Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 18.541
Filtrer
1.
ACS Appl Bio Mater ; 7(8): 5359-5368, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39102354

RÉSUMÉ

We have studied the endocytic mechanisms that determine subcellular localization for three carrier-free chemotherapeutic-photothermal (chemo-PTT) combination ionic nanomedicines (INMs) composed of doxorubicin (DOX) and an near-infrared (NIR) dye (ICG, IR820, or IR783). This study aims to understand the cellular basis for previously published enhanced toxicity results of these combination nanomedicines toward MCF-7 breast cancer cells. The active transport mechanism of INMs, unlike free DOX, which is known to employ passive transport, was validated by conducting temperature-dependent cellular uptake of the drug in MCF-7 cells using confocal microscopy. The internalization pathway of these INMs was further probed in the presence and absence of different endocytosis inhibitors. Detailed examination of the mode of entry of the carrier-free INMs in MCF-7 cells revealed that they are primarily internalized through clathrin-mediated endocytosis. In addition, time-dependent subcellular localization studies were also investigated. Examination of time-dependent confocal images indicated that the INMs targeted multiple organelles, in contrast to free DOX that primarily targets the nucleus. Collectively, the high cellular endocytic uptake in cancerous cells (EPR effect) and the multimode targeting ability demonstrated the main reason for the low half-maxima inhibitory concentration (IC50) value (the high cytotoxicity) of these carrier-free INMs as compared to their respective parent chemo and PTT drugs.


Sujet(s)
Doxorubicine , Endocytose , Nanomédecine , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Humains , Endocytose/effets des médicaments et des substances chimiques , Cellules MCF-7 , Taille de particule , Organites/métabolisme , Organites/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Test de matériaux , Tests de criblage d'agents antitumoraux , Survie cellulaire/effets des médicaments et des substances chimiques , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/composition chimique , Ions/composition chimique
2.
J Nanobiotechnology ; 22(1): 461, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090622

RÉSUMÉ

BACKGROUND: The combination of programmed cell death ligand-1 (PD-L1) immune checkpoint blockade (ICB) and immunogenic cell death (ICD)-inducing chemotherapy has shown promise in cancer immunotherapy. However, triple-negative breast cancer (TNBC) patients undergoing this treatment often face obstacles such as systemic toxicity and low response rates, primarily attributed to the immunosuppressive tumor microenvironment (TME). METHODS AND RESULTS: In this study, PD-L1-targeted theranostic systems were developed utilizing anti-PD-L1 peptide (APP) conjugated with a bio-orthogonal click chemistry group. Initially, TNBC was treated with azide-modified sugar to introduce azide groups onto tumor cell surfaces through metabolic glycoengineering. A PD-L1-targeted probe was developed to evaluate the PD-L1 status of TNBC using magnetic resonance/near-infrared fluorescence imaging. Subsequently, an acidic pH-responsive prodrug was employed to enhance tumor accumulation via bio-orthogonal click chemistry, which enhances PD-L1-targeted ICB, the pH-responsive DOX release and induction of pyroptosis-mediated ICD of TNBC. Combined PD-L1-targeted chemo-immunotherapy effectively reversed the immune-tolerant TME and elicited robust tumor-specific immune responses, resulting in significant inhibition of tumor progression. CONCLUSIONS: Our study has successfully engineered a bio-orthogonal multifunctional theranostic system, which employs bio-orthogonal click chemistry in conjunction with a PD-L1 targeting strategy. This innovative approach has been demonstrated to exhibit significant promise for both the targeted imaging and therapeutic intervention of TNBC.


Sujet(s)
Antigène CD274 , Chimie click , Immunothérapie , Pyroptose , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/traitement médicamenteux , Antigène CD274/métabolisme , Animaux , Femelle , Immunothérapie/méthodes , Souris , Pyroptose/effets des médicaments et des substances chimiques , Humains , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/usage thérapeutique , Imagerie optique/méthodes , Promédicaments/composition chimique , Promédicaments/pharmacologie
3.
Sci Rep ; 14(1): 18181, 2024 08 06.
Article de Anglais | MEDLINE | ID: mdl-39107323

RÉSUMÉ

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks an actionable target with limited treatment options beyond conventional chemotherapy. Therapeutic failure is often encountered due to inherent or acquired resistance to chemotherapy. Previous studies implicated PI3K/Akt/mTOR signaling pathway in cancer stem cells (CSCs) enrichment and hence chemoresistance. The present study aimed at investigating the potential effect of piperine (PIP), an amide alkaloid isolated from Piper nigrum, on enhancing the sensitivity of TNBC cells to doxorubicin (DOX) in vitro on MDA-MB-231 cell line and in vivo in an animal model of Ehrlich ascites carcinoma solid tumor. Results showed a synergistic interaction between DOX and PIP on MDA-MB-231 cells. In addition, the combination elicited enhanced suppression of PI3K/Akt/mTOR signaling that paralleled an upregulation in this pathway's negative regulator, PTEN, along with a curtailment in the levels of the CSCs surrogate marker, aldehyde dehydrogenase-1 (ALDH-1). Meanwhile, in vivo investigations demonstrated the potential of the combination regimen to enhance necrosis while downregulating PTEN and curbing PI3K levels as well as p-Akt, mTOR, and ALDH-1 immunoreactivities. Notably, the combination failed to change cleaved poly-ADP ribose polymerase levels suggesting a pro-necrotic rather than pro-apoptotic mechanism. Overall, these findings suggest a potential role of PIP in decreasing the resistance to DOX in vitro and in vivo, likely by interfering with the PI3K/Akt/mTOR pathway and CSCs.


Sujet(s)
Alcaloïdes , Benzodioxoles , Doxorubicine , Cellules souches tumorales , Phosphatidylinositol 3-kinases , Pipéridines , Amides gras polyinsaturés N-alkylés , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Tumeurs du sein triple-négatives , Doxorubicine/pharmacologie , Amides gras polyinsaturés N-alkylés/pharmacologie , Pipéridines/pharmacologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Humains , Alcaloïdes/pharmacologie , Benzodioxoles/pharmacologie , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Femelle , Lignée cellulaire tumorale , Sérine-thréonine kinases TOR/métabolisme , Synergie des médicaments , Souris , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
4.
J Am Chem Soc ; 146(32): 22675-22688, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39088029

RÉSUMÉ

Redox-responsive homodimer prodrug nanoassemblies (RHPNs) have emerged as a significant technology for overcoming chemotherapeutical limitations due to their high drug-loading capacity, low excipient-associated toxicity, and straightforward preparation method. Previous studies indicated that α-position disulfide bond bridged RHPNs exhibited rapid drug release rates but unsatisfactory assembly stability. In contrast, γ-disulfide bond bridged RHPNs showed better assembly stability but low drug release rates. Therefore, designing chemical linkages that ensure both stable assembly and rapid drug release remains challenging. To address this paradox of stable assembly and rapid drug release in RHPNs, we developed carbon-spaced double-disulfide bond (CSDD)-bridged RHPNs (CSDD-RHPNs) with two carbon-spaces. Pilot studies showed that CSDD-RHPNs with two carbon-spaces exhibited enhanced assembly stability, reduction-responsive drug release, and improved selective toxicity compared to α-/γ-position single disulfide bond bridged RHPNs. Based on these findings, CSDD-RHPNs with four and six carbon-spaces were designed to further investigate the properties of CSDD-RHPNs. These CSDD-RHPNs exhibited excellent assembly ability, safety, and prolonged circulation. Particularly, CSDD-RHPNs with two carbon-spaces displayed the best antitumor efficacy on 4T1 and B16-F10 tumor-bearing mice. CSDD chemical linkages offer novel perspectives on the rational design of RHPNs, potentially overcoming the design limitations regarding contradictory assembly ability and drug release rate.


Sujet(s)
Carbone , Disulfures , Promédicaments , Disulfures/composition chimique , Promédicaments/composition chimique , Animaux , Souris , Carbone/composition chimique , Humains , Libération de médicament , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Conception de médicament , Lignée cellulaire tumorale , Nanostructures/composition chimique , Dimérisation , Doxorubicine/composition chimique , Doxorubicine/pharmacologie
5.
Sci Rep ; 14(1): 18311, 2024 08 07.
Article de Anglais | MEDLINE | ID: mdl-39112669

RÉSUMÉ

Finding a novel drug delivery system (DDS) represents one of the most challenging endeavors in cancer therapy. Hence, in this study, we developed a new biocompatible and biodegradable zinc-based nanoscale metal-organic framework (Zn-NMOF) coated with folic acid (FA) functionalized chitosan (CS) to facilitate targeted delivery of doxorubicin (D), a standard chemotherapeutic agent, into breast cancer cells. The synthesis of the NMOF-CS-FA-D nanocomposite preceded its comprehensive characterization via FT-IR, DLS, XRD, SEM, and TEM analyses. Subsequent in vitro studies were conducted on MCF-7 breast cancer cells and HFF-1 normal cells, encompassing assessments of cell viability, expression levels of apoptotic and autophagy genes, cell cycle arrest, and apoptotic analyses. The size of the NMOF-CS-FA-D particles was determined to be less than 80 nm, with a drug loading efficiency of 72 ± 5%. The release kinetics of DOX from the nanocomposite were investigated, revealing controlled release behavior at pH 7.4 and accelerated release at pH 5.0, which is conducive to drug delivery into cancer cells. In vitro results indicated a 17.39% ± 6.34 cell viability after 24 h of treatment with a 40 nM concentration of the NMOF-CS-FA-D nanocomposite. Furthermore, the expression levels of Caspase-9 and BAX, key apoptotic genes, along with BECLIN1, an autophagy gene, were found to increase by two-fold, four-fold, and two-fold, respectively, following 5 h of treatment with the nanocomposite. Additionally, analysis of cell cycle distribution revealed 15.4 ± 2% of cells in the sub-G1 phase, indicative of apoptotic cells, and 31.9% of cells undergoing early and late apoptosis in MCF-7 cells. Collectively, these findings underscore the potential of the NMOF-CS-FA-D nanocomposite in inhibiting cancer cell proliferation with low side effects.


Sujet(s)
Apoptose , Tumeurs du sein , Chitosane , Doxorubicine , Réseaux organométalliques , Nanocomposites , Zinc , Humains , Nanocomposites/composition chimique , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Cellules MCF-7 , Zinc/composition chimique , Zinc/pharmacologie , Chitosane/composition chimique , Femelle , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Acide folique/composition chimique , Acide folique/pharmacologie , Systèmes de délivrance de médicaments , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Libération de médicament , Vecteurs de médicaments/composition chimique , Caspase-9/métabolisme , Caspase-9/génétique , Autophagie/effets des médicaments et des substances chimiques
6.
Sci Rep ; 14(1): 18544, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39122867

RÉSUMÉ

Breast cancer is one of cancer's most deadly varieties. Its variability makes the development of personalized therapies very difficult. Therefore, improvement of classic chemotherapy is still one of the important challenges of cancer research. We addressed this issue applying nanotechnology to verify the influence of silver nanoparticles (AgNPs) on doxorubicin (DOX) anticancer activity and assess if the size of AgNPs affects their interactions with DOX. We employed a broad spectrum of biophysical methods, characterizing 5 and 50 nm AgNPs interactions with DOX using UV-Vis spectroscopy, dynamic light scattering, fluorescence spectroscopy, and atomic force microscopy imaging. Biological effects of observed AgNPs-DOX interactions were assessed utilizing MTT and 3D Matrigel assays on SKBR3 and MDA-MB-231 breast cancer cell lines. Obtained results indicate direct interactions between AgNPs and DOX. Furthermore, AgNPs size influences their interactions with DOX, as evidenced by differences in the heteroaggregates formation observed in biophysical experiments and further supported by in vitro biological assays. We detected reduction of tumor cell viability and/or colony sizes of the analyzed cancer cell lines, registering differences linked to the observed phenomenon. However, the effects may be limited to the outer borders of the tumor microenvironment as evidenced by the 3D model. Summing up, we observed diverse patterns of interactions and biological effects for different sizes of AgNPs with DOX providing insight how the nanoparticles' size affects their interactions with other biologically active compounds. Moreover, obtained data can be further used in experiments on the reduction of tumor size i.e. before the surgical intervention.


Sujet(s)
Doxorubicine , Nanoparticules métalliques , Taille de particule , Argent , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Humains , Argent/composition chimique , Argent/pharmacologie , Nanoparticules métalliques/composition chimique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie
7.
Int J Nanomedicine ; 19: 8071-8090, 2024.
Article de Anglais | MEDLINE | ID: mdl-39130685

RÉSUMÉ

Purpose: Chemotherapy mediated by Reactive oxygen species (ROS)-responsive drug delivery systems can potentially mitigate the toxic side effects of chemotherapeutic drugs and significantly enhance their therapeutic efficacy. However, achieving precise targeted drug delivery and real-time control of ROS-responsive drug release at tumor sites remains a formidable challenge. Therefore, this study aimed to describe a ROS-responsive drug delivery system with specific tumor targeting capabilities for mitigating chemotherapy-induced toxicity while enhancing therapeutic efficacy under guidance of Fluorescence (FL) and Magnetic resonance (MR) bimodal imaging. Methods: Indocyanine green (ICG), Doxorubicin (DOX) prodrug pB-DOX and Superparamagnetic iron oxide (SPIO, Fe3O4) were encapsulated in poly(lactic-co-glycolic acid) (PLGA) by double emulsification method to prepare ICG/ pB-DOX/ Fe3O4/ PLGA nanoparticles (IBFP NPs). The surface of IBFP NPs was functionalized with mammaglobin antibodies (mAbs) by carbodiimide method to construct the breast cancer-targeting mAbs/ IBFP NPs (MIBFP NPs). Thereafter, FL and MR bimodal imaging ability of MIBFP NPs was evaluated in vitro and in vivo. Finally, the combined photodynamic therapy (PDT) and chemotherapy efficacy evaluation based on MIBFP NPs was studied. Results: The multifunctional MIBFP NPs exhibited significant targeting efficacy for breast cancer. FL and MR bimodal imaging clearly displayed the distribution of the targeting MIBFP NPs in vivo. Upon near-infrared laser irradiation, the MIBFP NPs loaded with ICG effectively generated ROS for PDT, enabling precise tumor ablation. Simultaneously, it triggered activation of the pB-DOX by cleaving its sensitive moiety, thereby restoring DOX activity and achieving ROS-responsive targeted chemotherapy. Furthermore, the MIBFP NPs combined PDT and chemotherapy to enhance the efficiency of tumor ablation under guidance of bimodal imaging. Conclusion: MIBFP NPs constitute a novel dual-modality imaging-guided drug delivery system for targeted breast cancer therapy and offer precise and controlled combined treatment options.


Sujet(s)
Tumeurs du sein , Doxorubicine , Vert indocyanine , Imagerie par résonance magnétique , Photothérapie dynamique , Copolymère d'acide poly(lactique-co-glycolique) , Espèces réactives de l'oxygène , Vert indocyanine/composition chimique , Vert indocyanine/pharmacocinétique , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/administration et posologie , Doxorubicine/pharmacocinétique , Espèces réactives de l'oxygène/métabolisme , Animaux , Femelle , Humains , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Photothérapie dynamique/méthodes , Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Imagerie par résonance magnétique/méthodes , Souris , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments/méthodes , Promédicaments/composition chimique , Promédicaments/pharmacocinétique , Promédicaments/pharmacologie , Souris de lignée BALB C , Nanoparticules magnétiques d'oxyde de fer/composition chimique , Souris nude , Nanoparticules de magnétite/composition chimique , Libération de médicament , Nanoparticules/composition chimique , Imagerie optique/méthodes
8.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126001

RÉSUMÉ

Breast cancer is the most diagnosed type of cancer worldwide and the second cause of death in women. Triple-negative breast cancer (TNBC) is the most aggressive, and due to the lack of specific targets, it is considered the most challenging subtype to treat and the subtype with the worst prognosis. The present study aims to determine the antitumor effect of beta-D-glucose-reduced silver nanoparticles (AgNPs-G) in a murine model of TNBC, as well as to study its effect on the tumor microenvironment. In an airbag model with 4T1 tumor cell implantation, the administration of AgNPs-G or doxorubicin showed antitumoral activity. Using immunohistochemistry it was demonstrated that treatment with AgNPs-G decreased the expression of PCNA, IDO, and GAL-3 and increased the expression of Caspase-3. In the tumor microenvironment, the treatment increased the percentage of memory T cells and innate effector cells and decreased CD4+ cells and regulatory T cells. There was also an increase in the levels of TNF-α, IFN-γ, and IL-6, while TNF-α was increased in serum. In conclusion, we suggest that AgNPs-G treatment has an antitumor effect that is demonstrated by its ability to remodel the tumor microenvironment in mice with TNBC.


Sujet(s)
Glucose , Nanoparticules métalliques , Argent , Tumeurs du sein triple-négatives , Microenvironnement tumoral , Animaux , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Argent/composition chimique , Nanoparticules métalliques/composition chimique , Femelle , Souris , Glucose/métabolisme , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Souris de lignée BALB C , Doxorubicine/pharmacologie , Humains
9.
Cancer Med ; 13(15): e70079, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39118454

RÉSUMÉ

BACKGROUND: Cancer remains a formidable global health challenge, currently affecting nearly 20 million individuals worldwide. Due to the absence of universally effective treatments, ongoing research explores diverse strategies to combat this disease. Recent efforts have concentrated on developing combined drug regimens and targeted therapeutic approaches. OBJECTIVE: This study aimed to investigate the anticancer efficacy of a conjugated drug system, consisting of doxorubicin and cisplatin (Dox-Cis), encapsulated within niosomes and modified with MUC-1 aptamers to enhance biocompatibility and target specific cancer cells. METHODS: The chemical structure of the Dox-Cis conjugate was characterized using Fourier Transform Infrared Spectroscopy (FTIR) and Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-Q-TOF/MS). The zeta potential and morphological parameters of the niosomal vesicles were determined through Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM). In vitro assessments of cell viability and apoptosis were conducted on MUC-1 positive HeLa cells and MUC-1 negative U87 cells. RESULTS: The findings confirmed the successful conjugation of Dox and Cis within the niosomes. The Nio/Dox-Cis/MUC-1 formulation demonstrated enhanced efficacy compared to the individual drugs and their unencapsulated combination in both cell lines. Notably, the Nio/Dox-Cis/MUC-1 formulation exhibited greater effectiveness on HeLa cells (38.503 ± 1.407) than on U87 cells (46.653 ± 1.297). CONCLUSION: The study underscores the potential of the Dox-Cis conjugate as a promising strategy for cancer treatment, particularly through platforms that facilitate targeted drug delivery to cancer cells. This targeted approach could lead to more effective and personalized cancer therapies.


Sujet(s)
Aptamères nucléotidiques , Survie cellulaire , Cisplatine , Doxorubicine , Liposomes , Mucine-1 , Humains , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Mucine-1/métabolisme , Mucine-1/composition chimique , Liposomes/composition chimique , Cisplatine/pharmacologie , Cisplatine/composition chimique , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cellules HeLa , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Préparation de médicament/méthodes
10.
Bull Exp Biol Med ; 177(2): 266-270, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39093476

RÉSUMÉ

The efficiency of combinations of cytostatics cisplatin and adriamycin with antioxidant sodium 3-(3'-tert-butyl-4-hydroxyphenyl)propyl thiosulfate (TS-13), and nitric oxide (NO) donor NaNO2 was evaluated on two drug-resistant strains of leukemia P388 with changed redox-status of cells. Simultaneous use of both NO donor and TS-13 in combinations with the cytostatics did not increase the efficiency of therapy. In addition, antioxidant activity of TS-13, NaNO2, and their combinations was studied by the method of luminol-dependent chemiluminescence on the model systems with the use of the homogenized cells of sensitive strain and two drug-resistant strains of leukemia P388. It was shown that TS-13 and NO donor produced opposite effects: TS-13 decreased, while NO donor increased the content of free radicals in the model system. Combinations of antioxidant TS-13 and NO donor should be used with consideration for the redox-status of tumor treated.


Sujet(s)
Antioxydants , Cisplatine , Doxorubicine , Résistance aux médicaments antinéoplasiques , Leucémie P388 , Donneur d'oxyde nitrique , Oxydoréduction , Animaux , Souris , Oxydoréduction/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Doxorubicine/pharmacologie , Leucémie P388/traitement médicamenteux , Leucémie P388/anatomopathologie , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Donneur d'oxyde nitrique/pharmacologie , Thiosulfates/pharmacologie , Nitrite de sodium/pharmacologie , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/pharmacologie
11.
Int J Nanomedicine ; 19: 7799-7816, 2024.
Article de Anglais | MEDLINE | ID: mdl-39099794

RÉSUMÉ

Background: At present, the few photothermal/chemotherapy studies about retinoblastoma that have been reported are mainly restricted to ectopic models involving subcutaneous implantation. However, eyeball is unique physiological structure, the blood-retina barrier (BRB) hinders the absorption of drug molecules through the systemic route. Moreover, the abundant blood circulation in the fundus accelerates drug metabolism. To uphold the required drug concentration, patients must undergo frequent chemotherapy sessions. Purpose: To address these challenges above, we need to develop a secure and effective drug delivery system (FA-PEG-PDA-DOX) for the fundus. Methods: We offered superior therapeutic efficacy with minimal or no side effects and successfully established orthotopic mouse models. We evaluated cellular uptake performance and targeting efficiency of FA-PEG-PDA-DOX nanosystem and assessed its synergistic antitumor effects in vitro and vivo. Biodistribution assessments were performed to determine the retention time and targeting efficiency of the NPs in vivo. Additionally, safety assessments were conducted. Results: Cell endocytosis rates of the FA-PEG-PDA-DOX+Laser group became 5.23 times that of the DOX group and 2.28 times that of FA-PEG-PDA-DOX group without irradiation. The fluorescence signal of FA-PEG-PDA-DOX persisted for more than 120 hours at the tumor site. The number of tumor cells (17.2%) in the proliferative cycle decreased by 61.6% in the photothermal-chemotherapy group, in contrast to that of the saline control group (78.8%). FA-PEG-PDA-DOX nanoparticles(NPs) exhibited favorable biosafety and high biocompatibility. Conclusion: The dual functional targeted nanosystem, with the effects of DOX and mild-temperature elevation by irradiation, resulted in precise chemo/photothermal therapy in nude mice model.


Sujet(s)
Doxorubicine , Indoles , Thérapie photothermique , Polymères , Rétinoblastome , Animaux , Rétinoblastome/thérapie , Doxorubicine/composition chimique , Doxorubicine/pharmacocinétique , Doxorubicine/pharmacologie , Doxorubicine/administration et posologie , Souris , Thérapie photothermique/méthodes , Humains , Indoles/composition chimique , Indoles/pharmacocinétique , Indoles/pharmacologie , Lignée cellulaire tumorale , Polymères/composition chimique , Distribution tissulaire , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacocinétique , Souris nude , Nanoparticules/composition chimique , Systèmes de délivrance de médicaments/méthodes , Tumeurs de la rétine/thérapie , Tumeurs de la rétine/traitement médicamenteux , Souris de lignée BALB C , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/composition chimique , Antibiotiques antinéoplasiques/pharmacocinétique , Antibiotiques antinéoplasiques/administration et posologie , Modèles animaux de maladie humaine , Tests d'activité antitumorale sur modèle de xénogreffe , Système d'administration de médicaments à base de nanoparticules/composition chimique , Système d'administration de médicaments à base de nanoparticules/pharmacocinétique
12.
Sensors (Basel) ; 24(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39123900

RÉSUMÉ

Nanotechnology has ushered in significant advancements in drug design, revolutionizing the prevention, diagnosis, and treatment of various diseases. The strategic utilization of nanotechnology to enhance drug loading, delivery, and release has garnered increasing attention, leveraging the enhanced physical and chemical properties offered by these systems. Polyamidoamine (PAMAM) dendrimers have been pivotal in drug delivery, yet there is room for further enhancement. In this study, we conjugated PAMAM dendrimers with chitosan (CS) to augment cellular internalization in tumor cells. Specifically, doxorubicin (DOX) was initially loaded into PAMAM dendrimers to form DOX-loaded PAMAM (DOX@PAMAM) complexes via intermolecular forces. Subsequently, CS was linked onto the DOX-loaded PAMAM dendrimers to yield CS-conjugated PAMAM loaded with DOX (DOX@CS@PAMAM) through glutaraldehyde crosslinking via the Schiff base reaction. The resultant DOX@CS@PAMAM complexes were comprehensively characterized using Fourier-transform infrared (FTIR) spectroscopy, transmission electron microscopy (TEM), and dynamic light scattering (DLS). Notably, while the drug release profile of DOX@CS@PAMAM in acidic environments was inferior to that of DOX@PAMAM, DOX@CS@PAMAM demonstrated effective acid-responsive drug release, with a cumulative release of 70% within 25 h attributed to the imine linkage. Most importantly, DOX@CS@PAMAM exhibited significant selective cellular internalization rates and antitumor efficacy compared to DOX@PAMAM, as validated through cell viability assays, fluorescence imaging, and flow cytometry analysis. In summary, DOX@CS@PAMAM demonstrated superior antitumor effects compared to unconjugated PAMAM dendrimers, thereby broadening the scope of dendrimer-based nanomedicines with enhanced therapeutic efficacy and promising applications in cancer therapy.


Sujet(s)
Chitosane , Dendrimères , Doxorubicine , Dendrimères/composition chimique , Chitosane/composition chimique , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Polyamines/composition chimique , Vecteurs de médicaments/composition chimique , Libération de médicament , Survie cellulaire/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments/méthodes , Lignée cellulaire tumorale
13.
Molecules ; 29(15)2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39124936

RÉSUMÉ

Multidrug resistance is a serious problem in modern medicine and the reason for the failure of various therapies. A particularly important problem is the occurrence of multidrug resistance in cancer therapies which affects many cancer patients. Observations on the effect of metformin-a well-known hypoglycemic drug used in the treatment of type 2 diabetes-on cancer cells indicate the possibility of an interaction of this substance with drugs already used and, as a result, an increase in the sensitivity of cancer cells to cytostatics. The aim of this study was to evaluate the effect of metformin on the occurrence of multidrug resistance of breast cancer cells. The MCF-7-sensitive cell line and the MCF-7/DX cytostatic-resistant cell line were used for this study. WST-1 and LDH assays were used to evaluate the effects of metformin and doxorubicin on cell proliferation and viability. The effect of metformin on increasing the sensitivity of MCF-7 and MCF-7/DX cells to doxorubicin was evaluated in an MDR test. The participation of metformin in increasing the sensitivity of resistant cells to the effect of the cytostatic (doxorubicin) has been demonstrated.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Cytostatiques , Doxorubicine , Résistance aux médicaments antinéoplasiques , Metformine , Humains , Metformine/pharmacologie , Cellules MCF-7 , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytostatiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Hypoglycémiants/pharmacologie
14.
Molecules ; 29(15)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39125024

RÉSUMÉ

Dimeric prodrugs have been investigated intensely as carrier-free drug self-delivery systems (DSDSs) in recent decades, and their stimuli-responsive drug release has usually been controlled by the conjugations between the drug molecules, including the stimuli (pH or redox) and responsive sensitivity. Here, an acid-triggered dimeric prodrug of doxorubicin (DOX) was synthesized by conjugating two DOX molecules with an acid-labile ketal linker. It possessed high drug content near the pure drug, while the premature drug leakage in blood circulation was efficiently suppressed. Furthermore, its aggregation structures were controlled by fabricating nanomedicines via different approaches, such as fast precipitation and slow self-assembly, to regulate the drug release performance. Such findings are expected to enable better anti-tumor efficacy with the desired drug release rate, beyond the molecular structure of the dimeric prodrug.


Sujet(s)
Doxorubicine , Systèmes de délivrance de médicaments , Libération de médicament , Promédicaments , Promédicaments/composition chimique , Promédicaments/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Concentration en ions d'hydrogène , Vecteurs de médicaments/composition chimique , Structure moléculaire
15.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39126039

RÉSUMÉ

Multidrug resistance (MDR) remains the most difficult problem facing conventional chemotherapy for cancers. Astragalus membranaceus is a historically traditional Chinese medicine. One of its bioactive components, formononetin, exhibits antitumor effects on various cancers. However, the effects of formononetin on MDR cancers have not been evaluated. Therefore, we investigated the defense's effects of formononetin on MDR. We used rhodamine 123 and doxorubicin efflux assays to analyze the inhibition kinetics of P-glycoprotein (P-gp) mediated-efflux. Cell viability was detected by sulforhodamine B assay, and the synergistic effects of formononetin combined with chemotherapeutic agents were further calculated using CompuSyn software. Molecular docking was performed with iGEMDOCK. We discovered that formononetin considerably induced oxidative stress and the disruption of mitochondrial membrane potential in MDR cancer cells. Furthermore, formononetin inhibits the P-gp efflux function by ATPase stimulation and the uncompetitive inhibition of P-gp-mediated effluxes of rhodamine 123 and doxorubicin. The molecular docking model indicates that formononetin may bind to P-gp by strong hydrogen bonds at Arginine (Arg) 489 and Glutamine (Gln) 912. Formononetin exhibits significant synergistic effects with vincristine and doxorubicin toward MDR cancer cells, and it synergistically suppressed tumor growth in vivo with paclitaxel. These results suggest that formononetin should be seen as a potential candidate for the adjuvant therapy of MDR cancers.


Sujet(s)
Doxorubicine , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Isoflavones , Simulation de docking moléculaire , Stress oxydatif , Isoflavones/pharmacologie , Isoflavones/composition chimique , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Souris , Doxorubicine/pharmacologie , Lignée cellulaire tumorale , Glycoprotéine P/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Synergie des médicaments
16.
Med Oncol ; 41(9): 220, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39115587

RÉSUMÉ

Breast cancer (BC) is the leading commonly diagnosed cancer in the world, with complex mechanisms underlying its development. There is an urgent need to enlighten key genes as potential therapeutic targets crucial to advancing BC treatment. This study sought to investigate the influence of doxorubicin (DOX) on identified key genes consistent across numerous BC datasets obtained through bioinformatic analysis. To date, a meta-analysis of publicly available coding datasets for expression profiling by array from the Gene Expression Omnibus (GEO) has been carried out. Differentially Expressed Genes (DEGs) identified using GEO2R revealed a total of 23 common DEGs, including nine upregulated genes and 14 downregulated genes among the datasets of three platforms (GPL570, GPL6244, and GPL17586), and the commonly upregulated DEGs, showed significant enrichment in the cell cycle in KEGG analysis. The top nine genes, NUSAP1, CENPF, TPX2, PRC1, ANLN, BUB1B, AURKA, CCNB2, and CDK-1, with higher degree values and MCODE scores in the cytoscape program, were regarded as hub genes. The hub genes were activated in disease states commonly across all the subclasses of BC and correlated with the unfavorable overall survival of BC patients, as verified by the GEPIA and UALCAN databases. qRT-PCR confirmed that DOX treatment resulted in reduced expression of these genes in BC cell lines, which reinforces the evidence that DOX remains an effective drug for BC and suggests that developing modified formulations of doxorubicin to reduce toxicity and resistance, could enhance its efficacy as an effective therapeutic option for BC.


Sujet(s)
Tumeurs du sein , Doxorubicine , Régulation de l'expression des gènes tumoraux , Humains , Doxorubicine/pharmacologie , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Antibiotiques antinéoplasiques/pharmacologie , Analyse de profil d'expression de gènes , Cycle cellulaire/effets des médicaments et des substances chimiques , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Biologie informatique/méthodes
17.
Biomacromolecules ; 25(8): 4991-5007, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39087761

RÉSUMÉ

The GPS-Nanoconveyor (MA-NV@DOX-Cas13a) is a targeted nanoplatform designed for the imaging and gene/chemotherapy synergistic treatment of melanoma. It utilizes rolling circle amplification (RCA) products as a scaffold to construct a DNA "Nanoconveyor" (NV), which incorporates a multivalent aptamer (MA) as a "GPS", encapsulates doxorubicin (DOX) in the transporter, and equips it with CRISPR/Cas13a ribonucleoproteins (Cas13a RNP). Carrying MA enhances the ability to recognize the overexpressed receptor nucleolin on B16 cells, enabling targeted imaging and precise delivery of MA-NV@DOX-Cas13a through receptor-mediated endocytosis. The activation of signal transducer and activator of transcription 3 (STAT3) in cancer cells triggers cis-cleavage of CRISPR/Cas13a, initiating its trans-cleavage function. Additionally, deoxyribonuclease I (DNase I) degrades MA-NV, releasing DOX for intracellular imaging and as a chemotherapeutic agent. Experiments demonstrate the superior capabilities of this versatile nanoplatform for cellular imaging and co-treatment while highlighting the advantages of these nanodrug delivery systems in mitigating DOX side effects.


Sujet(s)
Systèmes CRISPR-Cas , Doxorubicine , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Animaux , Souris , Humains , Aptamères nucléotidiques/composition chimique , Techniques d'amplification d'acides nucléiques/méthodes , Lignée cellulaire tumorale , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/administration et posologie , Antibiotiques antinéoplasiques/composition chimique
18.
FASEB J ; 38(15): e23876, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39120539

RÉSUMÉ

Breast cancer is a common malignant tumor in women. Ferroptosis, a programmed cell death pathway, is closely associated with breast cancer and its resistance. The transferrin receptor (TFRC) is a key factor in ferroptosis, playing a crucial role in intracellular iron accumulation and the occurrence of ferroptosis. This study investigates the influence and significance of TFRC and its upstream transcription factor hypoxia-inducible factor-1α (HIF1α) on the efficacy of neoadjuvant therapy in breast cancer. The differential gene obtained from clinical samples through genetic sequencing is TFRC. Bioinformatics analysis revealed that TFRC expression in breast cancer was significantly greater in breast cancer tissues than in normal tissues, but significantly downregulated in Adriamycin (ADR)-resistant tissues. Iron-responsive element-binding protein 2 (IREB2) interacts with TFRC and participates in ferroptosis. HIF1α, an upstream transcription factor, positively regulates TFRC. Experimental results indicated higher levels of ferroptosis markers in breast cancer tissue than in normal tissue. In the TAC neoadjuvant regimen-sensitive group, iron ion (Fe2+) and malondialdehyde (MDA) levels were greater than those in the resistant group (all p < .05). Expression levels of TFRC, IREB2, FTH1, and HIF1α were higher in breast cancer tissue compared to normal tissue. Additionally, the expression of the TFRC protein in the TAC neoadjuvant regimen-sensitive group was significantly higher than that in the resistant group (all p < .05), while the difference in the level of expression of IREB2 and FTH1 between the sensitive and resistant groups was not significant (p > .05). The dual-luciferase assay revealed that HIF1α acts as an upstream transcription factor of TFRC (p < .05). Overexpression of HIF1α in ADR-resistant breast cancer cells increased TFRC, Fe2+, and MDA content. After ADR treatment, the cell survival rate decreased significantly, and ferroptosis could be reversed by the combined application of Fer-1 (all p < .05). In conclusion, ferroptosis and chemotherapy resistance are correlated in breast cancer. TFRC is a key regulatory factor influenced by HIF1α and is associated with chemotherapy resistance. Upregulating HIF1α in resistant cells may reverse resistance by activating ferroptosis through TFRC overexpression.


Sujet(s)
Tumeurs du sein , Doxorubicine , Résistance aux médicaments antinéoplasiques , Ferroptose , Sous-unité alpha du facteur-1 induit par l'hypoxie , Récepteurs à la transferrine , Femelle , Humains , Adulte d'âge moyen , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Traitement médicamenteux adjuvant/méthodes , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Ferroptose/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Cellules MCF-7 , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Régulation positive
19.
PLoS One ; 19(8): e0308619, 2024.
Article de Anglais | MEDLINE | ID: mdl-39121041

RÉSUMÉ

BACKGROUND: This study aimed to determine the roles of interleukin (IL)-17, TAO kinase 1 (TAOK1), and NOD-like receptor protein 3 (NLRP3) in cardiomyocyte pyroptosis and proliferation. METHODS: The IL-17-treated H9C2 cells were used as in vitro heart failure (HF) models. These cells were subjected to TAOK1 overexpression or knockdown and treated with BMS-986299 (NLRP3 inflammasome agonist), MCC950 (NLRP3 inflammasome inhibitor), or verteporfin (Yes-associated protein [YAP] inhibitor). Thereafter, their pyroptosis, proliferative capacity, and gene and protein expression levels were detected. Doxorubicin-induced HF rats were used as in vivo models and subjected to TAOK1 overexpression. Thereafter, their myocardial pathology, NLRP3 inflammasome-mediated pyroptosis, and YAP/TEAD pathway function were evaluated. RESULTS: IL-17 treatment increased the pyroptosis and decreased the proliferative capacity of H9C2 cells. Additionally, IL-17 treatment inducedto the activation of the NLRP3 inflammasomes and inhibition of the YAP/TEAD pathway in the H9C2 cells. Moreover, the IL-17-mediated effects on the H9C2 cells were alleviated by TAOK1 overexpression and augmented by TAOK1 knockdown. Furthermore, treatment with BMS-986299 or verteporfin affected the pyroptosis, proliferative capacity, and NLRP3 inflammasome activation of the H9C2 cells independently of TAOK1 expression. In the doxorubicin-induced HF rat model, TAOK1 overexpression mitigated myocardial injury, suppressed NLRP3 inflammasome pathway activation, and restored the YAP/TEAD pathway activity. CONCLUSION: TAOK1 played a crucial role in regulating IL-17-mediated increase in the pyroptosis and decrease in the proliferation of cardiomyocytes by regulating the activities of the NLRP3 inflammasomes and the YAP/TEAD pathway.


Sujet(s)
Prolifération cellulaire , Défaillance cardiaque , Myocytes cardiaques , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Protéines de signalisation YAP , Animaux , Rats , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Doxorubicine/pharmacologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/traitement médicamenteux , Inflammasomes/métabolisme , Interleukine-17/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Pyroptose/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques , Protéines de signalisation YAP/métabolisme
20.
J Transl Med ; 22(1): 757, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39135106

RÉSUMÉ

BACKGROUND: Multi-drug resistance of poly(morpho)nuclear giant cells (PGCs) determines their cytoprotective and generative potential in cancer ecosystems. However, mechanisms underlying the involvement of PGCs in glioblastoma multiforme (GBM) adaptation to chemotherapeutic regimes remain largely obscure. In particular, metabolic reprogramming of PGCs has not yet been considered in terms of GBM recovery from doxorubicin (DOX)-induced stress. METHODS: Long-term proteomic and metabolic cell profiling was applied to trace the phenotypic dynamics of GBM populations subjected to pulse DOX treatment in vitro, with a particular focus on PGC formation and its metabolic background. The links between metabolic reprogramming, drug resistance and drug retention capacity of PGCs were assessed, along with their significance for GBM recovery from DOX-induced stress. RESULTS: Pulse DOX treatment triggered the transient formation of PGCs, followed by the appearance of small expanding cell (SEC) clusters. Development of PGCs was accompanied by the mobilization of their metabolic proteome, transient induction of oxidative phosphorylation (OXPHOS), and differential intracellular accumulation of NADH, NADPH, and ATP. The metabolic background of PGC formation was confirmed by the attenuation of GBM recovery from DOX-induced stress following the chemical inhibition of GSK-3ß, OXPHOS, and the pentose phosphate pathway. Concurrently, the mobilization of reactive oxygen species (ROS) scavenging systems and fine-tuning of NADPH-dependent ROS production systems in PGCs was observed. These processes were accompanied by perinuclear mobilization of ABCB1 and ABCG2 transporters and DOX retention in the perinuclear PGC compartments. CONCLUSIONS: These data demonstrate the cooperative pattern of GBM recovery from DOX-induced stress and the crucial role of metabolic reprogramming of PGCs in this process. Metabolic reprogramming enhances the efficiency of self-defense systems and increases the DOX retention capacity of PGCs, potentially reducing DOX bioavailability in the proximity of SECs. Consequently, the modulation of PGC metabolism is highlighted as a potential target for intervention in glioblastoma treatment.


Sujet(s)
Doxorubicine , Glioblastome , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Humains , Doxorubicine/pharmacologie , Lignée cellulaire tumorale , Stress physiologique/effets des médicaments et des substances chimiques , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Noyau de la cellule/métabolisme , Noyau de la cellule/effets des médicaments et des substances chimiques , Protéomique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Phosphorylation oxydative/effets des médicaments et des substances chimiques ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE