Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
ACS Appl Mater Interfaces ; 16(35): 45901-45916, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39169670

RESUMEN

Immune checkpoint blockade (ICB) therapy, while showing promise in various cancers, exhibits limited effectiveness in hepatic carcinoma due to the tumor's immunosuppressive microenvironment (TME) and challenges associated with immune cell infiltration. Efforts to transform the "cold" TME into an "inflamed" state, notably through chemo-immunotherapy, have sparked interest due to their potential to induce immunogenic cell death and augment the infiltration of cytotoxic T lymphocytes (CTLs). Nonetheless, the efficacy of chemo-immunotherapy is often compromised by suboptimal pharmacokinetics, poor tumor accumulation, and off-target toxicity. Herein, in response, we introduce an innovative, milder thermal therapeutic approach leveraging gold nano frameworks with mesopores for the targeted delivery of the immunostimulant imiquimod and NIR-II photothermal therapy. This strategy employs targeted molecule modifications to ensure precise tumor targeting, guided by photoacoustic imaging. Subsequent to mild thermal treatment, there is a release of immunogenic proteins (CRT and HSP90), enhancing tumor immunogenicity. Assisted by imiquimod, substantial CTL infiltration occurs, accompanied by pro-inflammatory factor release (TNF-α, IL-6), transforming M2 macrophages into the M1 phenotype. Ultimately, the proposed strategy combines PD-L1/PD-1 blockade, imiquimod and mild thermal treatment to synergistically enhance tumor immunogenicity, remodel the TME, and restrain hepatic carcinoma, making strides in ICB synergistic immune-thermal therapy.


Asunto(s)
Oro , Imiquimod , Inhibidores de Puntos de Control Inmunológico , Neoplasias Hepáticas , Terapia Fototérmica , Oro/química , Animales , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Imiquimod/química , Imiquimod/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/química , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Humanos , Inmunoterapia , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Línea Celular Tumoral
2.
Int J Nanomedicine ; 19: 6577-6588, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975319

RESUMEN

Introduction: Hepatocellular carcinomas (HCC) have a high morbidity and mortality rate, and is difficult to cure and prone to recurrence when it has already developed. Therefore, early detection and efficient treatment of HCC is necessary. Methods: In this study, we synthesized a novel NDI polymer with uniform size, long-term stability, and high near-infrared two-zone (NIR-II) absorption efficiency, which can greatly enhance the effect of photothermal therapy (PTT) after intravenous injection into Huh-7-tumor bearing mice. Results: The in vitro and in vivo studies showed that NDI polymer exhibited excellent NIR-guided PTT treatment, and the antitumor effect was approximately 88.5%, with obvious antimetastatic effects. Conclusion: This study developed an NDI polymer-mediated integrated diagnostic and therapeutic modality for NIR-II fluorescence imaging and photothermal therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Terapia Fototérmica , Polímeros , Animales , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Terapia Fototérmica/métodos , Polímeros/química , Ratones , Humanos , Línea Celular Tumoral , Rayos Infrarrojos , Ratones Desnudos , Imagen Óptica , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto , Fototerapia/métodos
3.
J Mater Chem B ; 12(24): 5812-5822, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38856691

RESUMEN

Immunotherapy is an important approach in cancer treatment. Transdermal administration is emerging as a promising method for delivering immunotherapeutics. Dissolving microneedles are made mainly of soluble or biodegradable polymers and have garnered widespread attention due to their painlessness, safety, convenience, excellent drug loading capacity, and easy availability of various materials, making them an ideal transdermal delivery system. This review comprehensively summarized the preparation methods, materials, and applications of dissolving microneedles in cancer vaccines, immune checkpoint inhibitors, and adoptive cell therapy. Additionally, the challenges and perspectives associated with their future clinical translation are discussed.


Asunto(s)
Administración Cutánea , Sistemas de Liberación de Medicamentos , Inmunoterapia , Agujas , Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Animales , Vacunas contra el Cáncer/administración & dosificación
4.
Adv Sci (Weinh) ; 11(17): e2306076, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38445883

RESUMEN

Earthworms, long utilized in traditional medicine, serve as a source of inspiration for modern therapeutics. Lysenin, a defensive factor in the coelom fluid of the earthworm Eisenia fetida, has multiple bioactivities. However, the inherent toxicity of Lysenin as a pore-forming protein (PFP) restricts its application in therapy. Here, a gene therapy strategy based on Lysenin for cancer treatment is presented. The formulation consists of polymeric nanoparticles complexed with the plasmid encoding Lysenin. After transfection in vitro, melanoma cells can express Lysenin, resulting in necrosis, autophagy, and immunogenic cell death. The secretory signal peptide alters the intracellular distribution of the expressed product of Lysenin, thereby potentiating its anticancer efficacy. The intratumor injection of Lysenin gene formulation can efficiently kill the transfected melanoma cells and activate the antitumor immune response. Notably, no obvious systemic toxicity is observed during the treatment. Non-viral gene therapy based on Lysenin derived from Eisenia foetida exhibits potential in cancer therapy, which can inspire future cancer therapeutics.


Asunto(s)
Terapia Genética , Melanoma , Oligoquetos , Animales , Ratones , Línea Celular Tumoral , Modelos Animales de Enfermedad , Terapia Genética/métodos , Melanoma/terapia , Melanoma/genética , Nanopartículas/química , Oligoquetos/genética , Toxinas Biológicas/genética , Femenino , Humanos
5.
Cancer Cell ; 40(9): 1044-1059.e8, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36099882

RESUMEN

Cisplatin-based chemotherapy remains the primary treatment for unresectable and metastatic muscle-invasive bladder cancers (MIBCs). However, tumors frequently develop chemoresistance. Here, we established a primary and orthotopic MIBC mouse model with gene-edited organoids to recapitulate the full course of chemotherapy in patients. We found that partial squamous differentiation, called semi-squamatization, is associated with acquired chemoresistance in both mice and human MIBCs. Multi-omics analyses showed that cathepsin H (CTSH) is correlated with chemoresistance and semi-squamatization. Cathepsin inhibition by E64 treatment induces full squamous differentiation and pyroptosis, and thus specifically restrains chemoresistant MIBCs. Mechanistically, E64 treatment activates the tumor necrosis factor pathway, which is required for the terminal differentiation and pyroptosis of chemoresistant MIBC cells. Our study revealed that semi-squamatization is a type of lineage plasticity associated with chemoresistance, suggesting that differentiation via targeting of CTSH is a potential therapeutic strategy for the treatment of chemoresistant MIBCs.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Vejiga Urinaria , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Diferenciación Celular , Cisplatino , Humanos , Ratones , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
6.
Int J Pharm ; 626: 122187, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36100145

RESUMEN

Detoxification of glutathione (GSH) and insufficient cellular uptake of cisplatin (CDDP) severely compromised the therapeutic efficacy of CDDP. Here, a nano-delivery system (BT-4@PtPPNPs) for CDDP prodrug (C16-Pt(Ⅳ)-PEG) based on a novel sulfhydryl blocking reagent methyl 2-(methylsulfonyl) benzothiazole-6-carboxylate (BT-4) was developed. On the one hand, BT-4 can deplete GSH in tumor cells by directly interacting with reactive sulfhydryl group on GSH, thereby increasing the cytotoxicity of CDDP. On the other hand, the CDDP prodrug carrier C16-Pt(IV)-PEG can promote the distribution of CDDP in tumors, reduce the probability of unexpected inactivation of CDDP, and reduce the content of GSH in tumor cells during the conversion to CDDP, thereby making CDDP more effective for treatment. The results showed that the optimized BT-4@PtPPNPs with a small particle size (130 nm) exhibited notable cytotoxicity and apoptosis of 4T1 cells. BT-4@PtPPNPs not only significantly improved the uptake of drugs by tumor cells, but also rapidly targeted and accumulated in the tumors for a long time. Moreover, in vivo efficacy studies showed that BT-4@PtPPNPs could effectively inhibit tumor growth, inhibiting 60.85 % of tumors in a 4T1 breast cancer mice model, showing superior antitumor activity, which can be attributed to GSH-triggered CDDP tolerance reversal. Overall, this study provides an attractive and simple strategy to combine novel sulfhydryl blockers and CDDP prodrugs to potentiate the efficacy of CDDP in breast cancer.


Asunto(s)
Antineoplásicos , Neoplasias , Profármacos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Benzotiazoles , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Glutatión , Ratones , Micelas , Neoplasias/tratamiento farmacológico , Profármacos/farmacología , Profármacos/uso terapéutico
7.
Int J Nanomedicine ; 16: 7697-7709, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34819728

RESUMEN

INTRODUCTION: ß-lactamase (LACTB) is a tumor suppressor gene in various tumors including melanoma. However, it remains challenging to efficiently deliver the LACTB gene into melanoma. Recently, we designed a nonviral nanocarrier iRGD/DOTAP/MPEG-PDLLA (iDPP) that could deliver gene targetedly to melanoma efficiently without obvious adverse effects. METHODS: In this study, the tumor-targeted nanoparticle iDPP was prepared to deliver LACTB gene to treat melanoma in vitro and in vivo. First, the expression level of LACTB in 6 clinical specimens of melanoma patients was evaluated. Subsequently, the characteristics of iDPP/LACTB nanocomplexes were studied. Afterwards, the in vitro and in vivo anti-tumor efficacy of the iDPP/LACTB nanocomplexes were explored utilizing the B16-F10 mouse melanoma cell line and the B16-F10 subcutaneous melanoma model. RESULTS: Compared with the normal epithelium, the expression level of LACTB in melanoma tissues was significantly downregulated. In vitro B16-F10 cell tests showed iDPP/LACTB nanocomplexes could increase the mRNA levels of P21, Bid, Bax, Pidd1, and Sival genes and up-regulate the p53 signaling pathway of melanoma cells, thus promoting cell apoptosis and blocking the cell cycle. Injected intravenously, iDPP nanoparticles could deliver DNA to the subcutaneous melanoma targetedly. Based on in vivo mouse xenograft model, iDPP/LACTB nanocomplexes could effectively inhibit tumor proliferation and induce tumor apoptosis, thus significantly inhibiting melanoma growth (tumor inhibition rate is about 68%) in the subcutaneous B16-F10 melanoma model. CONCLUSION: The downregulated LACTB might be a potential target for melanoma therapy. The iDPP/LACTB nanocomplexes could inhibit the growth of the mouse melanoma without obvious side effects, which provide a new option for melanoma gene therapy research.


Asunto(s)
Antineoplásicos , Melanoma Experimental , Nanopartículas , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Terapia Genética , Humanos , Melanoma Experimental/tratamiento farmacológico , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , beta-Lactamasas/farmacología , beta-Lactamasas/uso terapéutico
8.
Front Oncol ; 11: 680402, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34660263

RESUMEN

BACKGROUND: Microtubule-associated proteins (MAPs) have been considered to play significant roles in the tumor evolution of non-small cell lung cancer (NSCLC). Nevertheless, mRNA transcription levels and prognostic value of distinct MAPs in patients with NSCLC remain to be clarified. METHODS: In this study, the Oncomine database, Gene Expression Profiling Interactive Analysis (GEPIA) database, and Human Protein Atlas were utilized to analyze the relationship between mRNA/protein expression of different MAPs and clinical characteristics in NSCLC patients, including tumor type and pathological stage. The correlation between the transcription level of MAPs and overall survival (OS) of NSCLC patients was analyzed by Kaplan-Meier plotter. Besides, 50 frequently altered neighbor genes of the MAPs were screened out, and a network has been constructed via the cBioPortal and Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) dataset. Meanwhile, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis on the expression data of MAPs and their 50 frequently altered neighbor genes in NSCLC tissues. Furthermore, The Cancer Immunome Atlas (TCIA) was utilized to analyze the relationship between MAP expression and the response to immunotherapy. Finally, we used reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to verify the expression of MAPs in 20 patients with NSCLC. RESULTS: The present study discovered that the mRNA transcription levels of MAP7/7D2 were enriched in NSCLC tissues, while those of the MAP2/4/6/7D3 were lower in NSCLC specimens than those in control specimens. The mRNA transcription level of MAP6 was significantly associated with the advanced stage of NSCLC. Besides, survival analysis indicated that higher mRNA expressions of MAP2/4/6/7/7D3 were correlated considerably with favorable OS of NSCLC patients, whereas increased mRNA expression levels of MAP1A/1S were associated with poor OS. Moreover, the expression of MAP1A/1B/1S/4/6/7D1/7D3 was significantly correlated with immunophenoscore (IPS) in NSCLC patients. CONCLUSIONS: Our analysis indicated that MAP1A/1S could serve as potential personalized therapeutic targets for patients with NSCLC, and the enriched MAP2/4/6/7/7D3 expression could serve as a biomarker for favorable prognosis in NSCLC. Besides, the expression of MAP1A/1B/1S/4/6/7D1/7D3 was closely related to the response to immunotherapy. Taken together, MAP expression has potential application value in the clinical treatment and prognosis assessment of NSCLC patients, and further verifiable experiments can be conducted to verify our results.

9.
Blood Adv ; 5(23): 5269-5282, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34592762

RESUMEN

Multiple myeloma, a plasma cell malignancy in the bone marrow, remains largely incurable with currently available therapeutics. In this study, we discovered that the activated leukocyte cell adhesion molecule (ALCAM) interacted with epidermal growth factor receptor (EGFR), and regulated myelomagenesis. ALCAM was a negative regulator of myeloma clonogenicity. ALCAM expression was positively correlated with patients' survival. ALCAM-knockdown myeloma cells displayed enhanced colony formation in the presence of bone marrow stromal cells (BMSCs). BMSCs supported myeloma colony formation by secreted epidermal growth factor (EGF), which bound with its receptor (EGFR) on myeloma cells and activated Mek/Erk cell signaling, PI3K/Akt cell signaling, and hedgehog pathway. ALCAM could also bind with EGFR, block EGF from binding to EGFR, and abolish EGFR-initiated cell signaling. Hence, our study identifies ALCAM as a novel negative regulator of myeloma pathogenesis.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado , Proteínas Hedgehog , Antígenos CD , Moléculas de Adhesión Celular Neuronal , Receptores ErbB/genética , Proteínas Fetales , Humanos , Fosfatidilinositol 3-Quinasas , Transducción de Señal
10.
Int J Nanomedicine ; 16: 2833-2847, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33883894

RESUMEN

INTRODUCTION: Peptides can be rationally designed as non-covalent inhibitors for molecularly targeted therapy. However, it remains challenging to efficiently deliver the peptides into the targeted cells, which often severely affects their therapeutic efficiency. METHODS: Herein, we created a novel non-covalent peptide inhibitor against nuclear export factor CRM1 by a structure-guided drug design method and targetedly delivered the peptide into cancer cells by a nanoparticle-mediated gene expression system for use as a cancer therapy. RESULTS: The nuclear export signal (NES)-optimized CRM1 peptide inhibitor colocalized with CRM1 to the nuclear envelope and inhibited nuclear export in cancer cell lines in vitro. The crystal structures of the inhibitors complexed with CRM1 were solved. In contrast to the covalent inhibitors, the peptides were similarly effective against cells harboring the CRM1 C528S mutation. Moreover, a plasmid encoding the peptides was delivered by a iRGD-modified nanoparticle to efficiently target and transfect the cancer cells in vivo after intravenous administration. The peptides could be selectively expressed in the tumor, resulting in the efficient inhibition of subcutaneous melanoma xenografts without obvious systemic toxicity. DISCUSSION: This work provides an effective strategy to design peptide-based molecularly targeted therapeutics, which could lead to the development of future targeted therapy.


Asunto(s)
Espacio Intracelular/metabolismo , Carioferinas/antagonistas & inhibidores , Melanoma Experimental/tratamiento farmacológico , Nanopartículas/química , Péptidos/farmacología , Péptidos/uso terapéutico , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Células A549 , Transporte Activo de Núcleo Celular/efectos de los fármacos , Secuencia de Aminoácidos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Carioferinas/química , Carioferinas/metabolismo , Melanoma Experimental/patología , Proteínas Mutantes/metabolismo , Mutación/genética , Nanopartículas/ultraestructura , Señales de Exportación Nuclear , Péptidos/química , Unión Proteica/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas no Estructurales Virales/química , Proteína Exportina 1
11.
Sci Adv ; 6(23): eaba7406, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32537512

RESUMEN

Three-dimensional (3D) printing technology has great potential in advancing clinical medicine. Currently, the in vivo application strategies for 3D-printed macroscale products are limited to surgical implantation or in situ 3D printing at the exposed trauma, both requiring exposure of the application site. Here, we show a digital near-infrared (NIR) photopolymerization (DNP)-based 3D printing technology that enables the noninvasive in vivo 3D bioprinting of tissue constructs. In this technology, the NIR is modulated into customized pattern by a digital micromirror device, and dynamically projected for spatially inducing the polymerization of monomer solutions. By ex vivo irradiation with the patterned NIR, the subcutaneously injected bioink can be noninvasively printed into customized tissue constructs in situ. Without surgery implantation, a personalized ear-like tissue constructs with chondrification and a muscle tissue repairable cell-laden conformal scaffold were obtained in vivo. This work provides a proof of concept of noninvasive in vivo 3D bioprinting.

14.
Chem Commun (Camb) ; 55(87): 13128-13131, 2019 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-31616871

RESUMEN

We report here a novel light-triggered nanosystem based on co-assembling nanoaggregates (NAs) of lipophilic photosensitizers and lipophilic prodrugs containing multiple thioethers. Upon laser irradiation, the oxidization of the multiple thioethers by photosensitizer-generated singlet oxygen could rapidly destroy the NA structure, resulting in faster drug release than those containing a single thioether.


Asunto(s)
Luz , Nanopartículas/química , Fármacos Fotosensibilizantes/química , Profármacos/química , Sulfuros/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Humanos , Estructura Molecular , Tamaño de la Partícula , Fármacos Fotosensibilizantes/farmacología , Profármacos/farmacología , Oxígeno Singlete/química , Sulfuros/farmacología , Propiedades de Superficie
15.
ACS Appl Mater Interfaces ; 11(40): 36371-36382, 2019 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-31490057

RESUMEN

Combination of chemotherapeutics and immunomodulators can generate synergistic anticancer efficacy, exerting efficient chemoimmunotherapy for cancer treatment. Nanoparticulate delivery systems hold great promise to promote synergistic anticancer efficacy for the codelivery of drugs. However, there remain challenges to precisely coencapsulate and deliver combinational drugs at designed ratios due to the difference of compatibility between drugs and nanocarriers. In this study, coassembled nanoparticles of lipophilic prodrugs (LPs) were designed to codeliver chemotherapeutics and immunomodulators for cancer treatment. Such nanoassemblies (NAs) could act as platforms to ratiometrically coencapsulate chemotherapeutics and immunomodulators. Based on this method, NAs formed by the self-assembly of iRGD peptide derivatives, paclitaxel (PTX) LPs, and imiquimod (R837) LPs were demonstrated to target the tumor at unified pharmacokinetics, further inducing the effective tumor inhibition and tumor recurrence prevention. This work provided an alternative to prepare chemoimmunotherapeutic NAs with advantages of ratiometric drug coencapsulation and unified pharmacokinetics, which may advance the future cancer chemoimmunotherapy.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Quimioterapia , Inmunoterapia , Nanopartículas/química , Neoplasias/terapia , Animales , Presentación de Antígeno , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Liberación de Fármacos , Femenino , Imiquimod/administración & dosificación , Imiquimod/farmacocinética , Imiquimod/uso terapéutico , Ratones Endogámicos BALB C , Nanopartículas/ultraestructura , Recurrencia Local de Neoplasia/prevención & control , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Ratas Sprague-Dawley , Distribución Tisular
16.
J Mater Chem B ; 7(36): 5563-5572, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31465067

RESUMEN

Nanoassemblies (NAs) of small-molecule lipophilic prodrugs have been widely investigated for efficient drug delivery in cancer therapy, but their kinetic stability has not attracted sufficient attention in the past studies. Herein, we reported that kinetic stability has a great influence on the drug release from the NAs of lipophilic prodrugs in physiologically relevant media. Based on the co-assembled FRET nanosystems of two lipophilic fluorescent prodrugs, we demonstrated that NAs constructed by lipophilic prodrugs containing shorter alkyl chains or those with higher unsaturated degrees displayed poorer kinetic stability, which further resulted in remarkably faster drug release in mouse plasma and various tissue homogenates. More importantly, these kinetically unstable NAs also induced rapid intracellular drug release, resulting in much more potent cytotoxicity. These findings highlight the crucial role of kinetic stability in determining the drug release from the NAs of lipophilic prodrugs, which would effectively guide their rational designs for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Nanopartículas/química , Profármacos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Transferencia Resonante de Energía de Fluorescencia , Humanos , Cinética , Estructura Molecular , Tamaño de la Partícula , Profármacos/síntesis química , Profármacos/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Propiedades de Superficie , Células Tumorales Cultivadas
17.
J Biomed Nanotechnol ; 15(9): 1923-1936, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31387679

RESUMEN

Injectable hydrogels possess huge potential as localized drug carriers in breast cancer chemotherapy, owing to several advantages, including easy target administration, enhanced therapeutic efficiency, and less systemic side effects. Herein, we describe an injectable, near-infrared (NIR) light-responsive click cross-linked azobenzene hydrogel (AzoGel) that displays NIR irradiation-mediated smart drug release. The hydrogel can be formed in situ via click cross-linking by mixing two kinds of gelatin derivatives functioned with dibenzylcyclooctyne (DBCO) and azidated azobenzene (N3-Azo) respectively. The polyacrylic acid (PAA)-coated upconversion nanoparticles (UCNP@PAA)-encapsulated AzoGel has NIR light-responsive characteristics owing to the photoisomerization of azobenzene in the networks. The amount of an anticancer drug doxorubicin (DOX), released from the hydrogel can be efficiently controlled by tuning the exposure time and intensity of 980 nm NIR light. Results of the in vivo study using DOX and UCNP@PAA-loaded AzoGel controlled by NIR light in the 4T1 breast cancer xenograft mouse model demonstrated an enhanced anti-cancer effect. To conclude, the injectable, NIR light-responsive, click cross-linked AzoGel exhibits a high potential as a localized drug delivery platform for cancer therapy.


Asunto(s)
Neoplasias de la Mama , Animales , Compuestos Azo , Doxorrubicina , Humanos , Hidrogeles , Rayos Infrarrojos , Ratones
18.
Int J Nanomedicine ; 14: 3455-3468, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31190803

RESUMEN

Background: Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a novel tumor suppressor. However, whether LHPP is effective to melanoma has not been investigated. Gene therapy provides a new strategy for the treatment of melanoma. Currently, it suffers from the lack of safe and effective gene delivery systems. Methods: A CRGDKGPDC peptide (iRGD) modified hybrid monomethoxy poly(ethylene glycol)-poly(D,L-lactide) nanoparticle (iDPP) was prepared and complexed with a LHPP plasmid, forming an iDPP/LHPP nanocomplex. The iDPP/LHPP nanocomplex was characterized by particle size distribution, zeta potential, morphology, cytotoxicity, and transfection efficiency. The antitumor efficacy of the nanocomplex against melanoma was studied both in vitro and in vivo. Further, the potential epigenetic changes in melanoma induced by iDPP/LHPP nanocomplex were evaluated. Results: The iDPP/LHPP nanocomplex showed high transfection efficiency and low toxicity. Moreover, the nanocomplex displayed a neutral charge that can meet the requirement of intravenous injection for targeted gene therapy. In vitro and in vivo experiments indicated that the iDPP/LHPP nanocomplex significantly inhibited the melanoma growth without causing notable adverse effects. We also found that LHPP played an important role in epigenetics. It regulated the expression of genes related to the proliferation and apoptosis chiefly at the level of transcription. Conclusion: This work demonstrates that the iDPP nanoparticle-delivered LHPP gene has a potential application in melanoma therapy through regulation of the genes associated with epigenetics.


Asunto(s)
Pirofosfatasa Inorgánica/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Nanopartículas/química , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Epigénesis Genética/efectos de los fármacos , Humanos , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Nanopartículas/ultraestructura , Oligopéptidos/química , Especificidad de Órganos/efectos de los fármacos , Poliésteres/química , Polietilenglicoles/química
19.
J Biomed Nanotechnol ; 14(12): 2092-2101, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30305216

RESUMEN

Gene therapy is emerging as a promising tool for cancer treatment. Down-regulation of survivin gene can lead to the cancer inhibition. However, the lack of efficient and safe gene delivery system is still a critical obstacle to clinical gene therapy. In this study, we use a biodegradable nanoparticle to deliver human survivin-T34A (T34A) to dominant-negatively regulate survivin gene for ovarian cancer therapy. This nanoparticle, self-assembled from monomethoxy poly(ethylene glycol)-poly(D,L-lactide) (MPEG-PLA) copolymer and N-[1-(2,3-dioleoyloxy) propyl]-N,N,N-trimethylammonium chloride (DOTAP), has high transfection capability and negligible cytotoxicity. The nanoparticle-delivered T34A gene can efficiently inhibit the growth of SKOV3 ovarian cancer cells through induction of apoptosis in vitro. After intraperitoneal injection, the nanoparticle-delivered T34A gene significantly inhibited the growth of intraperitoneal metastasis of SKOV3 ovarian cancer, with no obvious adverse effects. Our data suggest that the nanoparticle-delivered T34A gene has promising clinical applications in ovarian cancer treatment.


Asunto(s)
Nanopartículas , Neoplasias Ováricas , Apoptosis , Línea Celular Tumoral , Femenino , Terapia Genética , Humanos , Proteínas Inhibidoras de la Apoptosis , Survivin , Transfección
20.
Adv Drug Deliv Rev ; 132: 235-251, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29935988

RESUMEN

3D bioprinting is emerging as a promising technology for fabricating complex tissue constructs with tailored biological components and mechanical properties. Recent advances have enabled scientists to precisely position materials and cells to build functional tissue models for in vitro drug screening and disease modeling. This review presents state-of-the-art 3D bioprinting techniques and discusses the choice of cell source and biomaterials for building functional tissue models that can be used for personalized drug screening and disease modeling. In particular, we focus on 3D-bioprinted liver models, cardiac tissues, vascularized constructs, and cancer models for their promising applications in medical research, drug discovery, toxicology, and other pre-clinical studies.


Asunto(s)
Bioimpresión , Enfermedades Cardiovasculares/patología , Evaluación Preclínica de Medicamentos , Modelos Biológicos , Medicina de Precisión , Impresión Tridimensional , Ingeniería de Tejidos , Materiales Biomiméticos/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA