Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.574
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Med Oncol ; 41(5): 111, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38592504

RESUMEN

The use of doxorubicin (Dox) in the treatment of breast cancer negatively affects the intestines and other tissues. Many studies have proven that probiotics and vitamin D3 have antitumor and intestinal tissue-protecting properties. To achieve effectiveness and minimize side effects, the current study aims to administer Dox together with probiotics (Lactobacillus acidophilus and Lactobacillus casei) and vitamin D3. Forty-two female BALB/c inbred mice were divided into six groups: Group 1 (Control), Group 2 (Dox), Group 3 (Dox and probiotics), Group 4 (Dox and vitamin D3), Group 5 (Dox, probiotics, and vitamin D3), and Group 6 (probiotics and vitamin D3). The 4T1 mouse carcinoma cell line was injected into the mammary fat pad of each mouse. Gene expression was examined using quantitative real-time PCR. The treated groups (except group 6) showed significantly reduced tumor volume and weight compared to the control group (P < 0.05, P < 0.01). Probiotics/vitamin D3 with Dox reduced chemotherapy toxicity and a combination of supplements had a significant protective effect against Dox (P < 0.05, 0.01, 0.001). The treated groups (except 6) had significantly higher expression of Bax/Caspase 3 genes and lower expression of Bcl-2 genes than the control group (P < 0.05, 0.01). Coadministration of Dox with probiotics and vitamin D3 showed promising results in reducing tumor size, protecting intestinal tissue and influencing gene expression, suggesting a strategy to enhance the effectiveness of breast cancer treatment while reducing side effects.


Asunto(s)
Lacticaseibacillus casei , Neoplasias , Probióticos , Femenino , Animales , Ratones , Lactobacillus acidophilus , Doxorrubicina/farmacología , Probióticos/farmacología , Modelos Animales de Enfermedad , Colecalciferol/farmacología , Ratones Endogámicos BALB C
2.
ACS Appl Mater Interfaces ; 16(15): 18252-18267, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38581365

RESUMEN

Nitric oxide (NO) intervenes, that is, a potential treatment strategy, and has attracted wide attention in the field of tumor therapy. However, the therapeutic effect of NO is still poor, due to its short half-life and instability. Therapeutic concentration ranges of NO should be delivered to the target tissue sites, cell, and even subcellular organelles and to control NO generation. Mitochondria have been considered a major target in cancer therapy for their essential roles in cancer cell metabolism and apoptosis. In this study, mesoporous silicon-coated gold nanorods encapsulated with a mitochondria targeted and the thermosensitive lipid layer (AuNR@MSN-lipid-DOX) served as the carrier to load NO prodrug (BNN6) to build the near-infrared-triggered synergetic photothermal NO-chemotherapy platform (AuNR@MSN(BNN6)-lipid-DOX). The core of AuNR@MSN exhibited excellent photothermal conversion capability and high loading efficiency in terms of BNN6, reaching a high value of 220 mg/g (w/w), which achieved near-infrared-triggered precise release of NO. The outer biocompatible lipid layer, comprising thermosensitive phospholipid DPPC and mitochondrial-targeted DSPE-PEG2000-DOX, guided the whole nanoparticle to the mitochondria of 4T1 cells observed through confocal microscopy. In the mitochondria, the nanoparticles increased the local temperature over 42 °C under NIR irradiation, and a high NO concentration from BNN6 detected by the NO probe and DSPE-PEG2000-DOX significantly inhibited 4T1 cancer cells in vitro and in vivo under the synergetic photothermal therapy (PTT)-NO therapy-chemotherapy modes. The built NIR-triggered combination therapy nanoplatform can serve as a strategy for multimodal collaboration.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Fosfatidiletanolaminas , Polietilenglicoles , Doxorrubicina/farmacología , Óxido Nítrico , Fototerapia , Nanopartículas/uso terapéutico , Mitocondrias , Lípidos , Línea Celular Tumoral
3.
Asian Pac J Cancer Prev ; 25(3): 939-949, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38546076

RESUMEN

OBJECTIVES: This research aims to comparatively investigate the capability of resveratrol (RES) and RES analogues, oxyresveratrol (Oxy-RES) and dihydrooxyresveratrol (DHoxy-RES), to potentiate doxorubicin (DOX) effects against lung carcinoma epithelial cells. METHODS: All experiments were performed on lung carcinoma cell lines (A549) with DOX combination between DOX and RES or RES analogues. Cell viability or growth inhibitory effect was assessed by MTT assay and genes associated with survival and metastasis were monitored by real-time polymerase chain reaction (RT-PCR). RESULTS: DOX obviously demonstrated cytotoxic and anti-metastatic activities against A549 cells. Expression of gene-associated with both activities was potentiated by RES and RES analogues. Oxy-RES showed highest capability to potentiate DOX effects. DHoxy-RES showed nearly no effect to DOX activities. CONCLUSIONS: These results provided an important basis of DOX combination with RES analogues, especially Oxy-RES, for better therapeutic effect. Further studies in human should be performed on exploring combination of DOX and Oxy-RES.


Asunto(s)
Carcinoma , Neoplasias Pulmonares , Extractos Vegetales , Estilbenos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Resveratrol/farmacología , Doxorrubicina/farmacología , Pulmón/patología , Línea Celular Tumoral , Apoptosis
4.
J Mater Chem B ; 12(14): 3521-3532, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38525839

RESUMEN

The combination of photothermal therapy and chemotherapy has emerged as a promising strategy to improve cancer therapeutic efficacy. However, developing a versatile nanoplatform that simultaneously possesses commendable photothermal effect and high drug encapsulation efficiency remains a challenging problem yet to be addressed. Herein, we report a facile supramolecular self-assembly strategy to construct gold nanoparticle clusters (AuNCs) for synergistic photothermal-chemo therapy. By utilizing the functional polysaccharide as a targeted ligand, hyaluronic acid-enriched AuNCs were endowed with targeting CD44 receptor overexpressed on the B16 cancer cells. Importantly, these hyaluronic acid modified AuNCs can shelter therapeutic cargo of doxorubicin (DOX) to aggregate larger nanoparticles via a host-guest interaction with the anchored ß-cyclodextrin, as a "nanocluster-bomb" (DOX@AuNCs). The in vitro results revealed that these DOX@AuNCs showed light-triggered drug release behavior and synergistic photothermal-chemo therapy. The improved efficacy of synergistic therapy was further demonstrated by treating a xenografted B16 tumor model in vivo. We envision that our multipronged design of DOX@AuNCs provides a potent theranostic platform for precise cancer therapy and could be further enriched by introducing different imaging probes and therapeutic drugs as appropriate suitable guest molecules.


Asunto(s)
Hipertermia Inducida , Nanopartículas del Metal , Neoplasias , Humanos , Oro , Terapia Fototérmica , Ácido Hialurónico , Neoplasias/patología , Doxorrubicina/farmacología
5.
Int J Mol Sci ; 25(6)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38542073

RESUMEN

Modulated electro-hyperthermia (mEHT) is an adjuvant cancer therapy that enables tumor-selective heating (+2.5 °C). In this study, we investigated whether mEHT accelerates the tumor-specific delivery of doxorubicin (DOX) from lyso-thermosensitive liposomal doxorubicin (LTLD) and improves its anticancer efficacy in mice bearing a triple-negative breast cancer cell line (4T1). The 4T1 cells were orthotopically injected into Balb/C mice, and mEHT was performed on days 9, 12, and 15 after the implantation. DOX, LTLD, or PEGylated liposomal DOX (PLD) were administered for comparison. The tumor size and DOX accumulation in the tumor were measured. The cleaved caspase-3 (cC3) and cell proliferation were evaluated by cC3 or Ki67 immunohistochemistry and Western blot. The LTLD+mEHT combination was more effective at inhibiting tumor growth than the free DOX and PLD, demonstrated by reductions in both the tumor volume and tumor weight. LTLD+mEHT resulted in the highest DOX accumulation in the tumor one hour after treatment. Tumor cell damage was associated with cC3 in the damaged area, and with a reduction in Ki67 in the living area. These changes were significantly the strongest in the LTLD+mEHT-treated tumors. The body weight loss was similar in all mice treated with any DOX formulation, suggesting no difference in toxicity. In conclusion, LTLD combined with mEHT represents a novel approach for DOX delivery into cancer tissue.


Asunto(s)
Doxorrubicina/análogos & derivados , Hipertermia Inducida , Neoplasias , Ratones , Animales , Liposomas , Antígeno Ki-67 , Hipertermia Inducida/métodos , Doxorrubicina/farmacología , Hipertermia , Línea Celular Tumoral , Polietilenglicoles
6.
J Nanobiotechnology ; 22(1): 110, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38481281

RESUMEN

BACKGROUND: Breast cancer ranks first among malignant tumors, of which triple-negative breast cancer (TNBC) is characterized by its highly invasive behavior and the worst prognosis. Timely diagnosis and precise treatment of TNBC are substantially challenging. Abnormal tumor vessels play a crucial role in TNBC progression and treatment. Nitric oxide (NO) regulates angiogenesis and maintains vascular homeostasis, while effective NO delivery can normalize the tumor vasculature. Accordingly, we have proposed here a tumor vascular microenvironment remodeling strategy based on NO-induced vessel normalization and extracellular matrix collagen degradation with multimodality imaging-guided nanoparticles against TNBC called DNMF/PLGA. RESULTS: Nanoparticles were synthesized using a chemotherapeutic agent doxorubicin (DOX), a NO donor L-arginine (L-Arg), ultrasmall spinel ferrites (MnFe2O4), and a poly (lactic-co-glycolic acid) (PLGA) shell. Nanoparticle distribution in the tumor was accurately monitored in real-time through highly enhanced magnetic resonance imaging and photoacoustic imaging. Near-infrared irradiation of tumor cells revealed that MnFe2O4 catalyzes the production of a large amount of reactive oxygen species (ROS) from H2O2, resulting in a cascade catalysis of L-Arg to trigger NO production in the presence of ROS. In addition, DOX activates niacinamide adenine dinucleotide phosphate oxidase to generate and supply H2O2. The generated NO improves the vascular endothelial cell integrity and pericellular contractility to promote vessel normalization and induces the activation of endogenous matrix metalloproteinases (mainly MMP-1 and MMP-2) so as to promote extravascular collagen degradation, thereby providing an auxiliary mechanism for efficient nanoparticle delivery and DOX penetration. Moreover, the chemotherapeutic effect of DOX and the photothermal effect of MnFe2O4 served as a chemo-hyperthermia synergistic therapy against TNBC. CONCLUSION: The two therapeutic mechanisms, along with an auxiliary mechanism, were perfectly combined to enhance the therapeutic effects. Briefly, multimodality image-guided nanoparticles provide a reliable strategy for the potential application in the fight against TNBC.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias de la Mama Triple Negativas , Humanos , Óxido Nítrico , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Especies Reactivas de Oxígeno , Peróxido de Hidrógeno , Doxorrubicina/farmacología , Fototerapia/métodos , Colágeno , Línea Celular Tumoral , Microambiente Tumoral
7.
Int J Pharm ; 655: 124007, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38493844

RESUMEN

Gold core mesoporous silica shell (AuMSS) nanorods are multifunctional nanomedicines that can act simultaneously as photothermal, drug delivery, and bioimaging agents. Nevertheless, it is reported that once administrated, nanoparticles can be coated with blood proteins, forming a protein corona, that directly impacts on nanomedicines' circulation time, biodistribution, and therapeutic performance. Therefore, it become crucial to develop novel alternatives to improve nanoparticles' half-life in the bloodstream. In this work, Polyethylenimine (PEI) and Red blood cells (RBC)-derived membranes were combined for the first time to functionalize AuMSS nanorods and simultaneously load acridine orange (AO). The obtained results revealed that the RBC-derived membranes promoted the neutralization of the AuMSS' surface charge and consequently improved the colloidal stability and biocompatibility of the nanocarriers. Indeed, the in vitro data revealed that PEI/RBC-derived membranes' functionalization also improved the nanoparticles' cellular internalization and was capable of mitigating the hemolytic effects of AuMSS and AuMSS/PEI nanorods. In turn, the combinatorial chemo-photothermal therapy mediated by AuMSS/PEI/RBC_AO nanorods was able to completely eliminate HeLa cells, contrasting with the less efficient standalone therapies. Such data reinforce the potential of AuMSS nanomaterials to act simultaneously as photothermal and chemotherapeutic agents.


Asunto(s)
Antineoplásicos , Nanotubos , Neoplasias , Humanos , Células HeLa , Terapia Fototérmica , Membrana Eritrocítica , Dióxido de Silicio , Oro , Distribución Tisular , Fototerapia , Doxorrubicina/farmacología , Neoplasias/tratamiento farmacológico
8.
Biomed Pharmacother ; 173: 116397, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38479181

RESUMEN

Angiosarcoma is a rare soft tissue sarcoma originating from endothelial cells. Given that current treatments for advanced disease have shown limited efficacy, alternative therapies need to be identified. In rare diseases, patient-derived cell models are crucial for screening anti-tumour activity. In this study, cell line models were characterised in 2D and 3D cultures. The cell lines' growth, migration and invasion capabilities were explored, confirming them as useful tools for preclinical angiosarcoma studies. By screening a drug library, we identified potentially effective compounds: 8-amino adenosine impacted cell growth and inhibited migration and invasion at considerably low concentrations as a single agent. No synergistic effect was detected when combining with paclitaxel, gemcitabine or doxorubicin. These results suggest that this compound could be a potentially useful drug in the treatment of AGS.


Asunto(s)
Hemangiosarcoma , Sarcoma , Humanos , Hemangiosarcoma/tratamiento farmacológico , Células Endoteliales/patología , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sarcoma/tratamiento farmacológico , Paclitaxel/farmacología , Paclitaxel/uso terapéutico
9.
Colloids Surf B Biointerfaces ; 237: 113849, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38492413

RESUMEN

Oral colonic nano-drug delivery system has received more and more attention in the treatment of colon cancer due to local precision treatment and reduction of drug system distribution. However, the complex and harsh gastrointestinal environment and the retention of nanoparticles in the colon limit its development. To this end, we designed Eudragit S100 (ES) coated nanoparticles (ES@PND-PEG-TPP/DOX). Polydopamine coated nanodiamond (PND) was modified with amino-functionalized polyethylene glycol (NH2-PEG-NH2) and triphenylphosphine (TPP) successively. Due to the high specific surface area of PND, it can efficiently load the model drug doxorubicin hydrochloride (DOX). In addition, PND also has high photothermal conversion efficiency, generating heat to kill cancer cells under near infrared (NIR) laser, realizing the combination of chemotherapy and photothermal therapy (CT-PTT). TPP modification enhanced nanoparticle uptake by colon cancer cells and prolonged preparations retention time at the colon. ES shell protected the drug from being destroyed and prevented the nanoparticles from sticking to the small intestine. Ex vitro fluorescence imaging showed that TPP modification can enhance the residence time of nanoparticles in the colon. In vivo pharmacodynamics demonstrated that CT-PTT group has the greatest inhibitory effect on tumor growth, which means that the nanocarrier has potential clinical value in the in-situ treatment of colon cancer.


Asunto(s)
Neoplasias del Colon , Nanodiamantes , Nanopartículas , Ácidos Polimetacrílicos , Humanos , Fototerapia/métodos , Doxorrubicina/farmacología , Neoplasias del Colon/tratamiento farmacológico , Línea Celular Tumoral
10.
Int J Biol Macromol ; 264(Pt 2): 130729, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38460643

RESUMEN

Astrocyte elevated gene-1 (AEG-1) oncogene is a notorious and evolving target in a variety of human malignancies including osteosarcoma. The RNA interference (RNAi) has been clinically proven to effectively knock down specific genes. To successfully implement RNAi in vivo, protective vectors are required not only to protect unstable siRNAs from degradation, but also to deliver siRNAs to target cells with controlled release. Here, we synthesized a Zein-poly(l-lysine) dendrons non-viral modular system that enables efficient siRNA-targeted AEG-1 gene silencing in osteosarcoma and encapsulation of antitumor drugs for controlled release. The rational design of the ZDP integrates the non-ionic and low immunogenicity of Zein and the positive charge of the poly(l-lysine) dendrons (DPLL) to encapsulate siRNA and doxorubicin (DOX) payloads via electrostatic complexes and achieve pH-controlled release in a lysosomal acidic microenvironment. Nanocomplexes-directed delivery greatly improves siRNA stability, uptake, and AEG-1 sequence-specific knockdown in 143B cells, with transfection efficiencies comparable to those of commercial lipofectamine but with lower cytotoxicity. This AEG-1-focused RNAi therapy supplemented with chemotherapy inhibited, and was effective in inhibiting the growth in of osteosarcoma xenografts mouse models. The combination therapy is an alternative or combinatorial strategy that can produce durable inhibitory responses in osteosarcoma patients.


Asunto(s)
Neoplasias Óseas , Dendrímeros , Nanopartículas , Osteosarcoma , Zeína , Animales , Ratones , Humanos , Polilisina , Azidas , Preparaciones de Acción Retardada , Alquinos , Doxorrubicina/farmacología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , ARN Interferente Pequeño/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Línea Celular Tumoral , Microambiente Tumoral
11.
Molecules ; 29(6)2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38543040

RESUMEN

Doxorubicin is an effective chemotherapeutic agent in the treatment of solid hematological and non-hematological carcinoma. However, its long-term usage could result in side effects, such as cardiomyopathy, chronic heart failure, neurotoxicity and cancer cell resistance. In this study, we reported the sensitivity enhancement of A549 human lung cancer cells on doxorubicin at a low dose (0.1 ppm) in combination with 10-60 ppm of crude and alkaloid extracts derived from the leaves of Kratom (Mitragyna speciosa (Korth.) Havil. Rubiaceae). A549 cancer cell lines were insensitive to the crude extract containing low mitragynine (MG) (4-5%), while these cells were moderately inhibited by the alkaloid extract containing 40-45% MG (IC50 of 48-55 ppm). The alkaloid extract was found to inhibit A549 cancer cells via apoptosis as suggested by the higher relative fluorescence intensity with Annexin compared to that in propidium iodide (PI), i.e., a positive Annexin and a negative PI. The combination of crude extract and doxorubicin sensitized A549 cancer cells to doxorubicin by 1.3 to 2.4 times, while the combination with the alkaloid induced a 2.6- to 3.4-fold increase in sensitivity. The calculated combination index (CI) for doxorubicin with the crude and alkaloid extracts was 0.6 and 0.3, respectively, showing potential synergistic combinations to reduce the level of dosage of doxorubicin used in chemotherapy. In addition, the synergistic enhancement effect of crude extract on the cytotoxic activity of doxorubicin provides insights into the plausibility of non-alkaloids to influence the biological activities of Kratom.


Asunto(s)
Neoplasias Pulmonares , Mitragyna , Alcaloides de Triptamina Secologanina , Humanos , Extractos Vegetales/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inducido químicamente , Doxorrubicina/farmacología , Alcaloides de Triptamina Secologanina/farmacología , Anexinas
12.
Molecules ; 29(5)2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38474556

RESUMEN

Chemotherapy is a well-established method for treating cancer, but it has limited effectiveness due to its high dosage and harmful side effects. To address this issue, researchers have explored the use of photothermal agent nanoparticles as carriers for precise drug release in vivo. In this study, three different sizes of polydopamine nanoparticles (PDA-1, PDA-2, and PDA-3) were synthesized and evaluated. PDA-2 was selected for its optimal size, encapsulation rate, and drug loading rate. The release of the drug from PDA-2@TAX was tested at different pH and NIR laser irradiation levels. The results showed that PDA-2@TAX released more readily in an acidic environment and exhibited a high photothermal conversion efficiency when exposed to an 808 nm laser. In vitro experiments on ovarian cancer cells demonstrated that PDA-2@TAX effectively inhibited cell proliferation, highlighting its potential for synergistic chemotherapy-photothermal treatment.


Asunto(s)
Hipertermia Inducida , Indoles , Nanopartículas , Neoplasias Ováricas , Polímeros , Quercetina/análogos & derivados , Humanos , Femenino , Fototerapia/métodos , Hipertermia Inducida/métodos , Neoplasias Ováricas/tratamiento farmacológico , Doxorrubicina/farmacología
13.
Chem Commun (Camb) ; 60(26): 3535-3538, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38450703

RESUMEN

A Fe(III) intercalated montmorillonite nanoplatform (Fe-MMT) was engineered for doxorubicin (DOX) loading. The constructed Fe-MMT/DOX nanoplatform could not only improve the production of H2O2 to enhance chemodynamic therapy but interfere with DNA damage repair to amplify the efficacy of DOX, proving an ideal combination of chemotherapy and chemodynamic therapy.


Asunto(s)
Compuestos Férricos , Neoplasias , Humanos , Arcilla , Peróxido de Hidrógeno , Doxorrubicina/farmacología , Línea Celular Tumoral
14.
Carbohydr Polym ; 332: 121931, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38431421

RESUMEN

Lumpectomy plus radiation is a treatment option offering better survival than conventional mastectomy for patients with early-stage breast cancer. However, successive radioactive therapy remains tedious and unsafe with severe adverse reactions and secondary injury. Herein, a composite hydrogel with pH- and photothermal double-sensitive activity is developed via physical crosslinking. The composite hydrogel incorporated with tempo-oxidized cellulose nanofiber (TOCN), polyvinyl alcohol (PVA) and a polydopamine (PDA) coating for photothermal therapy (PTT) triggered in situ release of doxorubicin (DOX) drug was utilized to optimize postoperative strategies of malignant tumors inhibition. The incorporation of TOCN significantly affects the performance of composite hydrogels. The best-performing TOCN/PVA7 was selected for drug loading and polydopamine coating by rational design. In vitro studies have demonstrated that the composite hydrogel exhibited high NIR photothermal conversion efficiency, benign cytotoxicity to L929 cells, pH-dependent release profiles, and strong MCF-7 cell inhibitory effects. Then the TOCN/PVA7-PDA@DOX hydrogel is implanted into the tumor resection cavity for local in vivo chemo-photothermal synergistical therapy to ablate residue tumor tissues. Overall, this work suggests that such a chemo-photothermal hydrogel delivery system has great potential as a promising tool for the postsurgical management of breast cancer.


Asunto(s)
Neoplasias de la Mama , Celulosa Oxidada , Hipertermia Inducida , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Terapia Fototérmica , Hidrogeles/química , Fototerapia , Mastectomía , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Concentración de Iones de Hidrógeno
15.
Phytomedicine ; 127: 155473, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38422972

RESUMEN

BACKGROUND: Doxorubicin (DOX) is widely used for the treatment of a variety of cancers. However, its clinical application is limited by dose-dependent cardiotoxicity. Recent findings demonstrated that autophagy inhibition and apoptosis of cardiomyocytes induced by oxidative stress dominate the pathophysiology of DOX-induced cardiotoxicity (DIC), however, there are no potential molecules targeting on these. PURPOSE: This study aimed to explore whether aucubin (AU) acting on inimitable crosstalk between NRF2 and HIPK2 mediated the autophagy, oxidative stress, and apoptosis in DIC, and provide a new and alternative strategy for the treatment of DIC. METHODS AND RESULTS: We first demonstrated the protection of AU on cardiac structure and function in DIC mice manifested by increased EF and FS values, decreased serum CK-MB and LDH contents and well-aligned cardiac tissue in HE staining. Furthermore, AU alleviated DOX-induced myocardial oxidative stress, mitochondrial damage, apoptosis, and autophagy flux dysregulation in mice, as measured by decreased ROS, 8-OHdG, and TUNEL-positive cells in myocardial tissue, increased SOD and decreased MDA in serum, aligned mitochondria with reduced vacuoles, and increased autophagosomes. In vitro, AU alleviated DOX-induced oxidative stress, autophagy inhibition, and apoptosis by promoting NRF2 and HIPK2 expression. We also identified crosstalk between NRF2 and HIPK2 in DIC as documented by overexpression of NRF2 or HIPK2 reversed cellular oxidative stress, autophagy blocking, and apoptosis aggravated by HIPK2 or NRF2 siRNA, respectively. Simultaneously, AU promoted the expression and nuclear localization of NRF2 protein, which was reversed by HIPK2 siRNA, and AU raised the expression of HIPK2 protein as well, which was reversed by NRF2 siRNA. Crucially, AU did not affect the antitumor activity of DOX against MCF-7 and HepG2 cells, which made up for the shortcomings of previous anti-DIC drugs. CONCLUSION: These collective results innovatively documented that AU regulated the unique crosstalk between NRF2 and HIPK2 to coordinate oxidative stress, autophagy, and apoptosis against DIC without compromising the anti-tumor effect of DOX in vitro.


Asunto(s)
Cardiotoxicidad , Glucósidos Iridoides , Factor 2 Relacionado con NF-E2 , Ratones , Animales , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Doxorrubicina/farmacología , Miocitos Cardíacos , Apoptosis , Estrés Oxidativo , ARN Interferente Pequeño/farmacología , Autofagia
16.
ACS Appl Mater Interfaces ; 16(10): 12289-12301, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38418381

RESUMEN

The intricate tumor microenvironment (TME) always brings about unsatisfactory therapeutic effects for treatments, although nanomedicines have been demonstrated to be highly beneficial for synergistic therapies to avoid the side effects caused by the complexity and heterogeneity of cancer. Developing nanotheranostics with the functionalities of both synergistic therapies and TME regulation is a good strategy but is still in its infancy. Herein, an "all-in-one" nanoplatform for integrated diagnosis and treatment, namely, Carrier@ICG@DOX@FA (CIDF), is constructed. Benefiting from the bimetallic coordination of Eu3+-HTHA (4,4,4-trifluoro-1-(9-hexylcarbazol-3-yl)-1,3-butanedione) and Fe3+ with the ligands in UiO-67, CIDF can simultaneously achieve two-photon fluorescence imaging, fluorescent lifetime imaging in deep tumors, and regulation of TME. Owing to its porosity, CIDF can encapsulate indocyanine green as photosensitizers and doxorubicin as chemotherapeutic agent, further realizing light-controlled drug release. Moreover, CIDF exhibited good biocompatibility and tumor targeting by coating with folic-acid-modified polymers. Both in vitro and in vivo experiments demonstrate the excellent therapeutic efficacy of CIDF through dual-modal-imaging-guided synergistic photothermal-, photodynamic-, and chemotherapy. CIDF provides a new paradigm for the construction of TME-regulated synergistic nanotheranostics and realizes the complete elimination of tumors without recurrence.


Asunto(s)
Nanopartículas , Fototerapia , Fototerapia/métodos , Línea Celular Tumoral , Microambiente Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Verde de Indocianina , Imagen Óptica
17.
Phytomedicine ; 126: 155460, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38394731

RESUMEN

BACKGROUND: Multidrug resistance is the major obstacle to cancer chemotherapy. Modulation of P-glycoprotein and drug combination approaches have been considered important strategies to overcome drug resistance. PURPOSE: Aiming at generating a small library of Amaryllidaceae-type alkaloids to overcome drug resistance, two major alkaloids, isolated from Pancratium maritimum, lycorine (1), and 2α-10bα-dihydroxy-9-O-demethylhomolycorine (2), were derivatized, giving rise to nineteen derivatives (3 - 21). METHODS: The main chemical transformation of lycorine resulted from the cleavage of ring E of the diacetylated lycorine derivative (3) to obtain compounds that have carbamate and amine functions (5 - 16), while acylation of compound 2 provided derivatives 17 - 21. Compounds 1 - 21 were evaluated for their effects on cytotoxicity, and drug resistance reversal, using resistant human ovarian carcinoma cells (HOC/ADR), overexpressing P-glycoprotein (P-gp/ABCB1), as model. RESULTS: Excluding lycorine (1) (IC50 values of 1.2- 2.5 µM), the compounds were not cytotoxic or showed moderate/weak cytotoxicity. Chemo-sensitization assays were performed by studying the in vitro interaction between the compounds and the anticancer drug doxorubicin. Most of the compounds have shown synergistic interactions with doxorubicin. Compounds 5, 6, 9 - 14, bearing both carbamate and aromatic amine moieties, were found to have the highest sensitization rate, reducing the dose of doxorubicin 5-35 times, highlighting their potential to reverse drug resistance in combination chemotherapy. Selected compounds (4 - 6, 9 - 14, and 21), able of re-sensitizing resistant cancer cells, were further evaluated as P-gp inhibitors. Compound 11, which has a para­methoxy-N-methylbenzylamine moiety, was the strongest inhibitor. In the ATPase assay, compounds 9-11 and 13 behaved as verapamil, suggesting competitive inhibition of P-gp. At the same time, none of these compounds affected P-gp expression at the mRNA or protein level. CONCLUSIONS: This study provided evidence of the potential of Amaryllidaceae alkaloids as lead candidates for the development of MDR reversal agents.


Asunto(s)
Adenocarcinoma , Alcaloides , Alcaloides de Amaryllidaceae , Antineoplásicos , Fenantridinas , Humanos , Alcaloides de Amaryllidaceae/farmacología , Resistencia a Antineoplásicos , Antineoplásicos/farmacología , Doxorrubicina/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Alcaloides/farmacología , Carbamatos/farmacología , Línea Celular Tumoral
18.
ACS Appl Bio Mater ; 7(3): 1569-1578, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38349029

RESUMEN

The therapeutic efficacy of bone tumor treatment is primarily limited by inadequate tumor resection, resulting in recurrence and metastasis, as well as the deep location of tumors. Herein, an injectable doxorubicin (DOX)-loaded magnetic alginate hydrogel (DOX@MAH) was developed to evaluate the efficacy of an alternating magnetic field (AMF)-responsive, chemothermal synergistic therapy for multimodality treatment of bone tumors. The prepared hydrogel exhibits a superior drug-loading capacity and a continuous DOX release. This multifunctionality can be attributed to the combined use of DOX for chemotherapy and iron oxide nanoparticle-containing alginate hydrogels as magnetic hyperthermia agents to generate hyperthermia for tumor elimination without the limit on penetration depth. Moreover, the hydrogel can be formed when in contact with the calcium ions, which are abundant in bone tissues; therefore, this hydrogel could perfectly fit the bone defects caused by the surgical removal of the bone tumor tissue, and the hydrogel could tightly attach the surgical margin of the bone to realize a high efficacy residual tumor tissue elimination treated by chemothermal synergistic therapy. The hydrogel demonstrates excellent hyperthermia performance, as evidenced by in vitro cytotoxicity tests on tumor cells. These tests reveal that the combined therapy based on DOX@MAH under AMF significantly induces cell death compared to single magnetic hyperthermia or chemotherapy. In vivo antitumor effects in tumor-bearing mice demonstrate that DOX@MAH injection at the tumor site effectively inhibits tumor growth and leads to tumor necrosis. This work not only establishes an effective DOX@MAH system as a synergistic chemothermal therapy platform for treating bone tumors but also sheds light on the application of alginate to combine calcium ions of the bone to treat bone defect diseases.


Asunto(s)
Neoplasias Óseas , Hipertermia Inducida , Animales , Ratones , Hidrogeles/farmacología , Calcio , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Hipertermia , Hipertermia Inducida/métodos , Alginatos , Iones , Fenómenos Magnéticos
19.
J Colloid Interface Sci ; 663: 436-448, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38417295

RESUMEN

Phototherapy has garnered increasing attention as it allows for precise treatment of tumor sites with its accurate spatiotemporal control. In this study, we have successfully synthesized degradable bifunctional phototherapy agents (UCNPs@mSiO2@MPN-MC540/DOX) based on upconversion nanoparticle (UCNPs) and metal phenolic network (MPN), serving as a novel nanoplatform for multimodal tumor treatment in the near-infrared (NIR) biological window. To address the issue of low light penetration depth, the UCNPs we synthesized exhibited efficient light conversion ability under 808 nm laser irradiation to activate the photosensitizer Merocyanine 540 (MC540) for photodynamic therapy. Simultaneously, the 808 nm NIR light can also excite the MPN layer to achieve photothermal therapy for tumors. Additionally, the MPN layer possesses the capability of self-degradation under weakly acidic conditions. Within the tumor microenvironment, the MPN layer gradually degrades, facilitating the controlled release of the chemotherapy drug doxorubicin (DOX), thus achieving pH-responsive drug release and reducing the side effects of chemotherapy. This study provides an example of NIR-excited multimodal tumor treatment and pH-responsive drug release, offering a therapy model for precise tumor therapy.


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fototerapia , Neoplasias/tratamiento farmacológico , Terapia Combinada , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Metales , Línea Celular Tumoral , Microambiente Tumoral
20.
Biomaterials ; 307: 122511, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38401482

RESUMEN

Combination of different therapies is an attractive approach for cancer therapy. However, it is a challenge to synchronize different therapies for maximization of therapeutic effects. In this work, a smart composite scaffold that could synchronize magnetic hyperthermia and chemotherapy was prepared by hybridization of magnetic Fe3O4 nanoparticles and doxorubicin (Dox)-loaded thermosensitive liposomes with biodegradable polymers. Irradiation of alternating magnetic field (AMF) could not only increase the scaffold temperature for magnetic hyperthermia but also trigger the release of Dox for chemotherapy. The two functions of magnetic hyperthermia and chemotherapy were synchronized by switching AMF on and off. The synergistic anticancer effects of the composite scaffold were confirmed by in vitro cell culture and in vivo animal experiments. The composite scaffold could efficiently eliminate breast cancer cells under AMF irradiation. Moreover, the scaffold could support proliferation and adipogenic differentiation of mesenchymal stem cells for adipose tissue reconstruction after anticancer treatment. In vivo regeneration experiments showed that the composite scaffolds could effectively maintain their structural integrity and facilitate the infiltration and proliferation of normal cells within the scaffolds. The composite scaffold possesses multi-functions and is attractive as a novel platform for efficient breast cancer therapy.


Asunto(s)
Doxorrubicina/análogos & derivados , Hipertermia Inducida , Neoplasias , Animales , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Hipertermia , Fenómenos Magnéticos , Polietilenglicoles
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA