Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Nanobiotechnology ; 22(1): 319, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38849938

RESUMEN

Myeloid-derived suppressor cells (MDSCs) have played a significant role in facilitating tumor immune escape and inducing an immunosuppressive tumor microenvironment. Eliminating MDSCs and tumor cells remains a major challenge in cancer immunotherapy. A novel approach has been developed using gemcitabine-celecoxib twin drug-based nano-assembled carrier-free nanoparticles (GEM-CXB NPs) for dual depletion of MDSCs and tumor cells in breast cancer chemoimmunotherapy. The GEM-CXB NPs exhibit prolonged blood circulation, leading to the preferential accumulation and co-release of GEM and CXB in tumors. This promotes synergistic chemotherapeutic activity by the proliferation inhibition and apoptosis induction against 4T1 tumor cells. In addition, it enhances tumor immunogenicity by immunogenic cell death induction and MDSC-induced immunosuppression alleviation through the depletion of MDSCs. These mechanisms synergistically activate the antitumor immune function of cytotoxic T cells and natural killer cells, inhibit the proliferation of regulatory T cells, and promote the M2 to M1 phenotype repolarization of tumor-associated macrophages, considerably enhancing the overall antitumor and anti-metastasis efficacy in BALB/c mice bearing 4T1 tumors. The simplified engineering of GEM-CXB NPs, with their dual depletion strategy targeting immunosuppressive cells and tumor cells, represents an advanced concept in cancer chemoimmunotherapy.


Asunto(s)
Desoxicitidina , Gemcitabina , Inmunoterapia , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide , Nanopartículas , Animales , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Células Supresoras de Origen Mieloide/efectos de los fármacos , Ratones , Inmunoterapia/métodos , Femenino , Nanopartículas/química , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Proliferación Celular/efectos de los fármacos
2.
J Nanobiotechnology ; 21(1): 393, 2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-37898773

RESUMEN

Irinotecan (Ir) is commonly employed as a first-line chemotherapeutic treatment for colorectal cancer (CRC). However, tremendous impediments remain to be addressed to surmount drug resistance and ameliorate adverse events. Poly-ADP-Ribose Polymerase (PARP) participates in the maintenance of genome stability and the repair of DNA damage, thus playing a critical role in chemotherapy resistance. In this work, we introduce a novel curative strategy that utilizes nanoparticles (NPs) prepared by dynamic supramolecular co-assembly of Ir and a PARP inhibitor (PARPi) niraparib (Nir) through π-π stacking and hydrogen bond interactions. The Ir and Nir self-assembled Nano-Twin-Drug of (Nir-Ir NPs) could enhance the therapeutic effect on CRC by synergistically inhibiting the DNA damage repair pathway and activating the tumor cell apoptosis process without obvious toxicity. In addition, the Nir-Ir NPs could effectively reverse irinotecan-resistance by inhibiting the expression of multiple resistance protein-1 (MRP-1). Overall, our study underscores the distinctive advantages and potential of Nir-Ir NPs as a complementary strategy to chemotherapy by simultaneously overcoming the Ir resistance and improving the anti-tumor efficacy against CRC.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Humanos , Irinotecán/farmacología , Irinotecán/uso terapéutico , Antineoplásicos/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Apoptosis , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Línea Celular Tumoral
3.
Bioorg Med Chem ; 65: 116793, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35550978

RESUMEN

Inspired by antitumor natural product evodiamine, a series of novel bis-evodiamine derivatives were designed and synthesized, which showed potent antitumor activity. In particular, compound 13b effectively inhibited the proliferation and migration of HCT116 cells. Further mechanism studies revealed that compound 13b acted by inducing HCT116 cell apoptosis and arresting the cell cycle at the G2/M phase. Thus, compound 13b represents a promising lead compound for the discovery of novel antitumor agents.


Asunto(s)
Antineoplásicos , Diseño de Fármacos , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Quinazolinas , Relación Estructura-Actividad
4.
Mol Pharm ; 12(7): 2328-36, 2015 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-25996874

RESUMEN

We report here an amphiphilic twin drug strategy directly using small molecular hydrophilic and hydrophobic anticancer drugs to self-assemble into nanoparticles with a high and fixed drug content, which can solve problems of anticancer drug delivery including poor water solubility, low therapeutic indices, and severe side effects. The twin drug has been prepared by the esterification of the hydrophilic anticancer drug floxuridine (FdU) with the hydrophobic anticancer drug bendamustine (BdM). Due to its inherent amphiphilicity, the FdU-BdM twin drug can self-assemble into stable and well-defined nanoparticles. After FdU-BdM twin drug enters into cells, the ester linkage between hydrophilic and hydrophobic drugs is readily cleaved by hydrolysis to release free FdU and BdM. Since both FdU and BdM can kill cancer cells, the FdU-BdM twin drug nanoparticles can overcome the multidrug resistance (MDR) of tumor cells and present an excellent anticancer activity. This strategy can be extended to other hydrophilic and hydrophobic anticancer drugs to synthesize amphiphilic twin drugs which can form nanoparticles to self-deliver drugs for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Clorhidrato de Bendamustina/farmacología , Floxuridina/farmacología , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Antineoplásicos/química , Clorhidrato de Bendamustina/química , Línea Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Floxuridina/química , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células MCF-7 , Nanopartículas/química
5.
Biomed Mater ; 18(4)2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37201518

RESUMEN

Podophyllotoxin (PPT) is an active natural pharmaceutical component with potent anticancer activity. However, due to its poor water solubility and serious side effects, its medical applications are limited. In this work, we synthesized a series of PPT dimers, which can be self-assembled into stable nanoparticles of 124-152 nm in aqueous solution and can significantly increase the solubility of PPT in aqueous solution. In addition, PPT dimer nanoparticles exhibited high drug loading capacity (>80%) and could store at 4 °C in aqueous state with good stability for at least 30 d.In vitrorelease studies showed that nanoparticles with disulfide bonds (SS NPs) can quickly release (about 96.5% drug released within 24 h) the conjugated drug in PBS buffer (pH = 7.4) in the presence of DTT. Cell endocytosis experiments showed that SS NPs enhanced cell uptake (18.56 times higher than PPT for Molm-13, 10.29 times for A2780S and 9.81 times for A2780T) and maintained antitumor effect against human ovarian tumor cells (A2780S and resistant A2780T) and human breast cancer cells (MCF-7). In addition, the endocytosis mechanism of SS NPs was revealed that these nanoparticles were mainly up-taken by macropinocytosis-mediated endocytosis. We believe that these PPT dimer-based nanoparticles will become an alternative formula for PPT, moreover the assembly behavior of PPT dimer can be extended to other therapeutic drugs.


Asunto(s)
Antineoplásicos , Nanopartículas , Humanos , Podofilotoxina/química , Preparaciones Farmacéuticas , Sistemas de Liberación de Medicamentos , Polímeros/química , Nanopartículas/química , Oxidación-Reducción , Antineoplásicos/química
6.
China Pharmacy ; (12): 660-665, 2023.
Artículo en Zh | WPRIM | ID: wpr-965501

RESUMEN

OBJECTIVE To study the protective effects of ligustrazine-scutellarein twin drug ST-11 on rat adrenal medullary pheochromocytoma PC12 cell injury induced by oxygen-glucose deprivation/reperfusion (OGD/R) and its mechanism. METHODS PC12 cells were divided into blank group, model group, nimodipine group (positive control, 5 μmol/L) and different concentration groups of ST-11 (5, 10, 20 μmol/L). After 24 hours of pre-administration intervention, all the other groups except the blank group were cultured in glucose-free DMEM culture medium containing 10 mmol/L Na2S2O4 for 4 hours with glucose deficiency and hypoxia. After 4 hours of glucose and oxygen re-introduction, the survival rate of cells in each group, the contents of lactate dehydrogenase (LDH), catalase (CAT), glutathione (GSH), malondialdehyde (MDA) and superoxide dismutase (SOD) in cell supernatant, apoptosis rate, the levels of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP), the protein expressions of B-cell lymphoma 2 related X protein (Bax), B-cell lymphoma 2 (Bcl-2) and caspase-3 were all detected in each group. RESULTS Compared with blank group, the cell survival rate, the contents of CAT, GSH and SOD in cell supernatant, MMP level, relative expression of Bcl-2 and Bcl-2/Bax ratio in model group decreased significantly (P<0.05), while the contents of LDH and MDA, ROS level, apoptosis rate, relative expressions of Bax and caspase-3 were significantly increased (P<0.05). Compared with model group, above indexes of ST-11 groups (except for the protein expression of caspase-3 in 5 μmol/L ST-11 group) were reversed signifi-cantly (P<0.05). CONCLUSIONS ST-11 has a certain protec-tive effect on OGD/R-injured PC12 cells, and its effects may be related to reduction of oxidative stress and inhibition of cell apoptosis.

7.
Eur J Pharm Sci ; 122: 179-184, 2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-29981402

RESUMEN

Inflammation is a nonspecific response of tissues to diverse stimuli and/or insults associated with the release of various mediators that induce pain, fever, and general sense of illness. Such responses can be reduced by the administration of non-steroidal or steroidal anti-inflammatory drugs. These anti-inflammatory drugs are associated with various side effect. However, the combination of non-steroidal anti-inflammatory drugs (NSAIDs) with glucocorticoids provides a synergistic anti-inflammatory and pain relief with less side effects. In this study, we aimed to synthesize a novel twin-drug of Dexamethasone-Diclofenac formulated into polylactide (PLA) nanoparticles to improve their solubility and provide a sustained release system. The twin-drug was synthesized through an esterification reaction which was then encapsulated into PLA nanoparticles. The hydrolysis of the synthesized twin-drug and drug release from PLA nanoparticles were studied in vitro with and without esterase enzyme. The anti-inflammatory activity was studied in BALB/c mice. After the successful synthesis of the twin-drug, its water solubility was improved by its encapsulation into PLA nanoparticles with a loading capacity of 66%. The in vitro release showed a sustained release profile of the twin-drug up to 52 h. The esterase hydrolysis showed a rapid release with a maximum hydrolysis after 1.5 h. Moreover, the anti-inflammatory activity exhibited a synergistic effect with a higher percentage of inhibition for the TNF-α level in comparison to the parent drugs after 6 h treatment. In conclusion, the prepared nano twin-drug is a novel therapy that showed a sustained release profile with an excellent anti-inflammatory activity.


Asunto(s)
Antiinflamatorios no Esteroideos , Dexametasona , Diclofenaco , Glucocorticoides , Nanopartículas , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/química , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/química , Dexametasona/administración & dosificación , Dexametasona/química , Diclofenaco/administración & dosificación , Diclofenaco/química , Combinación de Medicamentos , Composición de Medicamentos , Liberación de Fármacos , Glucocorticoides/administración & dosificación , Glucocorticoides/química , Lipopolisacáridos/farmacología , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Nanopartículas/química , Poliésteres/administración & dosificación , Poliésteres/química
8.
Eur J Med Chem ; 144: 137-150, 2018 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-29268130

RESUMEN

Shikonin exhibits powerful anticancer activities for various cancer cells, but its poor solubility and strong toxicity hinder its development as clinical anticancer agent. We previously confirmed that shikonin and its derivatives can disturb mitosis through targeting tubulin. In this study, α-lipoic acid, the naturally-occurring co-factor of pyruvate dehydrogenase (PDH), was introduced into shikonin to design the twin drugs against both mitosis (tubulin) and glycolysis (PDK). 18 kinds of α-lipoic acid shikonin ester derivatives were achieved through three rounds of screening process performed by computer assistant drug design method, being designated as the outstanding compounds. Among them, 1c displayed the most potent cytotoxicity towards cervical cancer cells (HeLa) with an IC50 value of 3.14 ± 0.58 µM and inhibited xenotransplanted tumor growth in a dose-dependent manner. Further pharmacologic study demonstrated that 1c can cause cell cycle arrest in G2/M phase as tubulin polymerization inhibitor. Moreover, it also showed good PDK1 inhibitory activity, promoting PDH activity and forced HeLa cells to process more aerobic metabolism to undergo cell apoptosis. We reported here the first dual inhibitors of tubulin and PDK1 based on shikonin. It may form a basis for shikonin optimization through twin drug design framework for the discovery of new and potent shikonin derivatives in the study of targeted cancer therapy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Naftoquinonas/química , Naftoquinonas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diseño de Fármacos , Glucólisis/efectos de los fármacos , Células HeLa , Humanos , Mitosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA