Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell Immunol ; 365: 104376, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33984534

RESUMEN

Adoptive T cell transfer is one of the most promising ways to combat solid tumors. However, the weak infiltration of T cells into tumor sites has restricted their antitumor efficacy. To overcome this obstacle, we used the lipophilic protein painting strategy to improve tumor targeting and penetrating capacity of lymphocytes for the first time. We synthesized the lipid anchor consisting of a bispecific recombinant protein iRGD-antiEGFR and DSPE-PEG derivates, then successfully inserted it into the membranes of T cells. This surface modification was non-invasive and could efficiently improve the infiltration ability of T cells into multicellular spheroids and tumor masses. The surface modified T cells also displayed superior antitumor activities in EGFR-positive tumor xenografts via systematic infusion. Moreover, the permeability and antitumor efficacy of these surface painted T cells could be remarkably enhanced when used in combination with local low-dose irradiation.


Asunto(s)
Membrana Celular/metabolismo , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Anticuerpos de Dominio Único/metabolismo , Neoplasias Gástricas/terapia , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Ingeniería Genética , Humanos , Lípidos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfatidiletanolaminas , Polietilenglicoles , Receptores de Antígenos de Linfocitos T/genética , Anticuerpos de Dominio Único/genética , Neoplasias Gástricas/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Cell Biochem ; 121(2): 1260-1272, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31544991

RESUMEN

In the past decade, substantial evidence established that long noncoding RNAs are serious about mediating the evolution of malignancies. In previous studies, LINC00365, which has not been reported in colorectal cancer (CRC), was selected using the bioinformatics analysis in GSE109454 and GSE41655 data sets. However, the function and mechanism of LINC00365 are still obscure. In our study, LINC00365 was found upregulated in CRC specimens and intimately connected with the prognosis of patients with CRC. In addition, LINC00365 overexpression enhances the cell abilities of proliferation, migration, and invasion in vitro. Meanwhile, mechanistic studies showed that LINC00365 might involve in CRC cell progression by mediating the Wnt/ß-catenin pathway. Furthermore, LINC00365 upregulation increased CDK1 protein expression. In conclusion, this study suggests that LINC00365 acts as a vital part in facilitating CRC progression and might play as a therapeutic target for patients with CRC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Ratones Desnudos , Pronóstico , Células Tumorales Cultivadas , Proteína Wnt1/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
3.
J Cell Biochem ; 120(6): 9250-9263, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30525245

RESUMEN

Colorectal cancer (CRC) is one of the most frequently diagnosed digestive system cancer. The aim of the present study was to investigate the interactions among messenger RNAs (mRNAs), microRNAs (miRNAs), and long noncoding RNAs (lncRNAs) in CRC to reveal the mechanisms of CRC. Differentially expressed genes (DEGs) were identified from public gene expression data sets. One thousand eighty-one common dysregulated mRNAs in two data sets were identified. Gene function analysis and protein-protein interaction network analysis indicated that these DEGs might play important roles in CRC. LINC00365 was selected through coding- noncoding network analysis and its expression was validated upregulated in 22 paired clinical samples and four CRC cell lines. A competing endogenous RNA network composed of 70 miRNAs, nine mRNAs, and LINC00365 was constructed. Eight of nine mRNAs were validated upregulated in The Cancer Genome Atlas data set. Our results suggested that LINC00365 was an oncogene in CRC and it could regulate the expression of several mRNAs through sponging miRNAs.


Asunto(s)
Neoplasias Colorrectales/genética , Redes Reguladoras de Genes/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Neoplasias Colorrectales/patología , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Mapas de Interacción de Proteínas/genética , ARN Mensajero/genética
4.
Pharm Dev Technol ; 23(1): 33-40, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29069711

RESUMEN

Poor water solubility and side effects hampered the clinical application of gambogic acid (GA) in cancer therapy. Accordingly, GA-loaded polyethylene glycol-poly(ɛ-caprolactone) (PEG-PCL) nanoparticles (GA-NPs) were developed and administered peritumorally to evaluate their antitumor activity. The particle size, polydispersity index, encapsulation efficiency and loading capacity of GA-NPs were 143.78 ± 0.054 nm, 0.179 ± 0.004, 81.3 ± 2.5% and 14.8 ± 0.6%, respectively. In addition, GA-NPs showed excellent stability, good biocompatibility and sustained release profile. Endocytosis studies in vitro demonstrated that the GA-NPs were effectively taken up by tumor cells in a time-dependent manner. In vivo real-time imaging showed that the nanoparticles effectively accumulated within the tumor tissue after peritumoral administration. The cytotoxicity study revealed that the GA-NPs effectively inhibited the proliferation of gastric cancer cells. In vivo antitumor therapy with peritumoral injection of GA-NPs exhibited superior antitumor activity compared with free GA. Moreover, no toxicity was detected in any treatment group. Histological studies confirmed a lower cell density and a higher number of apoptotic cells in the GA-NPs group compared with the free GA group. Furthermore, the expression level of the cysteine proteases 3 precursor (pro-caspase3), a crucial component of cellular apoptotic pathways, was efficiently reduced in mice treated with GA-NPs. In conclusion, the GA-NPs system provided an efficient drug delivery platform for chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Caproatos/química , Lactonas/química , Nanopartículas/química , Polietilenglicoles/química , Neoplasias Gástricas/tratamiento farmacológico , Xantonas/farmacología , Animales , Antineoplásicos/química , Línea Celular , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Polímeros/química , Xantonas/química
5.
Zhongguo Zhong Yao Za Zhi ; 41(11): 2093-2097, 2016 Jun.
Artículo en Zh | MEDLINE | ID: mdl-28901106

RESUMEN

The objective of this study is to develop a new-type biodegradable, biocompatible curcumin-loaded nanoerythrosomes (Cur-RBC-NPs) by means of the sonication method. The size of Cur-RBC-NPs was optimized by varying drug loading parameters. The morphology, size distribution, stability, in vitro release pattern, cellular uptake of nanoparticles and in vitro anti-tumor effects were evaluated, respectively. The results showed the prepared Cur-RBC-NPs were nearly uniform spheres, with an average diameter of (245.7 ± 1.3) nm. Encapsulation efficiency (EE) and load efficiency (LE) of Cur-RBC-NPs were 50.65% ± 1.36% and 6.27% ± 0.29%. And the nanoparticles had a good sustained release property. According to the in vitro experiment, Cur-RBC-NPs were effectively taken in by tumor cells, and exhibited a significant anti-tumor effect. In conclusion, the method for preparing Cur-RBC-NPs is convenient, with a good sustained release behavior and anti-tumor efficacy, and so expected to be a new-type nano-drug delivery system in clinical practice.


Asunto(s)
Antineoplásicos/farmacología , Curcumina , Portadores de Fármacos , Nanopartículas , Línea Celular Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula
6.
Chin J Integr Med ; 28(10): 909-917, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34913148

RESUMEN

OBJECTIVE: To verrify the anti-tumor efficacy and toxicity between juglone (Jug) and Jug-loaded PLGA nanoparticles (Jug-PLGA-NPs). METHODS: Jug-PLGA-NPs were prepared by ultrasonic emulsification. The anti-tumor activity of Jug (2, 3, 4 µg/mL) and Jug-PLGA-NPs (Jug: 2, 3, 4 µg/mL) in vitro was measured by MTT assay and cell apoptosis analysis. The distribution, anti-tumor effect and biological safety in vivo was evaluated on A375 nude mice. RESULTS: With the advantage of good penetration and targeting properties, Jug-PLGA-NPs significantly inhibited proliferation and migration of melanoma cells both in vitro and in vivo (P<0.05 or P<0.01) with acceptable biocompatibility. CONCLUSIONS: Jug can inhibit the growth of melanoma but is highly toxic. With the advantage of sustained release, tumor targeting, anti-tumor activity and acceptable biological safety, Jug-PLGA-NPs provide a new pharmaceutical form for future application of Jug.


Asunto(s)
Melanoma , Nanopartículas , Animales , Línea Celular Tumoral , Preparaciones de Acción Retardada/uso terapéutico , Portadores de Fármacos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Desnudos , Naftoquinonas , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico
7.
Front Oncol ; 12: 800484, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35211402

RESUMEN

Some of the mutant peptides produced by gene mutation transcription and translation have the ability to induce specific T cells, which are called new antigens. Neoantigen-based peptide, DNA, RNA, and dendritic cell vaccines have been used in the clinic. In this paper, we describe a lung metastasis of a phyllodes tumor patient demonstrating pathological complete response following treatment containing personalized multi-epitope peptide neoantigen nano-vaccine. Based on whole-exome sequencing (WES), RNA sequencing, and new antigen prediction, several mutated peptide fragments were predicted to bind to the patient's human leukocyte antigen (HLA) allotypes, including ten peptides with high predicted binding affinity for six genes. The pulmonary metastases remained stable after the four cycles of anti-PD1 and anlotinib. After the addition of the multi-epitope peptide neoantigen nano-vaccine, the tumor began to collapse and contracture developed, accompanied by a decrease of tumor markers to normal, and complete pathological remission was achieved. With the use of the vaccination, recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) was used every time, and low-dose cyclophosphamide was injected every 3 weeks to improve efficacy. Peripheral blood immune monitoring demonstrated immune reactivity against a series of peptides, with the most robust post-vaccine T-cell response detected against the HLA-DRB1*0901-restricted SLC44A5 V54F peptide.

8.
Adv Sci (Weinh) ; : e2203298, 2022 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-36351249

RESUMEN

Personalized neoantigen vaccines have shown strong immunogenicity in clinical trial, but still face various challenges in facilitating an efficient antitumor immune response. Here, a personalized neoantigen nanovaccine (PNVAC) platform for adjuvant cancer immunotherapy is generated. PNVAC triggers superior protective efficacy against tumor recurrence and promotes longer survival than free neoantigens, especially when combined with anti-PD-1 treatment in a murine tumor model. A phase I clinical trial (ChiCTR1800017319) is initiated to evaluate the safety, immunogenicity, and prophylactic effect of PNVAC on preventing tumor recurrence in patients with high-risk gastric/gastroesophageal junction cancer after adjuvant chemotherapy of postsurgical resection. The one- and two-year disease-free survival rates are significantly higher than historical record. PNVAC induces both CD4+ and CD8+ T cell responses as well as antigen-experienced memory T cell phenotype. Furthermore, the immune response is persistent and remains evident one year after the vaccination. This work provides a safe and feasible strategy for developing neoantigen vaccines to delay gastric cancer recurrence after surgery.

9.
Biomater Sci ; 9(19): 6597-6608, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34582523

RESUMEN

Immunotherapy has emerged as one of the most promising treatments for cancer in recent years. However, it works only for a small proportion of patients, which can in part be attributed to the immunosuppressive tumor microenvironment (TME). Tumor associated macrophages (TAMs) are the critical components of tumors and play an important role in the development of the immunosuppressive TME. The transition of TAMs from the pro-tumor (M2) phenotype to anti-tumor (M1) phenotype is crucial for the immunotherapy of gastric cancer. Herein, we developed a shear-thinning, injectable hydrogel co-loaded with polyphyllin II (PP2) and resiquimod (R848) (PR-Gel) for potentiating localized immunotherapy of gastric cancer through the repolarization of TAMs. In this work, we evaluate the effects of PR-Gel on TAM repolarization and explored its therapeutic effect for localized immunotherapy. The hydrogels were synthesized through the Schiff base reactions between aldehyde-functionalized polyethylene glycol and the amino group of polylysine. A M2-to-M1 repolarization of TAMs and increased production of TNF-α and IL-6 were observed after treatment with PR-Gel in vitro. The anti-tumor efficacy of PR-Gel in a subcutaneous xenograft model of gastric cancer showed that the hydrogels possess good tumor growth suppression properties after a single injection. Furthermore, an increased iNOS/CD206 ratio in TAMs and enhanced CD8+ T cell infiltration were also observed within the TME after the treatment with PR-Gel. Hence, the biocompatible, shear-thinning, injectable hydrogels are a promising noninvasive drug-delivery platform for the regulation of the immunosuppressive TME and have great potential in localized immunotherapy against gastric cancer.


Asunto(s)
Polilisina , Neoplasias Gástricas , Humanos , Hidrogeles , Inmunoterapia , Neoplasias Gástricas/terapia , Microambiente Tumoral , Macrófagos Asociados a Tumores
10.
Oncol Rep ; 45(1): 404-405, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33416136

RESUMEN

Following the publication of the above article, the authors noticed that data shown in certain of the panels in Figs. 4 and 5 were selected incorrectly and presented wrongly in these figures. Essentially, in Fig. 4, the data shown for the Tunel, anti­EGFR­iRGD and Tunel, anti­EGFR­iGRD+IR data panels (i.e., the panels in the third row, columns 2 and 4), were chosen incorrectly, and in Fig. 5, the data panel for the Lung, IR experiment (fourth row, third column) was selected incorrectly. The revised versions of Figs. 4 and 5, featuring all the correct data panels, are shown on the next page. Furthermore, the results were re­analyzed based on the correct data. The errors made in the compilation of these Figures did not affect the overall conclusions reported in the paper. The authors are grateful to the Editor of Oncology Reports for allowing them the opportunity to publish this Corrigendum, and apologize to the readership for any inconvenience caused. [the original article was published in Oncology Reports 40: 1583-1591, 2018; DOI: 10.3892/or.2018.6532].

11.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33986122

RESUMEN

BACKGROUND: Poor infiltration and limited activation of transferred T cells are fundamental factors impeding the development of adoptive cell immunotherapy in solid tumors. A tumor-penetrating peptide iRGD has been widely used to deliver drugs deep into tumor tissues. CD3-targeting bispecific antibodies represent a promising immunotherapy which recruits and activates T cells. METHODS: T-cell penetration was demonstrated in tumor spheroids using confocal microscope, and in xenografted tumors by histology and in vivo real-time fluorescence imaging. Activation and cytotoxicity of T cells were assessed by flow cytometry and confocal microscope. Bioluminescence imaging was used to evaluate in vivo antitumor effects, and transmission electron microscopy was used for mechanistic studies. RESULTS: We generated a novel bifunctional agent iRGD-anti-CD3 which could immobilize iRGD on the surface of T cells through CD3 engaging. We found that iRGD-anti-CD3 modification not only facilitated T-cell infiltration in 3D tumor spheroids and xenografted tumor nodules but also induced T-cell activation and cytotoxicity against target cancer cells. T cells modified with iRGD-anti-CD3 significantly inhibited tumor growth and prolonged survival in several xenograft mouse models, which was further enhanced by the combination of programmed cell death protein 1 (PD-1) blockade. Mechanistic studies revealed that iRGD-anti-CD3 initiated a transport pathway called vesiculovacuolar organelles in the endothelial cytoplasm to promote T-cell extravasation. CONCLUSION: Altogether, we show that iRGD-anti-CD3 modification is an innovative and bifunctional strategy to overcome major bottlenecks in adoptive cell therapy. Moreover, we demonstrate that combination with PD-1 blockade can further improve antitumor efficacy of iRGD-anti-CD3-modified T cells.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Complejo CD3/antagonistas & inhibidores , Movimiento Celular/efectos de los fármacos , Inmunoterapia Adoptiva , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/trasplante , Oligopéptidos/farmacología , Neoplasias Gástricas/terapia , Linfocitos T/efectos de los fármacos , Linfocitos T/trasplante , Animales , Complejo CD3/inmunología , Complejo CD3/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Citotoxicidad Inmunológica/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Esferoides Celulares , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Langmuir ; 26(8): 5428-34, 2010 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-20095619

RESUMEN

Multifunctional nanocarriers based on chitosan/gold nanorod (CS-AuNR) hybrid nanospheres have been successfully fabricated by a simple nonsolvent-aided counterion complexation method. Anticancer drug cisplatin was subsequently loaded into the obtained hybrid nanospheres, utilizing the loading space provided by the chitosan spherical matrix. In vitro cell experiments demonstrated that the CS-AuNR hybrid nanospheres can not only be utilized as contrast agents for real-time cell imaging but also serve as a near-infrared (NIR) thermotherapy nanodevice to achieve irradiation-induced cancer cell death owing to the unique optical properties endowed by the encapsulated gold nanorods. In addition, an effective attack on the cancer cells by the loaded anticancer drug cisplatin has also been observed, rendering the obtained nanocarriers an all-in-one system possessing drug delivery, cell imaging, and photothermal therapy functionalities.


Asunto(s)
Quitosano/química , Nanosferas/química , Nanotubos/química , Fotoquímica/métodos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/química , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/terapia , Humanos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Modelos Teóricos , Nanosferas/uso terapéutico
13.
Int J Nanomedicine ; 15: 3669-3680, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32547021

RESUMEN

INTRODUCTION: The exhaustion and poor homing of activated lymphocytes are critical obstacles in adoptive cell immunotherapy for solid tumors. In order to effectively deliver immune cells into tumors, we encapsulated interferon-α2b (IFN-α2b) into macroporous hydrogels as an enhancement factor and utilized low-dose irradiation (LDI) as a tumoral attractor of T cells. METHODS: Hydroxypropyl cellulose hydrogels were prepared by irradiation techniques, and the cross-sectional microstructure was characterized by scanning electron microscopy. The synergistic antitumor mechanism of combination of IFN-α2b and CIK cells was evaluated by detecting the expression of activation marker CD69 on CIK cell surface and IFN-γ production by CIK cells. The in vivo antitumor activity of IFN-α2b-incorporated hydroxypropyl cellulose hydrogels combined with CIK and radiation was evaluated in an MKN-45 xenografted nude mice model. RESULTS: The bioactivity of IFN-α2b was well maintained in ultraviolet-reactive, rapidly cross-linkable hydroxypropyl cellulose hydrogels. In vitro studies demonstrated IFN-α2b-activated T cells, as evidenced by upregulating early activation marker CD69 and secretion inflammatory cytokine IFN-γ. In vivo real-time image showed our hydrogels kept a higher amount of drug delivery at the tumor site for a long time compared with free drug injection. Low-dose irradiation promoted T cell accumulation and infiltration in subcutaneous tumors. Combination of IFN-α2b-loaded hydrogels (Gel-IFN) with T cells and LDI exhibited higher efficacy to eradicate human gastric cancer xenograted tumors with less proliferating cells and more necrotic regions compared with IFN-α2b or T cells alone. DISCUSSION: HPC hydrogels kept the activity of IFN-α2b and stably release of IFN-α2b to stimulate T cells for a long time. At the same time, low-dose radiation recruits T cells into tumors. This innovative integration mode of IFN-α2b-loaded hydrogels and radiotherapy offers a potent strategy to improve the therapeutic outcome of T cell therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Reactivos de Enlaces Cruzados/química , Hidrogeles/química , Interferón-alfa/uso terapéutico , Luz , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/radioterapia , Linfocitos T/inmunología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Celulosa/análogos & derivados , Celulosa/química , Relación Dosis-Respuesta en la Radiación , Electrones , Humanos , Interferón alfa-2 , Interferón-alfa/farmacología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Linfocitos T/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Biomater Appl ; 35(1): 49-58, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32223499

RESUMEN

Here, we describe a novel microfilter device to capture circulating tumor cells in an efficient and low-cost manner. Then, we validated the safety and clinical utility of the novel microfilter device. We next performed mutation analysis from circulating tumor cells collected from lung cancer patients using this new device. Our results indicate that this microfilter system can be used to investigate the genome landscape of circulating tumor cells collected from lung cancer patients. Further, our results highlight a proof-of-concept demonstration indicating that circulating tumor cell can be used for mutation profiling during tumor evolution, therapy prediction, and monitoring, with immediate clinical applicability.


Asunto(s)
Separación Celular/instrumentación , Neoplasias Pulmonares/patología , Células Neoplásicas Circulantes/patología , Línea Celular Tumoral , Diseño de Equipo , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/genética , Filtros Microporos , Mutación , Células Neoplásicas Circulantes/metabolismo
15.
Int J Nanomedicine ; 15: 735-747, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32099362

RESUMEN

INTRODUCTION: Gambogic acid (GA) is proved to have anti-tumor effects on gastric cancer. Due to poor solubility, non-specific biological distribution, toxicity to normal tissues and short half-life, it is hard to be applied into the clinic. To overcome these issues, we developed a thermosensitive and injectable hydrogel composed of hydroxypropyl cellulose, silk fibroin and glycerol, with short gelling time, good compatibility and sustained release, and demonstrated that the hydrogel packaged with gambogic acid nanoparticles (GA-NPs) and tumor-penetrating peptide iRGD could improve the anti-tumor activity. METHODS: The Gelling time and micropore size of the hydrogels were regulated through different concentrations of glycerol. Controlled release characteristics of the hydrogels were evaluated with a real-time near-infrared fluorescence imaging system. Location of nanoparticles from different carriers was traced by confocal laser scanning microscopy. The in vivo antitumor activity of the hydrogels packaging GA-NPs and iRGD was evaluated by investigating tumor volume and tumor size. RESULTS: The thermo-sensitive properties of hydrogels were characterized by 3-4 min, 37°C, when glycerol concentration was 20%. The hydrogels physically packaged with GA-NPs and iRGD showed higher fluorescence intensity than other groups. The in vivo study indicated that the co-administration of GA-NPs and iRGD by hydrogels had higher antitumor activity than the GA-loaded hydrogels and free GA combining with iRGD. Free GA group showed few antitumor effects. Compared with the control group, the body weight in other groups had no obvious change, and the count of leukocytes and hemoglobin was slightly decreased. DISCUSSION: The hydrogel constructed iRGD and GA-NPs exerted an effective anti-tumor effect possibly due to retention effect, local administration and continuous sustained release of iRGD promoting the penetration of nanoparticles into a deep part of tumors. The delivery system showed little systemic toxicity and would provide a promising strategy to improve anti-gastric cancer efficacy.


Asunto(s)
Antineoplásicos/uso terapéutico , Hidrogeles/química , Nanopartículas/química , Oligopéptidos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Temperatura , Xantonas/uso terapéutico , Animales , Antineoplásicos/farmacología , Bombyx , Línea Celular Tumoral , Fibroínas/química , Glicerol/química , Humanos , Derivados de la Hipromelosa/química , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/ultraestructura , Porosidad , Neoplasias Gástricas/patología , Distribución Tisular , Xantonas/farmacología
16.
ACS Biomater Sci Eng ; 5(2): 1100-1112, 2019 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-33405800

RESUMEN

The local delivery of therapeutics in a long-term sustained manner at tumor sites is attractive for the therapy of gastric cancer with peritoneal metastasis. In this manuscript, an injectable hydrogel-encapsulating paclitaxel-loaded red blood cell membrane nanoparticles (PRNP-gel) is designed on the basis of temperature-induced phase transition of polyethylene-glycol-modified bovine serum albumin (PEG-BSA). Dynamic light scattering, ζ potential, and electron microscopy were utilized to characterize the nanoparticle-hydrogel hybrid system. It was found that the PRNP had a spherical morphology with a diameter of about 133 nm and negative surface potential. The drug loading efficiency and loading content are 85% and 22%, respectively. In situ gelation occurred within 12 min when the gel precursor was incubated at 37 °C or injected subcutaneously. The in-situ-forming hydrogel showed a sustained release profile, and the cumulative release of PTX was ∼30% after 6 days. The PRNP-gel exhibited high cytocompatibility and biodegradability in vitro and in vivo. This nanoparticle-hydrogel hybrid system is applied as a drug carrier for local chemotherapy to enhance therapeutic levels at tumor site and reduce the systemic toxicity. In vivo antitumor evaluation within a subcutaneous xenograft and peritoneal dissemination model showed that the hydrogel possesses good tumor growth suppression properties after a single injection. Hence, the as-prepared injectable hydrogel system could be a promising candidate for the local delivery of chemotherapeutic drugs.

17.
Pathol Res Pract ; 215(4): 755-760, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30718098

RESUMEN

Gastric cancer, one of the most common disease, has become a major public health problem worldwide. Cisplatin (DDP) has been a widely used drug for the treatment of cancer, also usually applied in gastric cancer in clinic. However, the side effects including toxicity and drug-resistance restricted the usage of DDP in clinic, so we prepared a DDP-complexed hydrogel (DDP-Gel) and investigated its efficacy in gastric cancer. For in vivo studies, MKN45-Luc cells were injected into BLAB/C node mice subcutaneously to establish gastric cancer with orthotopically grown tumors. Mice bearing tumors were treated with normal saline, DDP and DDP-Gel. Body weight and survival condition were observed and recorded. The treatment efficacy in vivo was detected by luciferase imaging and histological evaluation was performed by H&E staining of different organs. Additionally, normal ICR mice were treated with different doses of DDP/DDP-Gel to calculate their LD50 in vivo. The results showed that DDP-Gel prolonged survival time and ameliorated body weight changes of mice bearing tumors. DDP-Gel exhibited higher efficacy to inhibit tumor growth and metastasis, compared to DDP. Besides, LD50 of DDP-Gel was 166.0 mg/kg, 13.2 folds higher than DDP. As a conclusion, DDP-Gel showed a more effective and safer function than DDP in gastric cancer, which indicating that DDP-Gel might be a novel strategy for gastric cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Hidrogeles/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Hidrogeles/administración & dosificación , Ratones , Ratones Endogámicos ICR , Neoplasias Gástricas/patología , Resultado del Tratamiento
18.
J Biomater Appl ; 33(10): 1394-1406, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952195

RESUMEN

Docetaxel (TXT) is acknowledged as one of the most important chemotherapy agents for gastric cancer (GC). PI3K/AKT signaling is frequently activated in GC, and its inhibitor LY294002 exerts potent antitumor effects. However, the hydrophobicity of TXT and the poor solubility and low bioavailability of LY294002 limit their clinical application. To overcome these shortcomings, we developed poly(lactic acid/glycolic) (PLGA) nanoparticles loaded with TXT and LY294002. PLGA facilitated the accumulation of TXT and LY294002 at the tumor sites. The in vitro functional results showed that PLGA(TXT+LY294002) exhibited controlled-release and resulted in a markedly reduced proliferative capacity and an elevated apoptosis rate. An in vivo orthotopic GC mouse model and xenograft mouse model confirmed the anticancer superiority and tumor-targeting feature of PLGA(TXT+LY294002). Histological analysis indicated that PLGA(TXT+LY294002) was biocompatible and had no toxicity to major organs. Characterized by the combined slow release of TXT and LY294002, this novel PLGA-based TXT/LY294002 drug delivery system provides controlled release and tumor targeting and is safe, shedding light on the future of targeted therapy against GC.


Asunto(s)
Antineoplásicos/administración & dosificación , Cromonas/administración & dosificación , Docetaxel/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Morfolinas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cromonas/farmacocinética , Cromonas/uso terapéutico , Preparaciones de Acción Retardada/química , Docetaxel/farmacocinética , Docetaxel/uso terapéutico , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Morfolinas/farmacocinética , Morfolinas/uso terapéutico , Nanopartículas/química , Neoplasias Gástricas/patología
19.
Nanoscale Res Lett ; 14(1): 395, 2019 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-31889247

RESUMEN

In this study, the tumor-targeted MRI contrast agent was prepared with gelatinase-stimuli nanoparticles (NPs) and Omniscan (Omn) by double emulsion method. The size, distribution, morphology, stability, drug loading, and encapsulation efficiency of Omn-NPs were characterized. The macroscopic and microscopic morphological changes of NPs in response to gelatinases (collagenases IV) were observed. The MR imaging using Omn-NPs as a contrast agent was evaluated in the oral squamous cell carcinoma models with Omn as a control. We found clear evidence that the Omn-NPs were transformed by gelatinases and the signal of T1-weighted MRI sequence showed that the tumor-to-background ratio was significantly higher in Omn-NPs than in Omn. The peak point of time after injection was much later for Omn-NPs than Omn. This study demonstrates that Omn-NPs hold great promise as MRI contrast agent with improved specificity and prolonged circulation time based on a relatively simple and universal strategy.

20.
Cancer Biol Ther ; 20(6): 886-896, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30794031

RESUMEN

Long noncoding RNAs are widely acknowledged as a group of regulatory factors in various diseases, especially in cancers. KCNQ1 overlapping transcript 1 (KCNQ1OT1) has been reported as oncogene in human cancers. However, the role of KCNQ1OT1 in colorectal cancer (CRC) has not been fully explained. Based on the database analysis, KCNQ1OT1 was highly expressed in CRC samples and predicted the poor prognosis for CRC patients. Functional experiments revealed that KCNQ1OT1 knockdown negatively affected the proliferation, migration and epithelial-mesenchymal transition (EMT) in CRC cells. Moreover, we identified the cytoplasmic localization of KCNQ1OT1 in CRC cells, indicating the post-transcriptional regulation of KCNQ1OT1 on gene expression. Mechanism experiments including RNA Immunoprecipitation (RIP) assay and dual luciferase reporter assays verified that KCNQ1OT1 acted as a competing endogenous RNA (ceRNA) in CRC by sponging microRNA-217 (miR-217) to up-regulate the expression of zinc finger E-box binding homeobox 1 (ZEB1). Further mechanism investigation revealed that ZEB1 enhanced the transcription activity of KCNQ1OT1 by acting as a transcription activator. Finally, rescue assays were designed to demonstrate the effect of KCNQ1OT1-miR-217-ZEB1 feedback loop on proliferation, migration, and EMT of CRC cells. In brief, our research findings revealed that ZEB1-induced upregulation of KCNQ1OT1 improved the proliferation, migration and EMT formation of CRC cells via regulation of miR-217/ZEB1 axis.


Asunto(s)
Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Noqueados , Canales de Potasio con Entrada de Voltaje/genética , Pronóstico , Interferencia de ARN , Transcripción Genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda