-derived microparticles (DOX-MPs) and aspirin (ASA) were developed as the implant materials (DOX-MPs/ASA@NF) for postoperative in-situ treatment. The characterization, cytotoxicity against tumor cells, inhibition in platelet activation-triggered proliferation, migration and metastasis of tumor cells and in vivo anti-recurrence and anti-metastasis activity induced by DOX-MPs/ASA@NF were systematically evaluated. Results: PLGA nanofibrous films facilitate the enhanced distribution of DOX-MPs as well as DOX-MPs and ASA release in a time-programmed manner within the tumor resection cavity. The released DOX-MPs efficiently kill the residual tumor cells, while ASA decreases platelet activation and inhibits platelet-promoted proliferation, migration and metastasis of tumor cells, resulting in the remarkable inhibition of postoperative tumor recurrence and metastasis. Conclusions: DOX-MPs/ASA@NF may be a promising candidate to prevent the recurrence and metastasis of resectable tumors.


Asunto(s)
Nanofibras , Línea Celular Tumoral , Doxorrubicina/farmacología , Humanos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/prevención & control , Neoplasia Residual/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria
13.
Nat Commun ; 13(1): 2794, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589680

RESUMEN

Insufficient tumor accumulation and distribution of photosensitizers as well as low antitumor immunity severely restrict the therapeutic efficacy of photothermal therapy (PTT). Cancer-associated fibroblasts (CAFs) play a key role in tumor extracellular matrix (ECM) remodeling and immune evasion. Reshaping tumor microenvironment via CAF regulation might provide a potential approach for complete tumor elimination in combination with PTT. Here, tumor cell-derived microparticles co-delivering calcipotriol and Indocyanine green (Cal/ICG@MPs) are developed to modulate CAFs for improved PTT efficacy. Cal/ICG@MPs efficiently target tumor tissues and regulate CAFs to reduce tumor ECM, resulting in enhanced tumor accumulation and penetration of ICG to generate strong PTT efficacy and activate CD8+ T cell-mediated antitumor immunity. In addition, Cal/ICG@MPs-triggered CAF regulation enhances tumor infiltration of CD8+ T cells and ameliorates CAF-induced antigen-mediated activation-induced cell death of tumor-specific CD8+ T cells in response to PTT, eliciting long-term antitumor immune memory to inhibit tumor recurrence and metastasis. Our results support Cal/ICG@MPs as a promising drug to improve PTT efficacy in cancer treatment.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Verde de Indocianina/farmacología , Neoplasias/terapia , Terapia Fototérmica , Recurrencia
15.
Theranostics ; 11(4): 1937-1952, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33408790

RESUMEN

Development of efficient therapeutic strategy to incorporate ultrasound (US)-triggered sonodynamic therapy (SDT) and ferroptosis is highly promising in cancer therapy. However, the SDT efficacy is severely limited by the hypoxia and high glutathione (GSH) in the tumor microenvironment, and ferroptosis is highly associated with reactive oxygen species (ROS) and GSH depletion. Methods: A manganese porphyrin-based metal-organic framework (Mn-MOF) was constructed as a nanosensitizer to self-supply oxygen (O2) and decrease GSH for enhanced SDT and ferroptosis. In vitro and in vivo analysis, including characterization, O2 generation, GSH depletion, ROS generation, lipid peroxidation, antitumor efficacy and tumor immune microenvironment were systematically evaluated. Results: Mn-MOF exhibited catalase-like and GSH decreasing activity in vitro. After efficient internalization into cancer cells, Mn-MOF persistently catalyzed tumor-overexpressed H2O2 to in-situ produce O2 to relieve tumor hypoxia and decrease GSH and GPX4, which facilitated the formation of ROS and ferroptosis to kill cancer cells upon US irradiation in hypoxic tumors. Thus, strong anticancer and anti-metastatic activity was found in H22 and 4T1 tumor-bearing mice after a single administration of Mn-MOF upon a single US irradiation. In addition, Mn-MOF showed strong antitumor immunity and improved immunosuppressive microenvironment upon US irradiation by increasing the numbers of activated CD8+ T cells and matured dendritic cells and decreaing the numbers of myeloid-derived suppressor cells in tumor tissues. Conclusions: Mn-MOF holds great potential for hypoxic cancer therapy.


Asunto(s)
Carcinoma Hepatocelular/terapia , Ferroptosis , Manganeso/química , Estructuras Metalorgánicas/farmacología , Porfirinas/química , Hipoxia Tumoral , Terapia por Ultrasonido/métodos , Animales , Apoptosis , Carcinoma Hepatocelular/patología , Proliferación Celular , Femenino , Glutatión/metabolismo , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Estructuras Metalorgánicas/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nat Commun ; 12(1): 440, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33469052

RESUMEN

The main challenges for programmed cell death 1(PD-1)/PD-1 ligand (PD-L1) checkpoint blockade lie in a lack of sufficient T cell infiltration, tumor immunosuppressive microenvironment, and the inadequate tumor accumulation and penetration of anti-PD-1/PD-L1 antibody. Resetting tumor-associated macrophages (TAMs) is a promising strategy to enhance T-cell antitumor immunity and ameliorate tumor immunosuppression. Here, mannose-modified macrophage-derived microparticles (Man-MPs) loading metformin (Met@Man-MPs) are developed to efficiently target to M2-like TAMs to repolarize into M1-like phenotype. Met@Man-MPs-reset TAMs remodel the tumor immune microenvironment by increasing the recruitment of CD8+ T cells into tumor tissues and decreasing immunosuppressive infiltration of myeloid-derived suppressor cells and regulatory T cells. More importantly, the collagen-degrading capacity of Man-MPs contributes to the infiltration of CD8+ T cells into tumor interiors and enhances tumor accumulation and penetration of anti-PD-1 antibody. These unique features of Met@Man-MPs contribute to boost anti-PD-1 antibody therapy, improving anticancer efficacy and long-term memory immunity after combination treatment. Our results support Met@Man-MPs as a potential drug to improve tumor resistance to anti-PD-1 therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Micropartículas Derivadas de Células/inmunología , Portadores de Fármacos/farmacología , Neoplasias/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Memoria Inmunológica , Masculino , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Células RAW 264.7 , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Control Release ; 328: 562-574, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32946875

RESUMEN

Extracellular vesicles (EVs), a heterogeneous population of membrane vesicles, are secreted by almost all living cells and contain different proteins, lipids and nucleic acid depending on their sources. Recently, numerous reports indicated EVs could interact with diverse cells in tumor microenvironment and influence both physiological and pathological conditions. Given that EVs regulate tumor immune microenvironment, EVs-based strategies are explored for anti-tumor immunotherapy. In this review, about nine kinds of EVs derived from diverse cells are indicated. The biological functions in tumor immunity, strategies for anti-tumor immunotherapy and further clinical trials of these EVs will be introduced in detail.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Sistemas de Liberación de Medicamentos , Humanos , Inmunoterapia , Neoplasias/terapia , Microambiente Tumoral
18.
Chem Commun (Camb) ; 56(46): 6171-6188, 2020 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-32478343

RESUMEN

Within the past few years, cell derived microparticles (MPs) have emerged as a highly potent natural drug delivery system for tumor therapy. We and others have engineered different cells to obtain eleven kinds of MPs, which efficiently delivered antitumor agents to recipient cells and achieved ideal treatment outcomes in a great number of rodent tumor models. More significantly, autologous tumor cell derived MPs packaging chemotherapeutic drugs were demonstrated to be safe and tolerable and accomplished decent objective clinical response in lung cancer patients in clinical settings, leading to their approval as a novel biotherapy for treating malignancies in China. In this feature article, we review MP mediated tumor targeting delivery. The impact of tumor cell derived MPs on tumor progression and metastasis will be briefly summarized. Different means to prepare, label and characterize MPs will be introduced. Advantages and limitations of distinctive cargo encapsulation strategies will be outlined and compared. Tumor cell MP mediated in vivo transport processes will be reviewed comprehensively. Representative MPs shed by platelets, endothelial cells, macrophages, and dendritic cells will be selected to showcase their advantages in tumor targeting delivery. Multifarious therapeutic agents, including chemotherapeutic drugs, oncolytic adenovirus, nucleic acids, antigens, antibodies, to name a few, have been transported to their targets for corresponding cancer therapies. Current challenges and future opportunities on translating MPs for tumor targeting delivery will be provided in the end.


Asunto(s)
Micropartículas Derivadas de Células , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Animales , Humanos
19.
J Control Release ; 322: 555-565, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32246977

RESUMEN

Extracellular vesicles (EVs) are membrane vesicles with size of nanometers to several micrometers, which are released by most eukaryotic and prokaryotic cells. Recently, EVs-based nanomedicines have achieved remarkable attentions. Five features need to be satisfied for EVs-based nanomedicine to achieve desirable anticancer activity after systemic administration, including long circulation, enhanced tumor accumulation, deep tumor penetration, efficient cellular internalization and drug release. In this review, we summarize recently reported EVs-based nanomedicines for efficient drug delivery based on our defined five features principle theory. Even though EVs-based nanomedicines are in infancy, it is expectable that they exhibit promising prospects for cancer treatment.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Sistemas de Liberación de Medicamentos , Humanos , Nanomedicina , Neoplasias/tratamiento farmacológico
20.
Chem Soc Rev ; 49(8): 2273-2290, 2020 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-32215407

RESUMEN

Modulating nanomedicine mechanical properties for enhanced drug delivery to tumors has attracted increasing attention in the past few decades. In this tutorial review, we analyze the impact of nanomedicine mechanical properties on in vivo transport processes and highlight the most recent advances in drug delivery efficiency and antitumor efficacy. Typical nanoparticles that have been explored for this purpose since 2000 are summarized while the methods to tune and the techniques to characterize nanomedicine mechanical properties are introduced. In the end, challenges and perspectives on tailoring nanomedicine mechanical properties for tumor targeting delivery are discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fenómenos Mecánicos , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...