Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
ACS Appl Mater Interfaces ; 15(42): 48855-48870, 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37823476

RESUMO

By means of a "live-cell" template strategy, silica replicas displaying the same morphology and topography of the mammalian cells used as templates are fabricated. The replicas are used as substrates to direct the differentiation of mesenchymal stem cells (MSCs) to predefined cell lineages. Upregulation of specific genes shows how the silica replica-based substrates have the ability to induce the molecular characteristics of the mature cell types from which they have been derived from. Thus, MSCs cultured in the presence of silica replicas of human osteoblasts (HObs) differentiate into HObs-like cells, as shown by the upregulation of specific osteogenic genes. Likewise, when MSCs are incubated with silica replicas derived from human chondrocytes, an enhanced expression of chondrogenic markers is observed. Importantly, the effects of the silica replicas are cell type-specific since the incubation of MSCs with HObs silica replicas does not result in upregulation of chondrogenic markers and vice versa. What is more, for both cases, the differentiation rate is enhanced when the silica replicas are used in combination with growth factors, suggesting a potential synergistic effect. These results demonstrate the potential of this innovative method as an efficient and cheap approach with the potential to eliminate, or at least reduce, the use of biochemically soluble compounds in stem cells research.


Assuntos
Células-Tronco Mesenquimais , Animais , Humanos , Linhagem da Célula , Diferenciação Celular , Condrócitos , Osteogênese , Células Cultivadas , Condrogênese , Mamíferos
2.
RSC Adv ; 13(23): 15540-15553, 2023 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-37228685

RESUMO

The development of hemoglobin (Hb)-based oxygen carriers (HBOCs) holds a lot of potential to overcome important drawbacks of donor blood such as a short shelf life or the potential risk of infection. However, a crucial limitation of current HBOCs is the autoxidation of Hb into methemoglobin (metHb), which lacks oxygen-carrying capacity. Herein, we address this challenge by fabricating a Hb and gold nanoclusters (AuNCs) composite (Hb@AuNCs) which preserves the exceptional features of both systems. Specifically, the Hb@AuNCs retain the oxygen-transporting properties of Hb, while the AuNCs provide antioxidant functionality as shown by their ability to catalytically deplete harmful reactive oxygen species (ROS). Importantly, these ROS-scavenging properties translate into antioxidant protection by minimizing the autoxidation of Hb into non-functional metHb. Furthermore, the AuNCs render Hb@AuNCs with auto-fluorescence properties which could potentially allow them to be monitored once administered into the body. Last but not least, these three features (i.e., oxygen transport, antioxidant and fluorescence properties) are well maintained following storage as a freeze-dried product. Thus, overall, the as-prepared Hb@AuNCs hold the potential to be used as a multifunctional blood surrogate in the near future.

3.
Nanoscale ; 13(31): 13328-13343, 2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34477739

RESUMO

Facing the barriers in each step of the in vivo delivery cascade, the low drug delivery efficiency remains problematic in tumor therapy. Although recently the nanofibril drug delivery systems have shown improved circulation and accumulation compared with nanoparticles, the poor deep penetration and cellular internalization hinder their application, especially for pancreatic cancer with dense stroma. To comprehensively address the hurdles in the delivery cascade, a matrix metalloproteinase 2 (MMP-2) responsive transformable beaded nanofibril, which integrates the merits of nanofibril and small-sized nanoparticles, is established. The beaded nanofibril (GD@PPF) is prepared by conjugating gemcitabine-loaded small-sized nanoparticles (GD) with fibrous PEG-PCL (PPF) via GPLGVRG, a substrate peptide of MMP-2. GD@PPF escapes the clearance of the reticuloendothelial system (RES), prolongs the circulation time, and increases the selective accumulation in the tumor as fibrous micelles. Once accumulated in the tumor, small positively-charged GD is released from the beaded nanofibrils in response to MMP-2 overexpression in the stroma of pancreatic cancer, enabling permeation in the dense tumor matrix and cellular internalization, which makes up for the shortcomings of fibrous micelles. Furthermore, the remaining fibrous PPF surround the tumor tightly to impede the efflux of drugs, leading to improved retention. GD@PPF is biocompatible and exhibits excellent antitumor effect in Pan 02 subcutaneous tumor models. Therefore, the MMP-2 responsive transformable beaded nanofibril, which enhances the delivery efficiency in multiple stage of the delivery cascade, presents a promising strategy for pancreatic cancer therapy.


Assuntos
Nanopartículas , Neoplasias Pancreáticas , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Metaloproteinase 2 da Matriz , Micelas , Neoplasias Pancreáticas/tratamento farmacológico
4.
Nanomaterials (Basel) ; 10(10)2020 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-33092104

RESUMO

Tissue engineering is a promising strategy to treat tissue and organ loss or damage caused by injury or disease. During the past two decades, mesenchymal stem cells (MSCs) have attracted a tremendous amount of interest in tissue engineering due to their multipotency and self-renewal ability. MSCs are also the most multipotent stem cells in the human adult body. However, the application of MSCs in tissue engineering is relatively limited because it is difficult to guide their differentiation toward a specific cell lineage by using traditional biochemical factors. Besides biochemical factors, the differentiation of MSCs also influenced by biophysical cues. To this end, much effort has been devoted to directing the cell lineage decisions of MSCs through adjusting the biophysical properties of biomaterials. The surface topography of the biomaterial-based scaffold can modulate the proliferation and differentiation of MSCs. Presently, the development of micro- and nano-fabrication techniques has made it possible to control the surface topography of the scaffold precisely. In this review, we highlight and discuss how the main topographical features (i.e., roughness, patterns, and porosity) are an efficient approach to control the fate of MSCs and the application of topography in tissue engineering.

5.
J Control Release ; 321: 564-575, 2020 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-32112854

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is rich in cancer-associated fibroblasts (CAFs), which participate in the formation of tumor stroma. However, the dense tumor stroma of PDAC presents major barriers to drug delivery, resulting in an obstacle for PDAC therapy. Considering the special tumor microenvironment of PDAC, we constructed a novel nanoparticle which is responsive to the membrane biomarker FAP-α on CAFs and near-infrared (NIR) laser irradiation. Small sized albumin nanoparticle of paclitaxel (HSA-PTX) with strong tumor-penetration ability was encapsulated into the CAP-(a FAP-α responsive cleavable amphiphilic peptide) modified thermosensitive liposomes (CAP-TSL). Moreover, IR-780, a photothermal agent, was incorporated into CAP-TSL to afford CAP-ITSL. The designed HSA-PTX@CAP-ITSL increased the drug retention of HSA-PTX in solid tumor and HSA-PTX was released via FAP-α (specifically expresses on CAFs) triggered. Under sequential stimulation of NIR laser irradiation, IR-780 produced hyperthermia to kill tumor cells and expand the tumor interstitial space at the same time, which further promoted the release of small sized HSA-PTX in deep tumor regions. Consequently, the excellent antitumor efficacy of HSA-PTX@CAP-ITSL was demonstrated in Pan 02 subcutaneous and orthotopic tumor mouse models. Therefore, HSA-PTX@CAP-ITSL well combined chemotherapy with photothermal therapy, providing a promising drug delivery strategy for PDAC treatment.


Assuntos
Albuminas , Fibroblastos Associados a Câncer , Liberação Controlada de Fármacos , Nanopartículas , Neoplasias Pancreáticas , Preparações Farmacêuticas , Animais , Linhagem Celular Tumoral , Lipídeos , Camundongos , Paclitaxel , Neoplasias Pancreáticas/tratamento farmacológico , Perfusão , Fototerapia , Microambiente Tumoral
6.
J Control Release ; 317: 43-56, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31758970

RESUMO

Myeloid-derived suppressor cells(MDSCs)are one of the most important immunosuppressive cells in tumor microenvironment, which also promote the development and progression of tumor cells. Nevertheless, due to the different distribution features of MDSCs and tumor cells, selective elimination of MDSCs and tumor cells in tumor microenvironment remain a great challenge. Here we have designed a dual-pH-sensitivity conjugated micelle system (PAH/RGX-104@PDM/PTX) that could deliver liver-X nuclear receptor (LXR) agonist RGX-104 and paclitaxel (PTX) to the perivascular region and tumor cells, respectively. Upon arrival at the acidic tumor microenvironment, the PAH/RGX-104@PDM/PTX undergo structure disintegration and capacitate coinstantaneous release of RGX-104 in the perivascular regions, leaving the intact PTX containing micelles PDM/PTX for tumor deep penetration. The released RGX-104 can be preferentially taken up by leukocytes, endothelial cells and macrophages which are nicely enriched in perivascular regions to active the LXR, and further reduces immunosuppressive MDSC levels. The remained small micelles carrying PTX enable deep tumor penetration as well as rapid specific drug release in the endosomal/lysosomal to kill tumor cells. PAH/RGX-104@PDM/PTX exhibits superior tumor accumulation as well as tumor penetration, and suppresses 74.88% in vivo tumor growth. More importantly, PAH/RGX-104@PDM/PTX has significantly alleviated tumor immunosuppression by eliminating MDSCs and increasing cytotoxic T lymphocytes infiltration. Our studies suggest that the dual-pH-sensitive codelivery nanocarrier not only cause apoptosis of cancer cells but also regulate the tumor immune environment to ultimately enhance the antitumor effect of CTLs through MDSCs depletion.


Assuntos
Células Supressoras Mieloides , Neoplasias , Células Endoteliais , Concentração de Íons de Hidrogênio , Imunoterapia , Micelas
7.
ACS Appl Mater Interfaces ; 11(43): 39545-39559, 2019 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-31617997

RESUMO

Tumor-associated fibroblasts (TAFs), which form a predominant stromal cellular component of the tumor microenvironment, hinder the delivery of nanomedicine to deep tumor cells and lead to poor prognosis of tumors. However, depletion of TAFs by therapeutic agents results in the secretion of damage response program (DRP) molecules to weaken the efficacy of tumor treatment. This paper reports a multifunctional size-switchable nanoparticle (denoted DGL (dendrigraft poly-l-lysine) (DGL)/GEM@PP/GA) for TAF-targeted regulation and deep tumor penetration. After accumulation at the tumor site, in response to overexpressed matrix metalloproteinase-2 (MMP-2) in the tumor microenvironment, gemcitabine (GEM)-conjugated small nanoparticles (DGL/GEM) are released from DGL/GEM@PP/GA, leaving 18ß-glycyrrhetinic acid (GA)-loaded large nanoparticles (PP/GA). The released DGL/GEM can penetrate to the deep region of the tumor as well as intracellularly release GEM to kill tumor cells. However, residual GA-loaded nanoparticles with lower tumor penetration ability could accumulate around tumor vessels and be preferentially absorbed by TAFs to regulate the secretion of Wnt 16, which is an important DRP molecule. By taking actions on both tumor cells and TAFs, DGL/GEM@PP/GA displayed significant and long-term antitumor effect in stroma-rich pancreatic cancer and breast cancer models.


Assuntos
Antineoplásicos , Fibroblastos Associados a Câncer/metabolismo , Ácido Glicirretínico/análogos & derivados , Nanopartículas , Neoplasias Experimentais/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Fibroblastos Associados a Câncer/patologia , Linhagem Celular Tumoral , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Preparações de Ação Retardada/farmacologia , Ácido Glicirretínico/química , Ácido Glicirretínico/farmacocinética , Ácido Glicirretínico/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Células NIH 3T3 , Nanopartículas/química , Nanopartículas/uso terapêutico , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Polilisina/química , Polilisina/farmacocinética , Polilisina/farmacologia , Proteínas Wnt/metabolismo
8.
J Control Release ; 304: 204-215, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31100311

RESUMO

The hypoxic tumor microenvironment of pancreatic cancer contributes to the progression and metastasis of tumor cells. Downregulation of hypoxia-inducible factor-1α (HIF-1α) with CRISPR/Cas9 is a promising approach to modulate tumor microenvironment and inhibit tumor metastasis. However, the in vivo delivery of CRISPR/Cas9 system remains a challenge. In the present manuscript, a tumor targeted lipid-based CRISPR/Cas9 delivery system was developed to suppress HIF-1α. Plasmids encoding Cas9 and HIF-1α-targeting sgRNA were successfully constructed and coencapsulated in R8-dGR peptide modified cationic liposome with PTX. R8-dGR-Lip exhibited enhanced BxPC-3 cell targeting and deep penetration into tumor spheroids. R8-dGR-Lip/PTX/pHIF-1α successfully downregulated HIF-1α and its downstream molecules VEGF and MMP-9, leading to enhanced antimetastatic effects. Besides, the blockade of HIF-1α also promoted the cytotoxicity of PTX on BxPC-3 cell lines. Compared with pegylated liposomes, R8-dGR-Lip enhanced the distribution in tumor tissues. The targeted delivery of CRISPR/Cas9-HIF-1α system and PTX significantly inhibited tumor growth. More importantly, inhibition of HIF-1α suppressed the metastasis of pancreatic cancer and prolonged survival time. Since CRISPR/Cas 9-HIF-1α hardly affected HIF-1α expression in normal hepatic cells, the designed R8-dGR-Lip/PTX/pHIF-1α did not induce severe toxicity in vivo. This strategy broadened the in vivo application of CRISPR/Cas9 system. Downregulation of HIF-1α may be a feasible approach for antimetastatic therapy.


Assuntos
Proteína 9 Associada à CRISPR/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pancreáticas/genética , Microambiente Tumoral/genética , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Lipídeos/química , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica/prevenção & controle , Neoplasias Pancreáticas/patologia , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
9.
AAPS J ; 21(2): 18, 2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30635795

RESUMO

Chemoimmunotherapy with chemotherapeutics and immunoadjuvant inhibits tumor growth by activating cytotoxic T cells. However, this process also upregulates the expression of PD-1/PD-L1 and consequently leads to immune suppression. To maximize the anti-tumor immune responses and alleviate immunosuppression, PD-L1 antibody was combined with paclitaxel (PTX) and the immunoadjuvant α-galactosylceramide (αGC), which were coencapsulated into pH-sensitive TH peptide-modified liposomes (PTX/αGC/TH-Lip) to treat melanoma and lung metastasis. Compared to treatment with PD-L1 antibody or PTX/αGC/TH-Lip alone, the combination of PD-L1 antibody and PTX/αGC/TH-Lip further elevated the tumor-specific cytotoxic T cell responses and promoted apoptosis in tumor cells, leading to enhanced anti-tumor and anti-metastatic effects. In adoptive therapy, PD-L1 antibody further alleviated immunosuppression and enhanced the anti-tumor effect of CD8+ T cells. The combination of PD-L1 antibody and chemoimmunotherapy PTX/αGC/TH-Lip provides a promising strategy for enhancing treatment for melanoma and lung metastasis.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Melanoma Experimental/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Adjuvantes Imunológicos/administração & dosagem , Animais , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral/transplante , Feminino , Galactosilceramidas/administração & dosagem , Tolerância Imunológica/efeitos dos fármacos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Paclitaxel/administração & dosagem , Paclitaxel/imunologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Linfócitos T Citotóxicos/efeitos dos fármacos
10.
Nanomedicine ; 14(8): 2541-2550, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30193815

RESUMO

Cholesterol facilitated the formation of T cell receptor on cytotoxic CD8+ T lymphocytes (CTLs). However, the activation of CD8+ T cells always resulted in the upregulation of acetyl-CoA acetyltransferase-1 (ACAT-1) and enhanced the esterification of cholesterol. To relieve the suppression on CD8+ T cells, an ACAT-1 inhibitor avasimibe was combined with chemo-immunotherapy. Paclitaxel and immunoadjuvant αGC were co-encapsulated in liposomes modified with pH sensitive TH peptide (PTX/αGC-TH-Lip). After intravenous injections, the combination of avasimibe significantly elevated the free cholesterol level and relieved the inhibition of CD8+ T cells caused by PTX/αGC-TH-Lip, leading to enhanced CTL responses and anti-tumor effects of PTX/αGC-TH-Lip in B16F10 melanoma xenograft and lung metastasis models. The adoptive immunotherapy further confirmed the enhanced anti-tumor immune responses of the combined strategy. The combination of avasimibe and PTX/αGC-TH-Lip was proven as a feasible approach to enhance the antitumor effects of chemo-immunotherapy by relieving the inhibition of CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Colesterol/metabolismo , Imunoterapia Adotiva , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Paclitaxel/farmacologia , Linfócitos T Citotóxicos/imunologia , Acetamidas , Acetatos/farmacologia , Acetil-CoA C-Acetiltransferase/antagonistas & inibidores , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Peptídeos Penetradores de Células/administração & dosagem , Peptídeos Penetradores de Células/química , Células Cultivadas , Colesterol/química , Indutores do Citocromo P-450 CYP3A/farmacologia , Esterificação , Feminino , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Sulfonamidas , Ácidos Sulfônicos/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/patologia
11.
Eur J Pharm Sci ; 124: 240-248, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30071282

RESUMO

Effective chemotherapy for clinical glioma treatment is still lacking due to the poor penetration of blood-brain barrier (BBB) and the poor internalization into tumor cells. To facilitate the transmigration across the BBB as well as the glioma targeting of chemotherapeutics, we constructed cell penetrating peptide dNP2 and tumor microenvironment-cleavable folic acid (FA) dual modified, paclitaxel (PTX) loaded liposome for the targeted delivery of glioma. The modification of dNP2 significantly enhanced the transmigration across the BBB in an in vitro BBB model. The acid-cleavable cFd-Lip/PTX exhibited sensitive cleavage of FA at pH 6.8, which led to enhanced cellular uptake mediated by both cell penetrating peptide dNP2 and the interaction between FA and folate receptor (FR) on the glioma cells. After intravenous injection, compared with non-cleavable Fd-Lip and single modified liposomes, cFd-Lip enhanced the accumulation in orthotropic glioma and improved the anti-tumor effect of glioma-bearing mice. The dual modified liposomes also facilitated deep penetration into tumor cells and consequently enhanced the cytotoxicity of PTX-loaded liposomes. The acid-cleavable dual modified strategy retained the BBB penetrating and tumor targeting ability, meanwhile, the cleavage of FA further maximized the cell permeability of dNP2, exhibiting enhanced tumor targeting effect. The multi-targeting strategy provides a promising approach towards targeted chemotherapy for glioma.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Peptídeos Penetradores de Células/administração & dosagem , Ácido Fólico/administração & dosagem , Glioma/tratamento farmacológico , Paclitaxel/administração & dosagem , Animais , Antineoplásicos Fitogênicos/química , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Feminino , Receptores de Folato com Âncoras de GPI/metabolismo , Ácido Fólico/química , Glioma/metabolismo , Concentração de Íons de Hidrogênio , Lipossomos , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular , Paclitaxel/química , Fosfatidiletanolaminas/administração & dosagem , Fosfatidiletanolaminas/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química
12.
Nanoscale ; 10(21): 9957-9970, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29770821

RESUMO

Cancer-related inflammation can promote tumorigenesis, tumor growth and tumor metastasis in many types of cancers. Therefore, inhibiting cancer-related inflammation significantly improves cancer therapy. It has been reported that metformin (MET) inhibits the nuclear translocation of nuclear factor-κB (NF-κB), a key factor in cancer-related inflammation. However, the short half-life and the lack of tumor targeting limit the anti-inflammatory efficacy of MET in vivo. Herein, using pH-sensitive imine bonds, MET and the chemotherapy drug doxorubicin (DOX) were loaded onto size-shrinkable RGD-DGL-GNP nanoparticles (RDG NPs) for combination therapy. The RGD-MET-DGL-GNP nanoparticles (RMDG NPs) penetrated deep into the tumor to deliver MET and inhibit the NF-κB activity in tumor cells, which further decreased tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) expressions in tumor tissues and suppressed tumor cell proliferation. As a result, the co-administration of RGD-DOX-DGL-GNP (RDDG NPs) and RMDG NPs induced an improved therapeutic effect in a xenograft tumor model and a lipopolysaccharide (LPS)-induced pulmonary metastasis model with murine 4T1 breast cancer and CT26 colon cancer cells. Combining RDDG and RMDG NPs to simultaneously target tumors and cancer-related inflammation is a very effective anti-cancer strategy.


Assuntos
Anti-Inflamatórios/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Doxorrubicina/administração & dosagem , Metformina/administração & dosagem , Nanopartículas , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/antagonistas & inibidores , Neoplasias Experimentais/tratamento farmacológico , Fator de Necrose Tumoral alfa/metabolismo
13.
Nanoscale ; 10(21): 9935-9948, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29770822

RESUMO

The complex tumor microenvironment (TME) in solid tumors forms physiological barriers to the efficient delivery of nanomedicine, leading to limited therapeutic efficacy. Herein, to overcome these physiological barriers and improve the therapeutic effect, we constructed a novel size-adjustable nanoplatform for efficient drug delivery into solid tumors. The smart size-switchable nanoplatform (DGL/DOX@PP) was prepared by conjugating small dendrigraft poly-l-lysine (DGL) to poly(ethylene glycol)-poly(caprolactone) micelles via a matrix metalloproteinase 2 (MMP-2)-sensitive peptide. DGL/DOX@PP had an initial size of 100 nm and a nearly neutral charge, rendering the system able to take advantage of the enhanced permeability and retention effect. After extravasation from the tumor vessels, small DGL/DOX nanoparticles (∼30 nm) were rapidly released from DGL/DOX@PP in response to MMP-2 in the TME. This process of particle size alteration greatly enhanced the nanoparticle penetration into both multicellular spheroids (MCSs) and solid tumors. In vivo results demonstrated that compared with small and non-switchable nanoparticles, particles from the size-switchable nanoplatform achieved excellent antitumor efficacy in 4T1 tumor-bearing mice. This size-adjustable nanoplatform provides a multifunctional strategy for TME modulation and tumor penetration.


Assuntos
Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Metaloproteinase 2 da Matriz/metabolismo , Nanopartículas , Neoplasias Experimentais/tratamento farmacológico , Microambiente Tumoral , Animais , Antibióticos Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Camundongos
14.
Nanomedicine ; 14(6): 1833-1843, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29800759

RESUMO

Cancer associated fibroblasts (CAFs) which shape the tumor microenvironment (TME) and the presence of blood brain barrier (BBB) remain great challenges in targeting breast cancer and its brain metastasis. Herein, we reported a strategy using PTX-loaded liposome co-modified with acid-cleavable folic acid (FA) and BBB transmigrating cell penetrating peptide dNP2 peptide (cFd-Lip/PTX) for enhanced delivery to orthotopic breast cancer and its brain metastasis. Compared with single ligand or non-cleavable Fd modified liposomes, cFd-Lip exhibited synergistic TME targeting and BBB transmigration. Moreover, upon arrival at the TME, the acid-cleavable cFd-Lip/PTX showed sensitive cleavage of FA, which reduced the hindrance effect and maximized the function of both FA and dNP2 peptide. Consequently, efficient targeting of folate receptor (FR)-positive tumor cells and FR-negative CAFs was achieved, leading to enhanced anti-tumor activity. This strategy provides a feasible approach to the cascade targeting of TME and BBB transmigration in orthotopic and metastatic cancer treatment.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Paclitaxel/administração & dosagem , Microambiente Tumoral/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Peptídeos Penetradores de Células/química , Feminino , Ácido Fólico/química , Humanos , Concentração de Íons de Hidrogênio , Ligantes , Lipossomos/administração & dosagem , Lipossomos/química , Camundongos , Camundongos Endogâmicos BALB C , Paclitaxel/química , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biomaterials ; 168: 64-75, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29626787

RESUMO

Chemotherapy remains restricted by poor drug delivery efficacy due to the heterogenous nature of tumor. Herein, we presented a novel nanoparticle that could not only response to the tumor microenvironment but also modulate it for deep tumor penetration and combination therapy. The intelligent nanoparticle (IDDHN) was engineered by hyaluronidase (HAase)-triggered size shrinkable hyaluronic acid shells, which were modified with NIR laser sensitive nitric oxide donor (HN), small-sized dendrimeric prodrug (IDD) of doxorubicin (DOX) as chemotherapy agent and indocyanine green (ICG) as photothermal agent into a single nanoparticle. IDDHN displayed synergistic deep penetration both in vitro and in vivo, owing to the enzymatically degradable HN shell mediated by HAase and laser-enhanced NO release triggered deep penetration upon strong hyperthermia effect of ICG under the NIR laser irradiation. The therapeutic effect of IDDHN was verified in 4T1 xenograft tumor model, and IDDHN showed a much better antitumor efficiency with few side effects upon NIR laser irradiation. Therefore, the valid of this study might provide a novel tactic for engineering nanoparticles both response to and modulate the tumor microenvironment for improving penetration and heterogeneity distribution of therapeutic agents in tumor.


Assuntos
Portadores de Fármacos , Hialuronoglucosaminidase , Nanopartículas/química , Óxido Nítrico , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Feminino , Humanos , Verde de Indocianina/uso terapêutico , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Tamanho da Partícula
16.
ACS Nano ; 10(11): 10086-10098, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27934068

RESUMO

The treatment of brain tumors remains a challenge due to the limited accumulation of drugs and nanoparticles. Here, we triggered the aggregation of gold nanoparticles (AuNPs) using legumain to enhance the retention of chemotherapeutics in brain tumors. This nanoplatform, AuNPs-A&C, is comprised of Ala-Ala-Asn-Cys-Lys modified AuNPs (AuNPs-AK) and 2-cyano-6-aminobenzothiazole modified AuNPs (AuNPs-CABT). AuNPs-AK could be hydrolyzed to expose the 1,2-thiolamino groups on AuNPs-AK in the presence of legumain, which occurs by a click cycloaddition with the contiguous cyano group on AuNPs-CABT, resulting in formation of AuNPs aggregates. This strategy led to an enhanced retention of the AuNPs in glioma cells both in vitro and in vivo due to the blocking of nanoparticle exocytosis and minimizing nanoparticle backflow to the bloodstream. After conjugation of doxorubicin (DOX) via a pH-sensitive linker to AuNPs-A&C, the efficiency for treating glioma was improved. The median survival time for the DOX-linked AuNPs-A&C increased to 288% in comparison to the saline group. We further show the use of the AuNPs-A&C for optical imaging applications. In conclusion, we provide a strategy to increase nanoparticle tumor accumulation with the potential to improve therapeutic outcome.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Ouro , Nanopartículas Metálicas , Antimetabólitos Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Humanos
17.
Sci Rep ; 6: 27669, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27273770

RESUMO

In recent years, active targeting strategies by ligand modification have emerged to enhance tumor accumulation of NP, but their clinical application was strictly restricted due to the complex preparation procedures, poor stability and serious toxicity. An effective and clinical translational strategy is required to satisfy the current problems. Interestingly, the internalization of NP is intimately related with cell cycle and the expression of receptors is not only related with cancer types but also cell cycle progression. So the cellular uptake of ligand modified NP may be related with cell cycle. However, few investigations were reported about the relationship between cell cycle and the internalization of ligand modified NP. Herein, cellular uptake of folic acid (FA) modified NP after utilizing chemotherapeutic to retain the tumor cells in G2/M phase was studied and a novel strategy was designed to enhance the active targeting effect. In our study, docetaxel (DTX) notably synchronized cells in G2/M phase and pretreatment with DTX highly improved in vitro and in vivo tumor cell targeting effect of FA decorated NP (FANP). Since FA was a most common used tumor active targeting ligand, we believe that this strategy possesses broader prospects in clinical application for its simplicity and effectiveness.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Nanopartículas , Neoplasias/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Docetaxel , Ácido Fólico , Humanos , Ligantes , Masculino , Camundongos , Imagem Molecular , Terapia de Alvo Molecular , Nanopartículas/química , Nanopartículas/ultraestrutura , Polietilenoglicóis , Polímeros , Taxoides/administração & dosagem , Taxoides/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Sci Rep ; 6: 19800, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26842655

RESUMO

[D]-H6L9, as a pH-responsive anti-microbial peptide (AMP), has been evidenced by us to be an excellent choice in tumor microenvironment-responsive delivery as it could render liposomes responsive to the acidified tumor microenvironment. However, [D]-H6L9-modified liposomes could not actively target to tumor area. Therefore, integrin αvß3-targeted peptide RGD was co-modified with [D]-H6L9 onto liposomes [(R + D)-Lip] for improved tumor delivery efficiency. Under pH 6.3, (R + D)-Lip could be taken up by C26 cells and C26 tumor spheroids (integrin αvß3-positive) with significantly improved efficiency compared with other groups, which was contributed by both RGD and [D]-H6L9, while RGD did not increase the cellular uptake performance on MCF-7 cells (integrin αvß3-negative). Results showed that RGD could decrease cellular uptake of (R + D)-Lip while [D]-H6L9 could increase it, implying the role of both RGD and [D]-H6L9 in cellular internalization of (R + D)-Lip. On the other hand, (R + D)-Lip could escape the entrapment of lysosomes. PTX-loaded (R + D)-Lip could further increase the cellular toxicity against C26 cells compared with liposomes modified only with RGD and [D]-H6L9 respectively, and achieve remarkable tumor inhibition effect on C26 tumor models.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Lipossomos/química , Oligopeptídeos/química , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Integrina alfaVbeta3/metabolismo , Lipossomos/metabolismo , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Oligopeptídeos/metabolismo , Paclitaxel/administração & dosagem , Paclitaxel/química , Paclitaxel/toxicidade , Tamanho da Partícula , Polietilenoglicóis/química , Distribuição Tecidual , Transplante Heterólogo , Microambiente Tumoral
19.
Drug Deliv ; 23(8): 2970-2979, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26758229

RESUMO

Tumor metastasis would seriously impair the efficacy of chemotherapy. Our previous studies showed losartan combined with paclitaxel-loaded pH-sensitive cleavable liposomes (PTX-Cl-Lip) facilitated paclitaxel accumulation and led to enhanced antitumor efficacy in 4T1 bearing mice. Since losartan could inhibit the level of collagen I which was related to tumor metastasis, this strategy was further applied to suppress tumor metastasis this time. Our in vivo anti-metastatic study manifested losartan could lower the colonies occupied in lungs by 76.4% compared with that of saline group. When losartan and PTX-Cl-Lip were combined, anti-metastatic efficiency reached to 88.2%, which was the best among all the groups. In vitro 3D tumor spheroids studies proved losartan could significantly suppress the invasion of tumor cells. Losartan plus PTX-Cl-Lip could further weaken the metastasis of tumor cells. Mechanism study showed the declination of collagen I level via losartan was caused by inhibition of active transforming growth factor-ß1. Western-blot study showed losartan could decrease the level of lysyl oxidase, then inhibit the cross-linking of collagen I, finally weakened the cell signaling transmit via integrin and the metastasis of tumor cells was restrained. All above studies illustrated this combined tactic could achieve favorable effect on suppression of lung tumor metastasis.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Colágeno Tipo I/metabolismo , Lipossomos/química , Losartan/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Paclitaxel/farmacologia , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Concentração de Íons de Hidrogênio , Losartan/química , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica/tratamento farmacológico , Paclitaxel/química
20.
Acta Biomater ; 31: 186-196, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26675124

RESUMO

Although development of nanomedicines has been a promising direction in tumor treatment, the therapeutic outcome of current nanomedicines is unsatisfying, partly because of the poor retention and penetration in tumors. Recently, a kind of tumor microenvironment sensitive size shrinkable nanoparticles (DOX-AuNPs-GNPs) has been developed by our lab, which could enhance the tumor penetration and retention depending on the size shrinking. However, the further enhancement is still restricted by dense collagen network in tumors. Thus in this study, we combined DOX-AuNPs-GNPs with losartan to deplete tumor collagen (constituted up to 90% of extracellular matrix) to further improve tumor penetration. In vitro, DOX-AuNPs-GNPs can shrink from over 117.8nm to less than 50.0nm and release DOX-AuNPs under the triggering of tumor overexpressed matrix metalloproteinases-2 (MMP-2). In vivo, pretreatment with losartan significantly decrease the collagen level and improve the tumor penetration. In combination, losartan combined with DOX-AuNPs-GNPs showed the best drug delivery efficiency, striking penetration efficiency and best 4T1 breast tumor inhibition effect. In conclusion, this study provided a promising synergetic strategy to improve the tumor treatment efficiency of nanomedicines. STATEMENT OF SIGNIFICANCE: We have developed a dual strategy for deep tumor penetration through combining size shrinkable DOX-AuNPs-GNPs with depleting tumor collagen by losartan. Additionally, we demonstrate therapeutic efficacy in breast tumor bearing mouse model. DOX-AuNPs-GNPs co-administration with losartan is a novel and highly attractive strategy for anti-tumor drug delivery with the potential for broad applications in clinic.


Assuntos
Colágeno/química , Losartan/administração & dosagem , Neoplasias Mamárias Animais/tratamento farmacológico , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Ouro/química , Losartan/química , Espectroscopia de Ressonância Magnética , Neoplasias Mamárias Animais/patologia , Metaloproteinase 2 da Matriz/metabolismo , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Tamanho da Partícula
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...