Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Nat Med ; 77(4): 817-828, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37354258

RESUMEN

Prostate cancer is one of the most prevalent lethal diseases among men globally. In the treatment of prostate cancer, the limited therapeutic efficacy of the standard non-hormonal systemic therapy docetaxel (DTX) represents an important challenge. Cancer-associated fibroblasts (CAFs) play a crucial role in resistance to therapy because of their prevalence and functional pleiotropy in tumor environments. Our previous research revealed that MPSSS, a novel polysaccharide extracted from Lentinus edodes, could significantly attenuate the immunosuppressive function of myeloid suppressor cells and CAFs. In this study, we investigated whether MPSSS could potentiate the efficacy of DTX against prostate cancer by inhibiting CAF-induced chemoresistance and elucidated its underlying mechanisms. The sensitivity of PC-3 prostate cancer cells cultured with conditioned medium derived from CAFs (CAF-CM) to DTX was assessed. The resistance effect induced by CAF-CM was abolished when CAFs were pretreated with MPSSS. Bioinformatic analysis of datasets from the Gene Expression Omnibus database revealed the activation of the transforming growth factor ß1 (TGF-ß1) signaling pathway in DTX-resistant cells. Based on this finding, we demonstrated that treatment with the TGF-ß1 receptor inhibitor SB525334 reversed DTX resistance in CAFs, suggesting that TGF-ß1 secreted by CAFs was a crucial intermediary in the development of DTX resistance in PC3 cells. Further research revealed that MPSSS decreases the secretion of TGF-ß1 by inhibiting the JAK2/STAT3 pathway via Toll-like receptor 4 in CAFs. Overall, MPSSS might be a potential adjuvant treatment for DTX resistance in prostate cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias de la Próstata , Hongos Shiitake , Masculino , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Factor de Crecimiento Transformador beta1/metabolismo , Docetaxel/farmacología , Docetaxel/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Fibroblastos , Línea Celular Tumoral , Polisacáridos/farmacología , Polisacáridos/metabolismo
2.
Chem Biol Drug Des ; 102(3): 444-456, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36509697

RESUMEN

The natural products plinabulin, docetaxel, and vinblastine are microtubule targeting agents (MTAs). They have been used alone or in combination in cancer treatment. However, the exact nature of their effects on microtubule (MT) polymerization dynamics is poorly understood. To elucidate the longitudinal conformational and energetic changes during MT dynamics, a total of 140 ns molecular dynamic simulations combined with binding free energy calculations were performed on seven tubulin models. The results indicated that the drugs disrupted MT polymerization by altering both MT conformation and binding free energy of the neighboring tubulin subunits. The combination of plinabulin and docetaxel destabilized MT polymerization due to bending MT and weakening the polarity of tubulin polymerization. The new combination of docetaxel and vinblastine synergistically enhanced MT depolymerization and bending, while plinabulin and vinblastine had no synergistic inhibitory effects. The results were verified by the tubulin assembly assay. Our study obtained a comprehensive understanding of the action mechanisms of three natural drugs and their combinations on MT dynamic, provided theoretical guidance for new MTA combinations, and would promote the optimal use of MTA and contribute to developing new MTAs as anticancer agents.


Asunto(s)
Antineoplásicos , Moduladores de Tubulina , Tubulina (Proteína) , Antineoplásicos/farmacología , Docetaxel/farmacología , Docetaxel/metabolismo , Microtúbulos , Tubulina (Proteína)/metabolismo , Vinblastina/farmacología , Vinblastina/análisis , Vinblastina/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
3.
J Obstet Gynaecol ; 42(8): 3672-3678, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36484524

RESUMEN

Taxanes are important chemotherapeutic agents used to manage breast cancer and gynaecological malignancies. However, ovarian toxicity induced by the taxane docetaxel (DOC) is of great concern. We investigated DOC-induced toxicity in the ovaries of female CD1 strain mice. The mice were divided into control (saline), DOC-5 (5 mg/kg DOC), and DOC-10 (10 mg/kg DOC) groups and administered saline or DOC on the first day of the study and two weeks later. Two weeks after the second dose, the ovaries were removed for analysis after inducing superovulation. Ovary weight, the number of secondary follicles, and the total number of follicles were reduced after DOC administration. Additionally, the expression levels of caspase-3 and the pro-apoptotic protein Bcl-2 interacting mediator of cell death (BIM) increased. Our findings suggest that high-dose DOC induces damage to growing follicles; however, it may not affect primordial follicles.Impact statementWhat is already known on this subject? Docetaxel (DOC) is one of the most effective chemotherapeutic agents used to manage various cancers. Some in-vitro studies have examined paclitaxel-induced ovarian toxicity; however, limited research on DOC is available.What do the results of this study add? We investigated DOC-induced ovarian toxicity in female CD1 strain mice at 5 mg/kg and 10 mg/kg. We found that DOC reduced ovary weight, the number of secondary follicles, and the total number of follicles, with the higher dose having a higher effect.What are the implications of these findings for clinical practice and/or further research? We believe that our study makes a significant contribution to the knowledge about the effect of DOC on ovarian function.


Asunto(s)
Docetaxel , Folículo Ovárico , Ovario , Animales , Femenino , Ratones , Docetaxel/metabolismo , Docetaxel/farmacología , Folículo Ovárico/efectos de los fármacos , Ovario/efectos de los fármacos , Inyecciones Intraperitoneales
4.
Microb Pathog ; 169: 105665, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35781005

RESUMEN

Urinary tract infections (UTIs) are a serious health concern worldwide. Treatment of UTIs is becoming a challenge as uropathogenic Escherichia coli (UPEC), which is the most common etiological agent, has developed resistance to the main classes of antibiotics. Small molecules that interfere with metabolic processes rather than growth are attractive alternatives to conventional antibiotics. Repurposing of already known drugs for treating infectious diseases could be an attractive avenue for finding novel therapeutics against infections caused by UPEC. Virtual screenings enable the rapid and economical identification of target ligands from large libraries of compounds, reducing the cost and time of traditional drug discovery. Moreover, the drugs that have been approved by the FDA have low cytotoxicity and good pharmacological characteristics. In this work, we targeted the HisC enzyme of the histidine biosynthetic pathway as enzymes of this pathway are absent in humans. We screened the library of FDA-approved drugs against HisC via molecular docking, and four hits (Docetaxel, Suramin, Digitoxin, and Nystatin) showing the highest binding energy were selected. These were further tested for antibacterial activity, which was observed only for Docetaxel (MIC value of 640 µg/ml); therefore, Docetaxel was further tested for its efficacy in vivo in murine catheter UTI model and antibiofilm activity using crystal violet staining and scanning electron microscopy. Docetaxel inhibited biofilm formation and reduced the bacterial load in urine, kidney, and bladder. Docking studies revealed that Docetaxel acts by blocking the binding site of HisC to the native substrate by competitive inhibition. Docetaxel may be a potential new inhibitor for UPEC with antibacterial and antibiofilm capability.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Docetaxel/metabolismo , Reposicionamiento de Medicamentos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Humanos , Ratones , Simulación del Acoplamiento Molecular , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/microbiología
5.
Mol Ther ; 30(3): 1171-1187, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35051615

RESUMEN

Therapy induced senescence (TIS) in tumors and TIS cancer cells secrete proinflammatory senescence-associated secretory phenotype (SASP) factors. SASP factors promote TIS cancer cells to re-enter the growth cycle with stemness characteristics, resulting in chemo-resistance and disease relapse. Herein, we show that the immunotherapeutic HCW9218, comprising transforming growth factor-ß (TGF-ß) receptor II and interleukin (IL)-15/IL-15 receptor α domains, enhances metabolic and cytotoxic activities of immune cells and reduces TIS tumor cells in vivo to improve the efficacy of docetaxel and gemcitabine plus nab-paclitaxel against B16F10 melanoma and SW1990 pancreatic tumors, respectively. Mechanistically, HCW9218 treatment reduces the immunosuppressive tumor microenvironment and enhances immune cell infiltration and cytotoxicity in the tumors to eliminate TIS cancer cells. Immuno-depletion analysis suggests that HCW9218-activated natural killer cells play a pivotal role in TIS cancer cell removal. HCW9218 treatment following docetaxel chemotherapy further enhances efficacy of tumor antigen-specific and anti-programmed death-ligand 1 (PD-L1) antibodies in B16F10 tumor-bearing mice. We also show that HCW9218 treatment decreases TIS cells and lowers SASP factors in off-target tissues caused by chemotherapy of tumor-bearing mice. Collectively, HCW9218 has the potential to significantly enhance anti-tumor efficacy of chemotherapy, therapeutic antibodies, and checkpoint blockade by eliminating TIS cancer cells while reducing TIS-mediated proinflammatory side effects in normal tissues.


Asunto(s)
Antígeno B7-H1 , Células Asesinas Naturales , Animales , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Senescencia Celular , Docetaxel/metabolismo , Docetaxel/farmacología , Inmunoterapia/métodos , Células Asesinas Naturales/metabolismo , Ratones , Microambiente Tumoral
6.
J Nanobiotechnology ; 19(1): 432, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930301

RESUMEN

BACKGROUND: Distant metastasis to vital organs is the major contributor to breast cancer mortality, and regional lymph node metastasis is an important facilitator of distant metastasis and recurrence in this cancer. The early diagnosis and precise treatment of lymph node metastasis are crucial for staging and prognosis in breast cancer. Herein, we report a visualized precision medicine nanoplatform of metastatic lymph nodes for ultrasonic/photoacoustic (US/PA) dual modal imaging-guided in situ targeted hyperthermia-combined chemotherapy. RESULTS: Carbon nanoparticles (CNs), approved by the China Food and Drug Administration, were loaded with docetaxel and rationally combined with anti-hypoxia-inducible factor 1α antibody-modified poly (lactic-co-glycolic acid) (PLGA) nanoparticles to achieve the combination of passive targeting at the lymph nodes and intracellular targeting at HIF 1α factor. The accumulation and retention of nanoparticles in metastatic lymph nodes via lymphatic delivery were enhanced. Docetaxel could be effectively offloaded by CNs that have active carbon nanoparticles, and the PLGA membrane prevented drug leakage. The nanoparticles exhibited excellent photothermal performance with a photothermal conversion efficiency of 28.9%, killing tumor cells in metastatic lymph nodes through hyperthermia. In vitro and in vivo systematic evaluations revealed that hyperpyrexia triggered the rupture of nanoparticles caused by the phase transition of perfluorohexane, resulting in docetaxel release for achieving in situ hyperthermia-combined chemotherapy. CONCLUSIONS: The laser-triggered highly efficient in situ chemotherapy nanosystem achieves targeted synergistic chemo-hyperthermia treatment of metastatic lymph nodes, and lymphatic delivery represents a strategy to avoid additional injury caused by drugs entering the blood circulation.


Asunto(s)
Antineoplásicos/uso terapéutico , Hipertermia Inducida/métodos , Ganglios Linfáticos/metabolismo , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos/química , Anticuerpos/inmunología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carbono/química , Línea Celular Tumoral , Docetaxel/química , Docetaxel/metabolismo , Docetaxel/farmacología , Docetaxel/uso terapéutico , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Rayos Infrarrojos , Metástasis Linfática , Nanomedicina , Nanopartículas/metabolismo , Neoplasias/patología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ratas , Trasplante Heterólogo
7.
J Nanobiotechnology ; 19(1): 446, 2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-34949198

RESUMEN

The integrin αvß3 receptor and Lactoferrin receptor (LfR) are over-expressed in both cerebral microvascular endothelial cells and glioma cells. RGD tripeptide and Lf can specifically bind with integrin αvß3 receptor and LfR, respectively. In our study, RGD and Lf dual-modified liposomes loaded with docetaxel (DTX) were designed to enhance the brain targeting effect and treatment of glioma. Our in vitro studies have shown that RGD-Lf-LP can significantly enhance the cellular uptake of U87 MG cells and human cerebral microvascular endothelial cells (hCMEC/D3) when compared to RGD modified liposomes (RGD-LP) and Lf modified liposomes (Lf-LP). Free RGD and Lf competitively reduced the cellular uptake of RGD-Lf-LP, in particular, free RGD played a main inhibitory effect on cellular uptake of RGD-Lf-LP in U87 MG cells, yet free Lf played a main inhibitory effect on cellular uptake of RGD-Lf-LP in hCMEC/D3 cells. RGD-Lf-LP can also significantly increase penetration of U87 MG tumor spheroids, and RGD modification plays a dominating role on promoting the penetration of U87 MG tumor spheroids. The results of in vitro BBB model were shown that RGD-Lf-LP-C6 obviously increased the transport of hCMEC/D3 cell monolayers, and Lf modification plays a dominating role on increasing the transport of hCMEC/D3 cell monolayers. In vivo imaging proved that RGD-Lf-LP shows stronger targeting effects for brain orthotopic gliomas than that of RGD-LP and Lf-LP. The result of tissue distribution confirmed that RGD-LF-LP-DTX could significantly increase brain targeting after intravenous injection. Furthermore, RGD-LF-LP-DTX (a dose of 5 mg kg-1 DTX) could significantly prolong the survival time of orthotopic glioma-bearing mice. In summary, RGD and LF dual modification are good combination for brain targeting delivery, RGD-Lf-LP-DTX could enhance brain targeting effects, and is thus a promising chemotherapeutic drug delivery system for treatment of glioma.


Asunto(s)
Antineoplásicos/farmacología , Docetaxel/química , Integrina alfaVbeta3/antagonistas & inhibidores , Liposomas/química , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Docetaxel/metabolismo , Docetaxel/farmacología , Docetaxel/uso terapéutico , Glioma/diagnóstico por imagen , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Integrina alfaVbeta3/metabolismo , Liposomas/farmacocinética , Ratones , Ratones Desnudos , Oligopéptidos/química , Tamaño de la Partícula , Receptores de Superficie Celular/metabolismo , Tasa de Supervivencia , Distribución Tisular
8.
Int J Mol Sci ; 22(18)2021 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-34576262

RESUMEN

Cannabidiol (CBD), a nonpsychoactive phytocannabinoid, has recently emerged as a potential cytotoxic agent in addition to its ameliorative activity in chemotherapy-associated side effects. In this work, the potential interactions of CBD with docetaxel (DOC), doxorubicin (DOX), paclitaxel (PTX), vinorelbine (VIN), and 7-ethyl-10-hydroxycamptothecin (SN-38) were explored in MCF7 breast adenocarcinoma cells using different synergy quantification models. The apoptotic profiles of MCF7 cells after the treatments were assessed via flow cytometry. The molecular mechanisms of CBD and the most promising combinations were investigated via label-free quantification proteomics. A strong synergy was observed across all synergy models at different molar ratios of CBD in combination with SN-38 and VIN. Intriguingly, synergy was observed for CBD with all chemotherapeutic drugs at a molar ratio of 636:1 in almost all synergy models. However, discording synergy trends warranted the validation of the selected combinations against different models. Enhanced apoptosis was observed for all synergistic CBD combinations compared to monotherapies or negative controls. A shotgun proteomics study highlighted 121 dysregulated proteins in CBD-treated MCF7 cells compared to the negative controls. We reported the inhibition of topoisomerase II ß and α, cullin 1, V-type proton ATPase, and CDK-6 in CBD-treated MCF7 cells for the first time as additional cytotoxic mechanisms of CBD, alongside sabotaged energy production and reduced mitochondrial translation. We observed 91 significantly dysregulated proteins in MCF7 cells treated with the synergistic combination of CBD with SN-38 (CSN-38), compared to the monotherapies. Regulation of telomerase, cell cycle, topoisomerase I, EGFR1, protein metabolism, TP53 regulation of DNA repair, death receptor signalling, and RHO GTPase signalling pathways contributed to the proteome-wide synergistic molecular mechanisms of CSN-38. In conclusion, we identified significant synergistic interactions between CBD and the five important chemotherapeutic drugs and the key molecular pathways of CBD and its synergistic combination with SN-38 in MCF7 cells. Further in vivo and clinical studies are warranted to evaluate the implementation of CBD-based synergistic adjuvant therapies for breast cancer.


Asunto(s)
Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Cannabidiol/química , Proteómica/métodos , Adenocarcinoma/metabolismo , Antineoplásicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis , Neoplasias de la Mama/metabolismo , Cannabidiol/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Dactinomicina/análogos & derivados , Dactinomicina/farmacología , Docetaxel/química , Docetaxel/metabolismo , Doxorrubicina/química , Doxorrubicina/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Humanos , Irinotecán/química , Irinotecán/metabolismo , Células MCF-7 , Paclitaxel/química , Paclitaxel/metabolismo , Proteoma , Vinorelbina/química , Vinorelbina/metabolismo
9.
Carbohydr Polym ; 267: 118160, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34119134

RESUMEN

To achieve target delivery of anti-tumor drugs with great biocompatibility into tumor tissues, a stimuli-responsive dendronized hyaluronic acid (HA)-docetaxel conjugate (HA-DTX-Dendron, HADD) was designed and prepared. The incorporation of HA in HADD improved the delivery of DTX to tumor cells with rich CD44 receptors. Enhanced biocompatibility and therapeutic outcomes were achieved using glyodendrons-modified HA and tumor microenvironment-responsive linkers in HADD. The glycodendron was connected with HA via GSH-responsive disulfide bonds, and the drug DTX was linked to the carrier via a cathepsin B-responsive tetrapeptide GFLG. This design resulted in self-assembly nanostructures for facilitating uptake of HADD by tumor cells and rapid release of DTX to exert its therapeutic effect. Compared to free DTX, HADD showed much higher tumor growth inhibition in the MDA-MB-231 tumor-bearing mice model (up to 99.71%), and no toxicity was observed. Therefore, HADD could be employed as an efficacious nano-agent for treating triple negative breast cancer (TNBC).


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Docetaxel/análogos & derivados , Docetaxel/uso terapéutico , Portadores de Fármacos/química , Ácido Hialurónico/análogos & derivados , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Dendrímeros/síntesis química , Dendrímeros/química , Dendrímeros/metabolismo , Docetaxel/metabolismo , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/síntesis química , Ácido Hialurónico/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Nanoestructuras/química , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Chem Biol Drug Des ; 98(3): 466-479, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107169

RESUMEN

Noscapine, an opium alkaloid, was discovered to bind tubulin, arrest dividing cells at mitosis, and selectively induce apoptosis to cancer cells. N-3-Br-Benzyl-Noscapine (Br-Bn-Nos), one of the derivatives of noscapine, was demonstrated to have improved anticancer potential compared with noscapine. We approached to evaluate the single and combined effect of Br-Bn-Nos and docetaxel (DOX) based on molecular modeling and cellular study. The individual predicted free energy of binding (∆Gbind,pred ) for Br-Bn-Nos and DOX with tubulin was found to be -28.89 and -36.07 kcal/mol based on molecular mechanics generalized Born solvation area (MM-GBSA) as well as -26.21 and -34.65 kcal/mol based on molecular mechanics Poisson Boltzmann solvation area (MM-PBSA), respectively. However, the ∆Gbind,pred of Br-Bn-Nos was significantly reduced (-33.02 and -30.24 kcal/mol using MM-GBSA and MM-PBSA) in the presence of DOX on its binding pocket. Parenthetically, the ∆Gbind,pred of DOX was significantly reduced (-37.17 and -32.80 kcal/mol using MM-GBSA and MM-PBSA) in the presence of Br-Bn-Nos on its binding pocket. The reduced ∆Gbind,pred in the presence of Br-Bn-Nos and DOX together indicated a combination effect of both the ligands. The combined interaction of both the agents onto tubulin dimmer was also determined experimentally using purified tubulin, in which a combination regimen of Br-Bn-Nos and DOX reduced the fluorescence intensity of tubulin to a higher value (68%) compared with the single regimen. Further, isobologram analysis revealed the synergistic effect of Br-Bn-Nos and DOX in antiproliferative activity using MCF-7 cell line at 48 hr (sum FIC = 0.49) and at 72 hr (sum FIC = 0.62). The combination dose regimen of Br-Bn-Nos and DOX blocks the cell cycle progression at the G2/M phase and induces apoptosis to cancer cells more effectively compared with the single regimen. Taken together, our study provides compelling evidence that the anticancer potential of noscapine derivatives may be substantially improved when it is used in a combined application with DOX for breast cancer.


Asunto(s)
Antineoplásicos/química , Docetaxel/química , Noscapina/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Docetaxel/metabolismo , Docetaxel/farmacología , Sinergismo Farmacológico , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Simulación del Acoplamiento Molecular , Noscapina/metabolismo , Noscapina/farmacología , Termodinámica , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
11.
AAPS PharmSciTech ; 22(5): 165, 2021 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-34046797

RESUMEN

In the present study, different in situ hydrogel formulations of docetaxel (DTX) based on biocompatible polymers such as Hyaluronic Acid (HA), poloxamer-407, chitosan and gellan gum were formulated to increase its therapeutic efficacy and reduce toxicity. DTX was loaded in nanovesicles (20 mg/mL equivalent to commercial strength) and further incorporated into the hydrogel bases to possess a dual rationale of protection against burst release and enhanced solubility of the drug. The optimized hydrogel formulation (NV-TPGS-3-GG-4) showed ideal rheological behavior and in situ characteristics at 37±0.5°C with sustained release of more than 144 h. The optimized formulation had instant in vitro gelation (2.8±0.3 min) with good injectability in comparison to the conventional commercial DTX injectable formulation having instant release (<2 h). Additionally, developed formulation exhibited an improved biodisponibility (25.1±0.2 h) in comparison to the commercially available formulation (1.7±0.1 h). The Solid Tumor Carcinoma model in Swiss albino mice revealed that the optimized formulation (based on gellan gum) showed better tumor reduction (85.7±1.2%) and lower toxicity as compared to the commercial formulation (77.3±1.3%). Pharmacokinetic and biodistribution studies demonstrated 3 to 4 times higher localization of drug in tumors. Our findings suggested that injectable gellan gum-based in situ hydrogel formulation can be an effective delivery system for DTX with enhanced solubility, reduced toxicity, and better targeting to the tumors for improved therapeutics.Graphical abstract.


Asunto(s)
Antineoplásicos/administración & dosificación , Docetaxel/administración & dosificación , Composición de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/métodos , Nanocápsulas/administración & dosificación , Polisacáridos Bacterianos/administración & dosificación , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Línea Celular Tumoral , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/metabolismo , Docetaxel/química , Docetaxel/metabolismo , Femenino , Hidrogeles/administración & dosificación , Hidrogeles/química , Hidrogeles/metabolismo , Ratones , Nanocápsulas/química , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/metabolismo , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
12.
Carbohydr Polym ; 255: 117393, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33436222

RESUMEN

In this paper, a novel redox-responsive nanoparticles has been designed for targeted delivery of docetaxel (DTX). Chondroitin sulfate (CS) was used to construct the nanoparticles due to the ability of tumor targeting through binding with CD44 receptor that overexpresses on the surfaces of various tumor cells. A redox-responsive small-molecular DTX prodrug was prepared through modifying with cystamine containing disulfide bonds (Cys-DTX). Then the DTX prodrug was grafted to the CS to construct the amphiphilic polymer (CS-ss-DTX). Further, Cys-DTX/CS-ss-DTX nanoparticles were formed by self-assembly of amphiphilic polymer and incorporation of free Cys-DTX prodrug. This category of nanosized DTX delivery system was expected for not only exhibiting high permeability and cytotoxicity of Cys-DTX prodrug, but also targeting transportation of encapsulated redox-responsive Cys-DTX prodrug. According to results of related researches on physicochemical properties and biological evaluation, the novel redox-responsive Cys-DTX/CS-ss-DTX nanoparticles increased amount of DTX released from the nanoparticles in reductive environment, improved permeability in tumor tissues, enhanced cytotoxicity and decreased side effects compared with free DTX. All of these results showed that this kind of Cys-DTX/CS-ss-DTX nanoparticles were worthy of being expectation in tumor chemotherapy in future.


Asunto(s)
Antineoplásicos/farmacología , Sulfatos de Condroitina/química , Docetaxel/farmacología , Glicoconjugados/farmacología , Melanoma Experimental/tratamiento farmacológico , Profármacos/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Supervivencia Celular/efectos de los fármacos , Docetaxel/química , Docetaxel/metabolismo , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacología , Liberación de Fármacos , Femenino , Glicoconjugados/química , Glicoconjugados/metabolismo , Humanos , Células MCF-7 , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Micelas , Nanopartículas/administración & dosificación , Nanopartículas/química , Nanopartículas/ultraestructura , Oxidación-Reducción , Profármacos/química , Profármacos/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Carga Tumoral/efectos de los fármacos
13.
Mater Sci Eng C Mater Biol Appl ; 111: 110760, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32279783

RESUMEN

Nanoparticle-protein interactions under conditions mimicking physiology determine how nanoparticles (NPs) will behave inside blood vessels and, therefore, the overall outcome of the drug-delivery system. Here, for the first time, we explore the effects of bio-mimicking shear stress and protein corona conditions on novel active targeting of clickable fattigation nanoparticles (NPs) for cancer therapy. Active targeting dibenzocyclooctyne-functionalized biocompatible gelatin-oleic NPs (GON-DBCOs) via a bioorthogonal click reaction were prepared by the desolvation method for delivery of docetaxel (DTX) to lung and breast cancer models. The effect of shear stress (5 dyne/cm2) and human serum albumin (HSA) protein corona on the cellular behavior of NPs was explored under a dynamic microfluidic system in lung (A549) and breast (MCF-7) cancer cell lines. The developed drug-loaded NPs had a particle size of 300 nm, a narrow size distribution, positive zeta potential, high encapsulation efficacy (72.4%), and spherical morphology. The particle size of the protein corona-coated NPs increased to 341 nm with a negative zeta potential. The inhibitory dose (IC50) increased approximately 3- and 42-fold in A549 and MCF-7 cells, respectively, under dynamic microfluidic conditions compared to static conditions. Cellular uptake was significantly decreased in the presence of shear stress and a protein corona, compared with static conditions, in both lung (A549, **p < 0.01) and breast (MCF-7, *p < 0.05) cancer cell lines. Clathrin-and energy-dependent pathways were found to be involved in the cellular uptake of NPs. This study could serve as a vital tool for the evaluation of NPs under aggressive bio-mimicking conditions comprising shear stress and a protein corona to predict the in vivo performance of NPs and support the preclinical and clinical translation of NP drug delivery systems.


Asunto(s)
Docetaxel/química , Portadores de Fármacos/química , Gelatina/química , Nanopartículas/química , Corona de Proteínas/química , Materiales Biocompatibles/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Química Clic , Docetaxel/metabolismo , Docetaxel/farmacología , Liberación de Fármacos , Humanos , Microscopía Confocal , Tamaño de la Partícula , Albúmina Sérica Humana/química , Resistencia al Corte
14.
Theranostics ; 10(4): 1619-1632, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32042326

RESUMEN

Until recently, there have been limited options for patients with bone metastatic castration-resistant prostate cancer (BmCRPC) following the failure of or development of resistance to docetaxel (DTX), which is one of the frontline treatments. Sterol regulatory element-binding protein 1 (SREBP1) is reported to regulate abnormal lipid metabolism and to promote the progression and metastasis of prostate cancer (PCa). The siRNA interferes SREBP1 may provide an efficient treatment when combined with DTX. Methods: In this study, lipoic acid (LA) and cross-linked peptide-lipoic acid micelles were cross-linked (LC) for DTX and siSREBP1 delivery (LC/D/siR). Then, cell membrane of PCa cells (Pm) and bone marrow mesenchymal stem cells (Bm) were fused for cloaking LC/D/siR (PB@LC/D/siR). Finally, the synthesized PB@LC/D/siR was evaluated in vitro and in vivo. Results: PB@LC/D/siR is internalized in PCa cells by a mechanism of lysosome escape. Tumor targeting and bone homing studies are evaluated using bone metastatic CRPC (BmCRPC) models, both in vitro and in vivo. Moreover, the enhanced anti-proliferation, anti-migration and anti-invasion capacities of DTX- and siSREBP1- loaded PB@LC (PB@LC/D/siR) were observed in vitro. Furthermore, PB@LC/D/siR was able to suppress the growth of the tumor effectively with deep tumor penetration, high safety and good protection of the bone at the tumor site. Additionally, the mRNA levels and protein levels of SREBP1 and SCD1 were able to be significantly downregulated by PB@LC/D/siR. Conclusion: This study presented a bone-cancer dual-targeting biomimetic nanodelivery system for bone metastatic CRPC.


Asunto(s)
Neoplasias Óseas/secundario , Docetaxel/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Interferente Pequeño/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Animales , Biomimética/métodos , Neoplasias Óseas/tratamiento farmacológico , Docetaxel/administración & dosificación , Docetaxel/uso terapéutico , Regulación hacia Abajo , Sistemas de Liberación de Medicamentos/métodos , Resistencia a Antineoplásicos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Micelas , Nanomedicina/métodos , Neoplasias de la Próstata Resistentes a la Castración/complicaciones , Ratas , Ratas Sprague-Dawley , Ácido Tióctico/administración & dosificación , Moduladores de Tubulina/administración & dosificación , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/uso terapéutico
15.
Xenobiotica ; 50(2): 125-134, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30865477

RESUMEN

1. Docetaxel (DTX) liposome powder was stable over three months, and the liposome suspension was stable within 8 h.2. Rabbits, rats and mice were intravenously treated with DTX-LP or with a DTX injection (DTX-IN). Four major metabolites of DTX were identified in feces: M1, M2, M3 and M4. However, M4 was not found in the bile.3. The most abundant metabolite in the feces was M2 followed by M1/M3, with only a small amount of M4 observed. The most abundant metabolite in bile was also M2, followed by M1/M3.4. The liposomal delivery of DTX did not alter the in vivo drug metabolism, and there were no significant differences among the three species tested. This suggested that this formulation is pharmaceutically safe for clinical use. In contrast to the traditional injected formula, DTX-LP administration significantly delayed drug metabolism, as observed in feces and bile. This property will greatly enhance the DTX therapeutic efficacy and reduce the systemic side effects of NSCLC treatment.


Asunto(s)
Antineoplásicos/metabolismo , Docetaxel/metabolismo , Liposomas/metabolismo , Animales , Humanos , Metaboloma , Ratones , Conejos , Ratas
16.
J Control Release ; 318: 197-209, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31672622

RESUMEN

With the in-depth research of organelles, the microenvironment characteristics of their own, such as the acid environment of lysosomes and the high temperature environment of mitochondria, could be used as a natural and powerful condition for tumor therapy. Based on this, we constructed a two-step precise targeting nanoplatform which can realize the drug release and drug action triggered by the microenvironment of lysosomes (endosomes) and mitochondria, respectively. To begin with, the mesoporous silica nanoparticles (MSNs) were modified with triphenylphosphonium (TPP) and loaded with 2,2'-azobis[2-(2-imidazolin-2-yl) propane] dihydrochloride (AIPH). Then, folic acid (FA) targeted pH-sensitive liposomes containing docetaxel (Lipo/DTX-FA) were prepared by thin-film dispersion method, and the core-shell AIPH/MSN-TPP@Lipo/DTX-FA nanoparticles were constructed by self-assembly during the hydration of the liposomes. When this nanoplatform entered into the tumor cells through FA receptor-mediated endocytosis, the pH-sensitive liposomes were destabilized in the lysosomes, resulting in the release of DTX and AIPH/MSN-TPP nanoparticles. After that, AIPH was delivered to mitochondria by AIPH/MSN-TPP, and the alkyl radicals produced by AIPH under the high temperature environment can cause oxidative damage to mitochondria. Not only that, the DTX could enhance the anti-tumor effect of AIPH by downregulating the expression of anti-apoptotic Bcl-2 protein. The in vitro and in vivo results demonstrate that this delivery system could induce apoptosis based on organelles' s own microenvironment, which provides a new approach for tumor therapy.


Asunto(s)
Antineoplásicos , Nanopartículas , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Docetaxel/metabolismo , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Mitocondrias/metabolismo , Dióxido de Silicio/metabolismo
17.
J Cell Physiol ; 235(5): 4618-4630, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31674023

RESUMEN

Prostate cancer (PCa) is one of the most prevalent non-drug delivery system cutaneous malignancies. Undoubtedly, introducing novel treatment options to achieve higher therapeutic index will be worthwhile. In this study, we report for the first time, a novel targeted self-assembled based on PEG-PLA nanoparticles (PEG-PLA NPs) containing galbanic acid (GBA) and docetaxel, which was targeted using ((S)-2-(3-((S)-5-amino-1-carboxypentyl) ureido) pentanedioic acid (ACUPA), a small molecule inhibitor targeting prostate-specific membrane antigen (PSMA), in prostate cancer cell line. The prepared NPs were characterized by different analytical methods. The MTT assay was used to compare the anti-proliferation of drugs-loaded PEG-PLA NPs and ACUPA-PEG-PLA against LNCaP (PSMA+ ) and PC3 (PSMA- ) cells. PEG-PLA NPs with an average size of 130-140 nm had an enhanced release of GBA and docetaxel at pH 5.5 compared with pH 7.5. Spectrofluorometric analysis suggested that ACUPA-modified PEG-PLA could effectively enhance the drug uptake in PSMA+ prostate cancer cells. Cytotoxicity studies showed that the targeted NPs loaded with different concentrations of GBA and fixed concentration of docetaxel (4 nM) have shown higher toxicity (IC50 30 ± 3 µM) than both free GBA (80 ± 4.5 µM) and nontargeted NPs (IC50 40 ± 4.6 µM) in LNCaP cells. Collectively, these findings suggest that ACUPA-conjugated PEG-PLA nanosystem containing GBA and docetaxel is a viable delivery carrier for various cancer-targeting PSMA that suffer from short circulation half-life and limited therapeutic efficacy.


Asunto(s)
Antígenos de Superficie/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cumarinas/farmacología , Docetaxel/farmacología , Portadores de Fármacos , Glutamato Carboxipeptidasa II/metabolismo , Glutaratos/química , Nanopartículas , Polietilenglicoles/química , Neoplasias de la Próstata/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cumarinas/química , Cumarinas/metabolismo , Docetaxel/química , Docetaxel/metabolismo , Composición de Medicamentos , Liberación de Fármacos , Glutaratos/metabolismo , Humanos , Ligandos , Masculino , Células PC-3 , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
18.
Spectrochim Acta A Mol Biomol Spectrosc ; 228: 117528, 2020 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-31718965

RESUMEN

DNA stands as the primary purpose of many anticancer drugs and according to the performed research on this field, some certain changes contain crucial functionalities in the regulated transcription of DNA. Therefore, the interaction between anticancer drugs and DNA play an important role in understanding their function and also provide a better groundwork for producing more efficient and newer drugs. Here, the interaction between Docetaxel (DO) and calf thymus DNA (ct DNA), in the presence and absence of Anastrozole (AN), has been examined through the usage of different methods that include isothermal titration calorimetry, multi-spectroscopic, viscometry, and molecular docking techniques. Interaction studies have been performed by preparing different molar ratios of DO with the constant ct DNA and AN concentration at pH = 6.8. The binding constants have been calculated to be 7.93 × 104 M-1 and 6.27 × 104 M-1, which indicate the strong binding of DO with ct DNA double helix in the absence and presence of AN, respectively. Thermodynamic parameters, which were obtained from fluorescence spectroscopy and isothermal titration calorimetry, have suggested that the binding of DO and AN to ct DNA as binary and ternary systems have been mainly driven by the electrostatic interactions. The relative viscosity of ct DNA has increased upon the addition of DO and AN, which confirms the interaction mode. A competitive binding study has reported that the enhanced emission intensity of ethidium bromide (EB) and acridine orange (AO), in the presence of ct DNA, have been quenched through the addition of DO and Anastrozole as binary and ternary systems. As it is indicated by these findings, DO is capable of displacing EB and AO from their binding site in ct DNA; hence, it can be concluded that DO and AN are able to intercalate into the base pairs of ct DNA in binary and ternary systems. Molecular docking studies have corroborated the mentioned experimental results.


Asunto(s)
Anastrozol/metabolismo , Simulación por Computador , ADN/metabolismo , Docetaxel/metabolismo , Anastrozol/química , Unión Competitiva , Calorimetría , ADN/química , Docetaxel/química , Cinética , Modelos Moleculares , Desnaturalización de Ácido Nucleico , Concentración Osmolar , Dispersión de Radiación , Espectrometría de Fluorescencia , Termodinámica , Viscosidad
19.
Biomater Sci ; 7(3): 1117-1131, 2019 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-30638237

RESUMEN

Oral chemotherapy of docetaxel (DTX) is restricted by active P-glycoprotein (P-gp) efflux, hepatic first-pass metabolism and then poor oral absorption. Herein, a lipophilic thioether-bridged oleate prodrug (DTX-S-OA) and an ester-bond linked oleate prodrug of docetaxel (DTX-OA) were synthesized and efficiently incorporated into a self-nanoemulsifying drug delivery system (SNEDDS) using core-matching technology with a high drug-loading rate. DTX-S-OA SNEDDS produced a uniform droplet size of about 30 nm and a significantly high drug loading capability (60 mg mL-1), compared with DTX SNEDDS (20 mg mL-1). Additionally, DTX-S-OA SNEDDS exhibited a markedly slower drug release property and higher (>2-fold) drug solubilization in the aqueous phase after 60 min lipolysis compared with DTX SNEDDS. In situ single-pass intestinal perfusion and intestinal biodistribution studies demonstrated that the membrane permeability and intestinal bioadhesion of SNEDDS were significantly increased. Moreover, DTX-S-OA showed a comparable ability with verapamil in inhibiting P-gp efflux. Lymphatic transport studies confirmed that DTX-S-OA SNEDDS could significantly enhance intestinal lymphatic transport. Notably, the bioavailability of DTX-S-OA SNEDDS was 6.2-fold and 2.0-fold higher than that of the DTX solution and DTX SNEDDS, respectively. Furthermore, DTX-S-OA achieved a more rapid release of free DTX from the prodrug in systemic circulation than DTX-OA. Therefore, such a unique combination strategy of the single thioether-bridged DTX-oleate prodrug and SNEDDS is a promising platform to enable effective oral delivery of DTX.


Asunto(s)
Docetaxel/química , Portadores de Fármacos/química , Profármacos/química , Sulfuros/química , Administración Oral , Animales , Cumarinas/química , Docetaxel/metabolismo , Liberación de Fármacos , Emulsiones/química , Mucosa Intestinal/metabolismo , Intestinos/patología , Ratones , Ácido Oléico/química , Tamaño de la Partícula , Profármacos/metabolismo , Ratas , Ratas Sprague-Dawley , Solubilidad , Tiazoles/química
20.
J Control Release ; 285: 187-199, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30017721

RESUMEN

The anticancer efficacy of chemotherapy is greatly limited by short blood circulation and poor tumor selectivity. Thus, anticancer prodrugs with prolonged systemic circulation, tumor-specific distribution and bioactivation, could significantly strengthen the chemotherapy efficacy. Herein, we design two novel tumor cell reduction/oxidation-responsive docetaxel (DTX) prodrugs, DTX-maleimide conjugates with disulfide bond (DSSM) or thioether bond (DSM) linkages, to evaluate the roles of different sensitive linkages in drug release, pharmacokinetics and therapeutic efficacy. An ester bond-linkage prodrug (DM) is utilized as a non-sensitive control. DSSM and DSM show reduction- or oxidation-sensitive release behavior, respectively, and exhibit hyperselective bioactivation and cytotoxicities between cancerous and normal cells. They could instantly hitchhike blood circulating albumin after i.v. administration with albumin-binding half-lives as short as 1 min, resulting in prolonged systemic circulation, increased tumor accumulation. In response to the upregulated reduction/oxidation environment within tumor cells, DSSM and DSM exhibit selectively release capacity in tumor tissues, their TAITumor/Liver values are over 30-fold greater than DM. Combining the above delivery advantages into one, DSSM and DSM achieve enhanced antitumor efficacy of DTX. Such a uniquely developed strategy, integrating high albumin-binding capability and reduction/oxidation-sensitive drug superselective release in tumors, has great potential to be applied in clinical cancer therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Docetaxel/análogos & derivados , Docetaxel/uso terapéutico , Profármacos/química , Profármacos/uso terapéutico , Albúmina Sérica/metabolismo , Animales , Línea Celular Tumoral , Docetaxel/metabolismo , Docetaxel/farmacocinética , Liberación de Fármacos , Femenino , Humanos , Masculino , Ratones Endogámicos BALB C , Oxidación-Reducción , Profármacos/metabolismo , Profármacos/farmacocinética , Unión Proteica , Ratas Sprague-Dawley , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...