Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
ACS Appl Mater Interfaces ; 15(19): 22843-22853, 2023 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-37133278

RESUMEN

Cancer nanomedicine treatment aims to achieve highly specific targeting and localization to cancer cells. Coating of nanoparticles with cell membranes endows them with homologous cellular mimicry, enabling nanoparticles to acquire new functions and properties, including homologous targeting and long circulation in vivo, and can enhance internalization by homologous cancer cells. Herein, we fused a human-derived HCT116 colon cancer cell membrane (cM) with a red blood cell membrane (rM) to fabricate an erythrocyte-cancer cell hybrid membrane (hM). Oxaliplatin and chlorin e6 (Ce6) co-encapsulated reactive oxygen species-responsive nanoparticles (NPOC) were camouflaged by hM and obtained a hybrid biomimetic nanomedicine (denoted as hNPOC) for colon cancer therapy. hNPOC exhibited prolonged circulation time and recognized homologous targeting ability in vivo since both rM and HCT116 cM proteins were maintained on the hNPOC surface. hNPOC showed enhanced homologous cell uptake in vitro and considerable homologous self-localization in vivo, producing effective synergistic chemophotodynamic therapy efficacy under irradiation with a homologous HCT116 tumor compared to that with a heterologous tumor. Together, the biomimetic hNPOC nanoparticles showed prolonged blood circulation and preferential cancer cell-targeted function in vivo to provide a bioinspired strategy for chemophotodynamic synergistic therapy of colon cancer.


Asunto(s)
Neoplasias del Colon , Nanopartículas , Humanos , Biónica , Membrana Eritrocítica/metabolismo , Fototerapia , Neoplasias del Colon/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral
2.
Acta Biomater ; 162: 98-109, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36931417

RESUMEN

Sorafenib is the first line drug for hepatocellular carcinoma (HCC) therapy. However, HCC patients usually acquire resistance to sorafenib treatment within 6 months. Recent evidences have shown that anticancer drugs with antiangiogenesis effect (e.g., sorafenib) can aggravate the hypoxia microenvironment and promote the infiltration of more tumor-associated macrophages (TAMs) into the tumor tissues. Therefore, repolarization of TAMs phenotype could be expected to not only eliminate the influence of TAMs on sorafenib lethality to HCC cells, but also provide an additional anticancer effect to achieve combination therapy. However, immune side effects remain a great challenge due to the non-specific macrophage repolarization in normal tissues. We herein employed a tumor microenvironment (TME) pH-responsive nanoplatform to concurrently transport sorafenib and modified resiquimod (R848-C16). This nanoparticle (NP) platform is made with a TME pH-responsive methoxyl-poly(ethylene glycol)-b-poly(lactic-co-glycolic acid) copolymer. After intravenous administration, the co-delivery NPs could highly accumulate in the tumor tissues and then respond to the TME pH to detach their surface PEG chains. With this PEG detachment to enhance uptake by TAMs and HCC cells, the co-delivery NPs could combinatorially inhibit HCC tumor growth via sorafenib-mediated lethality to HCC cells and R848-mediated repolarization of TAMs into tumoricidal M1-like macrophages. STATEMENT OF SIGNIFICANCE: Anticancer drugs with antiangiogenesis effect (e.g., sorafenib) can aggravate the hypoxia microenvironment and promote the infiltration of more tumor-associated macrophages (TAMs) into the tumor tissues to restrict the anticancer effect. In this work, we designed and developed a tumor microenvironment (TME) pH-responsive nanoplatform for systemic co-delivery of sorafenib and resiquimod in hepatocellular carcinoma (HCC) therapy. These co-delivery NPs show high tumor accumulation and could respond to the TME pH to enhance uptake by TAMs and HCC cells. With the sorafenib-mediated lethality to HCC cells and R848-mediated repolarization of TAMs, the co-delivery NPs show a combinational inhibition of HCC tumor growth in both xenograft and orthotopic tumor models.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Humanos , Sorafenib , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Inhibidores de la Angiogénesis/uso terapéutico , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Macrófagos/patología , Microambiente Tumoral , Nanopartículas/uso terapéutico
3.
Nanoscale ; 15(1): 365-375, 2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36508179

RESUMEN

Multifunctional magnet-fluorescent nanocomposites are widely applied in biomedical applications. Incorporating biocompatible quantum dots with highly ferrimagnetic magnetic nanoparticles into one nanoplatform for achieving efficient magnetic hyperthermia therapy (MHT) is very important. Herein, we reported an amphiphilic block copolymer with a flowable hydrophobic chain to encapsulate highly ferrimagnetic magnetic nanoparticles and ZnS/InP quantum dots via a facile self-assembly method. The obtained ferrimagnetic fluorescent micelle (FMFM) exhibited a uniform diameter of about 180 nm. In stark contrast, larger aggregation (400 nm in diameter) inevitably occurred using common poly(D,L-lactide) (PLA)-based amphiphilic block copolymer with a rigid hydrophobic chain, which was readily cleared by the reticuloendothelial system (RES). The flowable FMFM exhibited long-term colloidal stability within one month and desired fluorescent stability within 84 h. Benefiting from the high ferrimagnetism, the FMFM revealed excellent magnetic heating effect and magnetic resonance imaging capability. With accurate manipulation under an external magnetic field, FMFM realized in vitro enhanced fluorescence imaging sensitivity and accumulation efficiency at the tumor region, achieving in vitro and vivo improved MHT efficacy.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Puntos Cuánticos , Micelas , Polímeros/química
4.
Biomater Sci ; 9(9): 3516-3525, 2021 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-33949443

RESUMEN

Local administration of therapeutic agents with long-term retention capabilities efficiently avoids nonspecific distribution in normal organs with an increased drug concentration in pathological tissue. Herein, we developed an injectable and degradable alginate-calcium (Ca2+) hydrogel for the local administration of corn-like Au/Ag nanorods (NRs) and doxorubicin hydrochloride (DOX·HCl). The immobilized Au/Ag NRs with strong absorbance in the near-infrared II (NIR-II) window efficiently ablated the majority of tumor cells after 1064 nm laser irradiation and triggered the release of DOX to kill residual tumor cells. As a result, injectable hydrogel-mediated NIR-II photothermal therapy (PTT) and chemotherapy efficiently inhibited tumor growth, resulting in the complete eradication of tumors in most of the treated mice. Furthermore, owing to the confinement of the Au/Ag NRs and DOX·HCl within the hydrogel, such treatment exhibited excellent biocompatibility.


Asunto(s)
Hipertermia Inducida , Neoplasias , Animales , Línea Celular Tumoral , Doxorrubicina , Oro , Hidrogeles , Hipertermia , Ratones , Neoplasias/terapia
5.
Biomaterials ; 268: 120582, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33302120

RESUMEN

Immune checkpoint blocking (ICB) antibodies have shown great success in the clinic, but their low response rate in patients with immunosuppressive cold tumors remains a huge challenge. Inspired by the capability of immunogenic cell death (ICD) to convert tumors from cold to hot, we developed a corn-like Au/Ag nanorod (NR) that can induce the ICD of tumor cells under 1064-nm light irradiation. The corn-like Au/Ag NRs plus NIR-II light irradiation strikingly increased the tumor infiltration of T cells and provoked a systemic immune response to reprogram the immunosuppressive cold tumor microenvironment; these NRs synergized with ICB antibodies to efficiently inhibit distant tumor growth. Encouragingly, the combination of aCTLA4 and Au/Ag NRs plus 1064-nm light irradiation elicited a strong immunological memory effect that protected against tumor recurrence.


Asunto(s)
Nanotubos , Fotoquimioterapia , Oro , Humanos , Plata , Microambiente Tumoral , Zea mays
6.
Nat Commun ; 11(1): 1126, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111847

RESUMEN

The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery.


Asunto(s)
Quimiotaxis de Leucocito , Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Neoplasias/terapia , Neutrófilos/fisiología , Fototerapia , Animales , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/inmunología , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/química , Cisplatino/farmacocinética , Liberación de Fármacos , Vesículas Extracelulares/química , Vesículas Extracelulares/inmunología , Inmunoterapia Adoptiva , Inflamación/etiología , Ratones , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fototerapia/efectos adversos , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biomaterials ; 237: 119835, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32035321

RESUMEN

Hollow nanostructures have been evoked considerable attention owing to their intriguing hollow interior for important and potential applications in drug delivery, lithium battery, catalysis and etc. Herein, Bi2S3 hollow microspheres with rod-based urchin-like nanostructures (denoted as U-BSHM) were synthesized through a facile and rapid ion exchanging method using a particular hard template. The growth mechanism of the U-BSHM has been investigated and illustrated by the morphological evolution of the different samples at early stages. The obtained U-BSHM exhibited strong and wide UV-vis-NIR absorption ability and outstanding photothermal conversion efficiency. Thus, the U-BSHM can be used as spatio-temporal precisely controlled carrier by loading the mixture of 1-tetradecanol (phase change material, PCM) with melting point around 38 °C and hydrophilic chemotherapeutic doxorubicin hydrochloride (denoted as DOX) into the hollow interior to form (PCM + DOX)@Bi2S3 nanocomposites (denoted as PD@BS) for photoacoustic (PA) imaging and chemo-photothermal therapy of the tumors. When exposed to 808 nm near infrared light (NIR) laser irradiation, this nanocomposites could elevate the temperature of the surroundings by absorption and conversion of the NIR photons into heat energy, which inducing the triggered release of DOX from the hollow interior once the temperature reach up to the melting point of PCM. The killing efficiency of the chemo-photothermal therapy was systematically validated both in vitro and in vivo. In the meanwhile, the implanted tumor was completely restrained through PA imaging and combined therapies. Therefore, this kind of urchin-like hollow nanostructures would be used as important candidates for the multimodal bioimaging and therapy of tumors.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Preparaciones Farmacéuticas , Técnicas Fotoacústicas , Línea Celular Tumoral , Doxorrubicina , Liberación de Fármacos , Humanos , Microesferas , Fototerapia , Terapia Fototérmica
8.
Biomaterials ; 240: 119845, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32085974

RESUMEN

Photodynamic therapy (PDT) can destroy local tumor cells and induce effective antitumor immune responses, and has been applied in the treatment of patients with superficial solid tumors. Numerous systemic side effects of PDT, such as pain and skin photosensitivity, however, limit this therapeutic option. In addition, the immunosuppressive tumor microenvironment has been found to be another critical barrier for the antitumor immunity induced by PDT. Therefore, effectively enhancing the cytotoxicity to tumor cells of low-dose PDT and inhibiting the tumor immunosuppressive tumor microenvironment may be a feasible strategy to overcome these drawbacks of PDT. Here, a sorafenib and chlorin e6 co-loaded reactive oxygen species (ROS)-responsive nanoparticle (NP-sfb/ce6) is developed to improve antitumor responses by intratumoral release of sorafenib at the time of PDT. Under 660-nm laser irradiation, ROS produced by chlorin e6 (ce6) destruct the nanoparticles, resulting in boosted sorafenib cascade release. The rapidly released sorafenib acts synergistically with the low-dose PDT to inhibit tumor growth by inducing strong T cell-dependent local and systemic antitumor immune responses, reprograming the tumor immune microenvironment, and limiting the interaction between cytotoxic CD8+ T cells and immunosuppressive cells. This study provides new avenues for cascade-amplifying antitumor effects of photodynamic therapy.


Asunto(s)
Nanopartículas , Fotoquimioterapia , Porfirinas , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Inmunoterapia , Fármacos Fotosensibilizantes/uso terapéutico , Sorafenib
9.
Biomaterials ; 155: 103-111, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29175079

RESUMEN

Near-infrared (NIR) light-induced photothermal therapy (PTT) has attracted much interest in recent years. In the NIR region, tissue penetration ability of the second biological near-infrared window (1000-1350 nm) is recognized to be stronger than that of the first window (650-950 nm). However, NIR light absorbers in the second NIR region (NIR-II) have been scant even though various NIR light absorbers in the first NIR region (NIR-I) have been widely explored. In this work, a thieno-isoindigo derivative-based semiconducting polymer, PBTPBF-BT, were formulated into PEGylated nanoparticles. The obtained nanoparticle NPPBTPBF-BT exhibited strong absorption in NIR-II region, inherent high photothermal conversion efficacy, and excellent photostability. The in vitro and in vivo PTT study employing 1064 nm laser in NIR-II window revealed that NPPBTPBF-BT could efficiently ablate tumor cell at a power density of 0.42 W/cm2 (the skin tolerance threshold value). Moreover, NPPBTPBF-BT with excellent photostability exhibited enhanced photoacoustic (PA) imaging of tumor in living mice, suggesting the great probability of using NPPBTPBF-BT for in vivo PA imaging-guided PTT in the NIR-II window.


Asunto(s)
Nanopartículas/química , Polímeros/química , Semiconductores , Animales , Línea Celular Tumoral , Hipertermia Inducida , Rayos Infrarrojos , Rayos Láser , Ratones , Técnicas Fotoacústicas/métodos
10.
Colloids Surf B Biointerfaces ; 152: 449-458, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28187379

RESUMEN

Recently, combined photothermal-chemo therapy has attracted great attention due to its enhanced anti-tumor efficiency via synergistic effects. Herein, PEGylated cuprous telluride nanocrystals (PEGylated Cu2Te NCs) were developed as novel drug nanocarriers for combined photothermal-chemo treatment of cancer cells. PEGylated Cu2Te NCs were fabricated through a simple two-step process, comprised of hot injection and thin-film hydration. The as-prepared PEGylated Cu2Te NCs (average diameter of 5.21±1.05nm) showed a noticeable photothermal conversion efficiency of 33.1% and good capacity to load hydrophobic anti-cancer drug. Due to the protonated amine group at low pH, the doxorubicin (DOX)-loaded PEGylated Cu2Te NCs (PEGylated Cu2Te-DOX NCs) exhibited an acidic pH promoted drug release profile. Moreover, a three-parameter model, which considers the effects of drug-carrier interactions on the initial burst release and the sustained release of drug from micro- and nano-sized carriers, was used to gain insight into how pH and laser irradiation affect drug release from PEGylated Cu2Te-DOX NCs. Based on the results from in vitro cell study, PEGylated Cu2Te-DOX NCs revealed remarkably photothermal-chemo synergistic effect to HeLa cells, attributed to both the PEGylated Cu2Te NCs mediated photothermal ablation and enhanced cellular uptake of the drug. Thus, our results encourage the usage of Cu2Te-DOX drug nanocarriers for enhanced treatment of cancer cells by combined photothermal-chemo therapy.


Asunto(s)
Antineoplásicos/química , Doxorrubicina/química , Portadores de Fármacos/química , Polietilenglicoles/química , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Doxorrubicina/farmacología , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas/química , Fototerapia
11.
Hepatology ; 59(2): 385-94, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23907803

RESUMEN

UNLABELLED: Infection with hepatitis B virus (HBV) is the most common cause of liver disease worldwide. However, because the current interferon (IFN)-based treatments have toxic side effects and marginal efficacy, improved antivirals are essential. Here we report that unmethylated cytosine-phosphate-guanosine oligodeoxynucleotides (CpG ODNs) from the HBV genome (HBV-CpG) induced robust expression of IFN-α by plasmacytoid dendritic cells (pDCs) in a Toll-like receptor 9 (TLR9)-dependent manner. We also identified inhibitory guanosine-rich ODNs in the HBV genome (HBV-ODN) that are capable of inhibiting HBV-CpG-induced IFN-α production. Furthermore, nanoparticles containing HBV-CpG, termed NP(HBV-CpG), reversed the HBV-ODN-mediated suppression of IFN-α production and also exerted a strong immunostimulatory effect on lymphocytes. Our results suggest that NP(HBV-CpG) can enhance the immune response to hepatitis B surface antigen (HBsAg) and skew this response toward the Th1 pathway in mice immunized with rHBsAg and NP(HBV-CpG). Moreover, NP(HBV-CpG)-based therapy led to the efficient clearance of HBV and induced an anti-HBsAg response in HBV carrier mice. CONCLUSION: Endogenous HBV-CpG ODNs from the HBV genome induce IFN-α production so that nanoparticle-encapsulated HBV-CpG may act as an HBsAg vaccine adjuvant and may also represent a potent therapeutic agent for the treatment of chronic HBV infection.


Asunto(s)
Citosina , Guanosina , Virus de la Hepatitis B/genética , Hepatitis B/tratamiento farmacológico , Inmunoterapia/métodos , Nanopartículas/uso terapéutico , Oligodesoxirribonucleótidos/uso terapéutico , Fosfatos , Animales , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Hepatitis B/inmunología , Hepatitis B/prevención & control , Antígenos de Superficie de la Hepatitis B/farmacología , Antígenos de Superficie de la Hepatitis B/uso terapéutico , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Técnicas In Vitro , Interferón-alfa/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfocitos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA