Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(22): e2319880121, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38768353

RESUMEN

Elevated interstitial fluid pressure (IFP) within pathological tissues (e.g., tumors, obstructed kidneys, and cirrhotic livers) creates a significant hindrance to the transport of nanomedicine, ultimately impairing the therapeutic efficiency. Among these tissues, solid tumors present the most challenging scenario. While several strategies through reducing tumor IFP have been devised to enhance nanoparticle delivery, few approaches focus on modulating the intrinsic properties of nanoparticles to effectively counteract IFP during extravasation and penetration, which are precisely the stages obstructed by elevated IFP. Herein, we propose an innovative solution by engineering nanoparticles with a fusiform shape of high curvature, enabling efficient surmounting of IFP barriers during extravasation and penetration within tumor tissues. Through experimental and theoretical analyses, we demonstrate that the elongated nanoparticles with the highest mean curvature outperform spherical and rod-shaped counterparts against elevated IFP, leading to superior intratumoral accumulation and antitumor efficacy. Super-resolution microscopy and molecular dynamics simulations uncover the underlying mechanisms in which the high curvature contributes to diminished drag force in surmounting high-pressure differentials during extravasation. Simultaneously, the facilitated rotational movement augments the hopping frequency during penetration. This study effectively addresses the limitations posed by high-pressure impediments, uncovers the mutual interactions between the physical properties of NPs and their environment, and presents a promising avenue for advancing cancer treatment through nanomedicine.


Asunto(s)
Sistemas de Liberación de Medicamentos , Líquido Extracelular , Nanopartículas , Presión , Nanopartículas/química , Líquido Extracelular/metabolismo , Animales , Sistemas de Liberación de Medicamentos/métodos , Ratones , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Línea Celular Tumoral , Extravasación de Materiales Terapéuticos y Diagnósticos , Simulación de Dinámica Molecular , Antineoplásicos/farmacocinética , Antineoplásicos/administración & dosificación , Antineoplásicos/química
2.
Nano Lett ; 21(14): 6031-6041, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34240603

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease with a high recurrence rate and poor outcomes in clinic. In this study, inspired by the enriched innate immune cell type tumor-associated macrophages (TAMs) in TNBC, we proposed a matrix metalloprotease 2 (MMP2) responsive integrated immunochemotherapeutic strategy to deliver paclitaxel (PTX) and anti-CD47 (aCD47) by detachable immune liposomes (ILips). In the TNBC microenvironment, the "two-in-one" ILips facilitated MMP2-responsive release of aCD47 to efficiently polarize M2 macrophages toward the M1 phenotype to enhance phagocytosis against tumor cells and activate the systemic T cell immune response. Together with the immune effect, the detached PTX-loaded liposomes were internalized in MDA-MB-231 cells to synergistically inhibit tumor cell proliferation and metastasis. In the TNBC-bearing mouse model, PTX-loaded ILips demonstrated superior antitumor efficacy against TNBC and inhibited tumor recurrence. Our integrated strategy represents a promising approach to synchronously enhance immune response and tumor-killing effects, improving the therapeutic efficacy against TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Humanos , Liposomas , Ratones , Paclitaxel/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Microambiente Tumoral , Macrófagos Asociados a Tumores
3.
Acta Pharmacol Sin ; 42(10): 1714-1722, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33469196

RESUMEN

Lung cancer is one of the leading causes of cancer-related death worldwide. Various therapeutic failed in the effective treatment of the lung cancer due to their limited accumulation and exposure in tumors. In order to promote the chemotherapeutics delivery to lung tumor, we introduced chitosan oligosaccharide (CSO) modification on the liposomes. CSO conjugated Pluronic P123 polymers with different CSO grafting amounts, called as CP50 and CP20, were synthesized and used to prepare CSO modified liposomes (CP50-LSs and CP20-LSs). CP50-LSs and CP20-LSs displayed significantly enhanced cellular uptake in A549 cells in vitro as well as superior tumor accumulation in vivo compared with non-CSO modified liposomes (P-LSs). This phenomenon was related to the increased affinity between CSO modified liposomes and tumor cells following massive adsorption of collagen, which was highly expressed in lung tumors. In the A549 tumor-bearing mouse model, intravenous injection of paclitaxel (PTX)-loaded CP50-LSs every 3 days for 21 days resulted in optimal antitumor therapeutic performance with an inhibition rate of 86.4%. These results reveal that CSO modification provides promising applicability for nanomedicine design in the lung cancer treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Quitosano/química , Portadores de Fármacos/química , Liposomas/química , Neoplasias Pulmonares/tratamiento farmacológico , Paclitaxel/uso terapéutico , Células A549 , Animales , Antineoplásicos/química , Quitosano/metabolismo , Portadores de Fármacos/metabolismo , Liberación de Fármacos , Humanos , Liposomas/metabolismo , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Oligosacáridos/química , Oligosacáridos/metabolismo , Paclitaxel/química
4.
Acta Pharmacol Sin ; 41(9): 1261, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32081943

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nano Lett ; 18(4): 2411-2419, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29561622

RESUMEN

RNA interference (RNAi) technology has shown great promise for the treatment of cancer and other genetic disorders. Despite the efforts to increase the target tissue distribution, the safe and effective delivery of siRNA to the diseased cells with sufficient cytosolic transport is another critical factor for successful RNAi clinical application. Here, the constructed lipid-based liquid crystalline nanoparticles, called nano-Transformers, can transform thestructure in the intracellular acidic environment and perform high-efficient siRNA delivery for cancer treatment. The developed nano-Transformers have satisfactory siRNA loading efficiency and low cytotoxicity. Different from the traditional cationic nanocarriers, the endosomal membrane fusion induced by the conformational transition of lipids contributes to the easy dissociation of siRNA from nanocarriers and direct release of free siRNA into cytoplasm. We show that transfection with cyclin-dependent kinase 1 (CDK1)-siRNA-loaded nano-Transformers causes up to 95% reduction of relevant mRNA in vitro and greatly inhibits the tumor growth without causing any immunogenic response in vivo. This work highlights that the lipid-based nano-Transformers may become the next generation of siRNA delivery system with higher efficacy and improved safety profiles.


Asunto(s)
Lípidos/química , Cristales Líquidos/química , Nanopartículas/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , Animales , Proteína Quinasa CDC2/genética , Células Hep G2 , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/genética , Neoplasias/terapia , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia
6.
Acta Pharmacol Sin ; 38(2): 290-300, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27917871

RESUMEN

Acetylthevetin B (ATB), a cardiac glycoside from the seed of Thevetia peruviana (Pers) K Schum (yellow oleander), exhibits not only antitumor activity but also potential cardiac toxicity. In the present study, we attempted to enhance its antitumor action and decrease its adverse effects via chitosan-Pluronic P123 (CP) micelle encapsulation. Two ATB-loaded CP micelles (ATB-CP1, ATB-CP2) were prepared using an emulsion/solvent evaporation technique. They were spherical in shape with a particle size of 40-50 nm, showed a neutral zeta potential, and had acceptable encapsulation efficiency (>90%). Compared to the free ATB (IC50=2.94 µmol/L), ATB-loaded CP micelles exerted much stronger cytotoxicity against human lung cancer A549 cells with lower IC50 values (0.76 and 1.44 µmol/L for ATB-CP1 and ATB-CP2, respectively). After administration of a single dose in mice, the accumulation of ATB-loaded CP1 micelles in the tumor and lungs, respectively, was 15.31-fold and 9.49-fold as high as that of free ATB. A549 xenograft tumor mice treated with ATB-loaded CP1 micelles for 21 d showed the smallest tumor volume (one-fourth of that in the control group) and the highest inhibition rate (85.6%) among all the treatment groups. After 21-d treatment, no significant pathological changes were observed in hearts and other main tissues. In summary, ATB may serve as a promising antitumor chemotherapeutic agent for lung cancer, and its antitumor efficacy was significantly improved by CP micelles, with lower adverse effects.


Asunto(s)
Glicósidos Cardíacos/administración & dosificación , Glicósidos Cardíacos/farmacología , Portadores de Fármacos/química , Micelas , Poloxaleno/química , Animales , Glicósidos Cardíacos/uso terapéutico , Línea Celular Tumoral , Quitosano/química , Humanos , Ratones , Tamaño de la Partícula , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Control Release ; 371: 298-312, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38815703

RESUMEN

Wound management is a critical clinical challenge due to the dynamic and complex pathological characteristics of inflammation, proliferation, and matrix remodeling. To address this challenge, the regulation and management of this multi-stage pathological microenvironment may provide a feasible approach to wound healing. In this work, we synthesized a new lipid material (DA) with reactive oxygen species (ROS) scavenging effect to prepare DA-based liquid crystalline (DALC). Then, DALC was incorporated with adipose mesenchymal stem cells-derived extracellular vesicles (AMSC-EVs) to fabricate a novel scaffold dressing (EVs@DALC) for the treatment of the wound. DALC not only endowed EVs@DALC with ROS scavenging sites for relieving the oxidative stress and inflammation in the microenvironment of the wound site, but also facilitated cellular uptake and transfection of microRNA and growth factors contained in AMSC-EVs. Benefiting from DALC, AMSC-EVs effectively transferred microRNA and growth factors into the skin cells to induce cell proliferation and migration and accelerate angiogenesis. The results of wound healing effect in vivo indicate EVs@DALC achieved multi-stage pathological modulation for accelerating wound healing through alleviating inflammation, promoting cell proliferation and migration, and angiogenesis. Taken together, this work provides an effective strategy based on antioxidant lipid liquid crystalline delivering extracellular vesicles in treating skin wounds and paves a way for stem cell extracellular vesicles clinical translation.


Asunto(s)
Proliferación Celular , Vesículas Extracelulares , Lípidos , Cristales Líquidos , Células Madre Mesenquimatosas , Especies Reactivas de Oxígeno , Cicatrización de Heridas , Cicatrización de Heridas/efectos de los fármacos , Cristales Líquidos/química , Animales , Especies Reactivas de Oxígeno/metabolismo , Humanos , Lípidos/química , Proliferación Celular/efectos de los fármacos , Masculino , Movimiento Celular/efectos de los fármacos , MicroARNs/administración & dosificación , Piel/metabolismo , Depuradores de Radicales Libres/administración & dosificación , Tejido Adiposo/citología , Ratones
8.
Nat Commun ; 15(1): 1159, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326312

RESUMEN

The dynamic protein corona formed on nanocarriers has been revealed to strongly affect their in vivo behaviors. Precisely manipulating the formation of protein corona on nanocarriers may provide an alternative impetus for specific drug delivery. Herein, we explore the role of glycosylated polyhydroxy polymer-modified nanovesicles (CP-LVs) with different amino/hydroxyl ratios in protein corona formation and evolution. CP-LVs with an amino/hydroxyl ratio of approximately 0.4 (CP1-LVs) are found to efficiently suppress immunoglobulin adsorption in blood and livers, resulting in prolonged circulation. Moreover, CP1-LVs adsorb abundant tumor distinctive proteins, such as CD44 and osteopontin in tumor interstitial fluids, mediating selective tumor cell internalization. The proteins corona transformation specific to the environment appears to be affected by the electrostatic interaction between CP-LVs and proteins with diverse isoelectric points. Benefiting from surface modification-mediated protein corona regulation, paclitaxel-loaded CP1-LVs demonstrate superior antitumor efficacy to PEGylated liposomes. Our work offers a perspective on rational surface-design of nanocarriers to modulate the protein corona formation for efficient drug delivery.


Asunto(s)
Nanopartículas , Corona de Proteínas , Polímeros , Corona de Proteínas/metabolismo , Nanopartículas/metabolismo , Sistemas de Liberación de Medicamentos , Osteopontina
9.
Sci Bull (Beijing) ; 69(9): 1263-1274, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38418300

RESUMEN

Metabolic reprogramming is a mechanism by which cancer cells alter their metabolic patterns to promote cell proliferation and growth, thereby enabling their resistance to external stress. 2-Deoxy-D-glucose (2DG) can eliminate their energy source by inhibiting glucose glycolysis, leading to cancer cell death through starvation. However, a compensatory increase in mitochondrial metabolism inhibits its efficacy. Herein, we propose a synergistic approach that combines photodynamic therapy (PDT) with starvation therapy to address this challenge. To monitor the nanodrugs and determine the optimal triggering time for precise tumor therapy, a multifunctional nano-platform comprising lanthanide-doped nanoparticle (LnNP) cores was constructed and combined with mesoporous silicon shells loaded with 2DG and photosensitizer chlorin e6 (Ce6) in the mesopore channels. Under 980 nm near-infrared light excitation, the downshifted 1550 nm fluorescence signal in the second near-infrared (NIR-II, 1000-1700 nm) window from the LnNPs was used to monitor the accumulation of nanomaterials in tumors. Furthermore, upconverted 650 nm light excited the Ce6 to generate singlet oxygen for PDT, which damaged mitochondrial function and enhanced the efficacy of 2DG by inhibiting hexokinase 2 and lactate dehydrogenase A expressions. As a result, glucose metabolism reprogramming was inhibited and the efficiency of starvation therapy was significantly enhanced. Overall, the proposed NIR-II bioimaging-guided PDT-augmented starvation therapy, which simultaneously inhibited glycolysis and mitochondria, facilitated the effects of a cancer theranostic system.


Asunto(s)
Clorofilidas , Glucosa , Nanopartículas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Porfirinas , Fotoquimioterapia/métodos , Humanos , Animales , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/farmacología , Porfirinas/uso terapéutico , Glucosa/metabolismo , Nanopartículas/uso terapéutico , Desoxiglucosa/farmacología , Ratones , Rayos Infrarrojos , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias/diagnóstico por imagen , Hexoquinasa/metabolismo , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Glucólisis/efectos de los fármacos , Reprogramación Metabólica
10.
Bioact Mater ; 29: 116-131, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37456582

RESUMEN

Chemotherapy can induce a robust T cell antitumor immune response by triggering immunogenic cell death (ICD), a process in which tumor cells convert from nonimmunogenic to immunogenic forms. However, the antitumor immune response of ICD remains limited due to the low immunogenicity of tumor cells and the immunosuppressive tumor microenvironment. Although autophagy is involved in activating tumor immunity, the synergistic role of autophagy in ICD remains elusive and challenging. Herein, we report an autophagy amplification strategy using an ion-chelation reaction to augment chemoimmunotherapy in cancer treatments based on zinc ion (Zn2+)-doped, disulfiram (DSF)-loaded mesoporous silica nanoparticles (DSF@Zn-DMSNs). Upon pH-sensitive biodegradation of DSF@Zn-DMSNs, Zn2+ and DSF are coreleased in the mildly acidic tumor microenvironment, leading to the formation of toxic Zn2+ chelate through an in situ chelation reaction. Consequently, this chelate not only significantly stimulates cellular apoptosis and generates damage-associated molecular patterns (DAMPs) but also activates autophagy, which mediates the amplified release of DAMPs to enhance ICD. In vivo results demonstrated that DSF@Zn-DMSNs exhibit strong therapeutic efficacy via in situ ion chelation and possess the ability to activate autophagy, thus enhancing immunotherapy by promoting the infiltration of T cells. This study provides a smart in situ chelation strategy with tumor microenvironment-responsive autophagy amplification to achieve high tumor chemoimmunotherapy efficacy and biosafety.

11.
ACS Nano ; 16(4): 6527-6540, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35426300

RESUMEN

Despite considerable advancements in cell membrane-camouflaged nanocarriers to leverage natural cell functions, artificial nanocarriers that can accurately mimic both the biological and physical properties of cells are urgently needed. Herein, inspired by the important effect of the stiffness and deformability of natural red blood cells (RBCs) on their life span and flowing through narrow vessels, we report the construction of RBC membrane-camouflaged nanocarriers that can mimic RBCs at different life stages and study how the deformability of RBC-derived nanocarriers affects their biological behaviors. RBC membrane-coated elastic poly(ethylene glycol) diacrylate hydrogel nanoparticles (RBC-ENPs) simulating dynamic RBCs exhibited high immunocompatibility with minimum immunoglobulin adsorption in the surface protein corona, resulting in reduced opsonization in macrophages and ultralong circulation. Furthermore, RBC-ENPs can deform like RBCs and achieve excellent diffusion in tumor extracellular matrix, leading to improved multicellular spheroid penetration and tumor tissue accumulation. In mouse cancer models, doxorubicin-loaded RBC-ENPs demonstrated superior antitumor efficacy to the first-line chemotherapeutic drug PEGylated doxorubicin liposomes. Our work highlights that tuning the physical properties of cell membrane-derived nanocarriers may offer an alternative approach for the bionic design of nanomedicines in the future.


Asunto(s)
Biomimética , Neoplasias , Ratones , Animales , Eritrocitos , Membrana Celular , Doxorrubicina/farmacología , Neoplasias/terapia
12.
J Extracell Vesicles ; 11(3): e12198, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35233952

RESUMEN

Extracellular vesicles (EVs) have been proved a promising small interfering RNA (siRNA) delivery vehicle to mediate gene-silencing. Tumour-derived extracellular vesicles (TDEVs) as genetic exchange vectors in the tumour microenvironment, enable intercellular communication for a wide range of endogenous cargo molecules, such as RNAs and proteins. However, the oncogenic cargo of TDEVs limits their application in siRNA delivery for cancer therapy. Herein, we isolated TDEVs from hepatocellular carcinoma (HCC) cells and derived TDEV membranes by abandoning their content. Innovative TDEV membrane hybrid lipid nanovesicles (LEVs) were then fabricated by fusion of TDEV membranes and phospholipids to realize precise delivery to tumours and highly efficient transfection of siRNA. The TDEV membranes endow LEVs with 'homing' targeting ability, facilitating specific internalisation into parent HCC cells primarily through heparan sulfate proteoglycan-mediated pathways. Unlike conventional lipid-based nanovesicles, LEVs can bypass the endosomal degradation pathway, boost the delivery of siRNA through the Golgi and endoplasmic reticulum (ER) intracellular 'freeway' transportation, achieving a 1.7-fold improvement in siRNA transfection efficiency compared with liposomes. Additionally, siRNA loaded LEVs were demonstrated to enhance the antitumour efficacy in HCC bearing mice through effective gene silencing in the tumour sites. Our results highlight the potential application of the TDEV membrane-derived nanovesicles as an advanced siRNA delivery strategy for cancer therapy.


Asunto(s)
Carcinoma Hepatocelular , Vesículas Extracelulares , Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular/genética , Vesículas Extracelulares/metabolismo , Neoplasias Hepáticas/genética , Lípidos de la Membrana/metabolismo , Ratones , ARN Interferente Pequeño , Microambiente Tumoral
13.
Cell Discov ; 8(1): 9, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35102138

RESUMEN

Safe, effective, and economical vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are needed to achieve adequate herd immunity and end the pandemic. We constructed a novel SARS-CoV-2 vaccine, CoVac501, which is a self-adjuvanting peptide vaccine conjugated with Toll-like receptor 7 (TLR7) agonists. The vaccine contains immunodominant peptides screened from the receptor-binding domain (RBD) and is fully chemically synthesized. It has been formulated in an optimized nanoemulsion formulation and is stable at 40 °C for 1 month. In non-human primates (NHPs), CoVac501 elicited high and persistent titers of protective neutralizing antibodies against multiple RBD mutations, SARS-CoV-2 original strain, and variants (B.1.1.7 and B.1.617.2). Specific peptides booster immunization against the B.1.351 variant has also been shown to be effective in improving protection against B.1.351. Meanwhile, CoVac501 elicited the increase of memory T cells, antigen-specific CD8+ T-cell responses, and Th1-biased CD4+ T-cell immune responses in NHPs. Notably, at an extremely high SARS-CoV-2 challenge dose of 1 × 107 TCID50, CoVac501 provided near-complete protection for the upper and lower respiratory tracts of cynomolgus macaques.

14.
Int J Pharm ; 583: 119354, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32348799

RESUMEN

Phosphatidylcholines (PCs) have been widely used in pharmaceutical research. Unfortunately, our understanding of how PCs influence the in vivo lipolysis process of drug delivery systems is still limited. In this study, PCs with fatty acid chains of varying lengths and saturability were used as emulsifiers to prepare curcumin-loaded nanoemulsions (Cur-NEs). The differences in particle size as well as drug and free fatty acid release during the lipolysis process were studied in a simulated blood environment. Furthermore, the pharmacokinetics and antitumor efficacy of Cur-NEs were evaluated in mice. The prepared 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC)-stabilized Cur-NEs showed similar particle size and stability during storage but exhibited different lipolysis behaviors in vitro and in vivo. Due to the gel state of DPPC in the physiological environment, DPPC-stabilized Cur-NEs had low binding affinity with proteins and maintained their integrity in plasma, leading to sustained drug release, prolonged circulation time and enhanced antitumor efficacy in 4T1 tumor-bearing mice. In contrast, DOPC and DSPC-stabilized Cur-NEs were prone to coalescence in the plasma, resulting in rapid drug release and elimination from circulation. Our findings demonstrated that proper use of PCs is beneficial for obtaining desired transport behavior and drug therapeutic effects, providing guiding principles for rational design of nanodelivery systems.


Asunto(s)
Antineoplásicos/química , Curcumina/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Fosfatidilcolinas/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Curcumina/administración & dosificación , Curcumina/farmacocinética , Estabilidad de Medicamentos , Emulsiones , Ácidos Grasos no Esterificados/metabolismo , Femenino , Lipólisis , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Fosfatidilcolinas/administración & dosificación , Fosfatidilcolinas/farmacocinética , Ratas Sprague-Dawley
15.
Theranostics ; 10(8): 3722-3736, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32206118

RESUMEN

Rationale: Antitumor drug delivery faces multiple barriers that require consecutively achieving tumor targeting, selective cellular uptake and sufficient intracellular drug dosage. Methods: Herein, we designed smart nanoparticles (GPDC-MSNs) that can accumulate stepwise in tumor tissues, selectively enter cancer cells by responding to the acidic tumor extracellular environment, and achieve considerable drug release in the intracellular microenvironment. The GPDC-MSNs comprise the synthesized material galactosyl-conjugated PEO-PPO-PEO (Gal-P123) for hepatocellular carcinoma (HCC) targeting, the tumor extracellular pH-responsive lipid (2E)-4-(dioleostearin)-amino-4-carbonyl-2-butenonic (DC) for selective cellular internalization, and antitumor drug irinotecan (CPT-11)-loaded mesoporous silica nanoparticles (MSNs) for on-demand intracellular drug release. Results: GPDC-MSNs are negatively charged at pH 7.4 and promote active HCC targeting mediated by the asialoglycoprotein receptor. Upon reaching the weakly acidic tumor microenvironment, the nanoparticles undergo charge conversion to neutral, enhancing cellular internalization. Moreover, the encapsulated CPT-11 can be retained within GPDC-MSNs in the blood circulation but undergo intracellular burst release, which facilitates the apoptosis of tumor cells. GPDC-MSNs significantly increased HCC selectivity in vivo and exhibited up to 25 times higher accumulation in tumor tissue than in normal hepatic tissue, thus achieving superior antitumor efficacy and low systemic toxicity. Conclusion: This stepwise-responsive nanoparticle should serve as a valuable platform and promising strategy for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Portadores de Fármacos/uso terapéutico , Liberación de Fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Irinotecán/administración & dosificación , Irinotecán/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Sprague-Dawley , Dióxido de Silicio/química , Microambiente Tumoral
16.
Acta Pharm Sin B ; 9(4): 858-870, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31384544

RESUMEN

Recently, liposomes have been widely used in cancer therapeutics, but their anti-tumor effects are suboptimal due to limited tumor penetration. To solve this problem, researchers have made significant efforts to optimize liposomal diameters and potentials, but little attention has been paid to liposomal membrane rigidity. Herein, we sought to demonstrate the effects of cholesterol-tuned liposomal membrane rigidity on tumor penetration and anti-tumor effects. In this study, liposomes composed of hydrogenated soybean phospholipids (HSPC), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000) and different concentrations of cholesterol were prepared. It was revealed that liposomal membrane rigidity decreased with the addition of cholesterol. Moderate cholesterol content conferred excellent diffusivity to liposomes in simulated diffusion medium, while excessive cholesterol limited the diffusion process. We concluded that the differences of the diffusion rates likely stemmed from the alterations in liposomal membrane rigidity, with moderate rigidity leading to improved diffusion. Next, the in vitro tumor penetration and the in vivo anti-tumor effects were analyzed. The results showed that liposomes with moderate rigidity gained excellent tumor penetration and enhanced anti-tumor effects. These findings illustrate a feasible and effective way to improve tumor penetration and therapeutic efficacy of liposomes by changing the cholesterol content, and highlight the importance of liposomal membrane rigidity.

17.
ACS Nano ; 13(7): 7676-7689, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31187973

RESUMEN

Small unilamellar vesicles (SUVs), ubiquitous in organisms, play key and active roles in various biological processes. Although the physical properties of the constituent lipid molecules (i.e., the acyl chain length and saturation) are known to affect the mechanical properties of SUVs and consequently regulate their biological behaviors and functions, the underlying mechanism remains elusive. Here, we combined theoretical modeling and experimental investigation to probe the mechanical behaviors of SUVs with different lipid compositions. The membrane bending rigidity of SUVs increased with increasing chain length and saturation, resulting in differences in the vesicle rigidity and deformable capacity. Furthermore, we tested the tumor delivery capacity of liposomes with low, intermediate, and high rigidity as typical models for SUVs. Interestingly, liposomes with intermediate rigidity exhibited better tumor extracellular matrix diffusion and multicellular spheroid (MCS) penetration and retention than that of their stiffer or softer counterparts, contributing to improved tumor suppression. Stiff SUVs had superior cellular internalization capacity but intermediate tumor delivery efficacy. Stimulated emission depletion microscopy directly showed that the optimal formulation was able to transform to a rod-like shape in MCSs, which stimulated fast transport in tumor tissues. In contrast, stiff liposomes hardly deformed, whereas soft liposomes changed their shape irregularly, which slowed their MCS penetration. Our findings introduce special perspectives from which to map the detailed mechanical properties of SUVs with different compositions, provide clues for understanding the biological functions of SUVs, and suggest that liposome mechanics may be a design parameter for enhancing drug delivery.


Asunto(s)
Antineoplásicos/farmacología , Camptotecina/análogos & derivados , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Estrés Mecánico , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Camptotecina/química , Camptotecina/farmacología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Portadores de Fármacos/química , Liberación de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Liposomas/sangre , Liposomas/síntesis química , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Moleculares , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Imagen Óptica , Neoplasias Pancreáticas/patología , Tamaño de la Partícula , Propiedades de Superficie , Células Tumorales Cultivadas
18.
J Control Release ; 286: 348-357, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30077738

RESUMEN

Exosomes secreted from cancer cells promote tumor progression through the transfection of containing microRNA (miRNA), mRNAs and proteins. Yet, little of this knowledge has translated into the therapeutic application. Herein, we propose a tumor therapeutic strategy via decreasing exosomal miRNA secretion. The study designed small interfering RNA (siRNA) loaded nanoparticles to downregulate sphingosine kinase 2 (Sphk2) and investigate their potential in decreasing exosomal oncogenic miRNA content and inhibiting tumor growth. The synthesized lipid (2E)-4-(dioleostearin)-amino-4­carbonyl-2-butenoic (DC) and chitosan were utilized to produce siRNA loaded nanoparticles (DC/CS-siRNA NPs), with optimal siRNA complexation and high transfection efficacy. We demonstrated that Sphk2 gene silencing induced by nanoparticles in hepatocellular carcinoma (HCC) cells could reduce miRNA-21 sorting into exosomes, contributing to the inhibition of tumor cell migration and tumorigenic function of exosomes to normal liver cells. Furthermore, in xenograft mouse model, Sphk2 siRNA loaded DC/CS NPs could significantly block tumor progression of malignancy HCC. These results suggest a new therapeutic approach for tumor treatment by ablating oncogenic miRNA in malicious exosomes.


Asunto(s)
Carcinoma Hepatocelular/terapia , Exosomas/genética , Neoplasias Hepáticas/terapia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Quitosano/análogos & derivados , Portadores de Fármacos/química , Exosomas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , MicroARNs/genética , Nanopartículas/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia/métodos
19.
Int J Nanomedicine ; 12: 2033-2044, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28352173

RESUMEN

The development of noninvasive imaging techniques for the accurate diagnosis of progressive hepatocellular carcinoma (HCC) is of great clinical significance and has always been desired. Herein, a hepatocellular carcinoma cell-targeting fluorescent magnetic nanoparticle (NP) was obtained by conjugating near-infrared fluorescence to the surface of Fe3O4 (NIRF-Fe3O4) NPs, followed by coating the lipids consisting of tumoral hepatocytes-targeting polymer (Gal-P123). This magnetic NP (GPC@NIRF-Fe3O4) with superparamagnetic behavior showed high stability and safety in physiological conditions. In addition, GPC@NIRF-Fe3O4 achieved more specific uptake of human liver cancer cells than free Fe3O4 NPs. Importantly, with superpara-magnetic iron oxide and strong NIR absorbance, GPC@NIRF-Fe3O4 NPs demonstrate prominent tumor-contrasted imaging performance both on fluorescent and T2-weighted magnetic resonance (MR) imaging modalities in a living body. The relative MR signal enhancement of GPC@NIRF-Fe3O4 NPs achieved 5.4-fold improvement compared with NIR-Fe3O4 NPs. Therefore, GPC@ NIRF-Fe3O4 NPs may be potentially used as a candidate for dual-modal imaging of tumors with information covalidated and directly compared by combining fluorescence and MR imaging.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Lípidos/química , Neoplasias Hepáticas/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita/química , Animales , Carcinoma Hepatocelular/patología , Compuestos Férricos/química , Fluorescencia , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/patología , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA