Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Pharm ; 16(11): 4530-4541, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31617723

RESUMO

Tumor metastasis is the most dangerous stage in tumorigenesis and its evolution, which causes about 80% clinical death. However, common therapies including chemotherapy may increase the risk of tumor metastasis while killing cancer cells. Tumor metastasis is closely related to many factors in the tumor microenvironment, especially hypoxia. As one of the characteristics of a malignant tumor microenvironment, hypoxia plays an important role in the growth, metabolism, and metastasis of tumors. Upregulation of the hypoxia-inducible factor (HIF) would stimulate the metastasis and migration of cancer cells. In this study, we developed an artificial oxygen carrier system, a hemoglobin-loaded liposome (Hb@lipo), which was capable of effectively delivering oxygen to tumor. The way of providing oxygen not only alleviated tumor hypoxia but also downregulated the expression of HIF, which is conducive to reducing tumor malignancy. Alleviating the tumor hypoxic microenvironment alone is not enough to inhibit tumor metastasis; thus, we prepared the liposome containing a chemotherapeutic agent cabazitaxel (CBZ@lipo). Our data indicated that the combination therapy of Hb@lipo and CBZ@lipo can efficiently kill cancer cells and inhibit tumor growth. At the same time, it can effectively entrap cancer cells in tumor sites by relieving the hypoxic microenvironment of tumors and reduce the metastasis of cancer cells during and after the chemotherapy. Our research may provide a clinical cancer chemotherapy reference that reduces the risk of cancer cell metastasis while inhibiting tumor growth.


Assuntos
Antineoplásicos/farmacologia , Metástase Neoplásica/tratamento farmacológico , Oxigênio/metabolismo , Hipóxia Tumoral/efeitos dos fármacos , Animais , Biomimética/métodos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Feminino , Células HT29 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipossomos/química , Células MCF-7 , Melanoma Experimental , Camundongos , Camundongos Endogâmicos BALB C , Microambiente Tumoral/efeitos dos fármacos
2.
J Control Release ; 341: 769-781, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34952044

RESUMO

As a research hotspot, immune checkpoint inhibitors (ICIs) is often combined with other therapeutics in order to exert better clinical efficacy. To date, extensive laboratory and clinical investigations into the combination of ICIs and chemotherapy have been carried out, demonstrating augmented effectiveness and broad application prospects in anti-tumor therapy. However, the administration of these two treatment modalities is usually randomized or fixed to a given chronological order. Nevertheless, the pharmacological effect of drug is closely related to its exposure behavior in vivo, which may consequently affect the synergistic outcomes of a combined therapy. In this study, we prepared a lipid nanoparticle encapsulating docetaxel (DTX-VNS), and associated it with the immune checkpoint inhibitor anti-PD-1 antibody (αPD-1) for the treatment of malignant tumors. To identify the optimum timing and sequencing for chemotherapy and immunotherapy, we designed three administration regimes, including the simultaneous delivery of DTX-VNS and αPD-1(DTX-VNS@αPD-1), DTX-VNS delivery before (DTX-VNS plus αPD-1) or post (αPD-1 plus DTX-VNS) PD-1 blockade with an interval of two days. Analysis from mass spectrometry, multi-factor detection and other techniques indicated that DTX-VNS plus αPD-1 initiated a powerful anti-tumor response in multiple tumor models, contributing to a remarkably reshaped tumor microenvironment landscape, which may attribute to the maximum therapeutic additive effects arise from a concomitant exposure of DTX-VNS and αPD-1 at the tumor site. By profiling the exposure kinetics of nanoparticles and αPD-1 in vivo, we defined the administration schedule with utmost therapeutic benefits, which may provide a valuable clinical reference for the rational administration of immunochemotherapy.


Assuntos
Imunoterapia , Nanopartículas , Linhagem Celular Tumoral , Lipossomos , Nanopartículas/química
3.
Nanoscale ; 12(35): 18249-18262, 2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-32857088

RESUMO

Gene therapy mediated by non-viral carriers is gaining an increasing popularity due to its high biosafety and the convenience of production on a large scale, yet inefficient gene delivery is a limiting obstacle. Few gene vectors can avoid the endosome-lysosome route, and as a result, their DNA payloads are easily decomposed during transfection. Herein, a peptide (pardaxin, PAR)-modified cationic liposome (PAR-Lipo) targeting the endoplasmic reticulum (ER) was developed for improving the gene delivery efficiency. Interestingly, compared to non-PAR-modified cationic liposomes (Non-Lipos) and Lipofectamine 2000 (Lipo 2000, a commercial genetic vector), PAR-Lipos showed remarkably higher gene delivery efficiency in vitro and better antitumor efficacy in vivo. It was demonstrated that PAR-Lipos could be accumulated into the ER via a non-lysosome intracellular route after cellular internalization, which induced the retention of the DNA payload in the ER close to the nucleus, while Non-Lipos, like most conventional cationic carriers, mainly presented lysosomal retention after their endocytosis. The unique intracellular transport behavior of PAR-Lipos can enhance the protection of the DNA payload, prolong their residence time in the cell, and promote their entry into the nucleus relying on the intimate relationship between the ER and nuclear membrane, which is the explanation for the enhanced gene-therapy effect mediated by PAR-Lipos. Our research may provide alternative means of efficiently delivering genes in cells.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , DNA/genética , Retículo Endoplasmático , Lipossomos , Transfecção
4.
ACS Appl Mater Interfaces ; 11(50): 46536-46547, 2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31751119

RESUMO

Local hypoxia in solid malignancies often results in resistance to radiotherapy (RT) and chemotherapy (CT), which may be one of the main reasons for their failure in clinical application. Especially, oxygen is an essential element for enhancing DNA damage caused by ionizing radiation in radiotherapy. Here, two biomimetic oxygen delivery systems were designed by encapsulating hemoglobin (Hb) alone into a liposome (Hb-Lipo) or co-encapsulating Hb and doxorubicin (DOX) into a liposome (DOX-Hb-Lipo). Our data indicated that both Hb-Lipo and DOX-Hb-Lipo could effectively alleviate hypoxia in tumors. We demonstrated that RT plus tumor-targeting delivery of oxygen mediated by Hb-Lipo could significantly overcome the tolerance of hypoxic cancer cells to RT, showing significantly enhanced cancer-cell killing and tumor growth inhibition ability, mainly attributing to hypoxia alleviation and increased reactive oxygen species production under RT in cancer cells. Furthermore, a melanoma model that was quite insensitive to both RT and CT was used to test the efficacy of chemoradiotherapy combined with hypoxia alleviation. RT plus Hb-Lipo only caused a limited increase in antitumor activity. However, extremely strong tumor inhibition could be obtained by RT combined with DOX-Hb-Lipo-mediated CT, attributed to radio-triggered DOX release and enhanced immunogenic cell death induced by RT under an oxygen supplement. Our study provided a valuable reference for overcoming hypoxia-induced radioresistance and a useful therapeutic strategy for cancers that are extremely insensitive to chemo- or radiotherapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/radioterapia , Sistemas de Liberação de Medicamentos , Oxigênio/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Neoplasias da Mama/patologia , Quimiorradioterapia/métodos , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Doxorrubicina/química , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos/efeitos dos fármacos , Liberação Controlada de Fármacos/efeitos da radiação , Feminino , Hemoglobinas/química , Hemoglobinas/farmacologia , Humanos , Morte Celular Imunogênica/efeitos dos fármacos , Morte Celular Imunogênica/efeitos da radiação , Lipossomos/química , Lipossomos/farmacologia , Células MCF-7 , Oxigênio/química , Radiação Ionizante , Espécies Reativas de Oxigênio/química , Hipóxia Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Drug Deliv ; 25(1): 585-599, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29461122

RESUMO

Photosensitizer, proper laser irradiation, and oxygen are essential components for effective photodynamic therapy (PDT) in clinical cancer therapy. However, native hypoxic tumoral microenvironment is a major barrier hindering photodynamic reactions in vivo. Thus, we have prepared biocompatible liposomes by loading complexes of oxygen-carrier (hemoglobin, Hb) and photosensitizer (indocyanine green, ICG) for enhanced PDT against hypoxic tumor. Ideal oxygen donor Hb, which is an oxygen-carried protein in red blood cells, makes such liposome which provide stable oxygen supply. ICG, as a photosensitizer, could transfer energy from lasers to oxygen to generate cytotoxic reactive oxygen species (ROS) for treatment. The liposomes loading ICG and Hb (LIH) exhibited efficient tumor homing upon intravenous injection. As revealed by T2-weighted magnetic resonance imaging and immunohistochemical analysis, the intratumoral hypoxia was greatly alleviated, and the level of hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) in tumor was obviously down-regulated. A weak PDT efficiency was found in cells incubated in simulated hypoxia condition in vitro, while PDT effect was dramatically enhanced in LIH treated hypoxia cells under near-infrared (NIR) laser, which was mainly attributed to massive generation of ROS with sufficient oxygen supply. ROS trigger oxidative damage of tumors and induce complete suppression of tumor growth and 100% survival rate of mice, which were also in good health condition. Our work highlights a liposome-based nanomedicine that could effectively deliver oxygen to tumor and alleviate tumor hypoxia state, inducing greatly improved efficacy compared to conventional cancer PDT and demonstrates the promise of modulating unfavorable tumor microenvironment with nanotechnology to overcome limitations of cancer therapies.


Assuntos
Hipóxia/tratamento farmacológico , Oxigênio/administração & dosagem , Fármacos Fotossensibilizantes/administração & dosagem , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Verde de Indocianina/administração & dosagem , Lipossomos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Fotoquimioterapia/métodos , Espécies Reativas de Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
ACS Nano ; 12(8): 7647-7662, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30020768

RESUMO

A convenient and feasible therapeutic strategy for malignant and metastatic tumors was constructed here by combining photothermal ablation (PTA)-based laser immunotherapy with perdurable PD-1 blockade immunotherapy. Hollow gold nanoshells (HAuNS, a photothermal agent) and AUNP12 (an anti PD-1 peptide, APP) were co-encapsulated into poly(lactic- co-glycolic) acid (PLGA) nanoparticles. Unlike monoclonal PD-1/PD-L1 antibodies, PD-1 peptide inhibitor shows lower cost and immunotoxicity but needs frequent administration due to its rapid clearance in vivo. Our data here showed that the formed HAuNS- and APP-loaded PLGA nanoparticles (AA@PN) could maintain release periods of up to 40 days for the peptide, and a single intratumoral injection of AA@PN could replace the frequent administration of free APP. After the administration of AA@PN and irradiation with a near-infrared laser at the tumor site, an excellent killing effect on the primary tumor cells was achieved by the PTA. The nanoparticles also played a vaccine-like role under the adjuvant of cytosine-phospho-guanine (CpG) oligodeoxynucleotide and generated a localized antitumor-immune response. Furthermore, sustained APP release with laser-dependent transient triggering could induce the blockage of PD-1/PD-L1 pathway to activate T cells, thus subsequently generating a systemic immune response. Our data demonstrated that the PTA combined with perdurable PD-1 blocking could efficiently eradicate the primary tumors and inhibit the growth of metastatic tumors as well as their formation. The present study provides a promising therapeutic strategy for the treatment of advanced cancer with metastasis and presents a valuable reference for obtaining better outcomes in clinical cancer immunotherapy.


Assuntos
Neoplasias da Mama/terapia , Imunoterapia , Lasers , Fototerapia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Neoplasias da Mama/imunologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptor de Morte Celular Programada 1/imunologia , Propriedades de Superfície , Células Tumorais Cultivadas
7.
Biomaterials ; 182: 145-156, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30121013

RESUMO

Chemotherapy has become a critical treatment for many cancer types. However, its efficacy is hindered by chemoresistance and limited drug accumulation induced by the hypoxic tumor environment. Therefore, there is an urgent need for useful strategies to alleviate tumor hypoxia and enhance chemotherapy response in solid tumors. Herein, we report the development of a multifunctional liposome simultaneously loading an oxygen carrier (hemoglobin, Hb) and an anti-tumor drug (doxorubicin, DOX) to enhance chemotherapeutic effects against hypoxic tumors. The liposomes, DOX-Hb-lipo (DHL), showed efficient loading of oxygen and site-specific oxygen delivery into tumors, inducing the reversal of tumor hypoxia. Furthermore, the O2 interference capacity increased the uptake of the drug into hypoxic cancer cells, inducing a remarkably increased toxicity of the drug against cancer cells. Interestingly, the obtained DHL showed a significantly enhanced internalization into cancer cells and accumulation in tumors compared to DL (DOX loaded liposomes without Hb), while the enhanced effect did not occur in normal cells. The specific delivery of DHL into cancer cells should be attributed to the mediation of Hb on the surface of the liposomes. In addition, DHL considerably increased reactive oxygen species (ROS) production in a hypoxic environment and promoted the ROS-mediated cytotoxicity of DOX. Based on the elevated drug accumulation in the tumor sites, increased internalization into cancer cells and enhanced oxygen levels in tumor regions, DHL reversed hypoxia-induced chemoresistance and exhibited stronger antitumor effects. Thus, DHL might be a promising alternative strategy for cancer treatment.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Hemoglobinas/administração & dosagem , Neoplasias/tratamento farmacológico , Hipóxia Tumoral/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Hemoglobinas/farmacocinética , Hemoglobinas/uso terapêutico , Humanos , Lipossomos/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Neoplasias/patologia , Espécies Reativas de Oxigênio/metabolismo
8.
J Control Release ; 277: 114-125, 2018 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-29408424

RESUMO

The targeted drug delivery with the help of nanocarriers and the controlled drug release at the lesion sites are the most effective ways to enhance therapeutic efficacy and reduce side effects. Here, we built a light sensitive liposome (Her2-I&D-LSL) which was formed by a special phospholipid (PLsPC) and a hydrophobically modified photosensitizer (ICG-ODA). DOX was employed as the therapeutic drug, encapsulating in the internal phase of the liposome whose surface was modified by Her2 antibodies for recognizing tumor cells with high Her2 receptor expression. Mediated by NIR light, Her2-I&D-LSL was proved to generate sufficient ROS to realize PDT, which then triggered the release of DOX for combined chemotherapy. The ROS generation and DOX release were verified to be strictly controlled by NIR light and the proportion of ICG-ODA. Thanks to the mediation of Her2 receptor, the specific DOX release and the combination of PDT-chemotherapy triggered by NIR light, Her2-I&D-LSL showed a significant accumulation in MCF7 and SKOV3 tumors, thus leading to the strongest tumor growth inhibition effect compared to PDT alone (I-LSL) or chemotherapy alone (D-LSL). Her2-I&D-LSL also possessed a great biocompatibility due to the targeted treatment, holding promise for future cancer therapy in clinic.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Doxorrubicina/metabolismo , Liberação Controlada de Fármacos/fisiologia , Estimulação Luminosa/métodos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/metabolismo , Células A549 , Animais , Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Humanos , Lipossomos , Células MCF-7 , Masculino , Camundongos , Camundongos Nus , Fármacos Fotossensibilizantes/administração & dosagem
9.
Theranostics ; 8(11): 3059-3073, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29896302

RESUMO

Recently, nano-sized ultrasound contrast agents encapsulating drugs for cancer diagnosis and therapy have attracted much attention. However, the ultrasound signal of these agents is too weak to obtain an ideal ultrasound imaging effect. Furthermore, conventional ultrasound contrast agents with strong echo signal are not suitable for drug delivery against cancer because of their large size. To circumvent this problem, phase-transition ultrasound contrast agents are believed to be an excellent choice. Methods: Liposomes co-encapsulating doxorubicin (DOX), hollow gold nanospheres (HAuNS), and perfluorocarbon (PFC) were synthesized by film dispersion method. The morphology, particle size, and stability of these liposomes (DHPL) were investigated. The photothermal effect, drug release, particle size change, cytotoxicity, and ultrasound imaging were studied by using the near infrared (NIR) light. Furthermore, tumor accumulation of DHPL was observed by in vivo fluorescence imaging and the antitumor effect was verified in a 4T1 tumor model. Results: The nanosystem displayed a homogeneous size distribution (~200 nm) and an efficient light-to-heat conversion effect under 808 nm NIR laser irradiation. The nanometer size enabled considerable accumulation of DHPL in the tumor sites. The localized hyperthermia resulting from the photothermal effect of HAuNS could trigger the size transformation of DHPL followed by significant DOX release. Due to the gasification of PFC, a remarkably enhanced ultrasound signal was detected. DHPL also exhibited a prominent photothermally reinforced chemotherapeutic effect under the control of NIR light both in vitro and in vivo. Importantly, no systemic toxicity was observed by DHPL treatment. Conclusion: In this study, we fabricated multi-functional perfluorocarbon liposomes for ultrasound imaging-guided photothermal chemotherapy which have the potential to serve as a prospective cancer treatment approach.


Assuntos
Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Nanosferas/química , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Animais , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Fluorocarbonos/química , Ouro/química , Humanos , Raios Infravermelhos , Lipossomos/química , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/diagnóstico por imagem , Tamanho da Partícula , Temperatura , Ultrassonografia
10.
Colloids Surf B Biointerfaces ; 160: 446-454, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28985606

RESUMO

Hemoglobin-based oxygen carriers were developed as an alternative for blood transfusion. However, the research progress for their further clinic applications was slow in recent several years. Hypoxia is found in most solid tumors, which is responsible for the tumor formation, increased metastasis, drug resistance during therapeutic process as well as poor patient survival. In this work, novel hemoglobin (Hb) loaded nanoliposomes, as artificial red cells for oxygen delivery, were optimized by screening various types of phospholipids and analyzing different mole ratio of phospholipid to cholesterol. The nanoliposomes presented a high encapsulating efficiency to hemoglobin and also significantly enhanced its stability. The obtained hemoglobin loaded nanoliposome (HLL) could be lyophilized for long term storage. HLL did not cause significant cell death in the concentration range of 0-100µg equivalent Hb/mL under normoxia and hypoxia incubation conditions, suggesting the low cytotoxicity and high biocompatibility of HLL. Importantly, HLL could efficiently accumulate into subcutaneous and deep orthotopic tumors, inducing a significant decrease of hypoxia-inducible factors 1α subunits (HIF-1α) in the tumors and remarkably reduced expression of vascular endothelial growth factor (VEGF). The study of acute and chronic toxicity indicated that HLL did not induce obvious damage to main organs of mice after intravenous injections with total Hb dose of 120mg/kg. We presented a promising method for relieving the hypoxia degree in solid tumors and down-regulating HIF-1α protein by directly delivering oxygen into tumors, which will be very helpful for subsequent cancer therapy.


Assuntos
Hemoglobinas/química , Lipossomos/química , Nanopartículas/química , Hipóxia Tumoral , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Hemoglobinas/farmacocinética , Hemoglobinas/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipossomos/farmacocinética , Lipossomos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Microscopia Eletrônica de Transmissão , Nanopartículas/ultraestrutura , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Distribuição Tecidual , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA